You are on page 1of 12

Molecular Cell, Vol.

9, 459–470, March, 2002, Copyright 2002 by Cell Press

Mechanisms of Caspase Activation Review


and Inhibition during Apoptosis

Yigong Shi1 which include caspase-3, -6, and -7 (Figure 1). An initia-
Department of Molecular Biology tor caspase is characterized by an extended N-terminal
Princeton University prodomain (⬎90 amino acids) important for its function,
Lewis Thomas Laboratory whereas an effector caspase contains 20–30 residues
Washington Road in its prodomain sequence. All caspases are produced
Princeton, New Jersey 08544 in cells as catalytically inactive zymogens and must un-
dergo proteolytic activation during apoptosis. The acti-
vation of an effector caspase (such as caspase-3 or -7)
is performed by an initiator caspase (such as caspase-9)
Caspases are central components of the machinery through cleavage at specific internal Asp residues that
responsible for apoptosis. Recent structural and bio- separate the large and small subunits (Figure 1). The
chemical studies on procaspases, IAPs, Smac/DIA- initiator caspases, however, are autoactivated. As the
BLO, and apoptosome have revealed a conserved activation of an initiator caspase in cells inevitably trig-
mechanism of caspase activation and inhibition. This gers a cascade of downstream caspase activation, it is
article reviews these latest advances and presents our tightly regulated and often requires the assembly of a
current understanding of caspase regulation during multicomponent complex under apoptotic conditions.
For example, the activation of procaspase-9 is facili-
apoptosis.
tated by Apaf-1 and cytochrome c (Cyt. c) (Li et al.,
1997), which form an ⵑ1.4 MDa complex dubbed an
apoptosome in the presence of dATP or ATP (Rodriguez
Introduction
and Lazebnik, 1999; Saleh et al., 1999; Zou et al., 1999).
Apoptosis, or programmed cell death, plays a central
Once activated, the effector caspases are responsible
role in the development and homeostasis of all multi-
for the proteolytic cleavage of a broad spectrum of cellu-
cellular organisms (Horvitz, 1999; Jacobson et al., 1997).
lar targets, leading ultimately to cell death. The known
In humans, both excessive and insufficient apoptosis
cellular substrates include structural components (such
can lead to severe pathological consequences. Sup- as actin and nuclear lamin), regulatory proteins (such
pression of the apoptotic machinery causes autoim- as DNA-dependent protein kinase), inhibitors of deoxyri-
mune diseases and is a hallmark of cancer (Hanahan bonuclease (such as DFF45 or ICAD), and other proapo-
and Weinberg, 2000; Thompson, 1995). For example, ptotic proteins and caspases. Cleavage of the DFF45
the apoptotic protease-activating factor 1 (Apaf-1) is (DNA fragmentation factor 45) leads to removal of its
frequently inactivated in cancers such as malignant mel- inhibition of DFF40 or CAD (caspase-activated deoxyri-
anoma (Soengas et al., 2001). On the other hand, abnor- bonuclease), which degrades the chromosomes into
mal upregulation of apoptosis contributes to neurologi- nucleosomal fragments during apoptosis (Enari et al.,
cal disorders (Yuan and Yankner, 2000). 1998; Liu et al., 1997, 1998).
The critical involvement of a cysteine protease CED-3 Caspases are specific proteases, recognizing at least
in apoptosis was first discovered in the nematode four contiguous amino acids, named P4-P3-P2-P1, and
Caenorhabditis elegans 8 years ago (Yuan et al., 1993). cleaving after the C-terminal residue (P1), usually an
Since then, compelling evidence has demonstrated that Asp. Although the P1 residue was thought to be exclu-
the mechanism of apoptosis is evolutionarily conserved, sively Asp, recent studies indicate that some caspases
executed by a family of cysteine proteases that cleave can also cleave after Glu (Hawkins et al., 2000; Sriniva-
after an Asp residue in their substrates, hence the name sula et al., 2001). Interestingly, the preferred P3 position
caspase (Alnemri et al., 1996). At least 14 distinct mam- is invariantly Glu for all mammalian caspases examined
malian caspases have been identified, with their or- (Thornberry et al., 1997). Thus the preferred specificity
thologs present in species ranging from the nematode of cleavage for caspases can be described as X-Glu-X-
to the dipteran Drosophila melanogaster and the lepi- Asp.
dopteran Spodoptera frugiperda. Although the first Structural characterization of caspase-1 reveals a ho-
modimeric structure (Walker et al., 1994; Wilson et al.,
mammalian caspase, caspase-1 or ICE (interleukin
1994). It is now believed that the functional caspase unit
1␤-converting enzyme), was identified to be an impor-
is a homodimer, with each monomer comprising a large
tant regulator of inflammatory response, at least 8 of
(20 kDa) and a small (10 kDa) subunit. Homodimerization
the 14 caspases play important roles during apoptosis is mediated by hydrophobic interactions, with 6 antipar-
(Budihardjo et al., 1999; Earnshaw et al., 1999; Fesik allel ␤ strands from each catalytic subunit forming a
and Shi, 2001; Salvesen and Dixit, 1997; Thornberry and single contiguous 12-stranded ␤ sheet (Figure 2A). Sev-
Lazebnik, 1998). eral ␣ helices and short ␤ strands are located on either
Caspases involved in apoptosis are generally divided side of the central ␤ sheet, giving rise to a globular fold.
into two categories, the initiator caspases, which include The active sites, formed by four protruding loops from
caspase-2, -8, -9, and -10, and the effector caspases, the scaffold, are located at two opposite ends of the ␤
sheet (Figure 2A). In addition to caspase-1, structural
information is available for caspase-3 (Mittl et al., 1997;
1
Correspondence: yshi@molbio.princeton.edu Rotonda et al., 1996), caspase-7 (Wei et al., 2000), cas-
Molecular Cell
460

Figure 1. Schematic Diagram of the Mamma-


lian Caspases
Except caspase-11 (mouse), -12 (mouse),
and -13 (bovine), all listed caspases are of
human origin. Their phylogenetic relationship
(left) appears to correlate with their function
in apoptosis or inflammation. The initiator and
effector caspases are labeled in purple and
red, respectively. The position of the first acti-
vation cleavage (between the large and small
subunits) is highlighted with a large arrow
while additional sites of cleavage are repre-
sented by medium and small arrows. In con-
trast to other protease zymogens, removal of
the prodomain of a caspase is unnecessary
for its catalytic activity. The four surface loops
(L1-L4) that shape the catalytic groove are
indicated. The catalytic residue Cys is shown
as a red line at the beginning of loop L2. This
diagram is scaled according to the lengths
of caspases and the location of functional
segments.

pase-8 (Blanchard et al., 1999; Watt et al., 1999), and All known caspases share a similar conformation at
more recently, caspase-9 (Renatus et al., 2001). Rather the substrate-binding groove (Figure 2B). The S1 and
than giving a general review on the structural biology S3 sites are nearly identical among all caspases whereas
of apoptosis (Fesik, 2000; Shi, 2001), this article focuses the location of the S2 and S4 sites is highly conserved.
on the latest developments and surveys our current un- The P1 residue (Asp) is coordinated by three invariant
derstanding of caspase regulation at a molecular level. residues at the S1 site, an Arg from L1, a Gln at the
beginning of L2, and an Arg at the end of L3 (Earnshaw
Conserved Features of the Active Site et al., 1999). The Arg residue on the L3 loop also coordi-
The substrate-binding groove is shaped by four sur- nates the P3 residue (Glu). