Você está na página 1de 37

Biochimica et Biophysica Acta 1551 (2001) F1^F37

www.bba-direct.com

Review
Apoptosis regulators and their role in tumorigenesis
a;
Martin Zo«rnig *, Anne-Odile Hueber b , Wiebke Baum a , Gerard Evan c

a
Georg-Speyer-Haus, Paul-Ehrlich-StraMe 42^44, 60596 Frankfurt, Germany
b
Institute of Signaling, Developmental Biology and Cancer Research CNRS UMR 6543, Centre A. Lacassagne, 33 Avenue Valombrose,
06189 Nice, France
c
UCSF Cancer Center, 2340 Sutter Street, San Francisco, CA 94143-0128, USA

Received 9 May 2001; received in revised form 12 July 2001; accepted 25 July 2001

Abstract

It has become clear that, together with deregulated growth, inhibition of programmed cell death (PCD) plays a pivotal role
in tumorigenesis. In this review, we present an overview of the genes and mechanisms involved in PCD. We then summarize
the evidence that impaired PCD is a prerequisite for tumorigenesis, as indicated by the fact that more and more neoplastic
mutations appear to act by interfering with PCD. This has made the idea of restoration of corrupted `death programs' an
intriguing new area for potential cancer therapy. ß 2001 Elsevier Science B.V. All rights reserved.

Keywords: Apoptosis; Tumorigenesis; Programmed cell death

1. Introduction cytes to delete autoreactive or non-reactive cells,


widespread cell death of neuronal cells during the
For many biologists it came as a surprise to realize self-assembly of the central nervous system, and the
that the death of a cell is not necessarily a bad thing. formation of digits by involution of interdigital cells
Indeed, in metazoans cell death is required for devel- in the primitive limb paddle (a more extensive survey
opment, maintenance and survival of the organism. of literature describing apoptosis occuring in vivo
Physiological cell death has been observed in di¡er- can be found in [2]).
ing tissues and in various organisms for more than Apoptosis is, by far, the most predominant form
100 years [1]. Cell death occurs throughout the life of physiological cell death. In contrast, unambiguous
span of multicellular organisms and arguably repre- examples of physiological cell necrosis are few. Be-
sents the only irreversible cell fate decision. Promi- cause it is a regulated process, controlled by a diver-
nent examples of physiological apoptosis include the sity of extracellular and intracellular signals, apopto-
hormonally regulated involution of the tadpole tail sis is used for the coordinated death of excess,
during development, negative selection of lympho- hazardous or damaged somatic cells. Moreover, the
apoptotic process includes mechanisms that organize
both packaging and disposal of cell corpses, thereby
preventing in£ammation of the surrounding tissue.
* Corresponding author. Tel.: +49-69-63395115; This is an essential requirement in metazoans which,
Fax: +49-69-63395297. for obvious reasons, need to be able to distinguish
E-mail address: zoernig@em.uni-frankfurt.de (M. Zo«rnig). cells that die as part of the normal process of main-

0304-419X / 01 / $ ^ see front matter ß 2001 Elsevier Science B.V. All rights reserved.
PII: S 0 3 0 4 - 4 1 9 X ( 0 1 ) 0 0 0 3 1 - 2

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F2 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

taining tissue homeostasis from cells that die as a


result of trauma.
Severe disturbance of homeostasis of any particu-
lar cell population or lineage can cause major path-
ologies in multicellular organisms. Not surprisingly,
therefore, substantial evidence indicates that altera-
tions in control of cell death/survival contribute to
the pathogenesis of many human diseases [3]. Dis-
eases linked with suppression of apoptosis include
cancer, autoimmune disorders (e.g. systemic lupus
erythematosus) and viral infections (e.g. herpesvi-
ruses, poxviruses, adenoviruses); diseases in which
increased apoptosis is an element include AIDS [4],
neurodegenerative disorders [5], myelodysplastic syn-
dromes, ischemic injury (e.g. stroke, myocardial in-
farction), toxin-induced liver disease (e.g. alcohol)
and some autoimmune disorders [6]. In many cases,
it is unclear whether perturbations in cell death
mechanisms are causal or merely a consequence of
the disease process. Nonetheless, e¡orts aimed at
treating these diseases by manipulating cell suicide
would seem to have great potential, although they
are thus far at a relatively early stage.

Fig. 1. Cells undergoing apoptosis. (A, B) Rat-1 ¢broblasts ex-


2. Cell death: de¢nition and signi¢cance pressing c-Myc in the presence (A) or absence (B) of serum.
(A) Normal growing cells. (B) Rat-1 cell in a late stage of apo-
ptotic cell death. Cell shrinkage, nuclear condensation and ¢nal
Historically, apoptosis was de¢ned on a morpho- fragmentation of the whole cell are obvious.
logical basis by contrast with another type of cell
death, necrosis ([7,8]. The necrotic cell swells and
its chromatin takes on the appearance of a £occulent shrunken apoptotic cytoplasm retain a largely nor-
mass that eventually disappears to leave a nuclear mal appearance save for some dilation of the endo-
ghost. Cell DNA is non-speci¢cally degraded and plasmic reticulum (ER) and swelling of the mito-
characteristically appears as a smear when size frac- chondria. The transition from normal to shrunken
tionated on an agarose gel. One of the most prom- and blebbing is rapid, typically taking only some
inent features of apoptosis involves the nucleus. 10^30 min, and it is at this point that apoptotic cells
Chromatin condenses and forms aggregates near are probably phagocytosed in vivo, either by their
the nuclear membrane which, in turn, becomes con- nearest neighbors or by professional macrophages
voluted, whilst the nucleolus becomes enlarged and [12]. Recognition and phagocytosis of apoptotic cells
appears abnormally granular. Chromatin is also sub- is mediated by a variety of independent receptor^li-
ject to the actions of di¡erent activated endonu- gand interactions which will not be discussed in de-
cleases that cleave the DNA initially into 300^50 tail further and have been reviewed elsewhere [13].
kb fragments and subsequently into 180 bp frag- However, one feature of apoptotic cells involved in
ments [9^11]. Also during apoptosis, the cell visibly their phagocytosis is commonly used as a marker for
shrinks, adherent cells round up, and distinct protu- apoptosis: activation of a £ippase in apoptotic cells
berances or membrane blebs become discernible (see leads them to express externalized phosphatidylser-
Fig. 1). Blebbing cells still exclude vital dyes indicat- ine, usually present only in the internal lea£et of
ing membrane integrity. Organelles within the the plasma membrane [14]. The rapidity of the apo-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F3

ptotic program and of the clearance of apoptotic in the adult. Such detailed knowledge of develop-
cells within the soma largely explains why apoptosis mental cell deaths enabled easy identi¢cation of mu-
was, until recently, largely overlooked as a major tants with aberrant patterns of cell death [20]. Their
homeostatic process. analysis has identi¢ed genes controlling four aspects
Another morphologically distinct form of pro- of the cell death process: (1) a determination step, (2)
grammed cell death (PCD) is the process of auto- the execution of cell death, (3) engulfment of the
phagy, or bulk degradation of cellular proteins dying cell, and (4) degradation of the engulfed cell
through an autophagosomic^lysosomal pathway. DNA.
Autophagy is important in normal growth control, Three genes are involved in the critical cell death
regulated by steroids during development and may execution step. Two of these, ced (cell death defec-
be defective in tumor cells [15,16]. tive)-3 and ced-4 are required for each cell death [21]:
if inactivated by mutation, none of the 131 normal
cell deaths occur. The other gene, ced-9, acts to an-
3. Evolution of PCD: the nematode Caenorhabditis tagonize the killing activity of ced-3 and -4: gain of
elegans as an invertebrate model function ced-9 mutants show absence of cell death
whereas mutations that inactivate ced-9 lead to wide-
Physiological PCD has been described in all multi- spread and lethal embryonic death [22]. In the last
cellular organisms so far studied, including plants, few years, many of the molecular functions of the
slime molds, nematodes, insects and vertebrates proteins encoded by the ced-3, -4 and -9 genes have
[17]. While the physiological role of apoptosis in been deduced [23]. The killer gene ced-3 encodes the
the shaping and rebuilding of complex tissues of mul- Ced-3 protein, a member of a class of cysteine pro-
ticellular organisms is plain to see, the biological ra- tease that cleave after aspartate residues ^ hence their
tionale for PCD is less clear for single cell organisms. name `caspase' (cysteine aspartyl protease [24]). Cas-
Furthermore, unicellular PCD seems not to involve pases are synthesized as inactive zymogens that are
the action of caspases ^ the cysteine proteases that activated by cleavage at sites that themselves con-
are the hallmarks of metazoan apoptosis. Nonethe- form to canonical caspase cleavage sites. Activation
less, there is growing evidence that some form of abscises an N-terminal prodomain and cleaves the
PCD does exist in unicellular organisms like Trypa- remaining polypeptide into small and large subunits
nosoma cruzi, Trypanosoma brucei rhodesiense, Dic- that then assemble as an K2 L2 active tetramer. The
tyostelium discoideum or Tetrahymena thermophila Ced-4 protein physically interacts with both Ced-3
[18] and perhaps even in bacteria [19]. Possibly, [25,26] and Ced-9 [26^29] proteins and appears to
PCD arose in unicellular organisms as a way of en- act as an adapter protein that facilitates Ced-3 au-
suring survival of at least some members of a clonal toactivation [30]. Ced-4 has a putative ATPase (nu-
colony during periods of privation. An alternative cleotide binding) domain that is required for its abil-
idea is that unicellular PCD evolved as a defense ity to activate Ced-3. The interaction of Ced-4 with
against the spread of virus infection. Indeed, many the death-inhibiting protein Ced-9 inhibits its ability
metazoan viruses actively suppress apoptosis in order to activate Ced-3.
to ensure their propagation, indicating that host cell As discussed below, the Ced-3, -4 and -9 basal
suicide is an e¡ective way of forestalling virus spread. apoptotic machinery is highly conserved amongst
The various known anti-apoptotic viral genes possess the metazoa. The Ced-3 caspase is homologous to
a variety of di¡ering structures and modes of action. similar enzymes identi¢ed in insects and in verte-
C. elegans is well suited for the study of cell death brates ^ the prototypical mammalian homologue
at the cellular, genetic, and molecular levels because being the eponymous interleukin-1L converting en-
it is both transparent and developmentally invariant. zyme (ICE) [31], although some 14 other mammalian
This has permitted the complete lineage description caspases are now known [32,33].
of every one of the 1090 cells born during develop- The mammalian homologues of Ced-9 are the Bcl-
ment of the hermaphrodite form. During C. elegans 2 proteins [34,35] ^ key regulators of cell survival
development, 131 cells die to leave a ¢nal total of 959 ¢rst identi¢ed by the oncogenic activity of Bcl-2 in

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F4 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

human follicular B cell lymphoma. Recent evidence was ICE [31], now called caspase-1. Caspase-1 and -
suggests that Ced-9 prevents apoptosis in C. elegans 11 (and possibly -4 and -5) are principally involved in
by two distinct mechanisms [36]: it may directly in- proteolytic maturation of cytokines such as interleu-
hibit the Ced-3 protease by an interaction involving kin-1L [41^43] and may have little to do with apo-
Ced-3 cleavage sites within Ced-9, or Ced-9 may also ptosis. However, subsequent studies have identi¢ed a
directly or indirectly inhibit Ced-3 by means of a further 10 ICE family members of which caspases-2,
protective mechanism similar to that used by mam- -3, -6, -7, -8, -9, -10, -12, -13 and -14 are all impli-
malian Bcl-2: cleavage of Ced-9 by Ced-3 generates a cated in regulation and/or execution of apoptosis:
carboxy-terminal product that resembles Bcl-2 in se- Nedd-2/ICH-1 (caspase-2), Yama/CPP32/apopain
quence and in function. (caspase-3), Tx /ICH-2/ICErelII (caspase-4), ICErelIII
A mammalian homologue of Ced-4 has been iden- (caspase-5), Mch-2 (caspase-6), ICE-Lap-3/Mch-3/
ti¢ed as Apaf-1 (apoptotic protease activating fac- CMH-1 (caspase-7), FLICE/MACH (caspase-8),
tor), a large protein implicated in regulating caspase ICE-LAP-6 (caspase-9), Mch-4/FLICE-2 (caspase-
activity through mediating cytochrome c-dependent 10), ICH-3 (caspase-11), caspase-12, ERICE (cas-
activation of caspase-9 [37]. Such tremendous evolu- pase-13) and MICE (caspase-14) [32,44].
tionary conservation of cell death machinery These apoptotic caspases undergo activating cleav-
amongst metazoans has the bene¢t that analysis of age during apoptosis (either through autoactivation,
cell death in `simple' genetic model systems like as part of a caspase cascade or by other non-caspase
C. elegans is very informative in developing our proteinases such as granzyme B) and between them
understanding of the control of apoptosis in mam- they cut a range of substrate proteins whose cleavage
malian cells. For example, the C. elegans egl-1 en- either mediates or attends the apoptotic process. Fur-
codes a small protein containing a nine amino acid thermore, caspase inhibitors, whether virus proteins
stretch similar to the BH3 domain, a domain found such as cowpoxvirus CrmA or baculovirus p35, or
in various pro-apoptotic Bcl-2-like mammalian cell aldehydes or £uoromethyl ketone-derivatized syn-
death regulators. Experimental analysis showed that thetic peptide inhibitors based on preferred substrate
egl-1 probably acts upstream of ced-3 and ced-4, and sequences (e.g. ZVAD-fmk), suppress many aspects
direct interaction between the Egl-1 and Ced-9 pro- of mammalian apoptosis.
teins was also demonstrated [38]. This led to the Caspases share many common structural and cat-
suggestion that Egl-1 may act by interfering with alytic features. All contain an active site pentapeptide
Ced-9 so that it can no longer inhibit Ced-4, which sequence with the general structure QACXG (where
is then free to activate Ced-3, leading to cell death. X is R, Q or G): the cysteine (together with a distant
Further genes required for removal of apoptotic histidine) is directly involved in catalysis. In the
cells have been identi¢ed in C. elegans [20] resulting main, caspases recognize a tetrameric primary se-
in mammalian homologues being identi¢ed and char- quence in their substrates with a distinctive require-
acterized. The recently cloned human homologue of ment for an aspartic acid residue in the substrate
the C. elegans engulfment protein Ced-6, for exam- P1 position. As tetrapeptides corresponding to the
ple, speci¢cally promotes phagocytosis of apoptotic substrate P4 ^P1 residues are su¤cient for speci¢c rec-
cells [39,40]. ognition by caspases [45], such substrates have pro-
vided the basis for design of a range of inhibitors
[46]. Phylogenetic analysis of the caspases shows
4. Molecular pathways of cell death they fall into three subfamilies, each with signi¢-
cantly di¡ering substrate speci¢city which generally
4.1. Central e¡ectors: caspases correlates with caspase function: the ICE subfamily
of cytokine processors (caspases-1, -4, -5 and -11;
It is widely accepted that caspases, the family of because of sequence homology to caspase-1 the cas-
related Ced-3-like cysteine proteases, are common pases-12, -13 and -14 are also grouped with the cy-
e¡ectors of classical metazoan apoptosis. The ¢rst tokine processors), the Ced-3/CPP32 subfamily of
mammalian homologue of Ced-3 identi¢ed in 1993 apoptotic executioners (caspases-3, -6, and -7) and

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F5

the ICH-1/Nedd-2 subfamily of apoptotic initiators APAF-1) are presumed to mediate speci¢c intermo-
(caspase-2, -8, -9 and -10). lecular interactions that regulate caspase activation.
Caspases are synthesized as precursor proenzymes Indeed, one strongly inactivating mutation of Ced-3
which are proteolytically processed to their active is located within its prodomain [53], indicating the
forms. Active caspases are composed of a hetero- importance of this region for Ced-3 activation.
dimer comprising a large subunit (P20 for caspase- CARDs and death e¡ector domains as well as death
1, P17 for caspase-3) that contains the catalytic cys- domains, all contain six anti-parallel K-helices ar-
teine residue, and a smaller subunit (P10 for caspase- ranged in a similar three-dimensional fold and asso-
1, P12 for caspase-3) that contains determinants ciate via like^like interactions [32].
which govern substrate speci¢city. The X-ray crystal Genetic knockout studies in mice have been used
structure of activated caspase-1 indicates that two to investigate the measure of redundancy amongst
independent functional P20/P10 heterodimers are in- caspases [54]. Available data suggest that apoptosis
timately associated to form a (P20/P10)2 tetramer in triggered by di¡ering stimuli frequently employ dif-
which the two active sites reside at opposite ends of ferent `subsets' of activated caspases. Thus, mice de-
the complex [47,48]. Procaspases are activated by ¢cient in caspase-1 develop normally, are fertile, ap-
cleavage at critical aspartate residues that themselves pear healthy and exhibit no apparent abnormalities,
conform to the substrate consensus for caspases. suggesting absence of any gross defects in normal
This indicates that caspases exist within hierarchies physiological processes involving apoptosis [41,55].
of families that undergo auto- and trans-cleavage. However, thymocytes from the caspase-1null mice ex-
For example, evidence supports the idea that cas- hibit partial resistance to CD95-induced apoptosis,
pase-8 autoactivates itself upon recruitment to a implying a role for caspase-1 in implementing apo-
death receptor signaling complex (the zymogene full ptosis in response to that particular trigger. Caspase-
length caspase-8 molecule obviously possesses some 11 knockout mice also show defective interleukin-1L
residual proteolytic activity su¤cient for this self- production but develop normally and have minimal
cleavage) and the active enzyme then in turn cleaves apoptotic defects [43].
and activates the e¡ector caspases-3 and -7 that ex- Mice lacking caspase-3 are smaller than their wild-
ecute the apoptotic program. Interestingly, caspase-9 type littermates and die at 1^3 weeks of age [56].
does not necessarily require proteolytic processing, Analysis shows that the development of the brain
but instead requires binding to APAF-1 with which in these animals is markedly a¡ected: a variety of
it forms an active holoenzyme [49,50]. central nervous system hyperplasias are observed
All procaspases have an N-terminal prodomain from embryonic day 12 on, indicative of defective
that is also removed during activation. For some apoptosis. However, thymocytes from caspase-3null
caspases (caspase-3, -6, -7 and -14) the prodomain mice exhibit normal sensitivity to apoptosis induced
is short (10^40 residues) whilst for the other caspases by a number of di¡erent stimuli and the rather tis-
it is extensive and contains recognizable domains. sue-restricted phenotype in caspase-3-de¢cient mice
The extensive prodomains play important roles in again demonstrates that other caspases can substitute
caspase regulation and function as signal integrators for caspase-3 in most tissue or cell types. Animals
for apoptotic or pro-in£ammatory signals [51,52]. de¢cient in caspase-8 or caspase-9 die perinatally be-
For example, the prodomains of caspase-8 and -10 cause of profound defects in developmental cell
each contain two so-called death e¡ector domains deaths [57^59]. Caspase-2-de¢cient mice develop nor-
that mediate the proenzymes' interaction, via an mally, but cells from these animals show diminished
adapter molecule, with the cytoplasmic tail of mem- or enhanced apoptosis, depending on their tissue of
bers of the TNF-R1/CD95 receptor family. This al- origin [60]. Caspase-12 is localized to the ER and
lows receptor-induced activation of the caspases in becomes activated by ER stress. Mice that are de¢-
response to ligand binding. Caspases-1, -2, -4, -9 cient in caspase-12 are resistant to ER stress-induced
and Ced-3 possess a distinct prodomain structure apoptosis, but their cells undergo apoptosis in re-
termed CARD (caspase recruitment domain). sponse to other death stimuli [61].
CARD domains (also found in the adapter protein Caspase substrates [62,63] can be grouped into dif-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F6 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

ferent classes according to their (putative) function. Huntington's disease and Alzheimer's disease: cas-
Amongst these are pro- and anti-apoptotic proteins, pase-3 and -12 seem to be involved in proteolytic
components of the apoptotic machinery, structural cleavage of Alzheimer amyloid-L precursor protein
proteins, homeostatic proteins and proteins impor- and formation of the apoptosis-inducing amyloido-
tant for signaling, cellular repair and macromolecu- genic AL peptide [61,77]. The polyglutamine repeats
lar synthesis [32]. The following examples represent associated with Huntington's disease induce neuronal
these di¡erent classes of caspase substrates. (1) cell death via caspase-8 [78]. Caspase-10 loss of func-
Transactivation of procaspases by already activated tion mutations have been linked to defective cell
caspases could generate su¤cient proteolytic activity death in autoimmune lymphoproliferative syndrome
to overwhelm endogenous caspase inhibitors such as type II [79]. Caspase inactivation may also promote
inhibitor of apoptosis proteins (IAPs). (2) Caspase-3 oncogenesis [80]. However, there is one caveat: some
cleaves Bcl-2 and Bcl-xL which destroys the anti-ap- triggers of apoptosis retain the ability to kill cells
optotic function of these proteins and releases C-ter- even when caspases are inhibited [81^84], although
minal fragments that are pro-apoptotic [64,65]. (3) it is not clear what mechanisms are involved in
Caspase-8 cleaves Bid, a pro-apoptotic Bcl-2 family such caspase-independent cell death. The CD95/
member, generating a C-terminal fragment that in- Fas/Apo-1 receptor is able to kill activated primary
duces release of mitochondrial cytochrome c [66,67]. T-cells in the absence of active caspases (see below);
(4) Caspase-3 cleaves ICAD/DFF45 allowing the this cell death involves necrotic morphological
ICAD-bound nuclease CAD to translocate to the changes and depends on the kinase Rip as e¡ector
nucleus and to cut DNA [68^70]. (5) Caspase-3 molecule [85].
also cleaves and activates Gelsolin, a protein that
regulates actin dynamics and promotes both cyto- 4.2. The ancestral pathway: the role of mitochondria,
plasmic and nuclear apoptosis, including DNA frag- cytochrome c and Apaf-1 in mammalian apoptosis
mentation [71]. (6) Lamins are major structural pro-
teins within the nuclear envelope, and their cleavage Ideas concerning the mechanism of activation and
by caspase-6 may be responsible for some of the control of apoptosis have been greatly in£uenced by
observed nuclear changes [72]. (7) Cleavage of L-cat- the recent discovery that cytochrome c is released
enin and FAK might interrupt cell^cell contacts and from mitochondria during cell death and is involved
cell^matrix focal adhesions thereby promoting cellu- in triggering the e¡ector machinery of apoptosis
lar packaging and phagocytosis [73,74]. (8) Poly(- [23,86^89]. Cytochrome c normally resides in the
ADP-ribose) polymerase (PARP) cleavage may inter- space between the outer and inner membranes of
fere with its key homeostatic function as a DNA mitochondria where it participates in the process of
double-strand break repair enzyme [75] which might oxidative phosphorylation. Upon exposure of cells to
facilitate or allow the DNA degradation character- apoptotic stimuli, cytochrome c is released from mi-
istic of apoptosis [33]. However, it is worth noting tochondria into the cytosol, where it is one of several
that PARPnull mice seem to develop normally [76]. factors implicated in the proteolytic activation of
Unfortunately, in the case of many caspase sub- caspase-3 by caspase-9 [90].
strates it is not always obvious what, if any, mecha- Biochemical analysis has identi¢ed two cytosolic
nistic role their cleavage plays in apoptosis. Many proteins, Apaf-1 and Apaf-3, that form the complex
caspase substrates are `plausible' candidate e¡ectors with cytochrome c that activates caspase-3. Apaf-1
for apoptosis, but so far no `key' target has been shares limited homology with the product of the
found whose cleavage appears to provide the ulti- C. elegans ced-4 gene product [37], although it is a
mate `killer' cut for the cell. Nonetheless, given the larger and more complex protein, while Apaf-3 is
central role of caspases in the apoptotic program, caspase-9 [91]. The Ced-4-like domain in Apaf-1 is
these proteases o¡er obvious therapeutic targets for £anked on one side by a region with strong homol-
the control of inappropriate apoptosis [46]. Caspase ogy to the CARD motif within the prodomains of
over-reactivity promotes cellular suicide, and this Ced-3 and mammalian caspases-2 and -9 and on the
may be the basis for degenerative disorders such as other side by several WD-40 repeats believed to me-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F7

diate protein^protein interactions. The CARD do- cytochrome c transfected into HeLa cells it was re-
mains in Apaf-1 and the prodomain of caspase-9 cently demonstrated that the release of cytochrome
interact and, in the presence of cytochrome c and c-GFP always precedes exposure of phosphatidylser-
either ATP or ADP, this induces autocatalytic acti- ine at the cell surface and the loss of plasma mem-
vation of the caspase which then activates the down- brane integrity [99]. The time interval between death
stream caspase e¡ector cascade involving caspases-2, stimulus and cytochrome c release can vary in indi-
-3, -6, -7, -8 and -10 [90]. vidual cells (and depending on the apoptotic insult)
Apaf-1 dimerization is repressed by its own C-ter- but once initiated, cytochrome c is released from all
minus containing the WD-40 repeats. It is speculated mitochondria in individual cells within 5 min. This
that cytochrome c binding to the WD repeats then study also showed that the drop in the mitochondrial
induces a conformational change that allows Apaf-1 membrane potential typically seen in apoptotic cells
to oligomerize and by promoting clustering of this occurs later than cytochrome c release from mito-
caspase to activate caspase-9 [92]. chondria and that this process is dependent on cas-
The Apaf-1/caspase-9 interaction is clearly reminis- pase activation, whereas cytochrome c release is not.
cent of the mechanism of Ced-3 activation by Ced-4 These results suggest a speci¢c permeability of the
in C. elegans. This similarity extends to the involve- outer mitochondrial membrane without alteration
ment of the mammalian anti-apoptotic Bcl-2 and of the inner mitochondrial membrane.
Bcl-xL , proteins which are homologues of Ced-9. So far, several competing models exist to explain
Bcl-2/Bcl-xL reside in the outer mitochondrial mem- exactly how permeabilization of mitochondrial mem-
brane, where they function to suppress apoptosis in branes is mediated during apoptosis (see Fig. 2) [88]:
both or either of two ways: blocking cytochrome c the outer mitochondrial membrane might rupture as
release and binding to Apaf-1 to prevent its activat- a result of swelling of the mitochondrial matrix. In
ing caspase-9. The mammalian pro-apoptotic Bcl-2 one model swelling is postulated to result from open-
family members, such as Bax, Bak and Bik, may ing of a megachannel called the permeability transi-
promote apoptosis by displacing Apaf-1 from Bcl- tion pore (PTP). The adenine nucleotide translocator
2/xL . Nevertheless, although a direct binding of (ANT; located in the inner mitochondrial mem-
Bcl-xL to Apaf-1 (shown by in vitro experiments) brane) and the voltage-dependent anion channel
has been reported [93,94], these data (or at least their (VDAC, found in the outer mitochondrial mem-
physiological relevance) have been questioned re- brane) are major components of the PTP which is
cently [95]. The anti-apoptotic protein Aven, which proposed to span both the inner and the outer mi-
was identi¢ed in a yeast two-hybrid screen, has been tochondrial membranes at sites at which the two
shown to bind to both Bcl-xL and Apaf-1 and this membranes are opposed. According to the PTP mod-
molecule might link the two molecules and target the el, PTP openers, including the pro-apoptotic Bcl-2
Bcl-2 family member to the apoptosome [96]. The family member Bax, cause permeabilization of the
fact that direct Bcl-xL or Bcl-2 binding to Apaf-1 inner membrane and mitochondrial depolarization
could not be observed in vivo might also indicate by binding to the ANT [100] (although this is not
that other Ced-4 homologues exist which could supported by the data mentioned above). This pro-
bind to anti-apoptotic Bcl-2 family members. The cess allows entry of water and solutes into the matrix
idea of further Ced-4 family members is supported and leads to mitochondrial swelling. Another model
by the murine Apaf-1 knockout which leads to an postulates that swelling is due to a defect in mito-
impairment of apoptosis in some, but not all circum- chondrial ATP/ADP exchange as a result of closure
stances and cell types [97,98]. Targeted inactivation of the VDAC thus leading to hyperpolarization of
of Apaf-1 in mice nevertheless leads to profound the inner mitochondrial membrane and subsequent
developmental abnormalities in cell number regula- matrix swelling. Again such a scenario contradicts
tion in the brain as well as in other tissues such as reports stating that the drop in membrane potential,
the peripheral nervous system, resulting in embryonic at least in some cell types, follows the release of
lethality. cytochrome c.
By using green £uorescent protein (GFP)-tagged Other models do not predict damage of the outer

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F8 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

Fig. 2. Di¡erent models explain the release of cytochrome c from mitochondria during apoptosis. The outer mitochondrial membrane
might rupture as a result of swelling of the mitochondrial matrix. This could be explained with opening of the permeability transition
pore (PTP) or with the closure of the voltage-dependent anion channel (VDAC). Other models relate cytochrome c release to pore
formation allowing the passage of cytochrome c into the cytosol.

mitochondrial membrane but rather the formation of tors ^ cleaves the pro-apoptotic Bcl-2 family member
a pore in this membrane allowing the passage of Bid [66,67]. Cleaved Bid (tBid) then binds to Bax
cytochrome c (and other mitochondrial proteins) leading to Bax oligomerization and integration into
into the cytosol. Bax is a candidate for the formation the outer mitochondrial membrane where it triggers
of this pore. Bax oligomers can form large conduc- cytochrome c release [102]. Similarly, tBid binds to
tance channels in lipid planar bilayers [88]. Addition and oligomerizes another pro-apoptotic Bcl-2 homo-
of Bax directly to isolated mitochondria triggers re- logue, Bak, resulting in cytochrome c release [103].
lease of cytochrome c through a mechanism that is While studies in bak knockout cells show that tBid
insensitive to PTP blockers and does not involve mi- does not require Bak for mitochondrial targeting,
tochondrial swelling. Bak proved necessary for tBid-induced cytochrome
Yet another model involves Bax cooperating with c release. Consequently, bax3/3bak3/3 double
the VDAC to form a cytochrome c-conducting chan- knockout cells are resistant to a wide range of apo-
nel [101]. Nevertheless, direct evidence for the forma- ptotic stimuli [104].
tion of such pore structures in mitochondria during Several proteins in addition to cytochrome c are
apoptosis is still missing. released from mitochondria in cells induced to
An interesting link between death receptor-acti- undergo apoptosis. Among them is the recently iden-
vated apical caspases such as caspase-8 and mito- ti¢ed Smac/Diablo molecule which binds to, and in-
chondrial cytochrome c release has been established activates, IAPs [105,106]. IAPs inhibit cell death by
in the form of the BH3-domain-only protein Bid: binding to procaspases and activated caspases, there-
caspase-8 ^ initially activated at the death-inducing by blocking their processing and their activity. Smac/
signaling complex (DISC) of cell surface death recep- Diablo is released from the mitochondria along with

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F9

cytochrome c during apoptosis and relieves inhibi- [111], Rank, CD95, DR6 [112] and the newly discov-
tion of caspase-9 activation by IAP inactivation ered TRAIL receptors TRAIL-R1 (DR4), -R2
[107]. It is also possible that in some cells (type II (DR5), -R3 (DcR-1) and DcR-2 [113]. Activation
cells) Smac/Diablo is required to inactivate an IAP of members of this receptor family triggers a variety
preventing direct caspase-3 activation by caspase-8. of cellular responses depending on cell type and con-
In this scenario cytochrome c release might not be text, amongst which are (T-cell) activation and stim-
relevant for the death process, but rather Smac/Dia- ulation, proliferation, di¡erentiation, survival and
blo liberation into the cytosol. Smac/Diablo and the apoptotic cell death [114^116].
pro-apoptotic Drosophila proteins Reaper, Grim and Mammalian TNF-R family members are type I
Hid seem to function in a similar way (by inhibiting membrane proteins characterized by conserved extra-
IAP activity) and a sequence similarity among these cellular cysteine-rich domains. A functional TNF
proteins (restricted to their N-terminal 14 amino superfamily receptor is typically a trimeric or multi-
acids) has been reported [108] suggesting that meric complex stabilized by disul¢de bonds,
Smac/Diablo and the insect apoptosis-inducing pro- although some, such as CD95, TNF-R1 and TNF-
teins might be structural as well as functional homo- R2, also exist in a soluble form generated by proteo-
logues. lytic cleavage [117]. The receptors' ligands comprise
Mitochondrial integrity is important not only for another related family that includes TNF, LT-K
sequestering cytochrome c and Smac/Diablo but also (lymphotoxin-K), CD95 ligand (FasL/CD95L),
for other ways to regulate caspase activation and TRAIL, OX40L, CD27L, CD30L, CD40L, 4-1BBL
apoptosis [87]. A fraction of both caspase-9 and cas- and LT-L. Each of the ligands is synthesized as a
pase-3 has been localized to the mitochondrial inter- nascent type II membrane-associated protein and
membrane space in some cell types, and caspase-2 shares a characteristic 150 amino acid region towards
has also been reported to reside in mitochondria. the C-terminus by which each ligand interacts with
These caspases ^ like cytochrome c ^ can be released its cognate receptor. For the most part, these ligands
from the mitochondria to the cytosol during apopto- exist as trimeric or multimeric membrane-bound pro-
sis induction. Another protein, AIF (apoptosis-in- teins that may function to induce receptor aggrega-
ducing factor), also redistributes from mitochondria tion. However, a few members, such as TNF and
and induces some of the nuclear morphology associ- CD95L, are also functional in soluble form. Interest-
ated with apoptosis in a caspase-independent manner ingly, a domain N-terminal to the ligand binding
[109]. Genetic inactivation of AIF renders embryonic domain in the extracellular region of TNF-R1,
stem cells resistant to cell death after serum depriva- TNF-R2 and CD95 was recently identi¢ed that
tion and disables PCD during caviation of embryoid mediated receptor self-association before ligand
bodies in early mouse morphogenesis [110]. binding [118]. This pre-ligand binding assembly do-
main (PLAD) is critical for assembly of functional
4.3. The death receptor pathway receptor complexes on the cell surface. Thus, TNF
receptor family members might function as pre-
Recently, a direct mechanistic link between a par- formed complexes rather than as individual receptor
ticular apoptotic stimulus and activation of the basal subunits that oligomerize after ligand binding.
caspase apoptotic machinery has been forged: acti- A detailed discourse on the multiple pleiotropic
vation of a speci¢c group of transmembrane recep- cellular and physiological activities provoked by liga-
tors of the tumor necrosis factor (TNF) receptor tion of TNF receptor family members in various cell
superfamily, either by ligand or (experimentally) by types is beyond the scope of this article. We will
binding an agonistic antibody, can lead to direct ac- con¢ne ourselves to those receptors whose ligation
tivation of caspases. The list of TNF receptor family has been shown to induce apoptosis ^ namely
members is growing and includes TNF-R1 (P55), TNF-R1, CD27 [119], CD30, CD40, LT-LR, CD95,
TNF-R2 (P75), TNF-R3 (TNF-RP), LT-KR, Ox- DR3, DR4, DR5 and DR6 [120]. A subgroup of
40, CD27, CD28, CD30, CD40, 4-1BB, p75 NGF- these receptors shares a common intracellular pro-
R (low a¤nity nerve growth factor receptor), GIT-R tein^protein interaction domain, the so-called death

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F10 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

domain. These receptors are referred to as death re- tors. Rather, signal transduction is mediated via di-
ceptors and they include TNF-R1, CD95, DR3, rect recruitment of, and intermolecular association
DR4, DR5, and DR6. with, various downstream signaling e¡ector mole-
In particular, we shall summarize current knowl- cules [51]. In this regard, a key intracellular interac-
edge concerning the CD95, TNF-R1 and the TRAIL tion domain present in the cytoplasmic tail of all
receptors and the attendant molecules mediating death receptors is the 65 amino acid `death domain'
their death signal transduction. (DD), a name deriving from its ability to recruit
downstream e¡ectors that can induce apoptosis
4.3.1. CD95/FAS/Apo-1 [125]. However, the term `death domain' is somewhat
CD95/Fas/Apo-1, henceforth called CD95, is ex- unfortunate, since it implies that cell death is the
pressed in activated lymphocytes as well as in all generic function of this type of motif. In fact, DDs
other tissues such as the liver, heart and lung. Liga- are domains that mediate homo- and heterotypic
tion of CD95, whether by its ligand CD95L or, ex- protein^protein interactions in order to propagate
perimentally, by agonistic antibody can induce apo- signals, and they have since been found in signaling
ptosis in several cell types. CD95L is predominantly pathways that have no obvious link with cell death
expressed on activated lymphocytes, NK cells, eryth- [126].
roblasts and immune privilege tissues, but also on The DDs in the ligated TNF-R1/CD95 receptors
certain tumors. The CD95/CD95L apoptotic path- recruit the C-terminal DD of the cytoplasmic adapter
way also functions to maintain homeostasis in vari- FADD/MORT-1. At its N-terminus, FADD/MORT
ous tissues ^ the liver being a particularly well docu- possesses a di¡erent protein binding domain, a
mented example [121]. However, the biological role `death e¡ector domain', that mediates interaction
of CD95 is probably best understood in the immune with the N-terminal prodomain of caspase-8
system, where it is implicated in peripheral clonal [127,128]. The recruitment of caspase-8 by FADD/
deletion of T-lymphocytes, activation-induced suicide MORT to the activated CD95 receptor generates a
of mature T-cells, cytotoxic response and induction DISC [129] that leads to proteolytic autoactivation
of apoptosis in B-cells. Constitutive cell surface of caspase-8. Caspase-8 then activates other caspases,
expression of CD95L also seems to contribute to including caspase-1 and caspase-3, which then are
immunological privilege of certain organs by killing presumed to execute the apoptotic dissolution of
in¢ltrating lymphocytes and in£ammatory cells the cell [130]. Members of the TNF receptor family
expressing CD95 receptor [122]. Mice carrying which lack a death domain (e.g. TNF-R2, CD27,
mutations in the genes for CD95 (lpr for lymphopro- CD30, CD40) are also under certain circumstances
liferation) and CD95L (gld for generalized lympho- able to induce cell death via alternative mechanisms
proliferative disease) have been identi¢ed. Mice ho- [120].
mozygous for either of these mutations accumulate CD95, through recruitment of the DISC, appears
an excess of non-malignant CD3‡ B220‡ CD43 CD83 to provide a direct link between external ligand and
T-cells in their spleens and lymph nodes and also the basal e¡ector machinery of apoptosis. However,
su¡er from an autoimmune systemic lupus erythema- it has recently become clear that this direct molecular
tosus-like condition. This demonstrates that CD95/ cantilever only seems to operate in certain cell types
CD95L signaling ful¢ls an important function in de- ^ type 1 cells [131]. In other (type 2) cells, CD95
leting autoreactive lymphocytes and maintaining pe- leads to changes in mitochondria that activate down-
ripheral tolerance. Mutations in the human CD95 stream caspases in a di¡erent way. The amount of
gene cause a similar lympho-accumulative syndrome receptor-activated caspase-8 in type 2 cells is much
[123]: patients with autoimmune lymphoproliferative lower than in type 1 cells [132] and probably insu¤-
syndrome type 1A have heterozygous CD95 germ- cient to induce downstream e¡ector caspase cleavage.
line mutations and their lymphocytes are resistant It nevertheless is enough to cleave Bid, a BH3 do-
to CD95-induced apoptosis [124]. main-only member of the Bcl-2 family [66,67]. Trun-
No identi¢able catalytic motifs are present in the cated Bid then translocates to the mitochondria
cytoplasmic domains of the CD95/TNF-R1 recep- where it induces cytochrome c release and conse-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F11

quently further caspase activation and ¢nally cell is the Fas-interacting serine/threonine kinase/homeo-
death. domain-interacting protein kinase FIST/HIPK3
The DDs of both CD95 and TNF-R1 interact with [140]. FIST not only binds to CD95 but also inter-
the C-terminal DD of a second receptor-associated acts with FADD in a trimolecular complex com-
protein, designated RIP [133]. The RIP N-terminus posed of CD95, FADD and FIST. FIST kinase in-
resembles a tyrosine kinase domain which is intrigu- duces FADD phosphorylation and inhibits CD95-
ing because experimental data implicate a tyrosine mediated JNK kinase activation. It is localized
kinase in CD95-mediated signal transduction. Phar- both in the cytoplasm and in the nucleus and is ca-
macologic inhibitors of protein kinases block, in a pable of binding to Daxx in a kinase activity-depen-
concentration-dependent manner, CD95-induced dent manner.
DNA fragmentation and prolong cell survival [134]. In addition to activation of caspases and JNKs,
The DD of RIP also binds to the C-terminal DD of both CD95 and TNF-R1 trigger other signaling ef-
another `death adapter protein', RAIDD (for RIP- fectors. CD95-generated apoptotic signals activate
associated ICH-1/Ced-3 homologous protein with a acidic sphingomyelinase causing accumulation of cer-
death domain). At its N-terminus, RAIDD is homol- amide [141] which is observed in both CD95- and
ogous to, and oligomerizes with, the prodomain of TNF-R1-induced apoptosis. Whether ceramide pro-
caspase-2 (Ich-1). Thus, caspase-2 can be recruited to duction is a major determinant of the apoptotic de-
the CD95 receptor through sequential interactions of cision is still a matter of debate.
RAIDD, RIP, FADD and CD95 [115]. A close rel- Naturally occurring inhibitors of the CD95/TNF-
ative of RAIDD is CRADD, which also interacts R1 death signaling pathways exist in the guise of the
with both RIP and caspase-2 [135]. RIP is required FLIPs (Fas-associated death domain-like ICE inhib-
for TNF-induced NF-UB activation. Cleavage of RIP itory proteins) which interfere with recruitment of
by caspase-8 results in the blockage of NF-UB-medi- caspases to the CD95/TNF-R1 signaling complexes.
ated anti-apoptotic signals [136]. A number of viruses encode FLIPs as part of their
Recently, another CD95 binding protein, Daxx, strategy for manipulating host cell suicide and viabil-
has been described [137]. Daxx also binds to the ity. For example, the Q-herpesviruses encode FLIPs
CD95 death domain but lacks a death domain of that comprise two death e¡ector domains which in-
its own. Overexpression of Daxx activates Jun teract with FADD/MORT and inhibit its recruitment
N-terminal kinase (JNK) and potentiates CD95-in- and activation of caspase-8 [142]. Recently, a cellular
duced apoptosis. On this basis, it has been proposed homologue of v-FLIP was identi¢ed by di¡erent
that CD95 engages two independent pathways that groups [143^150]. c-FLIP is structurally similar to
induce cell death: one pathway via FADD/caspase-8/ caspase-8 since it contains two death e¡ector do-
2 and the other via Daxx/JNK activation. Interest- mains and an inactive caspase-like domain lacking
ingly, Daxx is a nuclear protein that interacts and the conserved functional cysteine residue. c-FLIP is
colocalizes with the tumor-suppressive promyelocytic expressed in two isoforms (long and short form),
leukemia protein PML in nuclear bodies [138]. Re- both of which are recruited to the CD95 DISC in a
porter gene assays show that DAXX is able to re- stimulation-dependent fashion. c-FLIP blocks cas-
press basal transcription; SUMO-1-modi¢ed PML pase-8 activation at the DISC and thereby inhibits
sequesters DAXX to the nuclear bodies and inhibits CD95-mediated apoptosis [151]. During this process,
Daxx-mediated transcriptional repression. How pre- both caspase-8 and c-FLIP undergo cleavage be-
cisely CD95 activation acts on Daxx localization, tween the p18 and p10 subunits, generating two sta-
and its in£uence on transcription, is presently un- ble intermediates of 43 kDa that stay bound to the
clear. Strangely enough, rather than showing de- DISC.
creased apoptosis, inactivation of Daxx results in B- and T-cells downregulate c-FLIP upon activa-
extensive apoptosis and embryonic lethality in mice tion in vitro [152], providing a possible explanation
[139]. for the observation that resting peripheral T-cells are
Among the proteins which have been shown to resistant to CD95-induced apoptosis and become
bind to the cytosolic domain of the CD95 receptor susceptible only after their activation. By inhibiting

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F12 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

death receptor-mediated cell death, c-FLIP has been upregulating CD95 or CD95L [163,164] although the
identi¢ed as a tumor progression factor in mouse generality of this concept has been questioned
models [153,154]. Several groups found a pro-apo- [165,166].
ptotic function of c-FLIP in transient overexpression
studies [146], the physiological relevance of which is 4.3.2. TNF receptors
presently unclear [151]. TNF is well recognized as a cytokine produced by
Another way to inhibit death ligand-induced apo- activated T-cells and macrophages that orchestrates
ptosis is to quench the signal via decoy receptors. A aspects of the host in£ammatory response. It does so
soluble CD95 decoy receptor (DcR3) has been dis- by in£uencing the proliferation, di¡erentiation and
covered that binds to CD95L and inhibits CD95L- apoptosis of cells involved in in£ammation. TNF
induced apoptosis [155,156]. The physiological im- (together with the lymphotoxin LT) is the ligand
portance of such signal inhibition is underlined by for two receptors ^ TNF-R1 and TNF-R2. TNF-
the ¢nding that the DcR3 gene was ampli¢ed in R1 alone appears to be able to mediate most, if
about half of the primary lung and colon tumors not all, of the biological responses engendered by
studied. TNF, although TNF-R2 may provide an auxiliary
Knowledge of the downstream e¡ectors involved function in cooperating in the binding of TNF to
in CD95 death signaling has facilitated analysis of TNF-R1 [167]. Genetic deletions of both receptors
the role of CD95 in vivo. As discussed above, mice have demonstrated the di¡erences in biological func-
with inactivating mutations in the genes for either tionality of TNF-R1 and TNF-R2 in vivo [168^170].
CD95 (lpr) or CD95L (gld) exhibit generalized lym- Although both act to potentiate in£ammation/host
phoproliferative disease. The cowpoxvirus caspase defense and share the common ability to activate
inhibitor CrmA, which e¤ciently blocks caspase-8 the pleiotropic transcription factor NF-UB [171],
(as well as other caspases such as caspase-1), has TNF-R1 alone can clearly trigger apoptosis whereas
been expressed transgenically in peripheral T-lym- TNF-R2 mainly seems to promote cell survival,
phocytes via the CD2 promoter. Such CD2-crmA although it was shown to kill certain cells when over-
transgenic mice exhibit resistance to CD95-induced expressed [120]. However, substantial evidence indi-
apoptosis equivalent to that seen in lpr mice [157] cates that TNF-R1 can also promote cell survival
although neither Q-radiation- nor corticosteroid-in- under certain circumstances, although this anti-apo-
duced cell death is suppressed. However, in contrast ptotic activity, unlike activation of the caspase cas-
to lpr mice, CD2-crmA transgenic mice develop nei- cade, appears to be indirect and require de novo syn-
ther T-cell hyperplasia nor serum autoantibodies, im- thesis of survival proteins.
plying that the lpr phenotype is not merely due to TNF-R1 signaling, like CD95, is able to activate
failure of CD95 to trigger caspase-dependent T-cell the proteolytic caspase cascade by recruiting caspase-
apoptosis. Expression of a dominant negative mutant 8 via FADD/MORT. Although the FADD/MORT
of FADD in T-lymphocytes also severely repressed adapter molecule does not bind directly to TNF-R1,
CD95 killing yet failed to cause accumulation of pe- it is recruited to the activated receptor via an inter-
ripheral T-cells as seen in lpr and gld mice [158,159]. mediary cytoplasmic DD adapter called TRADD
Mice with a deletion in the FADD gene die at day (TNF-R-associated death domain). TRADD also
11.5 of embryogenesis; their phenotype suggests that binds RIP, thereby linking TNF-R1 to caspase-2 ac-
FADD is essential for embryo development and sig- tivation via RAIDD and CRADD. Both TNF-R1
naling from some (but not all) inducers of apoptosis and TNF-R2 recruit another class of signaling adapt-
[160]. Interestingly, inactivation of FADD by expres- er molecules, called TRAFs (TNFR-associated fac-
sion of a FADD dominant negative molecule or by tors) of which six are currently identi¢ed [172]. Cer-
gene targeting leads to impairment of activation-in- tain of the TRAFs mediate activation of JNK or
duced T-cell proliferation [158,159,161,162]. NF-UB [173], the latter by interaction with the down-
CD95 is also interesting in another aspect of tu- stream signaling kinase NIK. NIK, in turn, activates
mor therapy: several anticancer drugs have been the IUB kinases which phosphorylate and inactivate
shown to sensitize certain cell types to apoptosis by IUB, the endogenous cellular inhibitor of NF-UB

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F13

[174]. Substantial evidence supports the notion that ily conserved family of viral and cellular genes. IAP
NF-UB can act as a powerful suppressor of apoptosis proteins are characterized by the presence of so-
providing an explanation for how TRAFs might ex- called BIR (baculovirus IAP repeats) motifs. These
ert their anti-apoptotic activity. Based on their NF- 65 amino acid repeats are typically found in the
UB-dependent expression and anti-apoptotic func- N-terminal region of the IAPs and mediate various
tion, the cIAPs, TRAF-1 and TRAF-2 as well as types of protein^protein interactions. Certain IAPs
A20 have been proposed to play some role in NF- also contain a C-terminal ring ¢nger which is pre-
UB-mediated prevention of apoptosis [175,176]. sumed to mediate other speci¢c protein^protein in-
TRAFs are held in abeyance in the cytoplasm teractions and acts as a ubiquitin ligase, promoting
through their association in oligomeric complexes the degradation of the X-linked IAP (XIAP) [182].
with I-TRAF [177]. All TRAFs share a conserved Like their viral homologues, several cellular IAPs are
V230 amino acid `TRAF' domain which mediates implicated in suppression of apoptosis. The gene en-
their homo- or hetero-oligomerization with other coding one member of the human IAP family, NAIP
TRAFs, their interaction with the cytoplasmic tails (neuronal apoptosis inhibitory protein), is partially
of members of the TNF-R superfamily, and interac- deleted in individuals with spinal muscular atrophy,
tions with downstream signal transducers [173]. In a disease involving apoptosis of motor neurons [183].
addition to the TRAF domain, most of the TRAFs Moreover, human cIAP-1 and cIAP-2 have both
also contain an N-terminal ring ¢nger plus several been biochemically puri¢ed as part of the TNF-R2-
zinc ¢nger structures which appear to be important TRAF signaling complex [184], an interaction involv-
for their various e¡ector functions. ing the IAPs' BIR domains and the N-terminal do-
TRAF proteins are signal transduction adapter main of TRAF-2. Although interference with TNF
proteins. TRAF-2, -5, and -6 have been shown to signaling could partly explain cellular IAP anti-apo-
be mediators of both NF-UB activation and SAPK/ ptotic activity, at least one other potent anti-apopto-
JNK activation [178]. The activation processes in- tic human IAP homologue, hILP, exhibits no inter-
volve successions of protein^protein interactions action with any tested TRAF protein. Nonetheless, it
and phosphorylation of protein kinases. The pheno- is possible that some cellular IAPs act to modulate
types of Traf-2 knockout mice and transgenic mice TRAF activation of the anti-apoptotic NF-UB tran-
expressing a dominant negative mutant of TRAF-2 scription factor.
indicate that TRAF2 is important for regulation of Another member of the IAP family, survivin, has
lymphocyte proliferation and survival [179,180]. been found to be overexpressed in tumor cells while
They are also compromised for JNK activation, undetectable in terminally di¡erentiated adult tissues
although NF-UB activation is una¡ected. [185]. These observations potentially place survivin in
TRAF-2, TRAF-5, and TRAF-6 interact with the the same class of cell death-inhibiting oncogenes as
downstream kinase NF-UB-inducing kinase (Nik), bcl-2.
which in turn interacts with the kinases within the IAPs are conserved during evolution and have
IUB kinase complex [178]. In addition, the death do- been identi¢ed in Drosophila, C. elegans and in yeast.
main kinase RIP and the serine/threonine kinase Nevertheless, while the Drosophila caspase inhibitor
IRAK have also been reported to interact with DIAP-1 is essential for cell survival [186,187] as
TRAF proteins and mediate NF-UB activation. On mammalian IAPs are inhibited by Smac/Diablo
the other hand, apoptosis signal-regulating kinase (like cytochrome c released from mitochondria dur-
ASK-1, a TRAF-interacting kinase, was recently ing apoptosis [105,106]), the C. elegans and yeast
demonstrated to be a downstream target of TRAF- IAPs do not seem to play a role in the inhibition
2, TRAF-5, and TRAF-6 in the JNK signaling path- of cell death but are rather involved in the regulation
way. of cytokinesis and cell division, respectively
One particularly intriguing potential target of the [188,189]. Such a role has also been identi¢ed for
TRAFs are the IAP proteins. Originally discovered the mammalian survivin protein, in addition to its
in baculovirus as suppressors of host cell apoptosis, anti-apoptotic function [190,191].
the iap [181] genes now comprise a large evolutionar- A possible model to explain the contradictory ap-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F14 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

optotic and anti-apoptotic outcomes of TNF recep- (DcR1/TRID/LIT/TRAIL-R3) and decoy receptor 2
tor signaling emerges from the cloning of another (DcR2/TRUNDD), lack any functional death do-
TRAF-interacting protein, TRIP [173]. TRIP con- main and are therefore unable to transduce a death
tains a ring ¢nger motif and an extended coiled-coil signal (as is the third receptor, osteoprotegerin, a
domain and it associates with TNF receptor family secreted protein). They can therefore act as `decoy
members through its interaction with TRAF pro- receptors' by abstracting TRAIL ligand from pro-
teins. When so associated, TRIP inhibits TRAF- ductive interactions with DR4 or DR5 and their
mediated activation of the apoptosis suppressor presence could explain why some cells are preferen-
NF-UB. Thus, TRAF interactions with cIAPs would tially killed by TRAIL and others not. Indeed, DcR1
suppress apoptosis whilst interactions with TRIP and DcR2 are expressed in many tissues although
would promote it. Which outcome of TNF signaling not in most cancer lines examined. However,
prevails may depend upon the availability of cIAPs although apoptosis depends on the expression of
and TRIP and their relative a¤nities for whichever one or both of the death domain-containing recep-
TRAFs are available in any particular instance. tors DR4 and/or DR5, resistance to TRAIL-induced
In another report the authors showed that in the apoptosis does not correlate with the expression of
presence of the serine/threonine kinase RIP (required the `decoy' receptors. One possibility is that rather
for NF-UB activation by TNF-R1), TNF-R2 triggers than binding to all its receptors with equivalent high
cell death in T-cells whereas in the absence of RIP, a¤nities, TRAIL a¤nity di¡erences exist at physio-
TNF-R2 activates NF-UB [192]. RIP is induced dur- logical temperature, and the cell death-inducing DR5
ing interleukin (IL)-2-driven T-cell proliferation, and receptor seems to be the highest a¤nity receptor
its inhibition reduces susceptibility to TNF-depen- [197]. So the ability of TRAIL to speci¢cally kill
dent apoptosis. tumor cells still awaits a satisfying explanation.
Not much is known about TRAIL's normal bio-
4.3.3. TRAIL receptors logical function. A TRAIL-dependent mechanism of
Recently, an interesting subfamily of TNF recep- monocyte-induced cell cytotoxicity has been reported
tors has been identi¢ed ^ the TRAIL receptors suggesting that TRAIL might be an important e¡ec-
[113,193,194]. TRAIL (TNF-related apoptosis-induc- tor molecule in antitumor activity in vivo [198].
ing ligand), also called apo-2L, is a broadly ex- Preclinical studies with mice and non-human pri-
pressed TNF-related ligand which appears not to mates have shown that indeed TRAIL induced the in
bind either CD95 or TNF-R1. Like all members of vivo regression of tumors ^ in contrast to TNF or
the TNF/CD95L family, TRAIL is synthesized as a CD95 ligand/CD95 antibody ^ without severe side
membrane-bound proprotein which can be cleaved to e¡ects [199]. In particular, a combined treatment of
generate a soluble ligand. Many human tumor cells tumor cells with either chemotherapeutics or ionizing
and tumor cell lines are sensitive to induction of ap- radiation gave promising results [200,201]. Neverthe-
optosis by cell surface or soluble TRAIL [195]. How- less, a recent publication reported that human pri-
ever, normal cells, such as freshly isolated mouse mary hepatocytes (in contrast to mouse hepatocytes)
thymocytes or primary B- or T-cells, are insensitive. are also e¤ciently killed by TRAIL [202,203], a ¢nd-
Such a cell type-dependent response might be con- ing that has to be taken into account for future clin-
strued as indicative of a restricted receptor distribu- ical trials involving TRAIL. Human astrocytes seem
tion. However, the ¢rst two identi¢ed TRAIL bind- to represent another untransformed cell type sensi-
ing receptors (DR4/TRAIL-R1 and DR5/TRICK-2/ tive to TRAIL-mediated killing [199].
TRAIL-R2/KILLER/DR5) are expressed in most
human tissues and some tumor cell lines. TRAIL
induces apoptosis through these two death domain- 5. Cell death regulators
containing receptors requiring FADD and caspase-8,
just like CD95-mediated cell killing [196]. Neverthe- 5.1. Oncoprotein-induced cell death
less, three other TRAIL receptors have since been
identi¢ed. Two of these receptors, decoy receptor 1 A number of oncoproteins induce apoptosis when

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F15

overexpressed in cells [204]. The best characterized genes might hinder expansion of potentially malig-
examples are the transcription factor c-Myc [205] nant cells, proteins such as Myc or E2F may, in
and the adenovirus protein E1A [206], but the list certain circumstances, act as tumor suppressors.
also includes c-Jun [207] and c-Fos [208], both com- For example, E2F-1 knockout mice develop dyspla-
ponents of the AP-1 transcriptional complex, as well sias and lymphocytic hyperplasias [214,215], in part
as components of the G1 cell cycle progression ma- because of insu¤cient apoptosis (nevertheless, loss of
chinery such as E2F1 and cyclin E (while induction E2F-1 reduces tumorigenesis and extends the lifespan
of apoptosis seems to represent the general cellular of Rb-1(+/3) heterozygous knockout mice [216],
response upon oncogene overexpression, in certain demonstrating that E2F-1 also positively regulates
settings the reduction of c-Myc [209], Jun or Fos cell cycle progression).
[210] expression via an unknown mechanism leads Mutagenesis studies show that regions of the
to apoptosis). Because all such oncoproteins promote c-Myc protein necessary for apoptosis induction over-
cell proliferation, their pro-apoptotic activity was lap with regions important for co-transformation
often interpreted as arising from some kind of abor- and include the N-terminal transactivation domain
tive or failed attempt at going through the cell cycle and the C-terminal bHLH-LZ region (basic helix-
^ a mitotic or S phase catastrophe. However, with loop-helix domain with a leucine zipper) involved
the elucidation of its underlying mechanisms it be- in sequence-speci¢c DNA binding and dimerization
came clear that apoptosis is not merely a corrupted with Max [211]. The heterologous protein Max (re-
cell cycle. An alternative variant of the same idea is quired for Myc as a binding partner for DNA bind-
that oncogene-induced apoptosis arises through a ing) is absolutely required for induction of apoptosis
con£ict of growth signals: oncogenes activate apo- by c-Myc in ¢broblasts [217] while Mad-1 overex-
ptosis if their proliferative action is blocked in pression (antagonizing Myc functions by repressing
some way or if the cell's proliferative machinery is gene transcription) inhibits proliferation and apopto-
incompletely activated or coordinated. However, the sis [218]. These data strongly argue that c-Myc in-
idea of a con£ict of signals, whilst intuitively plausi- duces apoptosis through its action as a transcription
ble, does not o¡er any explanation as to the molec- factor ^ presumably by modulation of appropriate
ular nature of such a con£ict, how it arises or is target genes.
de¢ned, or why any con£ict (were it to exist) triggers Several candidate Myc-regulated genes have been
the machinery of apoptosis. identi¢ed [219,220]. Two genes suggested as Myc tar-
The current view of Myc-induced apoptosis is ex- gets, based on the pattern of expression, are orni-
pressed in the `dual signal' hypothesis [211]. Accord- thine decarboxylase (ODC) and cdc25A. ODC is a
ing to this hypothesis Myc promotes both pathways rate-limiting enzyme in polyamine biosynthesis, nec-
at the same time ^ proliferation and apoptosis. The essary for DNA synthesis, while Cdc25A encodes a
apoptotic pathway is suppressed as long as appropri- tyrosine phosphatase involved in activation of the
ate survival factors deliver anti-apoptotic signals. key inducers of mitosis, the cyclin-dependent kinase
Such a scenario would ¢t with a general model of (CDK) complexes. Both candidate Myc targets,
survival/cell death regulation according to which the ODC and cdc25A, when overexpressed, induce apo-
`default' fate of a cell would be cell death unless ptosis in cells lacking survival factors (as Myc over-
suppressed by anti-apoptotic cytokine signaling expression does). It therefore seems possible that the
[212]. Coupling of the `contradictory' pathways of cell proliferation and cell death pathways bifurcate
cell proliferation and cell death downstream of pro- `downstream' of c-Myc. In addition the TNF-R-as-
teins like c-Myc incorporates a potent safety mecha- sociated protein TRAP-1 [221] and the Bcl-2 family
nism for the suppression of carcinogenesis: any le- member Bax [222] have been revealed as further Myc
sion that activates the mitogenic pathway will prove targets possibly involved in Myc-induced apoptosis.
lethal should the a¡ected cell and its progeny out- The ¢nding that c-Myc (and E1A) activates P53
grow the paracrine environment enabling their sur- via p19ARF , a protein encoded by the alternate read-
vival [213]. ing frame of the p16 tumor suppressor gene, has
Since the cell death-promoting activity of onco- provided another possible mechanism linking c-Myc

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F16 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

with the induction of apoptosis [223,224]. In this genesis (many tumors show deregulated Myc expres-
context it is interesting to note that E2F-1 seems to sion and necessarily need to counteract Myc's pro-
induce apoptosis by upregulation of the P53 homo- death function), it has been di¤cult to prove phys-
logue P73 [225,226]. iological relevance of Myc-induced apoptosis. One
A novel Myc-interacting protein, Bin-1, has been reason for that is the di¤culty to delete Myc func-
shown to induce apoptosis and di¡erentiation in neu- tion (which is needed for proliferation) and thereby
roblastoma [227] but the functional relation to Myc- showing impairment of certain apoptotic processes.
induced apoptosis is unclear. The demonstrated dependence of Myc on CD95 sig-
Recently, it was revealed that c-Myc-induced apo- naling and vice versa for cell killing raises the possi-
ptosis requires interaction on the cell surface between bility that every physiological cell death requiring
CD95 and CD95L [228]. With this study the two CD95/CD95L also depends on Myc function. It
previously independent apoptosis pathways (Myc has also been shown that c-Myc (and E1A) sensitizes
and CD95/CD95L) have been interconnected. Dur- target cells for the cytotoxic action of activated NK
ing Myc-induced apoptosis in serum-deprived ¢bro- cells [230] thereby possibly directing NK cytotoxicity
blasts c-Myc acts downstream of the CD95 receptor towards virus-infected and cancer cells.
by sensitizing cells to the CD95 death signal, The precise consequences of Myc overexpression
although in general c-Myc may also act upstream in vivo seem to depend on the particular type of
of CD95/CD95L by inducing expression of their cog- tissue and its potency to suppress apoptosis via sur-
nate genes. vival signals. For example, transgenic expression of a
Expression of c-Myc sensitizes cells to a wide regulatable c-Myc protein in suprabasal keratino-
range of pro-apoptotic stimuli. The best hint towards cytes of the mouse epidermis results in premalignant
the mechanism underlying Myc-mediated sensitiza- papillomatous skin lesions accompanied by angio-
tion comes from studies showing that this pro-apo- genesis [231]. Apparently, ectopic Myc activation in
ptotic e¡ect is mediated through release of mitochon- skin causes proliferation with no detectable apopto-
drial holocytochrome c into the cytosol [229]. sis, although Myc potently triggers apoptosis in vitro
Cytochrome c release is caspase-independent and is in isolated serum-deprived keratinocytes from the
blocked by the survival factor insulin-like growth same transgenic animals. Myc-induced apoptosis in
factor (IGF)-1. While neither P53 nor CD95/Fas sig- intact skin might be suppressed by the presence of
naling was required for Myc-induced cytochrome excess survival signals such as cytokines or extracel-
c release, a dominant negative version of the adapter lular matrix attachments. The dominant loss of
protein FADD (inhibiting CD95 signaling) blocked c-Myc-activated keratinocytes through shedding
caspase activation subsequent to cytochrome c re- may account for the rarity of malignant progression
lease. The emerging model suggests that c-Myc pro- of actinic keratosis (although c-Myc alone is su¤-
motes apoptosis by causing the release of cytochrome cient to induce premalignant skin lesions).
c, but the ability of cytochrome c to promote apo- In contrast to skin, the predominant e¡ect of Myc
ptosis is critically dependent upon other signals such activation in the L-cells of transgenic murine pan-
as CD95 activation. Myc's cytochrome c-releasing creas is apoptosis [232] indicating that c-Myc would
activity might be mediated by upregulation of the be unlikely to initiate a L-cell tumor in the absence of
pro-apoptotic Bcl-2 family member Bax which has an anti-apoptotic lesion.
been suggested as a transcriptional target and medi-
ator of c-Myc-induced apoptosis [222]. 5.2. The Bcl-2 protein family
The fact that IGF-1 inhibits c-Myc-induced cyto-
chrome c release but not c-Myc-dependent prolifer- 5.2.1. The Bcl-2 protein
ation represents additional evidence that the mito- The proto-oncogene bcl-2 was ¢rst discovered as
genic and apoptotic pathways downstream of Myc the target gene present at the translocation site of the
are distinct and separate. t(14;18) chromosomal translocation breakpoint in
While the importance of Myc-induced apoptosis is the tumor cells of approximately 80% of patients
evident in an unphysiological situation like tumori- with human follicular B-cell lymphoma [233]. The

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F17

translocation places the bcl-2 gene under the aegis of (BH1^BH4). BH1, BH2 and BH4, together with
the 5P immunoglobulin heavy chain gene enhancer the carboxy-terminal hydrophobic transmembrane
(EW, chromosome 14), an element highly transcrip- region (membrane anchor) found in most family
tionally active in B-lymphoid cells. Functional stud- members, are important for functional activity. The
ies of the e¡ects of deregulated bcl-2 on lymphocytes BH3 domain seems to be dispensable for the death-
in culture indicated that bcl-2 exerted a novel type of suppressing function of anti-apoptotic Bcl-2 homo-
oncogenic function ^ rather than promoting cell pro- logues although its presence is essential for cytotoxic
liferation or inhibiting di¡erentiation, it suppressed activity in the pro-apoptotic family members [242].
lymphocyte apoptosis [234].
The anti-apoptotic activity of Bcl-2 explains the 5.2.2.1. Bcl-x. Bcl-xL is the closest mammalian
marked oncogenic synergy observed between Bcl-2 relative of Bcl-2 and it too acts as a death suppressor
and c-Myc. Co-expression of both c-Myc and Bcl-2 [243]. The 2.7 kb bcl-x transcript encodes a protein
induces very rapid genesis of lymphoma [235]. Sev- with 44% sequence identity to human and mouse bcl-
eral in vitro studies con¢rm that this synergy arises 2. However, unlike bcl-2, the bcl-x gene encodes two
because Bcl-2 e¡ectively suppresses c-Myc-induced polypeptides arising from alternative splicing. The
apoptosis without signi¢cantly a¡ecting the ability longer mRNA encodes a death-suppressing function-
of c-Myc to drive uncontrolled proliferation [236^ al Bcl-2 homologue (Bcl-xL ) comprising 233 amino
238]. acids with an apparent molecular weight of 28 kDa.
Bcl-2 is expressed in a wide variety of fetal tissues Bcl-xL contains all four BH domains (BH1^BH4)
but in the adult tends to be more restricted to cells found in Bcl-2. The shorter mRNA encodes the
that are rapidly dividing and di¡erentiating. 170 amino acid 18 kDa Bcl-xS protein. Compared
Bcl-2 knockout mice appear almost normal at to Bcl-xL , Bcl-xS lacks a 63 amino acid stretch which
birth but later in life develop hair hypopigmentation includes the BH1 and BH2 domains and it may act
(due to death of melanocytes), distortion of their as a dominant interfering regulator of either Bcl-xL
small intestines and polycystic kidney disease [239]. or Bcl-2 or both.
Kidneys from Bcl-2-de¢cient mice are small and con- Bcl-x is widely expressed and is particularly high in
tain fewer nephrons than those of wild-type animals, brain, kidney and adult thymus. Its expression pat-
their immune systems exhibit depletion of B- and tern in the di¡erent subsets of T-cells is clearly di¡er-
T-cells due to apoptosis and this leads eventually to ent from that of Bcl-2, whose expression is highest in
massive involution of spleen and thymus. In addi- CD43 CD83 (DN) T-cells, is downregulated in
tion, Bcl-2 knockout mice exhibit marked postnatal CD4‡ CD8‡ (DP) thymocytes and rises again in
degeneration of motor neurons, sensory and sympa- CD4‡ or CD8‡ (SP) T-cells. Bcl-xL is transcription-
thetic neurons. Nevertheless, the largely normal pro- ally activated by NF-UB [244] or by STAT 5 [245].
gression of Bcl-2-de¢cient embryos through develop- Transgenic deregulation of Bcl-xL in the lymphoid
ment (despite the critical nature of apoptosis in lineage has consequences very similar to those of Bcl-
development) attests to a signi¢cant functional re- 2 overexpression [246,247]: lymphocytes are pro-
dundancy that mitigates the e¡ects of loss of Bcl-2. tected to varying degrees from killing by dexametha-
sone, Q-irradiation, ionomycin and CD3 ligation but
5.2.2. Other members of the Bcl-2 family clonal deletion of T-cells is una¡ected. Moreover,
Bcl-2 is structurally and functionally conserved Bcl-xL will rescue mature T-cells in Bcl-2-de¢cient
throughout metazoan evolution and it is the proto- mice. The similar functionality of Bcl-2 and Bcl-xL
type of an extended family of related viral and mam- and the fact that both heterodimerize with pro-apo-
malian proteins. This family can be divided into in- ptotic Bcl-2 family members such as Bax and Bak all
hibitors (Bcl-2, Bcl-xL , Bcl-w, B£-1, Brag-1, Mcl-1, argue that Bcl-xL and Bcl-2 function in a common
A1, E1B19K, LMW5-HL and EBV BHRF1) and pathway to antagonize PCD. However, their di¡er-
promoters (Bax, Bak, Bcl-xS , Bad, Bid, Bik, Hrk, ent patterns of expression indicate that their physio-
Bim and Bok) of apoptosis [240,241]. Many members logical roles are distinct. This is most dramatically
of the Bcl-2 family share four conserved domains evidenced by the fact that Bcl-xL -de¢cient mice, un-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F18 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

like those lacking Bcl-2, exhibit embryonic lethality role for Bax in the promotion of apoptosis. Addition
at around day E13 which is accompanied by massive of Bax to isolated mitochondria induces cytochrome
apoptosis in the brain and hematopoietic tissues c release through a mechanism that is suppressible by
[248]. co-addition of Bcl-xL [257]. In vivo it has also been
shown that Q-radiation induces upregulation of Bax
5.2.2.2. Bax. Bax is a prototypic member of the protein with subsequent apoptosis in radiation-sensi-
Bcl-2 family that accelerates or activates apoptosis tive mouse cells. Such DNA damage-induced expres-
rather than suppressing it. Both Bax and its close sion of Bax is, in part, mediated by P53 for which a
relative Bak possess BH1, BH2 and BH3 regions consensus binding site exists in the human [258] and
but their BH4 is poorly preserved. Bax and Bak cy- in the mouse bax gene (although it was shown that
totoxic activity is dependent upon a small region of the putative P53 binding sites were not occupied by
each protein that comprises the BH3 domain. The protein in vivo in primary murine thymocytes either
same BH3 region mediates dimerization between before or after induction of P53 [259]). Thus, for
Bax/Bak and Bcl-2, Bcl-xL , E1B 19K, Bid or itself example, neurons from bax knockout mice show
[249]. Bax accelerates apoptosis in prolymphocytic much reduced apoptosis following DNA damage or
FL5.12 cells upon IL-3 withdrawal [241] and also excitotoxin exposure both of which activate P53 re-
counters the death-suppressive activity of Bcl-2: the sponses [260]. Moreover, Bax de¢ciency fosters drug
higher the expression levels of Bax compared to Bcl- resistance as well as oncogenic transformation by
2, the greater the suppression of Bcl-2 function. On attenuating P53-dependent apoptosis [261]. Indeed,
this basis, a `rheostat' model for Bcl-2/Bax function in a choroid plexus epithelial tumor model induced
has been proposed [250] in which the propensity to by transgenic expression of a truncated SV40 large
undergo PCD depends upon the relative ratios of T-oncoprotein that lacks the ability to inactivate
Bax/Bax homodimers, Bcl-2/Bax heterodimers and P53, absence of Bax greatly diminishes the ability
Bcl-2/Bcl-2 homodimers: an excess of Bax homo- of P53 to retard tumor growth [262]. Nevertheless,
dimers promotes cell death, whereas Bax complexed Bax cannot be the sole apoptotic e¡ector of P53
with Bcl-2 favors survival. It is still not clear, how- because thymocytes, and many other cell types,
ever, whether Bax promotes PCD, or whether it from Bax knockout mice behave normally with re-
merely suppresses the protective e¡ect of Bcl-2, or spect to P53-dependent DNA damage responses
whether Bax and Bcl-2 regulate cell viability through [241]. Moreover, transgenic expression of Bax is un-
interaction with discrete downstream e¡ectors [251]. able to substitute for the absence of P53 in thymo-
Nevertheless, some reports have shown that Bcl-2 cytes exposed to genotoxins [256].
and Bcl-x can inhibit apoptosis and Bax can promote bax knockout mice appear to develop normally,
apoptosis by a heterodimerization-independent indicating, as with bcl-2, a su¤cient amount of func-
mechanism [252^254] and that Bak can accelerate tional redundancy conferred by other cellular pro-
chemotherapy-induced cell death independently of teins (also bak3=3 mice were found to be develop-
its heterodimerization with Bcl-xL and Bcl-2 [255]. mentally normal while bax3=3 bak3=3 mice display
Bax is expressed in a wide variety of tissues includ- multiple developmental defects indicating overlap-
ing lymphoid organs, lung, stomach and kidney. ping roles for Bax and Bak in the regulation of ap-
Transgenic expression of Bax in T-cells of mice con- optosis during mammalian development [263]).
¢rms the pro-apoptotic activity of Bax [256]: a¡ected Nonetheless, Bax-de¢cient mice demonstrate a few
cells exhibit accelerated apoptosis in response to interesting phenotypes [264]: bax null mice display
Q-radiation, dexamethasone and etoposide. When thymocyte and B-cell hyperplasia ^ a role perfectly
T-cell-targeted bax transgenic mice are crossed with consistent with the idea of Bax being a positive ef-
their bcl-2 transgenic counterparts, the ratio of Bax fector of apoptosis. Likewise, neurons from bax
to Bcl-2 present in primary T-cells determines net knockout mice exhibit marked resistance towards ap-
sensitivity to apoptotic stimuli, as predicted by the optosis induced by trophic factor deprivation [265] as
`rheostat' model. well as insensitivity to toxins. Surprisingly, male bax
In vitro and transgenic data con¢rm an important knockout mice become sterile due to a block in

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F19

sperm production. In this case the pathology linked by caspase-8 and the truncated form then translo-
to loss of Bax seems to involve rather induction of cates to mitochondria [279] where it activates Bax
excess and ectopic apoptosis. Another interpretation [102,280] and Bak [103] by inducing oligomerization
is that Bax normally mediates elimination of certain of these pro-apoptotic Bcl-2 family members. Never-
cells during development which, when retained in theless, in HeLa cells treated with staurosporine to
bax3=3 mice, lead to inappropriate cell death during undergo apoptosis, it is the full length Bid that trans-
spermatogenesis, perhaps by a¡ecting the availability locates to the mitochondria suggesting that caspase-
of trophic signals. induced Bid cleavage is not an essential requirement
for its movement to mitochondria [280].
5.2.2.3. Bad and other BH3 domain only Bcl-2 fam- Recently, another BH3-only protein, Noxa, was
ily members. Egl-1 of C. elegans and at least seven described to be regulated by P53 [281]. Like other
mammalian proteins (Bad, Bik, Blk, Hrk, Bid, Bim, Bcl-2 family members it localizes to the mitochondria
Noxa) share only the short (nine to 16 residue) BH3 after an apoptotic stimulus and is able to interact
domain with the Bcl-2 family. This domain allows with further Bcl-2 family members.
them to bind to the pro-survival Bcl-2-like molecules
and neutralize their function. 5.2.3. Bcl-2 family function
Bim for example was shown to be required for Although Bcl-2 has long been recognized as an
hematopoietic homeostasis and as a barrier to auto- oncogene that acts as a suppressor of apoptosis,
immunity. Moreover, particular death stimuli appear the molecular basis for Bcl-2's anti-apoptotic action
to activate apoptosis through distinct BH3-only pro- has until recently been mysterious. Confocal and
teins [266]. electron microscopy studies show Bcl-2 to be local-
Bad was originally identi¢ed by yeast two-hybrid ized to the outer mitochondrial and nuclear mem-
screening and expression cloning as a 204 residue branes and the ER. The mitochondrial localization
(22.1 kDa) protein that binds Bcl-2/Bcl-xL and pro- of Bcl-2 is perhaps especially intriguing in the light of
motes apoptosis [267]. However, unlike many other the emerging role of the mitochondrion in apoptosis.
known Bcl-2 family members, Bad ^ like Bid ^ has At ¢rst this mitochondrial location fostered early
no identi¢able C-terminal membrane anchor se- speculation that Bcl-2 might have some role in reg-
quence and is probably not an integral membrane ulating oxidative phosphorylation [282,283], a notion
protein. Its location in cells is dynamically controlled reinforced by the ability of Bcl-2 to suppress cell
by its association/dissociation with other Bcl-2 family death induced by oxidative damage, possibly by se-
proteins [268]. The 1.1 kb bad mRNA is co-expressed questering free oxygen radicals. However, analysis of
with Bcl-xL in many mouse tissues and two-hybrid cells grown under highly anoxic conditions [284] and
analysis and co-immunoprecipitation analyses indi- cells lacking mitochondrial DNA (and, hence, mito-
cate that the Bad protein heterodimerizes with both chondrial respiration) [285] indicated there was no
Bcl-2 and Bcl-xL but not with Bcl-xS , Bax, Mcl-1, A1 direct link between the function of anti-apoptotic
or itself. Experiments indicate that Bad counters the activity of Bcl-2 and oxidative phosphorylation.
death repressor activity of Bcl-xL , and probably ex- Moreover, levels of cellular ATP or oxygen con-
erts its pro-apoptotic function by competing with sumption are not a¡ected by Bcl-2 [286,287].
Bax for Bcl-xL binding. Another possible mechanism by which Bcl-2 might
Recently, regulation of Bad function has been suppress apoptosis arose from observations that Bcl-
linked to phosphorylation [269]. In response to IL- 2 expression can a¡ect intracellular Ca2‡ homeosta-
3 and other growth factors, Bad is phosphorylated at sis [241]. Alterations in intracellular Ca2‡ concentra-
three sites (Ser-112, Ser-136 and Ser-155); it then tions are known to in£uence PCD so it remains pos-
binds to the cytosolic 14-3-3 protein and becomes sible that Bcl-2 either directly modulates calcium
sequestered and functionally inactivated [270^277]. channels or acts to protect lipid membranes from
In contrast, regulation of another BH3-only do- damage by peroxide radicals which is known to dis-
main protein, Bid [278], is regulated by proteolytic rupt Ca2‡ homeostasis.
cleavage rather than phosphorylation. Bid is cleaved Both Bcl-2 and Bcl-xL have been shown to interact

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F20 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

directly with a variety of intracellular proteins. C. elegans. In the nematode, the adapter protein
Known interactors include pro-apoptotic Bcl-2 fam- Ced-4 binds to Ced-3 [25,27,28] and induces ATP-
ily members (such as Bax [288], Bak and Bik), the dependent autoactivation of the Ced-3 zymogen
Raf-1 protein kinase, the protein phosphatase calci- [30,297]. Ced-9, the C. elegans homologue of Bcl-2,
neurin, R-Ras and H-Ras, the P53 binding protein physically interacts with Ced-4 and thereby blocks its
P53-BP2, the prion protein Pr-1 and several other activation of Ced-3. Although nematode genetics has
proteins with unclear functions. The functional rele- identi¢ed only Ced-3, Ced-4 and Ced-9 as players in
vance of many of these interactions is still a matter this process, it is possible that there are other com-
of debate, e.g. the observed binding of Bcl-2 to the ponents in such `apoptosome' complexes that cannot
C. elegans protein Ced-4 or its mammalian ortho- be discriminated genetically by virtue of the fact that
logue Apaf-1. While some groups could show such they are essential proteins involved in some other
an interaction [93], others failed to do so [95,289]. mandatory biological process ^ cytochrome c being
The interaction between Bcl-2 and the pro-apoptotic a candidate of this type. By analogy with the nem-
Bcl-2 family members is regarded as particularly im- atode, Bcl-2/Bcl-xL may inhibit activation of the api-
portant and will be discussed below. cal caspase-9 by the Ced-4 orthologue Apaf-1. In
Currently, three (non-exclusive) models are used to such a model, the role of pro-apoptotic Bcl-2 family
explain Bcl-2 function. These models describe Bcl-2 proteins is to displace Bcl-2/Bcl-xL from the Apaf-1/
proteins as ion channels, or as proteins that modu- cytochrome c/caspase-9 complex and so trigger cas-
late activation of caspases, or as inhibitors of cyto- pase-9 autoactivation [26].
chrome c export from mitochondria. In this context, the dynamics of Bax subcellular
The interesting possibility that Bcl-2 family pro- localization are particularly interesting. In contrast
teins act as membrane channels emerges from eluci- to Bcl-2/xL , Bax ^ at least in some cell types ^ ex-
dation of the detailed three-dimensional structure of hibits a di¡use localization throughout the cytosol.
Bcl-xL , which resembles the membrane insertion do- However, upon induction of apoptosis, it rapidly re-
main of bacterial toxins such as diphtheria toxin and localizes to a punctate distribution that partially co-
colicin [290]. Like Bcl-xL , these membrane insertion localizes with mitochondria [298] where it presum-
domains contain two central helices consisting of ably associates with Bcl-2/Bcl-xL . This builds a
apolar residues that are able to span a membrane. persuasive argument that relocalization of Bax is
Because diphtheria toxin can form membrane pores part of a trigger for apoptosis, although the molec-
in a pH-dependent manner, it has been suggested ular basis for its subcellular movement is still un-
that Bcl-2 and its homologues act in a similar way known.
to generate pores in cytoplasmic and mitochondrial Biochemical analysis has shown that release of mi-
membranes (possibly regulated by voltage- or pH- tochondrial cytochrome c is a central process in as-
dependent signals). Indeed, ion channel activity has sembly of the `apoptosome' ^ the multimeric com-
been detected with Bcl-2, Bcl-xL and with the pro- plex comprising caspase-9, Apaf-1, dATP and
apoptotic Bax protein in isolated lipid bilayers in cytochrome c that triggers autocatalytic cleavage
vitro, although at present there is no direct evidence and activation of caspase-9 [37]. Cytochrome c nor-
for in vivo channel formation [291,292]. Bax is able mally resides in the intermitochondrial membrane
to interact with VDAC [293] and/or the adenine nu- space, and the molecular mechanism of its transloca-
cleotide translocator, ANT [294], to release cyto- tion is not yet fully understood. One possibility is
chrome c. After oligomerization it may also form that Bcl-2/xL , which resides on the outer surface of
cytochrome c-releasing pores by itself [295]. This cy- the outer mitochondrial membrane, somehow acts to
tochrome c release can be blocked by BH4 domain- forestall cytochrome c release [299^301].
containing Bcl-2 family members such as Bcl-2 and It is possible that Bcl-2/xL proteins operate
Bcl-xL [296]. through all three of these mechanisms. Perhaps, the
A direct role for Bcl-2 proteins in regulating acti- channel-forming activity of Bcl-2/xL proteins modu-
vation of apical (regulatory) caspases is indicated by lates cytochrome c release whilst other regions of the
analogy with the basal apoptotic machinery of protein interact with Apaf-1. Interestingly, Bcl-2 it-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F21

self has been identi¢ed as a caspase target that is acterized activities of P53 are the induction of either
cleaved during apoptosis to generate a carboxy-ter- cell growth arrest or apoptosis [310]. Levels of P53
minal cleavage product that promotes apoptosis rapidly increase following DNA damage, mainly be-
[252]. This may be part of a positive feedback loop cause the normally short-lived P53 protein becomes
within the cell death machinery. stabilized, and this appears to be an important com-
For a long time the question has remained open ponent of the G1 arrest and apoptosis response that
whether pro-apoptotic Bcl-2 family members are pro- follows DNA damage [311]. The induction of growth
moting cell death as active molecules of the apoptotic arrest by P53 depends on its activity as a sequence-
machinery or whether they rather neutralize anti-ap- speci¢c transcriptional activator and the P21
optotic Bcl-2 homologues which themselves represent (WAF1/CIP-1) protein appears to be the major e¡ec-
proteins actively participating in default survival tor of P53-mediated G1 cell cycle arrest after DNA
pathways. Recent data support the ¢rst scenario: damage [311]. P21WAF1=CIPÿ1 binds to and inhibits
bax3=3 bak3=3 double knockout cells are resistant cyclin-dependent kinases, thereby blocking cell pro-
towards radiation- and drug-induced apoptosis (in- liferation.
terestingly they remain sensitive towards CD95-medi- How P53 mediates apoptosis is less clear and
ated cell killing) and overexpression of BH3-only seems to involve both transcriptional activation-de-
proteins that bind pro-survival Bcl-2 family members pendent and -independent pathways [312,313]. The
fail to induce apoptosis in the absence of Bax and P21 cyclin-dependent kinase inhibitor is dispensable
Bak [104,263]. If the apoptotic Bcl-2 homologues are for P53-dependent oncogene-induced apoptosis [312].
indeed the `active' molecules while the role of anti- A di¡erent set of P53 target genes might regulate
apoptotic Bcl-2 family members is just to inhibit growth arrest and apoptosis, respectively. Both func-
them by binding, then Bcl-2, Bcl-xL etc. may not tions seem to be genetically dissectable: a human
be needed in the absence of Bax- and Bak-like mol- tumor-derived P53 mutant was identi¢ed which has
ecules as seems to be the case in yeast. speci¢cally lost its apoptotic function but not its cell
Bcl-2 and other related family members not only cycle function [314]. A model has been proposed in
inhibit or promote apoptosis e¤ciently but also have which upstream e¡ectors (such as the atm gene prod-
an in£uence on cell cycle progression. While over- uct which is defective in ataxia telangiectasia and
expression of Bcl-2, Bcl-xL , Epstein^Barr virus pro- which is part of a pathway responding to DNA dam-
tein BHRF1, adenovirus E1B19kD or MCL-1 can age as a result of ionizing radiation) selectively acti-
slow entry of cells into the cell cycle and promote vate P53 to regulate speci¢c downstream pathways,
exit of cells from the cycle [302^305], overexpression providing a mechanism for controlling distinct cell
of the pro-apoptotic Bax protein can accelerate cell cycle and apoptotic responses [315].
cycle entry [306,307]. Some evidence suggests that the The decision whether an individual cell undergoes
anti-apoptotic function of Bcl-2 can be genetically growth arrest or apoptosis following P53 activation
separated from its inhibitory e¡ect on cell cycle entry appears to depend on a variety of factors, such as
[308]. The molecular basis for this cell cycle regula- environmental conditions and the cell type. It seems
tion by Bcl-2 family members remains unclear but in safe to assume that, in the context of cellular DNA
the case of MCL-1 the cell cycle regulatory function damage following P53 cell cycle arrest, some `control
is mediated through its binding of proliferating cell mechanism' evaluates whether the DNA can be re-
nuclear antigen (PCNA), an interaction which seems paired in a reasonable time or whether the damage is
to inhibit PCNA's ability to promote progression so severe that the cell undergoes apoptosis in a P53-
through S phase [305]. dependent manner.
Analysis of P53-regulated gene expression patterns
5.3. P53 and apoptosis using oligonucleotide arrays has demonstrated that
the nature of the P53 response in diverse mRNA
The p53 tumor suppressor gene is functionally in- species depends on the levels of P53 protein in a
activated in 70% of human tumors [309]. P53 can cell, the type of inducing agent or event, and the
function as a transcription factor and the best char- cell type employed [316]. Of 6000 genes examined

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F22 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

for P53 regulatory responses, 107 induced and 54 [325]. This molecule might link death receptors
repressed genes fell into categories of apoptosis and such as CD95 (which also contain a death domain
growth arrest, cytoskeletal functions, growth factors as a protein^protein interaction motif) to P53-medi-
and their inhibitors, extracellular matrix, and adhe- ated cell death. Through direct cloning of P53 bind-
sion genes. ing sequences from human genomic DNA, a novel
Several mechanisms have been suggested by which gene, designated p53AIP1 (P53-regulated apoptosis-
P53 protein might signal to the apoptotic machinery: inducing protein 1), has been isolated, which is in-
the pro-apoptotic Bcl-2 family member Bax appears ducible by wild-type P53 [326]. P53AIP1 is located
to be transcriptionally induced by P53 after DNA within mitochondria and its overexpression leads to
damage in certain cell types [258]. However, Bax ap- disruption of the mitochondrial membrane potential
pears to contribute only in part to P53-mediated cell and apoptotic cell death. Interestingly, phosphoryla-
death [241]: the pro-apoptotic P53 function seems tion of Ser-46 within P53 regulates the transcription-
not to be e¡ected in bax-de¢cient mice and apoptosis al activation of this apoptosis-inducing gene.
in transgenic Bax-expressing thymocytes is neither While in general oncogene-induced apoptosis
increased nor accelerated in a p533=3 background. seems to be P53-dependent [309] it is still a matter
In addition, genomic DMS footprinting has revealed of debate whether this is also true for Myc-induced
that the activity of the murine Bax promoter is regu- cell death. Although data from p53 knockout cells
lated by Sp1/3 and E-box binding proteins but not support a P53 dependence of Myc killing [327] there
by P53 [259]. are other examples for P53-independent c-Myc-in-
In another scenario suggested for P53-mediated duced apoptotic cell death [328].
cell death, both DNA damage and overexpression P53 induces apoptosis not only through transacti-
of a wild-type p53 transgene cause transcriptional vation-dependent but also through non-transcrip-
induction of the TRAIL receptor DR5 through an tional mechanisms. In human vascular smooth
intronic sequence-speci¢c DNA binding site [317]. muscle cells, P53 activation transiently increased sur-
DNA damage fails to induce DR5 expression in tu- face CD95 expression by transport from the Golgi
mor cells with mutated p53. Interestingly, overex- complex [329]. This cell surface redistribution of cy-
pression of DR5 (by transfection) appears to cause toplasmic CD95 transiently sensitizes cells to CD95-
apoptosis in a TRAIL ligand-independent manner, induced apoptosis and occurs without new RNA
implying that simply increasing the expression of synthesis.
DR5 following P53 induction could result in activa- P53 can also contribute to apoptosis by direct sig-
tion of the apoptotic pathway [318]. naling at the mitochondria [330]: a small fraction of
UV- and X-ray radiation-induced apoptosis is stress-induced P53 protein after DNA or hypoxic
P53-dependent and seems to be mediated by activa- damage tra¤cs to mitochondria. This mitochondrial
tion of CD95 [319^321] providing another potential localization precedes the release of cytochrome c and
link between P53 and a (cell surface) death receptor. procaspase-3 activation and is blocked by overex-
The CD95 receptor has already been suggested as a pression of anti-apoptotic Bcl-2 proteins. Redirecting
target gene for transcriptional activation by P53 P53 from the nucleus and targeting it to mitochon-
[322]. dria by using mitochondrial import leader peptides is
Other genes capable of inducing apoptosis have su¤cient to induce apoptosis in P53-de¢cient cells,
been suggested as P53 targets: c-fos proto-oncogene even with a transcriptionally inactive p53 mutant.
transcription is stimulated by P53 in cells undergoing As can be expected for an important protein such
P53-mediated apoptosis [323] and MCG10 has been as P53 and its pleiotropic signaling, regulation of P53
identi¢ed as a novel P53 target gene encoding a KH activity itself is rather complex and occurs at many
domain RNA binding protein capable of inducing di¡erent levels [331,332]. A few examples are summa-
apoptosis and cell cycle arrest in the G2 ^M phase rized in the following paragraphs.
of the cell cycle [324]. Very recently, Pidd, a new The oncoprotein Mdm2 binds to P53 and is a
death domain-containing protein, has been shown physiological negative modulator of P53 activity.
to be induced by P53 and to promote apoptosis Mdm2 can inhibit the function of P53 as a transcrip-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F23

tion factor by binding to the P53 N-terminus, there- cell cycle the Rb protein is phosphorylated by cyclin-
by preventing P53 from interacting with the tran- dependent kinases resulting in release of bound E2F
scriptional machinery and inhibiting activation of factors, which are necessary for the G1/S transition.
P53 responsive genes. Loss of Rb results in uncontrolled proliferation and
Mdm2 binding also targets P53 for ubiquitin- P53-dependent apoptosis; this might explain why
mediated degradation. Upon DNA damage disrup- many tumor types display mutations in both Rb
tion of the Mdm2/P53 interaction is seen as a mech- and p53 [340].
anism to increase P53's half-life in response to DNA The P53 homologue P73 seems to play a role in
damage [331]. Mdm2 itself is regulated by the small E2F-1-induced apoptosis [225,226]: activation of P73
ubiquitin-like modi¢er protein SUMO-1: conjuga- provides a means for E2F-1 to induce death in the
tion of Mdm2 with SUMO-1 excludes its self-ubiq- absence of P53 and might constitute a P53-indepen-
uitination and increases Mdm1's ability to ubiquiti- dent, anti-tumorigenic safeguard mechanism.
nate P53 [333]. Reduced Mdm2 sumoylation in
response to DNA damage contributes to P53 stabil-
ity. 6. Inhibition of apoptosis and tumorigenesis
The alternative reading frame (ARF) in the locus
encoding the Cdk inhibitor P16 can also bind to Since the discovery of bcl-2 as an oncogene that
Mdm2 and thus prevent Mdm2-mediated proteolytic promotes cell survival it has been widely acknowl-
destruction of P53, although ARF-dependent mech- edged that anti-apoptotic genetic lesions are neces-
anisms do not seem to be essential for the induction sary for tumors to arise. The net expansion of a
of P53 levels in response to genotoxic stress clone of transformed cells is not only achieved by
[334,335]. an increased proliferative index but also by a de-
Other mechanisms exist to increase P53 levels: the creased apoptotic rate. The evidence is mounting,
gene mutated in ataxia telangiectasia, ATM, is a ser- principally from studies in mouse models and cul-
ine/threonine kinase and P53 stabilization in re- tured cells, as well as from descriptive analysis of
sponse to ionizing radiation is believed to occur in biopsied stages in human carcinogenesis, that ac-
part by ATM-dependent phosphorylation of P53 at quired resistance toward apoptosis is a hallmark of
Ser-15, thereby disrupting the Mdm2/P53 complex most and perhaps all types of cancer [233,341,342].
and increasing the half-life and transcriptional prop- Enhanced cell survival is needed at several steps
erties of P53 in a DNA damage-dependent fashion during tumorigenesis: deregulated oncogene expres-
[315,336]. The DNA-dependent protein kinase DNA- sion not only leads to accelerated proliferation, but
PK is another candidate kinase for phosphorylating concomitantly induces apoptosis which needs to be
P53 on Ser-15 [337]. As mentioned before, phosphor- suppressed for the transformed cell to survive and
ylation of Ser-46 within P53 regulates transcriptional multiply. Having reached a certain tumor size, su¤-
activation of the pro-apoptotic P53 target gene cient nutrition for every tumor cell becomes re-
p53AIP1 [326]. P53-mediated cell growth arrest and stricted; starvation of (tumor) cells from cytokines
apoptosis is also modulated by acetylation and de- usually leads to apoptotic cell death. This selects
acetylation [338]. for further apoptosis resistance (and for angiogene-
Hypoxia (lack of oxygen in body tissue or tumor sis). Finally, the metastasizing tumor cells, deprived
mass) induces accumulation of wild-type P53 of cell^cell contact and of their normal (growth fac-
through hypoxia-inducible factor 1K (HIF-1K)-de- tor) environment, are prone to anoikis ^ further mu-
pendent increase in p53 protein activity [339]: HIF- tations can suppress this cell death which occurs
1K binds to and stabilizes P53 but has no direct e¡ect when untransformed adherent cells detach from the
on P53 transcriptional activity. extracellular matrix.
Loss of the tumor suppressor Rb function may An elegant transgenic mouse study provides in
contribute to P53-induced apoptosis [340]. In its hy- vivo insight into at which stages during multistep
pophosphorylated state, Rb binds to and inactivates tumorigenesis apoptosis is downregulated [343].
transcription factors of the E2F family. During the Mice expressing the SV40 large T-antigen oncogene

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F24 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

in the L-cells of the pancreatic islets develop islet cell 6.1. Bcl-2 family
carcinoma. Di¡erent stages during tumor develop-
ment can clearly be distinguished: an initial hyper- The Bcl-2 family has been implicated in tumori-
proliferation involving 50^70% of the islets in the genesis for a long time. Bcl-2 was identi¢ed in human
pancreas is followed by an angiogenic phenotype follicular B-cell lymphoma where it becomes overex-
(10%) before ¢nally a few (1^2%) solid, encapsulated pressed as a consequence of a t(14;18) chromosomal
tumors emerge. During this multistep process IGF-2 translocation. Bcl-2 synergizes e¡ectively with cell
expression concomitant with the switch to hyperpla- proliferation-promoting oncogenes such as Myc
sia was shown to have a survival (rather than a mi- [233]: overexpression of Myc normally leads to in-
togenic) function. Furthermore, transgenic overex- duction of apoptosis which is inhibited in vitro and
pression of Bcl-xL led to an increase in tumor in vivo by Bcl-2 expression [345]. In addition to fol-
incidence due to downregulation of apoptosis at the licular lymphoma, Bcl-2 levels are elevated in a
conversion from the late preneoplastic state of angio- broad range of other human cancers including carci-
genic islets to that of islet cell carcinoma. nomas of breast, prostate, ovary, colon, and lung,
Several other studies have shown that during tu- indicating that this molecule might have a role in
mor progression apoptosis resistance is increasing, raising the apoptotic threshold in a broad spectrum
for example in AKR lymphoma [344]. of cancerous disorders [346].
The following is a brief summary of data obtained In addition to its anti-apoptotic potential, Bcl-2
for the tumorigenic potential of some prominent pro- also inhibits entry into cell cycle, and the two Bcl-2
teins known to be important for the regulation of functions are genetically separable [308]. The anti-
apoptosis (Fig. 3 summarizes current knowledge proliferative behavior probably does not support tu-
about some apoptosis pathways involving proteins mor growth and indeed loss of antimitotic e¡ects of
which have been found in tumors to be deregulated). Bcl-2 with retention of anti-apoptotic activity during

Fig. 3. Signaling pathways involved in PCD. Overview of how di¡erent signaling pathways interconnect to regulate and determine the
apoptotic behavior of the cell. Molecules highlighted in red have been found mutated in tumor cells leading to increased resistance
against apoptotic stimuli.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F25

tumor progression has been observed in a mouse apoptosis ^ and complementary to Bcl-2 ^ Bax in-
model [347]. duces proliferation in T-cells, consequently Bax
Overexpression of the anti-apoptotic Bcl-2 homo- transgenic mice have an increased percentage of cells
logue Bcl-xL enhances SV40 large T-antigen-medi- in cycle. Bax-induced proliferation seems to cooper-
ated transformation of pancreatic L-cells in transgen- ate in tumorigenesis with increased apoptosis resis-
ic mice [343]. Bcl-xL is a transcriptional target gene tance due to P53 de¢ciency. Obviously, at least in
of the Stat-5 (and Stat-1 and Stat-3) transcription mice the dual roles of Bax can either accelerate or
factor [245,348]. Interestingly, Stat-5 is constitutively inhibit tumorigenesis depending on the genetic con-
phosphorylated and activated in Bcr/Abl-trans- text.
formed cells [349,350]. The bcr/abl oncogene is the For Bak, reduced protein expression (and elevated
product of the t(9;22) translocation in chronic mye- Bcl-xL expression) has been demonstrated in primary
logenous leukemia. The resulting chimeric Bcr/Abl colorectal adenocarcinomas [356], a tumor type for
protein displays enhanced tyrosine kinase activity which the transformation of colorectal epithelium to
which leads to increased activation of Stat-5 and carcinomas had always been linked to inhibition of
elevated levels of its target gene Bcl-xL [349,351]. apoptosis [357].
Expression of Bcr/Abl in hematopoietic cells induces Because of functional redundancy between the
resistance to apoptosis in cell^cell and cell^matrix many Bcl-2 homologues the overall ratio between
interactions, thereby contributing to malignant pro- the amount of pro- and anti-apoptotic Bcl-2 family
gression by conferring a survival advantage. members might represent the important `rheostat' for
Inactivation of pro-apoptotic Bcl-2 family mem- the cell's sensitivity towards apoptotic stimuli. Re-
bers may also contribute to tumorigenesis: a fraction lapse in childhood acute lymphoblastic leukemia
of human colon carcinomas with the mutator pheno- (ALL) for example is associated with a decrease in
type contain frameshift mutations in the bax gene the Bax/Bcl-2 ratio and with loss of spontaneous
[352]. It has also been shown that P53-dependent caspase-3 processing in vivo [358]. The decreased
expression of Bax is induced in slow-growing apo- Bax/Bcl-2 ratio contributes to the observed distur-
ptotic transgenic mouse brain tumors; tumor growth bance of apoptotic pathways in ALL relapse.
is accelerated and apoptosis drops by 50% in Bax-
de¢cient mice [262]. Another transgenic mouse study 6.2. TNF receptor family
showed that haploid loss of bax leads to accelerated
mammary tumor development in C3(1)/SV40 T-anti- Given the immunological importance and e¡ec-
gen transgenic mice and that this increase in tumori- tiveness of apoptosis induced by TNF receptor fam-
genesis is due to a reduction in the protective apo- ily members one would expect their functional inac-
ptotic response at the preneoplastic stage [353]. In tivation to occur regularly during tumorigenesis
human colorectal cancer cells inactivation of Bax [359]. For example, expression of the soluble form
completely abolished the apoptotic response to the of CD95, which competes with the membrane-bound
chemopreventive agent sulindac and other non-ste- receptor for natural ligand in human serum, corre-
roidal anti-in£ammatory drugs in vitro [354]. These lates with lymphoma (and autoimmune disease) and
results implicate Bax as a tumor suppressor. loss of CD95 receptor accelerates lymphomagenesis
On the other hand, Bax-de¢cient mice (without in EW L-Myc transgenic mice [360]. Of 150 cases of
any further tumorigenic transgene expression) do human non-Hodgkin's lymphoma examined, 16 of
not display an increased incidence of spontaneous the tumors (11%) showed somatic CD95 mutations
cancers and Bax de¢ciency does not further acceler- indicating a link between CD95 receptor mutation,
ate oncogenesis in mice also de¢cient in P53 [355]. cancer and autoimmunity [361]. Mutation analysis
Surprisingly, transgenic overexpression of Bax in for the entire coding regions of the CD95 gene
T-cells in p53 knockout mice leads to accelerated were performed in 43 cases of gastric cancer and
(rather than decreased) T-cell lymphomagenesis ¢ve (11%) missense mutations were detected, all of
although apoptosis was increased due to the pro-ap- them in the death domain of the receptor [362]. In 81
optotic function of Bax. In addition to promoting de novo childhood T-lineage ALLs (T-ALL) exam-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F26 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

ined for the presence of CD95 mutations, one patient CD95L expression is involved in another interest-
with a heterozygous mutation in exon 3 of CD95 ing aspect of tumor biology: several tumors express
associated with decreased CD95-mediated apoptosis CD95L on their surface having downregulated their
was found [363]. In another study, CD95-de¢cient own CD95 receptor expression. This might protect
transgenic mice overexpressing Bcl-2 develop acute the tumor cells against CD95L-induced apoptosis
myeloblastic leukemia which again suggests that while at the same time any activated T-cell bearing
CD95 may act as a tumor suppressor to control leu- CD95 on its surface and attacking the tumor would
kemogenic transformation in myeloid progenitor be killed [369]. The same mechanism is used to es-
cells [364]. tablish immune privilege and could explain why can-
The anti-apoptotic protein c-FLIP inhibits death cer patients' immune systems fail to eliminate the
receptor-induced apoptosis and was found to be tumor. Nevertheless, the `tumor strikes back (by ex-
overexpressed in human melanomas. Two groups pressing CD95L)' model has been challenged recently
have demonstrated that overexpression of c-FLIP [370].
leads to immune escape of tumors in vivo presum-
ably by blocking CD95-mediated cytotoxic T-cell 6.3. Caspases
killing [153,154,365]. Certain tumors may also escape
CD95 ligand-dependent immune-cytotoxic attack by Given the apparent central role of the caspase cas-
expressing a decoy receptor (DcR3) that blocks cade for the apoptotic pathway, it is striking that so
CD95 ligand and its killing activity [155]. The far functional inactivation of caspases has been iden-
DcR3 gene was ampli¢ed and expressed in about ti¢ed in only a few tumors (although other mutations
half of 35 primary lung and colon tumors studied. either prevent caspase cleavage or directly inhibit ac-
Ampli¢cation and expression of DcR3 was also tivated caspases, as discussed in this section). Cas-
found in Epstein^Barr virus- and HTLV-1-associated pase-8 is frequently inactivated in neuroblastoma, a
lymphomas, probably allowing the virus to escape childhood tumor of the peripheral nervous system
from the immune system during lymphomagenesis [371,372]. The gene is silenced through DNA meth-
[366]. ylation as well as through gene deletion, preferen-
Recently, a new component of the CD95-DISC tially in neuroblastoma with N-Myc ampli¢cations.
has been identi¢ed, SADS (small accelerator for Caspase-8 null tumor cells are resistant to death re-
death signaling), which enhances the interaction of ceptor- and doxorubicin-mediated apoptosis. And
FADD and procaspase-8 and which is downregu- the resistance to TRAIL-induced apoptosis in prim-
lated in patients with colon carcinoma [367,368]. itive neuroectodermal brain tumor cells correlates
This downregulation most likely leads to an in- with a loss of caspase-8 expression [373], again
creased resistance towards CD95-induced cell death showing that in principle caspase-8 can act as a tu-
of the tumor cells (as observed with colon carcinoma mor suppressor. Decreased caspase-1 protein levels
cells despite CD95 cell surface expression). have been reported as a potential step in the loss
Nevertheless, although lpr and CD95knockout of apoptotic control during prostate tumorigenesis
mice develop a severe lymphoaccumulation, they do [374].
not develop lymphoma and the relatively low percen- It is nevertheless worth noting that no cell type or
tages of tumors with CD95 mutations identi¢ed in tumor cell line has been identi¢ed in which it is not
the aforementioned studies do not support a role for possible to eventually induce apoptosis via a certain
CD95 as a strong tumor suppressor a¡ected in a stimulus. This could be explained by a huge redun-
large number of malignancies. Knockout mice for dancy in apoptosis-signaling pathways: for example,
other TNF receptor family members are not predis- mutation of any caspase would leave other caspases
posed to tumorigenesis [233]. Perhaps the emerging capable of taking over the pro-apoptotic function
role for TNF receptor family members in prolifera- (furthermore, caspase-independent death pathways
tion prevents selection of their functional inactiva- seem to exist [85]). Alternatively it is possible that
tion during tumor development at least in some types proteins essential for induction and execution of ap-
of cancer [159]. optosis are simultaneously coupled to another vital

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F27

cell function so that their inactivation leads to a le- expressed in 5^10% of human tumors. Its major
thal phenotype disallowing the selection for such mu- physiological role is to inhibit P53 and its anti-apo-
tations [375]. ptotic and growth-arresting activities [383].
Recent evidence indicates that in order for P53 to
6.4. IAPs exert its function as a growth arrest- and apoptosis-
inducing safety guard (i.e. as a tumor suppressor) it
The IAP protein family plays an evolutionarily needs to physically interact with another tumor sup-
conserved role in regulating PCD in diverse species pressor, ING-1 [384]. This interaction renders P53 an
ranging from insects to humans [181]. The central even more e¡ective transcriptional activator (ING-1
mechanisms of IAP apoptotic suppression appear can for example increase P53-dependent activation of
to be through direct caspase and procaspase inhibi- the p21/Waf-1 promoter). New data link histone ace-
tion (primarily caspases-9, -3 and -7) and modulation tyltransferase activity to three yeast ING-1 homo-
of and by the transcription factor NF-UB. IAPs may logues, which suggests a role in chromatin remodel-
contribute to cancer by facilitating the insurgence of ing for ING-1 [385]. Therefore, loss of ING-1
mutations and by promoting resistance to therapy function is another potential mechanism for the in-
[376]. The IAP family member survivin, while unde- activation of P53 in cancer cells.
tectable in terminally di¡erentiated adult tissues, be- Since Rb loss of function-induced apoptosis is
comes prominently expressed in transformed cell P53-dependent, both genes, p53 and Rb, are fre-
lines and in all the most common human cancers quently inactivated during cellular transformation.
of lung, colon, pancreas, prostate and breast [185]. Data on knockout mice have ¢rmly established that
Survivin is also found in approximately 50% of high- germ-line mutations in p53 and Rb can cooperate in
grade non-Hodgkin's lymphomas [185,377] and high tumorigenesis [386].
survivin expression is signi¢cantly associated with
poor prognostic factors and promotes cell survival 6.6. Other anti-apoptotic mutations implicated in
in human neuroblastomas [378]. Another IAP family tumorigenesis
member, cIAP-2, is known to undergo chromosomal
translocations and is activated in certain types of Apaf-1 expression is frequently lost due to meth-
lymphomas (MALTomas [379]). The caspase-inhibit- ylation in metastatic melanoma tumors. Apaf-1 acts
ing and anti-apoptotic abilities of XIAP are blocked with cytochrome c and caspase-9 to mediate P53-de-
by its binding partner, XIAP-associated factor 1 pendent apoptosis [387,388]. Although p53 mutations
(XAF1) [380]. Cancer cell lines tested exhibited rela- often occur in aggressive and chemoresistant cancers
tively low xaf1 and high xiap mRNA levels com- they are rarely observed in melanoma. Apaf-1-nega-
pared to normal tissue suggesting that a high level tive melanomas are invariably chemoresistant despite
of XIAP to XAF1 expression in cancer cells may their functional P53 and they are unable to execute a
provide a survival advantage [381]. typical apoptotic program in response to P53 activa-
tion.
6.5. P53 Gene silencing by methylation also plays a role in
the case of TMS1, a gene which is aberrantly meth-
P53 is mutated in most human tumors and the ylated and silenced in 40% (11 of 27) of primary
resulting defects in (DNA damage-induced) growth breast tumors [389]. TMS1 encodes a 22 kDa protein
arrest and apoptosis have already been discussed. containing a terminal caspase recruitment domain
Germ-line mutation of one p53 allele in human con- and its ectopic expression induces apoptosis in hu-
fers a predisposition to develop various malignancies, man breast cancer cells, placing TMS1 in the group
the Li^Fraumeni syndrome [233]. An impressive of pro-apoptotic tumor suppressor genes.
demonstration of P53's role as a tumor suppressor Overexpression of oncoproteins such as c-Myc,
came from the p53 knockout mice which develop a c-Fos or c-Jun not only leads to proliferation but
broad spectrum of tumors with a high incidence simultaneously induces apoptosis. Consequently, ec-
[382]. Consequently, the MDM2 oncoprotein is over- topic overexpression of E2F-1 in glioma triggers ap-

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F28 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

optosis and suppresses tumor growth in vitro and in cated in metastasis for a long time and it has been
vivo [390]. The c-myc oncogene collaborates strongly demonstrated that disruption of cell surface CD44
with bmi-1 in murine lymphomagenesis. While c-Myc function induces apoptosis in metastatic mammary
provides a strong proliferative stimulus, Bmi-1 inhib- carcinoma cells in vivo [396].
its c-Myc-induced apoptosis via P19ARF [391]. This
oncogenic collaboration is reminiscent of the Myc/ 6.7. Therapeutic approaches to induce apoptosis in
Bcl-2 collaboration in tumorigenesis where Bcl-2 tumor cells
also prevents Myc-mediated apoptosis [235].
In colon cancer, one of the earliest manifestations Identi¢cation of mutations in tumors that lead to
is the formation of polyps, caused by somatic and decreased apoptosis is not only of academic interest
inherited mutations of the adenomatous polyposis but rather an important goal in the light of cancer
coli (APC) tumor suppressor gene in both humans therapy. Clearly, mutations in cell death control do
and mice. Overexpression of APC in human colorec- a¡ect sensitivity of tumor cells to anti-cancer therapy
tal cancer cells (which contain inactivated APC al- which in most cases functions by inducing apoptosis
leles) stops cell growth by inducing apoptosis, imply- [354,397]. Thus, for example, the e¡ectiveness of che-
ing a role for suppression of apoptosis at the very motherapy might depend on the level of Bcl-2 ex-
earliest stages of neoplasia [392]. pression in the tumor cells. The approach to decrease
The promyelocytic leukemia (PML) gene encodes Bcl-2 protein levels in tumor cells by applying Bcl-2
another tumor suppressor gene involved in the con- antisense oligonucleotides has already achieved
trol of apoptosis. In most acute promyelocytic leuke- promising results in preclinical [398] and clinical
mia patients, PML is fused to the retinoic acid re- studies [399,400]. A similar antisense strategy tries
ceptor K (RARK) gene as a consequence of to induce apoptosis and to sensitize lung cancer cells
chromosomal translocations. PMLRARK antago- to chemotherapy by reducing survivin expression
nizes PML function by heterodimerizing with PML [401].
resulting in its delocalization from the nuclear body. Recently, a series of novel molecules have been
Inactivation of PML in hematopoietic cells leads to identi¢ed that inhibit the binding of the Bak BH3
increased resistance towards apoptotic stimuli and peptide to Bcl-xL [402]. These chemical inhibitors
lymphomagenesis [393]. speci¢cally block the BH3 domain-mediated hetero-
Cells respond to a variety of stressful stimuli by dimerization between Bcl-2 family members and in-
accumulating and/or activating a set of highly con- duce apoptosis. Such small molecule inhibitors are
served proteins known as heat shock proteins more stable than peptide inhibitors and are often
(HSPs). These proteins normally function as molec- cell-permeable. Therefore they might represent prom-
ular chaperones by assisting the folding of newly syn- ising new tools for cancer therapy.
thesized polypeptides, the assembly of multiprotein TRAIL is a ligand for the apoptosis-inducing
complexes, and the transport of proteins across cel- death receptors DR4 and DR5. Given the remark-
lular membranes [394]. Heat shock proteins such as able sensitivity of cancer cells compared with the re-
Hsp70, Hsp27 and Hsp90 can inhibit apoptosis sistance in normal cells, recombinant TRAIL has
through direct physical interaction with key com- appealing therapeutic potential for a variety of hu-
ponents of the apoptotic machinery [395]. Hsp70 man cancers [399]. Promising results have been ob-
for example is highly expressed in human breast tu- tained in preclinical animal models involving human
mors and tumor cell lines and antisense studies have tumor xenografts into SCID or nude mice. For glio-
revealed that tumorigenic breast cancer cells depend mas, injected recombinant TRAIL caused complete
on the constitutive high expression of Hsp70 to sup- disease regression and entirely ablated tumor mass.
press a transformation-associated death program For colon carcinomas the combination of subthresh-
[394]. old doses of recombinant TRAIL with existing che-
One molecule which could be responsible for sup- motherapeutic agents resulted in a substantial posi-
pressing anoikis in metastasizing tumor cells is the tive interaction, completely eliminating the tumors in
hyaluronan receptor CD44. CD44 has been impli- some animals.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F29

References [31] J. Yuan, S. Shaham, S. Ledoux, H. Ellis, H. Horvitz, Cell 75


(1993) 641^652.
[32] B.B. Wolf, D.R. Green, J. Biol. Chem. 274 (1999) 200049^
[1] G. Hacker, D.L. Vaux, Apoptosis 2 (1997) 247^256. 200052.
[2] N.J. Holbrook, G.R. Martin, R.A. Lockshin, in: N.J. Hol- [33] G.M. Cohen, Biochem. J. 326 (1997) 1^16.
brook, G.R. Martin, R.A. Lockshin (Eds.), Cellular Aging [34] M.O. Hengartner, H.R. Horvitz, Cell 76 (1994) 665^676.
and Cell Death, Wiley-Liss, New York, 1996. [35] D.L. Vaux, I.L. Weissman, S.K. Kim, Science 258 (1992)
[3] C.B. Thompson, Science 267 (1995) 1456^1462. 1955^1957.
[4] M.K. Hellerstein, J.M. McCune, Immunity 7 (1997) 583^ [36] D. Xue, H.R. Horvitz, Nature 390 (1997) 305^308.
589. [37] H. Zou, W.J. Henzel, X. Liu, A. Lutschg, X. Wang, Cell 90
[5] H. Iwahash, Y. Eguchi, N. Yasuhara, T. Hanafusa, Y. Mat- (1997) 405^413.
suzawa, Y. Tsujimoto, Nature 390 (1997) 413^417. [38] B. Conradt, H.R. Horvitz, Cell 93 (1998) 519^529.
[6] C. Benoist, D. Mathis, Cell 89 (1997) 1^3. [39] Q.A. Liu, M.O. Hengartner, Curr. Biol. 9 (1999) 1347^1350.
[7] G. Kroemer, P. Petit, N. Zamzami, J.L. Vayssiere, B. [40] E. Smits, W. van Criekinge, G. Plaetinck, T. Bogaert, Curr.
Mignotte, FASEB J. 9 (1995) 1277^1287. Biol. 9 (1999) 1351^1354.
[8] N.J. McCarthy, Apoptosis. A Review, SCRIP Reports, PJB, [41] P. Li, H. Allen, S. Banerjee, S. Franklin, L. Herzog, C.
Richmond, 1996. Johnston, J. McDowell, M. Paskind, L. Rodman, J. Salfeld,
[9] D.A. Brown, J.K. Rose, Cell 68 (1992) 533^544. Cell 80 (1995) 401^411.
[10] F. Oberhammer, J. Wilson, C. Dive, I. Morris, J. Hickman, [42] N.A. Thornberry, H.G. Bull, J.R. Calaycay, K.T. Chapman,
A. Wakeling, P. Walker, M. Sikorska, EMBO J. 12 (1993) A.D. Howard, M.J. Kostura, D.K. Miller, S.M. Molineaux,
3679^3684. J.R. weidner, J. Aunins, Nature 356 (1992) 768^774.
[11] A.H. Wyllie, Nature 284 (1980) 555^556. [43] S. Wang, M. Miura, Y.K. Jung, H. Zhu, E. Li, J. Yuan, Cell
[12] J. Savill, V. Fadok, P. Henson, C. Haslett, Immunol. Today 92 (1998) 501^509.
14 (1993) 131^136. [44] N.A. Thornberry, Y. Lazebnik, Science 281 (1998) 1312^
[13] J. Savill, Br. Med. Bull. 53 (1997) 491^508. 1316.
[14] V.A. Fadok, J.S. savill, C. Haslett, D.L. Bratton, D.E. Doh- [45] N.A. Thornberry, T.A. Rano, E.P. Peterson, D.M. Rasper,
erty, P.A. Campbell, P.M. Henson, J. Immunol. 149 (1992) T. Timkey, M. Garcia-Calvo, V.M. Houtzager, P.A. Nord-
4029^4035. strom, S. Roy, J.P. Vaillancourt, K.T. Chapman, D.W.
[15] X.H. Liang, S. Jackson, M. Seaman, K. Brown, B. Nicholson, J. Biol. Chem. 272 (1997) 17907^17911.
Kempkes, H. Hibshoosh, B. Levine, Nature 402 (1999) [46] D.W. Nicholson, Nature Biotechnol. 14 (1996) 297^301.
672^676. [47] N.P. Walker, R.V. Talanian, K.D. Brady, L.C. Dang, N.J.
[16] C.Y. Lee, E.H. Baehrecke, Development 128 (2001) 1443^ Bump, C.R. Ferenz, S. Franklin, T. Ghayur, M.C. Hackett,
1455. L.D. Hamill, Cell 78 (1994) 343^352.
[17] D.L. Vaux, G. Haecker, A. Strasser, Cell 76 (1994) 777^779. [48] K.P. Wilson, J.A. Black, J.A. Thomson, E.E. Kim, J.P. Grif-
[18] J.C. Ameisen, Science 272 (1996) 1278^1279. ¢th, M.A. Navia, M.A. Murcko, S.P. Chambers, R.A. Al-
[19] M.B. Yarmolinsky, Science 267 (1995) 836^837. dape, S.A. Raybuck, P.J. Livingston, Nature 370 (1994) 270^
[20] M.M. Metzstein, G.M. Stan¢eld, H.R. Horvitz, Trends Gen- 275.
et. 14 (1998) 410^416. [49] J. Rodriguez, Y. Lazebnik, Genes Dev. 13 (1999) 3179^3184.
[21] H.M. Ellis, H.R. Horvitz, Cell 44 (1986) 817^829. [50] H.R. Stennicke, Q.L. Deveraux, E.W. Humke, J.C. Reed,
[22] M.O. Hengartner, R.E. Ellis, H.R. Horvitz, Nature 356 V.M. Dixit, G.S. Salvesen, J. Biol. Chem. 274 (1999) 8359^
(1992) 494^499. 8362.
[23] D.L. Vaux, Cell 90 (1997) 389^390. [51] A. Fraser, G. Evan, Cell 85 (1997) 781^784.
[24] E.S. Alnemri, D.J. Livingston, D.W. Nicholson, G. Salvesen, [52] K. Hofmann, P. Bucher, J. Tschopp, Trends Biochem. Sci.
N.A. Thornberry, W.W. Wong, J. Yuan, Cell 87 (1996) 171. 22 (1997) 155^156.
[25] M. Irmler, K. Hofmann, D. Vaux, J. Tschopp, FEBS Lett. [53] S. Shaham, H.R. Horvitz, Genes Dev. 10 (1996) 578^591.
406 (1997) 189^190. [54] M. Los, S. Wesselborg, K. Schulze-Ostho¡, Immunity 10
[26] A.M. Chinnaiyan, K. O'Rourke, B.R. Lane, V.M. Dixit, (1999) 629^639.
Science 275 (1997) 1122^1126. [55] K. Kuida, J.A. Lippke, G. Ku, M.W. Harding, D.J. Living-
[27] C. James, S. Gschmeissner, A. Fraser, G.I. Evan, Curr. Biol. ston, M.S.-S. Su, R.A. Flavell, Science 267 (1995) 2000^
7 (1997) 246^252. 2003.
[28] M.S. Spector, S. Desnoyers, D.J. Hoeppner, M. Hengartner, [56] K. Kuida, T.S. Zheng, S.Q. Na, C.Y. Kuan, D. Yang, H.
Nature 385 (1997) 653^656. Karasuyama, P. Rakic, R.A. Flavell, Nature 384 (1996) 368^
[29] D. Wu, H.D. Wallen, G. Nunez, Science 275 (1997) 1126^ 372.
1129. [57] E.E. Varfolomeev, M. Schuchmann, V. Luria, N. Chiannil-
[30] A.M. Chinnaiyan, D. Chaudhary, K. O'Rourke, E.V. Koo- kulchai, J.S. Beckmann, I.L. Mett, D. Rebrikov, V.M. Bro-
nin, V.M. Dixit, Nature 388 (1997) 728^729. dianski, O.C. Kemper, O. Kollet, T. Lapidot, D. So¡er, T.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F30 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

Sobe, K.B. Avraham, T. Goncharov, H. Holtmann, P. Lo- [79] J. Wang, L. Zheng, A. Lobito, F.K. Chan, J. Dale, M.
nai, D. Wallach, Immunity 9 (1998) 267^276. Sneller, X. Yao, J.M. Puck, S.E. Straus, M.J. Lenardo,
[58] R. Hakem, A. Hakem, G.S. Duncan, J.T. Henderson, M. Cell 98 (1999) 47^58.
Woo, M.S. Soengas, A. Elia, J.L. de la Pompa, D. Kagi, [80] M. Resnico¡, B. Valentinis, D. Herbert, D. Abraham, P.D.
W. Khoo, J. Potter, R. Yoshida, S.A. Kaufman, S.W. Friesen, E.S. Alnemri, R. Baserga, J. Biol. Chem. 273
Lowe, J.M. Penninger, T.W. Mak, Cell 94 (1998) 339^352. (1998) 10376^10380.
[59] K. Kuida, T.F. Haydar, C.Y. Kuan, Y. Gu, C. Taya, H. [81] N.J. McCarthy, M.K.B. Whyte, C. Gilbert, G. Evan,
Karasuyama, M.S. Su, P. Rakic, R.A. Flavell, Cell 94 (1998) J. Cell Biol. 136 (1997) 1^13.
325^337. [82] J. Xiang, D.T. Chao, S.J. Korsmeyer, Proc. Natl. Acad.
[60] L. Bergeron, G.I. Perez, G. Macdonald, L. Shi, Y. Sun, A. Sci. USA 93 (1996) 14559^14563.
Jurisicova, S. Varmuza, K.E. Latham, J.A. Flaws, J.C. Salt- [83] M. Chautan, G. Chazal, F. Cecconi, P. Gruss, P. Golstein,
er, H. Hara, M.A. Moskowitz, E. Li, A. Greenberg, J.L. Curr. Biol. 9 (1999) 967^970.
Tilly, J. Yuan, Genes Dev. 12 (1998) 1304^1314. [84] C. Borner, L. Monney, Cell Death Di¡er. 6 (1999) 497^507.
[61] T. Nakagawa, H. Zhu, N. Morishima, E. Li, J. Xu, B.A. [85] N. Holler, R. Zaru, O. Micheau, M. Thome, A. Attinger, S.
Yankner, J. Yuan, Nature 403 (2000) 98^103. Valitutti, J.L. Bodmer, P. Schneider, B. Seed, J. Tschopp,
[62] V. Cryns, J. Yuan, Genes Dev. 12 (1998) 1551^1570. Nature Immunol. 1 (2000) 489^495.
[63] D.W. Nicholson, Cell Death Di¡er. 6 (1999) 1028^1042. [86] J.C. Reed, Cell 91 (1997) 559^562.
[64] E.H. Cheng, D.G. Kirsch, R.J. Clem, R. Ravi, M.B. Kastan, [87] M.G. Vander Heiden, C.B. Thompson, Nature Cell Biol. 1
A. Bedi, K. Ueno, J.M. Hardwick, Science 278 (1997) 1966^ (1999) E209^E216.
1968. [88] J.C. Martinou, S. Desagher, B. Antonsson, Nature Cell
[65] R.J. Clem, E.H. Cheng, C.L. Karp, D.G. Kirsch, K. Ueno, Biol. 2 (2000) E41^E43.
A. Takahashi, M.B. Kastan, D.E. Gri¤n, W.C. Earnshaw, [89] G. Kroemer, J.C. Reed, Nature Med. 6 (2000) 513^519.
M.A. Veliuona, J.M. Hardwick, Proc. Natl. Acad. Sci. USA [90] E.A. Slee, M.T. Harte, R.M. Kluck, B.B. Wolf, C.A. Ca-
95 (1998) 554^559. siano, D.D. Newmeyer, H.G. Wang, J.C. Reed, D.W.
[66] H. Li, H. Zhu, C.J. Xu, J. Yuan, Cell 94 (1998) 491^501. Nicholson, E.S. Alnemri, D.R. Green, S.J. Martin,
[67] X. Luo, I. Budihardjo, H. Zou, C. Slaughter, X. Wang, Cell J. Cell. Biol. 144 (1999) 281^292.
94 (1998) 481^490. [91] P. Li, D. Nijhawan, I. Budihardjo, S.M. Srinivasula, M.
[68] H. Sakahira, M. Enari, S. Nagata, Nature 391 (1998) 96^99. Ahmad, E.S. Alnemri, X. Wang, Cell 91 (1997) 479^489.
[69] X. Liu, H. Zou, C. Slaughter, X. Wang, Cell 89 (1997) 175^ [92] S.M. Srinivasula, M. Ahmad, T. Fernandes-Alnemri, E.S.
184. Alnemri, Mol. Cell 1 (1998) 949^957.
[70] M. Enari, H. Sakahira, H. Yokoyama, K. Okawa, A. Iwa- [93] G. Pan, K. O'Rourke, V.M. Dixit, J. Biol. Chem. 273
matsu, S. Nagata, Nature 391 (1998) 43^50. (1998) 5841^5845.
[71] S. Kothakota, T. Azuma, C. Reinhard, A. Klippel, J. Tang, [94] Y. Hu, M.A. Benedict, D. Wu, N. Inohara, G. Nunez,
K. Chu, T.J. McGarry, M.W. Kirschner, K. Koths, D.J. Proc. Natl. Acad. Sci. USA 95 (1998) 4386^4391.
Kwiatkowski, L.T. Williams, Science 278 (1997) 294^298. [95] K. Moriishi, D.C. Huang, S. Cory, J.M. Adams, Proc.
[72] A. Takahashi, E.S. Alnemri, Y.A. Lazebnik, T. Fernandes- Natl. Acad. Sci. USA 96 (1999) 9683^9688.
Alnemri, G. Litwack, R.D. Moir, R.D. Goldman, G.G. Poi- [96] B.N. Chau, E.H. Cheng, D.A. Kerr, J.M. Hardwick, Mol.
rier, S.H. Kaufmann, W.C. Earnshaw, Proc. Natl. Acad. Sci. Cell 6 (2000) 31^40.
USA 93 (1996) 8395^8400. [97] H. Yoshida, Y.Y. Kong, R. Yoshida, A.J. Elia, A. Hakem,
[73] C. Brancolini, D. Lazarevic, J. Rodriguez, C. Schneider, R. Hakem, J.M. Penninger, T.W. Mak, Cell 94 (1998) 739^
J. Cell Biol. 139 (1997) 759^771. 750.
[74] L.P. Wen, J.A. Fahrni, S. Troie, J.L. Guan, K. Orth, G.D. [98] F. Cecconi, G. Alvarez-Bolado, B.I. Meyer, K.A. Roth, P.
Rosen, J. Biol. Chem. 272 (1997) 26056^26061. Gruss, Cell 94 (1998) 727^737.
[75] L. Casciola-Rosen, D.W. Nicholson, T. Chong, K.R. Row- [99] J.C. Goldstein, N.J. Waterhouse, P. Juin, G.I. Evan, D.R.
an, N.A. Thornberry, D.K. Miller, A. Rosen, J. Exp. Med. Green, Nature Cell Biol. 2 (2000) 156^162.
183 (1996) 1957^1964. [100] I. Marzo, C. Brenner, N. Zamzami, J.M. Jurgensmeier,
[76] Z.Q. Wang, B. Auer, L. Stinl, H. Berghammer, D. Haida- S.A. Susin, H.L. Vieira, M.C. Prevost, Z. Xie, S. Matsu-
cher, M. Schweiger, E.F. Wagner, Genes Dev. 9 (1995) 509^ yama, J.C. Reed, G. Kroemer, Science 281 (1998) 2027^
520. 2031.
[77] F.G. Gervais, D. Xu, G.S. Robertson, J.P. Vaillancourt, Y. [101] S. Shimizu, M. Narita, Y. Tsujimoto, Nature 399 (1999)
Zhu, J. Huang, A. LeBlanc, D. Smith, M. Rigby, M.S. 483^487.
Shearman, E.E. Clarke, H. Zheng, L.H. Van Der Ploeg, [102] R. Eskes, S. Desagher, B. Antonsson, J.C. Martinou, Mol.
S.C. Ru¡olo, N.A. Thornberry, S. Xanthoudakis, R.J. Zam- Cell. Biol. 20 (2000) 929^935.
boni, S. Roy, D.W. Nicholson, Cell 97 (1999) 395^406. [103] M.C. Wei, T. Lindsten, V.K. Mootha, S. Weiler, A. Gross,
[78] I. Sanchez, C.J. Xu, P. Juo, A. Kakizaka, J. Blenis, J. Yuan, M. Ashiya, C.B. Thompson, S.J. Korsmeyer, Genes Dev.
Neuron 22 (1999) 623^633. 14 (2000) 2060^2071.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F31

[104] W.X. Zong, T. Lindsten, A.J. Ross, G.R. MacGregor, C.B. [127] M. Boldin, T. Goncharov, Y. Golstev, D. Wallach, Cell 85
Thompson, Genes Dev. 15 (2001) 1481^1486. (1996) 803^815.
[105] A.M. Verhagen, P.G. Ekert, M. Pakusch, J. Silke, L.M. [128] M. Muzio, A. Chinnaiyan, F.C. Kischel, K. O'Rourke, A.
Connolly, G.E. Reid, R.L. Moritz, R.J. Simpson, D.L. Shevchenko, J. Ni, C. Sca¤di, J.D. Bretz, M. Zhang, R.
Vaux, Cell 102 (2000) 43^53. Gentz, M. Mann, P. Krammer, M. Peter, V. Dixit, Cell 85
[106] C. Du, M. Fang, Y. Li, L. Li, X. Wang, Cell 102 (2000) 33^ (1996) 817^827.
42. [129] F.C. Kischkel, S. Hellbardt, I. Behrmann, M. Germer, M.
[107] D.R. Green, Cell 102 (2000) 1^4. Pawlita, P.H. Krammer, M.E. Peter, EMBO J. 14 (1995)
[108] J. Silke, A.M. Verhagen, P.G. Ekert, D.L. Vaux, Cell 5579^5588.
Death Di¡er. 7 (2000) 1275. [130] M. Enari, R.V. Talanian, W.W. Wong, S. Nagata, Nature
[109] S.A. Susin, H.K. Lorenzo, N. Zamzami, I. Marzo, B.E. 380 (1996) 723^726.
Snow, G.M. Brothers, J. Mangion, E. Jacotot, P. Costan- [131] C. Sca¤di, S. Fulda, A. Srinivasan, C. Friesen, F. Li, K.J.
tini, M. Loe¥er, N. Larochette, D.R. Goodlett, R. Aeber- Tomaselli, K.M. Debatin, P.H. Krammer, M.E. Peter,
sold, D.P. Siderovski, J.M. Penninger, G. Kroemer, Nature EMBO J. 17 (1998) 1675^1687.
397 (1999) 441^446. [132] C. Sca¤di, I. Schmitz, J. Zha, S.J. Korsmeyer, P.H.
[110] N. Joza, S.A. Susin, E. Daugas, W.L. Stanford, S.K. Cho, Krammer, M.E. Peter, J. Biol. Chem. 274 (1999) 22532^
C.Y. Li, T. Sasaki, A.J. Elia, H.Y. Cheng, L. Ravagnan, 22538.
K.F. Ferri, N. Zamzami, A. Wakeham, R. Hakem, H. [133] B.Z. Stanger, P. Leder, T.H. Lee, E. Kim, B. Seed, Cell 81
Yoshida, Y.Y. Kong, T.W. Mak, J.C. Zuniga-P£ucker, (1995) 513^523.
G. Kroemer, J.M. Penninger, Nature 410 (2001) 549^ [134] C.M. Eischen, C.J. Dick, P.J. Leibson, J. Immunol. 153
554. (1994) 1947^1954.
[111] G. Nacentini, L. Giunchi, S. Ronchetti, L.T. Krausz, A. [135] M. Ahmad, S.M. Srinivasula, L. Wang, R.V. Talanian, G.
Bartoli, R. Moraca, G. Migliorati, C. Riccardi, Proc. Litwack, T. Fernandes-Alnemri, E.S. Alnemri, Cancer Res.
Natl. Acad. Sci. USA 94 (1997) 6216^6221. 57 (1997) 615^619.
[112] G. Pan, J.H. Bauer, V. Haridas, S. Wang, D. Liu, G. Yu, [136] Y. Lin, A. Devin, Y. Rodriguez, Z.G. Liu, Genes Dev. 13
C. Vincenz, B.B. Aggarwal, J. Ni, V.M. Dixit, FEBS Lett. (1999) 2514^2526.
431 (1998) 351^356. [137] X. Yang, R. Khosravi-Far, H. Chang, D. Baltimore, Cell
[113] P. Golstein, Curr. Biol. 7 (1997) R750^R753. 89 (1997) 1067^1076.
[114] S.J. Baker, E.P. Reddy, Oncogene 12 (1996) 1^9. [138] H. Li, C. Leo, J. Zhu, X. Wu, J. O'Neil, E.J. Park, J.D.
[115] D. Wallach, Trends Biochem. Sci. 1997 (1997) 107^109. Chen, Mol. Cell. Biol. 20 (2000) 1784^1796.
[116] C.F. Ware, S. VanArsdale, T.L. VanArsdale, J. Cell Bio- [139] J.S. Michaelson, D. Bader, F. Kuo, C. Kozak, P. Leder,
chem. 60 (1996) 47^55. Genes Dev. 13 (1999) 1918^1923.
[117] D.P.M. Hughes, I.N. Crispe, J. Exp. Med. 182 (1995) 1395^ [140] V. Rochat-Steiner, K. Becker, O. Micheau, P. Schneider, K.
1401. Burns, J. Tschopp, J. Exp. Med. 192 (2000) 1165^1174.
[118] F.K. Chan, H.J. Chun, L. Zheng, R.M. Siegel, K.L. Bui, [141] M.G. Cifone, R. De Maria, P. Roncaioli, M.R. Rippo, M.
M.J. Lenardo, Science 288 (2000) 2351^2354. Azuma, L.L. Lanier, A. Santoni, R. Testi, J. Exp. Med. 180
[119] K.V. Prasad, Z. Ao, Y. Yoon, M.X. Wu, M. Rizk, S. (1994) 1547^1552.
Jacquot, S.F. Schlossman, Proc. Natl. Acad. Sci. USA 94 [142] M. Thome, P. Schneider, K. Hofmann, H. Fickenscher, E.
(1997) 6346^6351. Meinl, F. Neipel, C. Mattmann, K. Burns, J.L. Bodmer, M.
[120] M. Grell, G. Zimmermann, E. Gottfried, C.M. Chen, U. Schroter, C. Sca¤di, P.H. Krammer, M.E. Peter, J.
Grunwald, D.C. Huang, Y.H. Wu Lee, H. Durkop, H. Tschopp, Nature 386 (1997) 517^521.
Engelmann, P. Scheurich, H. Wajant, A. Strasser, EMBO [143] S.M. Srinivasula, M. Ahmad, S. Ottilie, F. Bullrich, S.
J. 18 (1999) 3034^3043. Banks, Y. Wang, T. Fernandes-Alnemri, C.M. Croce, G.
[121] M. Adachi, S. Suematsu, T. Kondo, J. Ogasawara, T. Ta- Litwack, K.J. Tomaselli, R.C. Armstrong, E.S. Alnemri,
naka, N. Yoshida, S. Nagata, Nature Genet. 11 (1995) 294^ J. Biol. Chem. 272 (1997) 18542^18545.
300. [144] M. Irmler, M. Thome, M. Hahne, P. Schneider, K. Hof-
[122] D.R. Green, C.F. Ware, Proc. Natl. Acad. Sci. USA 94 man, V. Steiner, J.L. Bodmer, M. Schroter, K. Burns, C.
(1997) 5986^5990. Mattmann, D. Rimoldi, L.E. French, J. Tschopp, Nature
[123] G.H. Fischer, F.J. Rosenberg, S.E. Straus, J.K. Dale, L.A. 388 (1997) 190^195.
Middleton, A.Y. Lin, W. Strober, M.J. Lenardo, J.M. [145] Y.V. Goltsev, A.V. Kovalenko, E. Arnold, E.E. Varfolo-
Puck, Cell 81 (1995) 935^946. meev, V.M. Brodianskii, D. Wallach, J. Biol. Chem. 272
[124] R.M. Siegel, J.K. Frederiksen, D.A. Zacharias, F.K. Chan, (1997) 19641^19644.
M. Johnson, D. Lynch, R.Y. Tsien, M.J. Lenardo, Science [146] H.B. Shu, D.R. Halpin, D.V. Goeddel, Immunity 6 (1997)
288 (2000) 2354^2357. 751^763.
[125] N. Itoh, S. Nagata, J. Biol. Chem. 268 (1993) 10932^10937. [147] N. Inohara, T. Koseki, Y. Hu, S. Chen, G. Nunez, Proc.
[126] K. Hofmann, J. Tschopp, FEBS Lett. 371 (1995) 321^323. Natl. Acad. Sci. USA 94 (1997) 10717^10722.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F32 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

[148] S. Hu, C. Vincenz, J. Ni, R. Gentz, V.M. Dixit, J. Biol. Schreiber, D.V. Goeddel, M.W. Moore, Nature 372
Chem. 272 (1997) 17255^17257. (1994) 560^563.
[149] D.K. Han, P.M. Chaudhary, M.E. Wright, C. Friedman, [169] K. Pfe¡er, T. Matsuyama, T.M. Ku«ndig, A. Wakeham, K.
B.J. Trask, R.T. Riedel, D.G. Baskin, S.M. Schwartz, L. Kishihara, A. Shahinian, K. Wiegmann, P.S. Ohashi, M.
Hood, Proc. Natl. Acad. Sci. USA 94 (1997) 11333^ Kro«nke, T.W. Mak, Cell 73 (1993) 457^467.
11338. [170] J. Rothe, W. Lesslauer, H. Lo«tscher, Y. Lang, P. Koebel,
[150] D.M. Rasper, J.P. Vaillancourt, S. Hadano, V.M. Houtz- F. Ko«ntgen, A. Althage, R. Zinkernagel, M. Steinmetz, H.
ager, I. Seiden, S.L. Keen, P. Tawa, S. Xanthoudakis, J. Bluethmann, Nature 364 (1993) 798^802.
Nasir, D. Martindale, B.F. Koop, E.P. Peterson, N.A. [171] M. Rothe, S.C. Wong, W.J. Henzel, D.V. Goeddel, Cell 78
Thornberry, J. Huang, D.P. MacPherson, S.C. Black, F. (1994) 681^692.
Hornung, M.J. Lenardo, M.R. Hayden, S. Roy, D.W. [172] J. Inoue, T. Ishida, N. Tsukamoto, N. Kobayashi, A. Nai-
Nicholson, Cell Death Di¡er. 5 (1998) 271^288. to, S. Azuma, T. Yamamoto, Exp. Cell Res. 254 (2000) 14^
[151] C. Sca¤di, I. Schmitz, P.H. Krammer, M.E. Peter, J. Biol. 24.
Chem. 274 (1999) 1541^1548. [173] S.Y. Lee, Y. Choi, J. Exp. Med. 185 (1997) 1275^1285.
[152] L. Van Parijs, Y. Refaeli, A.K. Abbas, D. Baltimore, Im- [174] N.L. Malinin, M.P. Boldin, A.V. Kovalenko, D. Wallach,
munity 11 (1999) 763^770. Nature 385 (1997) 540^544.
[153] M. Djerbi, V. Screpanti, A.I. Catrina, B. Bogen, P. Biber- [175] C.Y. Wang, M.W. Mayo, R.G. Korneluk, D.V. Goeddel,
feld, A. Grandien, J. Exp. Med. 190 (1999) 1025^1032. A.S. Baldwin Jr., Science 281 (1998) 1680^1683.
[154] J.P. Medema, J. de Jong, T. van Hall, C.J. Melief, R. O¡- [176] V. Sarma, Z. Lin, L. Clark, B.M. Rust, M. Tewari, R.J.
ringa, J. Exp. Med. 190 (1999) 1033^1038. Noelle, V.M. Dixit, J. Biol. Chem. 270 (1995) 12343^12346.
[155] R.M. Pitti, S.A. Marsters, D.A. Lawrence, M. Roy, F.C. [177] M. Rothe, J. Xiong, H.B. Shu, K. Williamson, D. God-
Kischkel, P. Dowd, A. Huang, C.J. Donahue, S.W. Sher- dard, D.V. Goeddel, Proc. Natl. Acad. Sci. USA 93
wood, D.T. Baldwin, P.J. Godowski, W.I. Wood, A.L. (1996) 8241^8246.
Gurney, K.J. Hillan, R.L. Cohen, A.D. Goddard, D. Bot- [178] X. Ye, P. Mehlen, S. Rabizadeh, T. VanArsdale, H. Zhang,
stein, A. Ashkenazi, Nature 396 (1998) 699^703. H. Shin, J.J. Wang, E. Leo, J. Zapata, C.A. Hauser, J.C.
[156] C. Bai, B. Connolly, M.L. Metzker, C.A. Hilliard, X. Liu, Reed, D.E. Bredesen, J. Biol. Chem. 274 (1999) 30202^
V. Sandig, A. Soderman, S.M. Galloway, Q. Liu, C.P. 30208.
Austin, C.T. Caskey, Proc. Natl. Acad. Sci. USA 97 [179] S.Y. Lee, A. Reichlin, A. Santana, K.A. Sokol, M.C. Nus-
(2000) 1230^1235. senzweig, Y. Choi, Immunity 7 (1997) 703^713.
[157] K.G.C. Smith, A. Strasser, D.L. Vaux, EMBO J. 15 (1996) [180] W.C. Yeh, A. Shahinian, D. Speiser, J. Kraunus, F. Billia,
5167^5176. A. Wakeham, J.L. de la Pompa, D. Ferrick, B. Hum, N.
[158] K. Newton, A.W. Harris, M.L. Bath, K.G.C. Smith, A. Iscove, P. Ohashi, M. Rothe, D.V. Goeddel, T.W. Mak,
Strasser, EMBO J. 17 (1998) 706^718. Immunity 7 (1997) 715^725.
[159] M. Zornig, A.O. Hueber, G.I.E. Evan, Curr. Biol. 8 (1998) [181] R.J. Clem, C.S. Duckett, Trends Cell Biol. 7 (1997) 337^
467^470. 340.
[160] W.C. Yeh, J.L. Pompa, M.E. Mc Currach, H.B. Shu, A.J. [182] Y. Yang, S. Fang, J.P. Jensen, A.M. Weissman, J.D. Ash-
Elia, A. Shahinian, M. Ng, A. Wakemam, W. Khoo, K. well, Science 288 (2000) 874^877.
Mitchell, W.S. El-Deiry, S.W. lowe, D.V. Goeddel, T.W. [183] N. Roy, M.S. Mahadevan, M. McLean, G. Shutler, Z.
Mak, Science 279 (1998) 1954^1958. Yaraghi, R. Farahani, S. Baird, A. Besner-Johnston, C.
[161] C.M. Walsh, B.G. Wen, A.M. Chinnaiyan, K. O'Rourke, Lefebvre, X. Kang, Cell 80 (1995) 167^178.
V.M. Dixit, S.M. Hedrick, Immunity 8 (1998) 439^449. [184] M. Rothe, M.G. Pan, W.J. Henzel, T.M. Ayres, D.V.
[162] J. Zhang, D. Cado, A. Chen, N.H. Kabra, A. Winoto, Goeddel, Cell 83 (1995) 1243^1252.
Nature 392 (1998) 296^300. [185] G. Ambrosini, C. Adida, D.C. Altieri, Nature Med. 3
[163] C. Friesen, I. Herr, P.H. Krammer, K.M. Debatin, Nature (1997) 917^921.
Med. 2 (1996) 574^577. [186] S.L. Wang, C.J. Hawkins, S.J. Yoo, H.A. Muller, B.A.
[164] X.X. Wu, Y. Mizutani, Y. Kakehi, O. Yoshida, O. Ogawa, Hay, Cell 98 (1999) 453^463.
Cancer Res. 60 (2000) 2912^2918. [187] L. Goyal, K. McCall, J. Agapite, E. Hartwieg, H. Steller,
[165] S. Wesselborg, I.H. Engels, E. Rossmann, M. Los, K. EMBO J. 19 (2000) 589^597.
Schulze-Ostho¡, Blood 93 (1999) 3053^3063. [188] A.G. Fraser, C. James, G.I. Evan, M.O. Hengartner, Curr.
[166] T. Wieder, F. Essmann, A. Prokop, K. Schmelz, K. Biol. 9 (1999) 292^301.
Schulze-Ostho¡, R. Beyaert, B. Dorken, P.T. Daniel, Blood [189] A.G. Uren, T. Beilharz, M.J. O'Connell, S.J. Bugg, R. van
97 (2001) 1378^1387. Driel, D.L. Vaux, T. Lithgow, Proc. Natl. Acad. Sci. USA
[167] L.A. Tartaglia, D. Pennica, D.V. Goeddel, J. Biol. Chem. 96 (1999) 10170^10175.
268 (1993) 18542^18548. [190] F. Li, G. Ambrosini, E.Y. Chu, J. Plescia, S. Tognin, P.C.
[168] S.L. Erickson, F.J. de Sauvage, K. Kikly, K. Carver- Marchisio, D.C. Altieri, Nature 396 (1998) 580^584.
Moore, S. Pitts-Meek, N. Gillett, K.C.F. Sheehan, R.D. [191] F. Li, E.J. Ackermann, C.F. Bennett, A.L. Rothermel, J.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F33

Plescia, S. Tognin, A. Villa, P.C. Marchisio, D.C. Altieri, [216] L. Yamasaki, R. Bronson, B.O. Williams, N.J. Dyson, E.
Nature Cell Biol. 1 (1999) 461^466. Harlow, T. Jacks, Nature Genet. 18 (1998) 360^364.
[192] F.X. Pimentel-Muinos, B. Seed, Immunity 11 (1999) 783^ [217] B. Amati, T.D. Littlewood, G.I. Evan, H. Land, EMBO J.
793. 12 (1993) 5083^5087.
[193] T. Gura, Science 277 (1997) 768. [218] S. Gehring, S. Rottmann, A.R. Menkel, J. Mertsching, A.
[194] A. Ashkenazi, V.M. Dixit, Science 281 (1998) 1305^1308. Krippner-Heidenreich, B. Luscher, J. Biol. Chem. 275
[195] R.M. Pitti, S.A. Marsters, S. Ruppert, C.J. Donahue, A. (2000) 10413^10420.
Moore, A. Ashkenazi, J. Biol. Chem. 271 (1996) 12687^ [219] C. Grandori, R.N. Eisenman, Trends Biochem. Sci. 1997
12690. (1997) 177^181.
[196] J.L. Bodmer, N. Holler, S. Reynard, P. Vinciguerra, P. [220] M. Zornig, G. Evan, Curr. Biol. 6 (1996) 1553^1556.
Schneider, P. Juo, J. Blenis, J. Tschopp, Nature Cell Biol. [221] H.A. Coller, C. Grandori, P. Tamayo, T. Colbert, E.S.
2 (2000) 241^243. Lander, R.N. Eisenman, T.R. Golub, Proc. Natl. Acad.
[197] A. Truneh, S. Sharma, C. Silverman, S. Khandekar, M.P. Sci. USA 97 (2000) 3260^3265.
Redy, K.C. Deen, M.M. McLaughlin, S.M. Srinivasula, [222] K.O. Mitchell, M.S. Ricci, T. Miyashita, D.T. Dicker, Z.
G.P. Livi, L.A. Marshall, E.S. Alnemri, W.V. Williams, Jin, J.C. Reed, W.S. El-Deiry, Cancer Res. 60 (2000) 6318^
M.L. Doyle, J. Biol. Chem. 197 (2000) 23319^23325. 6325.
[198] T.S. Gri¤th, S.R. Wiley, M.Z. Kubin, L.M. Sedger, C.R. [223] F. Zindy, C.M. Eischen, D.H. Randle, T. Kamijo, J.L.
Maliszewski, N.A. Fanger, J. Exp. Med. 189 (1999) 1343^ Cleveland, C.J. Sherr, M.F. Roussel, Genes Dev. 12
1354. (1998) 2424^2433.
[199] H. Walczak, R.E. Miller, K. Ariail, B. Gliniak, T.S. Grif- [224] E. de Stanchina, M.E. McCurrach, F. Zindy, S.Y. Shieh,
¢th, M. Kubin, W. Chin, J. Jones, A. Woodward, T. Le, C. G. Ferbeyre, A.V. Samuelson, C. Prives, M.F. Roussel, C.J.
Smith, P. Smolak, R.G. Goodwin, C.T. Rauch, J.C. Schuh, Sherr, S.W. Lowe, Genes Dev. 12 (1998) 2434^2442.
D.H. Lynch, Nature Med. 5 (1999) 157^163. [225] T. Stiewe, B.M. Putzer, Nature Genet. 26 (2000) 464^469.
[200] A.M. Chinnaiyan, U. Prasad, S. Shankar, D.A. Hamstra, [226] M. Irwin, M.C. Marin, A.C. Phillips, R.S. Seelan, D.I.
M. Shanaiah, T.L. Chenevert, B.D. Ross, A. Rehemtulla, Smith, W. Liu, E.R. Flores, K.Y. Tsai, T. Jacks, K.H.
Proc. Natl. Acad. Sci. USA 97 (2000) 1754^1759. Vousden, W.G. Kaelin Jr., Nature 407 (2000) 645^648.
[201] S.B. Gibson, R. Oyer, A.C. Spalding, S.M. Anderson, G.L. [227] U. Galderisi, G. Di Bernardo, M. Cipollaro, F.P. Jori, E.
Johnson, Mol. Cell. Biol. 20 (2000) 205^212. Piegari, A. Cascino, G. Peluso, M.A. Melone, J. Cell Bio-
[202] M. Jo, T.H. Kim, D.W. Seol, J.E. Esplen, K. Dorko, T.R. chem. 74 (1999) 313^322.
Billiar, S.C. Strom, Nature Med. 6 (2000) 564^567. [228] A.O. Hueber, M. Zornig, D. Lyon, T. Suda, S. Nagata,
[203] S. Nagata, Nature Med. 6 (2000) 502^503. G.I. Evan, Science 278 (1997) 1305^1308.
[204] E.A. Harrington, A. Fanidi, G. Evan, Curr. Opin. Genet. [229] P. Juin, A.O. Hueber, T. Littlewood, G. Evan, Genes Dev.
Dev. 4 (1994) 120^129. 13 (1999) 1367^1381.
[205] G.I. Evan, A.H. Wyllie, C.S. Gilbert, T.D. Littlewood, H. [230] J. Klefstrom, P.E. Kovanen, K. Somersalo, A.O. Hueber,
Land, M. Brooks, C.M. Waters, L.Z. Penn, D.C. Hancock, T. Littlewood, G.I. Evan, A.H. Greenberg, E. Saksela, T.
Cell 69 (1992) 119^128. Timonen, K. Alitalo, Oncogene 18 (1999) 2181^2188.
[206] E. White, Proc. Soc. Exp. Biol. Med. 204 (1993) 30^39. [231] S. Pelengaris, T. Littlewood, M. Khan, G. Elia, G. Evan,
[207] E. Bossy-Wetzel, L. Bakiri, M. Yaniv, EMBO J. 16 (1997) Mol. Cell 3 (1999) 565^577.
1695^1709. [232] S. Pelengaris, B. Rudolph, T. Littlewood, Curr. Opin. Gen-
[208] G.A. Preston, T.T. Lyon, Y. Yin, J.E. Lang, G. Solomon, et. Dev. 10 (2000) 100^105.
L. Annab, D.G. Srinivasan, D.A. Alcorta, J.C. Barrett, [233] A. Strasser, D.C. Huang, D.L. Vaux, Biochim. Biophys.
Mol. Cell. Biol. 16 (1996) 211^218. Acta 1333 (1997) F151^F178.
[209] E.B. Thompson, Annu. Rev. Physiol. 60 (1998) 575^600. [234] D.L. Vaux, S. Cory, J.M. Adams, Nature 335 (1988) 440^
[210] R. Wisdom, R.S. Johnson, C. Moore, EMBO J. 18 (1999) 442.
188^197. [235] A. Strasser, A.W. Harris, M.L. Bath, S. Cory, Nature 348
[211] E.A. Harrington, M.R. Bennett, A. Fanidi, G.I. Evan, (1990) 331^333.
EMBO J. 13 (1994) 3286^3295. [236] A. Fanidi, E.A. Harrington, G.I. Evan, Nature 359 (1992)
[212] Y. Ishizaki, L. Cheng, A.W. Mudge, M.C. Ra¡, Mol. Biol. 554^556.
Cell 6 (1995) 1443^1458. [237] R.P. Bissonnette, F. Echeverri, A. Mahboubi, D.R. Green,
[213] A.O. Hueber, G.I. Evan, Trends Genet. 14 (1998) 364^367. Nature 359 (1992) 552^554.
[214] L. Yamasaki, T. Jacks, R. Bronson, E. Goillot, E. Harlow, [238] A.J. Wagner, M.B. Small, N. Hay, Mol. Cell. Biol. 13
N.J. Dyson, Cell 85 (1996) 537^548. (1993) 2432^2440.
[215] S.J. Field, F.Y. Tsai, F. Kuo, A.M. Zubiaga, W.G. Kaelin [239] D.J. Veis, C.M. Sorenson, J.R. Shutter, S.J. Korsmeyer,
Jr., D.M. Livingston, S.H. Orkin, M.E. Greenberg, Cell 85 Cell 75 (1993) 229^240.
(1996) 549^561. [240] J.M. Adams, S. Cory, Science 281 (1998) 1322^1326.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F34 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

[241] T. Moroy, M. Zornig, J. Cell Physiol. Biochem. 6 (1997) [265] T.L. Deckwerth, J.L. Elliott, C.M. Knudson, E.M. Jonh-
312^336. son, W.D. Snider, S.J. Korsmeyer, Neuron 17 (1996) 401^
[242] A. Kelekar, C.B. Thompson, Trends Cell Biol. 8 (1998) 411.
324^330. [266] P. Bouillet, D. Metcalf, D.C. Huang, D.M. Tarlinton, T.W.
[243] L.H. Boise, M. Gonzalez-Garcia, C.E. Postema, L. Ding, Kay, F. Kontgen, J.M. Adams, A. Strasser, Science 286
T. Lindsten, L.A. Turka, X. Mao, G. Nunez, C.B. Thomp- (1999) 1735^1738.
son, Cell 74 (1993) 597^608. [267] E. Yang, J. Zha, J. Jockel, L.H. Boise, C.B. Thompson, S.J.
[244] H.H. Lee, H. Dadgostar, Q. Cheng, J. Shu, G. Cheng, Korsmeyer, Cell 80 (1995) 285^291.
Proc. Natl. Acad. Sci. USA 96 (1999) 9136^9141. [268] J.C. Reed, Oncogene 17 (1998) 3225^3236.
[245] M. Silva, A. Benito, C. Sanz, F. Prosper, D. Ekhterae, G. [269] T.F. Franke, L.C. Cantley, Nature 390 (1997) 116^117.
Nunez, J.L. Fernandez-Luna, J. Biol. Chem. 274 (1999) [270] K. Virdee, P.A. Parone, A.M. Tolkovsky, Curr. Biol. 10
22165^22169. (2000) 1151^1154.
[246] D.T. Chao, G.P. linette, L.H. Boise, L.S. White, C.B. [271] S.R. Datta, A. Katsov, L. Hu, A. Petros, S.W. Fesik, M.B.
Thompson, S.J. Korsmeyer, J. Exp. Med. 182 (1995) 821^ Ya¡e, M.E. Greenberg, Mol. Cell 6 (2000) 41^51.
828. [272] J.M. Lizcano, N. Morrice, P. Cohen, Biochem. J. 349
[247] D.A. Grillot, R. Merino, G. Nunez, J. Exp. Med. 182 (2000) 547^557.
(1995) 1973^1983. [273] X.M. Zhou, Y. Liu, G. Payne, R.J. Lutz, T. Chittenden,
[248] N. Motoyama, F. Wang, K.A. Roth, H. Sawa, K. Nakaya- J. Biol. Chem. 275 (2000) 25046^25051.
ma, I. Negishi, S. Senju, Q. Zhang, S. Fujii, D. Loh, Sci- [274] S.R. Datta, H. Dudek, X. Tao, S. Masters, H. Fu, Y.
ence 267 (1995) 1506^1510. Gotoh, M.E. Greenberg, Cell 91 (1997) 231^241.
[249] M. Sattler, H. Liang, D. Nettesheim, R.P. Meadows, J.E. [275] L. del Peso, M. Gonzalez-Garcia, C. Page, R. Herrera, G.
Harlan, M. Eberstadt, H.S. Yoon, S.B. Shuker, B.S. Nunez, Science 278 (1997) 687^689.
Chang, A.J. Minn, C.B. Thompson, S.W. Fesik, Science [276] H.G. Wang, U.R. Rapp, J.C. Reed, Cell 87 (1996) 629^
275 (1997) 983^986. 638.
[250] G. Kroemer, Nature Med. 3 (1997) 614^620. [277] J. Zha, H. Harada, E. Yang, J. Jockel, S.J. Korsmeyer, Cell
[251] Z.N. Oltvai, C.L. Milliman, S.J. Korsmeyer, Cell 74 (1993) 87 (1996) 619^628.
609^619. [278] K. Wang, X.M. Yin, D.T. Chao, C.L. Milliman, S.J. Kors-
[252] E.H. Cheng, B. Levine, L.H. Boise, C.B. Thompson, J.M. meyer, Genes Dev. 10 (1996) 2859^2869.
Hardwick, Nature 379 (1998) 554^556. [279] A. Gross, X.M. Yin, K. Wang, M.C. Wei, J. Jockel, C.
[253] E.G. St Clair, S.J. Anderson, Z.N. Oltvai, J. Biol. Chem. Milliman, H. Erdjument-Bromage, P. Tempst, S.J. Kors-
272 (1997) 29347^29355. meyer, J. Biol. Chem. 274 (1999) 1156^1163.
[254] H. Zha, J.C. Reed, J. Biol. Chem. 272 (1997) 31482^31488. [280] S. Desagher, A. Osen-Sand, A. Nichols, R. Eskes, S. Mon-
[255] P.L. Simonian, D.A. Grillot, G. Nunez, Oncogene 15 tessuit, S. Lauper, K. Maundrell, B. Antonsson, J.C. Mar-
(1997) 1871^1875. tinou, J. Cell Biol. 144 (1999) 891^901.
[256] H.J. Brady, G.S. Salomons, R.C. Bobeldijk, A.J. Berns, [281] E. Oda, R. Ohki, H. Murasawa, J. Nemoto, T. Shibue, T.
EMBO J. 15 (1996) 1221^1230. Yamashita, T. Tokino, T. Taniguchi, N. Tanaka, Science
[257] J.M. Jurgensmeier, Z. Xie, Q. Deveraux, L. Ellerby, D. 288 (2000) 1053^1058.
Bredesen, J.C. Reed, Proc. Natl. Acad. Sci. USA 95 [282] D. Hockenbery, Z.N. Oltvai, X.M. Yin, C.L. Milliman, S.J.
(1998) 4997^5002. Korsmeyer, Cell 75 (1993) 241^251.
[258] T. Miyashita, J.C. Reed, Cell 80 (1995) 293^299. [283] D.J. Kane, T.A. Sara¢an, R. Anton, H. Hahn, E.B. Gralla,
[259] T. Schmidt, K. Korner, H. Karsunky, S. Korsmeyer, R. J.S. Valentine, T. Ord, D.E. Bredesen, Science 262 (1993)
Muller, T. Moroy, Cell Death Di¡er. 6 (1999) 873^882. 1274^1277.
[260] H. Xiang, J. Neurosci. 18 (1998) 1363^1373. [284] M.D. Jacobson, M.C. Ra¡, Nature 374 (1995) 814^816.
[261] M.E. McCurrach, T.M.F. Connor, C.M. Knudson, S.J. [285] M.D. Jacobson, J.F. Burne, M.P. King, T. Miyashita, J.C.
Korsmeyer, S.W. lowe, Proc. Natl. Acad. Sci. USA 94 Reed, M.C. Ra¡, Nature 361 (1993) 365^369.
(1997) 2345^2349. [286] S.P. Mah, L.T. Zhong, Y. Liu, A. Roghani, R.H. Edwards,
[262] C. Yin, C.M. Knudson, S.J. Korsmeyer, T. Van Dyke, D.E. Bredesen, J. Neurochem. 60 (1993) 1183^1186.
Nature 385 (1997) 637^640. [287] L.T. Zhong, T. Sara¢an, D.J. Kane, A.C. Charles, S.P.
[263] T. Lindsten, A.J. Ross, A. King, W.X. Zong, J.C. Rath- Mah, R.H. Edwards, D.E. Bredesen, Proc. Natl. Acad.
mell, H.A. Shiels, E. Ulrich, K.G. Waymire, P. Mahar, K. Sci. USA 90 (1993) 4533^4537.
Frauwirth, Y. Chen, M. Wei, V.M. Eng, D.M. Adelman, [288] M. Hirotani, Y. Zhang, N. Fujita, M. Naito, T. Tsuruo,
M.C. Simon, A. Ma, J.A. Golden, G. Evan, S.J. Kors- J. Biol. Chem. 274 (1999) 20415^20420.
meyer, G.R. MacGregor, C.B. Thompson, Mol. Cell 6 [289] D.D. Newmeyer, E. Bossy-Wetzel, R.M. Kluck, B.B. Wolf,
(2000) 1389^1399. H.M. Beere, D.R. Green, Cell Death Di¡er. 7 (2000) 402^
[264] C.M. Knudson, K.S. Tung, W.G. Tourtellotte, G.A. 407.
Brown, S.J. Korsmeyer, Science 270 (1995) 96^99. [290] S.W. Muchmore, M. Sattler, H. Liang, R.P. Meadows, J.E.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F35

Harlan, H.S. Yoon, D. Nettesheim, B.S. Chang, C.B. [317] R. Takimoto, W.S. El-Deiry, Oncogene 19 (2000) 1735^
Thompson, S.L. Wong, s.L. Ng, S.W. Fesik, Nature 381 1743.
(1996) 335^341. [318] M. Kastan, Nature Genet. 17 (1997) 130^131.
[291] A. Gross, J.M. McDonnell, S.J. Korsmeyer, Genes Dev. 13 [319] Y. Aragane, D. Kulms, D. Metze, G. Wilkes, B. Poppel-
(1999) 1899^1911. mann, T.A. Luger, T. Schwarz, J. Cell Biol. 140 (1998) 171^
[292] J.C. Reed, Nature 387 (1997) 773^776. 182.
[293] S. Shimizu, T. Ide, T. Yanagida, Y. Tsujimoto, J. Biol. [320] A. Rehemtulla, C.A. Hamilton, A.M. Chinnaiyan, V.M.
Chem. 275 (2000) 12321^12325. Dixit, J. Biol. Chem. 272 (1997) 25783^25786.
[294] C. Brenner, H. Cadiou, H.L. Vieira, N. Zamzami, I. Mar- [321] A. Gutierrez del Arroyo, C. Gil-Lamagniere, I. Lazaro,
zo, Z. Xie, B. Leber, D. Andrews, H. Duclohier, J.C. Reed, M.C. de Marco, I. Layunta, A. Silva, Oncogene 19 (2000)
G. Kroemer, Oncogene 19 (2000) 329^336. 3647^3655.
[295] B. Antonsson, S. Montessuit, S. Lauper, R. Eskes, J.C. [322] L.B. Owen-Schaub, W. Zhang, J.C. Cusack, L.S. Angelo,
Martinou, Biochem. J. 345 (2000) 271^278. S.M. Santee, T. Fujiwara, J.A. Roth, A.B. Deisseroth,
[296] S. Shimizu, A. Konishi, T. Kodama, Y. Tsujimoto, Proc. W.W. Zhang, E. Kruzel, Mol. Cell. Biol. 15 (1995) 3032^
Natl. Acad. Sci. USA 97 (2000) 3100^3105. 3040.
[297] D. Wu, H.D. Wallen, N. Inohara, G. Nunez, J. Biol. Chem. [323] A. Elkeles, T. Juven-Gershon, D. Israeli, S. Wilder, A.
272 (1997) 21449^21454. Zalcenstein, M. Oren, Mol. Cell. Biol. 19 (1999) 2594^2600.
[298] K.G. Wolter, Y.T. Hsu, C.L. Smith, A. Nechustan, X.G. [324] J. Zhu, X. Chen, Mol. Cell. Biol. 20 (2000) 5602^5618.
XI, R.J. Youle, J. Cell Biol. 139 (1997) 1281^1292. [325] Y. Lin, W. Ma, S. Benchimol, Nature Genet. 26 (2000)
[299] R.M. Kluck, E. Bossy-Wetzel, D.R. Green, D.D. New- 122^127.
meyer, Science 275 (1997) 1132^1136. [326] K. Oda, H. Arakawa, T. Tanaka, K. Matsuda, C. Tanika-
[300] X. Liu, C.N. kim, J. Yang, R. Jemmerson, X. Wang, Cell wa, T. Mori, H. Nishimori, K. Tamai, T. Tokino, Y. Na-
86 (1996) 147^157. kamura, Y. Taya, Cell 102 (2000) 849^862.
[301] J. Yang, X. Liu, K. Bhalla, C.N. kim, A.M. Ibrado, J. Cai, [327] H. Hermeking, D. Eick, Science 265 (1994) 2091^2093.
T.I. Peng, D.P. Jones, X. Wang, Science 275 (1997) 1129^ [328] M. Trudel, J. Lanoix, L. Barisoni, M.J. Blouin, M. Des-
1132. forges, C. L'Italien, V. D'Agati, J. Exp. Med. 186 (1997)
[302] S. Mazel, D. Burtrum, H.T. Petrie, J. Exp. Med. 183 (1996) 1873^1884.
2219^2226. [329] M. Bennett, K. Macdonald, S.W. Chan, J.P. Luzio, R.
[303] G. Vairo, K.M. Innes, J.M. Adams, Oncogene 13 (1996) Simari, P. Weissberg, Science 282 (1998) 290^293.
1511^1519. [330] N.D. Marchenko, A. Zaika, U.M. Moll, J. Biol. Chem. 275
[304] L. O'Reilly, D. Huang, A. Strasser, EMBO J. 15 (1996) (2000) 16202^16212.
6979^6990. [331] N.D. Lakin, S.P. Jackson, Oncogene 18 (1999) 7644^7655.
[305] K. Fujise, D. Zhang, J.L. Liu, E.T. Yeh, J. Biol. Chem. 275 [332] B. Vogelstein, D. Lane, A.J. Levine, Nature 408 (2000)
(2000) 39458^39465. 307^310.
[306] G. Gil-Gomez, A. Berns, H.J. Brady, EMBO J. 17 (1998) [333] T. Buschmann, S.Y. Fuchs, C.G. Lee, Z.Q. Pan, Z. Ronai,
7209^7218. Cell 101 (2000) 753^762.
[307] H.J. Brady, G. Gil-Gomez, J. Kirberg, A.J. Berns, EMBO [334] Y. Zhang, Y. Xiong, W.G. Yarbrough, Cell 92 (1998) 725^
J. 15 (1996) 6991^7001. 734.
[308] D.C. Huang, L.A. O'Reilly, A. Strasser, S. Cory, EMBO J. [335] T. Kamijo, F. Zindy, M.F. Roussel, D.E. Quelle, J.R.
16 (1997) 4628^4638. Downing, R.A. Ashmun, G. Grosveld, C.J. Sherr, Cell 91
[309] G.E. Evan, L. Brown, M. Whyte, E. Harrington, Curr. (1997) 649^659.
Opin. Cell Biol. 7 (1995) 825^834. [336] S. Banin, L. Moyal, S. Shieh, Y. Taya, C.W. Anderson, L.
[310] C. Choisy-Rossi, E. Yonish-Rouach, Cell Death Di¡er. 5 Chessa, N.I. Smorodinsky, C. Prives, Y. Reiss, Y. Shiloh,
(1998) 129^131. Y. Ziv, Science 281 (1998) 1674^1677.
[311] P. May, E. May, Oncogene 18 (1999) 7621^7636. [337] S.Y. Shieh, M. Ikeda, Y. Taya, C. Prives, Cell 91 (1997)
[312] L.D. Attardi, S.W. lowe, J. Brugarolas, T. Jacks, EMBO J. 325^334.
15 (1996) 3693^3701. [338] J. Luo, F. Su, D. Chen, A. Shiloh, W. Gu, Nature 408
[313] E. Yonish-Rouach, C. Choisy, V. Deguin, C. Breugnot, E. (2000) 377^381.
May, Behring Inst. Mitt. 97 (1996) 60^71. [339] W.G. An, M. Kanekal, M.C. Simon, E. Maltepe, M.V.
[314] S. Rowan, R.L. Ludwig, Y. Haupt, S. Bates, X. Lu, M. Blagosklonny, L.M. Neckers, Nature 392 (1998) 405^408.
Oren, K.H. Vousden, EMBO J. 15 (1996) 827^838. [340] S.D. Morgenbesser, B.O. Williams, T. Jacks, R.A. De-
[315] C. Barlow, K.D. Brown, C.X. Deng, D.A. Tagle, A. Wyn- Pinho, Nature 371 (1998) 72^74.
shaw-Boris, Nature Genet. 17 (1997) 453^456. [341] M. Jaattela, Exp. Cell Res. 248 (1999) 30^43.
[316] R. Zhao, K. Gish, M. Murphy, Y. Yin, D. Notterman, [342] D. Hanahan, R.A. Weinberg, Cell 100 (2000) 57^70.
W.H. Ho¡man, E. Tom, D.H. Mack, A.J. Levine, Genes [343] P. Naik, J. Karrim, D. Hanahan, Genes Dev. 10 (1996)
Dev. 14 (2000) 981^993. 2105^2116.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


F36 M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37

[344] S. Kay, N. Donin, M. Michowitz, D. Katzenelson, J. Hiss, [369] N. Williams, Science 274 (1996) 1302.
G. Schibi, A. Pinchassov, J. Leibovici, Biochim. Biophys. [370] N. Favre-Felix, A. Fromentin, A. Hammann, E. Solary, F.
Acta 1497 (2000) 37^50. Martin, B. Bonnotte, J. Immunol. 164 (2000) 5023^5027.
[345] M.C. Marin, B. Hsu, L.C. Stephens, S. Brisbay, T.J. [371] T. Teitz, T. Wei, M.B. Valentine, E.F. Vanin, J. Grenet,
McDonnell, Exp. Cell Res. 217 (1998) 240^247. V.A. Valentine, F.G. Behm, A.T. Look, J.M. Lahti, V.J.
[346] E.M. Bruckheimer, S. Brisbay, D.J. Johnson, J.R. Ging- Kidd, Nature Med. 6 (2000) 529^535.
rich, N. Greenberg, T.J. McDonnell, Oncogene 19 (2000) [372] P. Juin, G. Evan, Nature Med. 6 (2000) 498^500.
5251^5258. [373] M.A. Grotzer, A. Eggert, T.J. Zuzak, A.J. Janss, S. Mar-
[347] P.A. Furth, U. Bar-Peled, M. Li, A. Lewis, R. Laucirica, R. waha, B.R. Wiewrodt, N. Ikegaki, G.M. Brodeur, P.C.
Jager, H. Weiher, R.G. Russell, Oncogene 18 (1999) 6589^ Phillips, Oncogene 19 (2000) 4604^4610.
6596. [374] R.N. Winter, A. Kramer, A. Borkowski, N. Kyprianou,
[348] M. Socolovsky, A.E. Fallon, S. Wang, C. Brugnara, H.F. Cancer Res. 61 (2001) 1227^1232.
Lodish, Cell 98 (1999) 181^191. [375] N.J. Kennedy, T. Kataoka, J. Tschopp, R.C. Budd, J. Exp.
[349] F. Gesbert, J.D. Gri¤n, Blood 96 (2000) 2269^2276. Med. 190 (1999) 1891^1896.
[350] R.P. de Groot, J.A. Raaijmakers, J.W. Lammers, L. Koen- [376] E.C. LaCasse, S. Baird, R.G. Korneluk, A.E. MacKenzie,
derman, Mol. Cell. Biol. Res. Commun. 3 (2000) 299^305. Oncogene 17 (1998) 3247^3259.
[351] M. Horita, E.J. Andreu, A. Benito, C. Arbona, C. Sanz, I. [377] C. Adida, C. Haioun, P. Gaulard, E. Lepage, P. Morel, J.
Benet, F. Prosper, J.L. Fernandez-Luna, J. Exp. Med. 191 Briere, H. Dombret, F. Reyes, J. Diebold, C. Gisselbrecht,
(2000) 977^984. G. Salles, D.C. Altieri, T.J. Molina, Blood 96 (2000) 1921^
[352] N. Rampino, H. Yamamoto, Y. Ionov, Y. Li, H. Sawai, 1925.
J.C. Reed, M. Perucho, Science 275 (1997) 967^969. [378] A. Islam, H. Kageyama, N. Takada, T. Kawamoto, H.
[353] M.A. Shibata, M.L. Liu, M.C. Knudson, E. Shibata, K. Takayasu, E. Isogai, M. Ohira, K. Hashizume, H. Kobaya-
Yoshidome, T. Bandey, S.J. Korsmeyer, J.E. Green, shi, Y. Kaneko, A. Nakagawara, Oncogene 19 (2000) 617^
EMBO J. 18 (1999) 2692^2701. 623.
[354] L. Zhang, J. Yu, B.H. Park, K.W. Kinzler, B. Vogelstein, [379] J. Dierlamm, M. Baens, I. Wlodarska, M. Stefanova-Ou-
Science 290 (2000) 989^992. zounova, J.M. Hernandez, D.K. Hossfeld, C. De Wolf-Pee-
[355] C.M. Knudson, G.M. Johnson, Y. Lin, S.J. Korsmeyer, ters, A. Hagemeijer, H. Van den Berghe, P. Marynen,
Cancer Res. 61 (2001) 659^665. Blood 93 (1999) 3601^3609.
[356] M. Krajewska, S.F. Moss, S. Krajewski, K. Song, P.R. [380] P. Liston, W.G. Fong, N.L. Kelly, S. Toji, T. Miyazaki, D.
Holt, J.C. Reed, Cancer Res. 56 (1996) 2422^2427. Conte, K. Tamai, C.G. Craig, M.W. McBurney, R.G. Kor-
[357] A. Bedi, P.J. Pasricha, A.J. Akhtar, J.P. Barber, G.C. Bedi, neluk, Nature Cell Biol. 3 (2001) 128^133.
F.M. Giardiello, B.A. Zehnbauer, S.R. Hamilton, R.J. [381] W.G. Fong, P. Liston, E. Rajcan-Separovic, M. St Jean, C.
Jones, Cancer Res. 55 (1995) 1811^1816. Craig, R.G. Korneluk, Genomics 70 (2000) 113^122.
[358] A. Prokop, T. Wieder, I. Sturm, F. Essmann, K. Seeger, C. [382] L.A. Donehower, Semin. Cancer Biol. 7 (1996) 269^278.
Wuchter, W.D. Ludwig, G. Henze, B. Dorken, P.T. Daniel, [383] G. Ganguli, J. Abecassis, B. Wasylyk, EMBO J. 19 (2000)
Leukemia 14 (2000) 1606^1613. 5135^5147.
[359] A. Villunger, A. Strasser, Oncol. Res. 10 (1998) 541^550. [384] M. Oren, Nature 391 (1998) 233^234.
[360] M. Zornig, A. Grzeschiczek, M.B. Kowalski, K.U. Hart- [385] R. Loewith, M. Meijer, S.P. Lees-Miller, K. Riabowol, D.
mann, T. Moroy, Oncogene 10 (1995) 2397^2401. Young, Mol. Cell. Biol. 20 (2000) 3807^3816.
[361] K. Gronbaek, P.T. Straten, E. Ralfkiaer, V. Ahrenkiel, [386] B.O. Williams, S.D. Morgenbesser, R.A. DePinho, T.
M.K. Andersen, N.E. Hansen, J. Zeuthen, K. Hou-Jensen, Jacks, Cold Spring Harbor Symp. Quant. Biol. 59 (1994)
P. Guldberg, Blood 92 (1998) 3018^3024. 449^457.
[362] W.S. Park, R.R. Oh, Y.S. Kim, J.Y. Park, S.H. Lee, M.S. [387] M.S. Soengas, P. Capodieci, D. Polsky, J. Mora, M. Estel-
Shin, S.Y. Kim, P.J. Kim, H.K. Lee, N.Y. Yoo, J.Y. Lee, ler, X. Opitz-Araya, R. McCombie, J.G. Herman, W.L.
J. Pathol. 193 (2001) 162^168. Gerald, Y.A. Lazebnik, C. Cordon-Cardo, S.W. Lowe, Na-
[363] C. Beltinger, E. Kurz, T. Bohler, M. Schrappe, W.D. Lud- ture 409 (2001) 207^211.
wig, K.M. Debatin, Blood 91 (1998) 3943^3951. [388] P.A. Jones, Nature 409 (2001) 141, 143^144.
[364] D. Traver, K. Akashi, I.L. Weissman, E. Lagasse, Immun- [389] K.E. Conway, B.B. McConnell, C.E. Bowring, C.D. Don-
ity 9 (1998) 47^57. ald, S.T. Warren, P.M. Vertino, Cancer Res. 60 (2000)
[365] L.E. French, J. Tschopp, J. Exp. Med. 190 (1999) 891^894. 6236^6242.
[366] K. Ohshima, S. Haraoka, M. Sugihara, J. Suzumiya, C. [390] J. Fueyo, C. Gomez-Manzano, W.K. Yung, T.J. Liu, R.
Kawasaki, M. Kanda, M. Kikuchi, Cancer Lett. 160 Alemany, T.J. McDonnell, X. Shi, J.S. Rao, V.A. Levin,
(2000) 89^97. A.P. Kyritsis, Nature Med. 4 (1998) 685^690.
[367] S. Obata, M. Hayashida, H. Kawano, T. Nakano, K. Shi- [391] J.J. Jacobs, B. Scheijen, J.W. Voncken, K. Kieboom, A.
raki, A. Suzuki, Nature Med. 7 (2001) 88^93. Berns, M. van Lohuizen, Genes Dev. 13 (1999) 2678^
[368] H. Takahashi, Nature Med. 7 (2001) 26^27. 2690.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart


M. Zo«rnig et al. / Biochimica et Biophysica Acta 1551 (2001) F1^F37 F37

[392] P.J. Morin, B. Vogelstein, K.W. Kinzler, Proc. Natl. Acad. [398] B. Jansen, H. Schlagbauer-Wadl, B.D. Brown, R.N. Bryan,
Sci. USA 93 (1996) 7950^7954. A. van Elsas, M. Muller, K. Wol¡, H.G. Eichler, H.
[393] E. Rego, Z. Wang, D. Peruzzi, L. He, C. Cordon-Cardo, P. Pehamberger, Nature Med. 4 (1998) 232^234.
Pandol¢, J. Exp. Med. 193 (2001) 521^530. [399] D.W. Nicholson, Nature 407 (2000) 810^816.
[394] J. Nylandsted, M. Rohde, K. Brand, L. Bastholm, F. El- [400] C.J. Reed, Semin. Hematol. 37 (2000) 9^16.
ling, M. Jaattela, Proc. Natl. Acad. Sci. USA 97 (2000) [401] R.A. Olie, A.P. Simoes-Wust, B. Baumann, S.H. Leech, D.
7871^7876. Fabbro, R.A. Stahel, U. Zangemeister-Wittke, Cancer Res.
[395] S. Xanthoudakis, D.W. Nicholson, Nature Cell Biol. 2 60 (2000) 2805^2809.
(2000) E163^E165. [402] A. Degterev, A. Lugovskoy, M. Cardone, B. Mulley, G.
[396] Q. Yu, B.P. Toole, I. Stamenkovic, J. Exp. Med. 186 (1997) Wagner, T. Mitchison, J. Yuan, Nature Cell Biol. 3
1985^1996. (2001) 173^182.
[397] E. Finkel, Science 286 (1999) 2256^2258.

BBACAN 87502 25-9-01 Cyaan Magenta Geel Zwart

Você também pode gostar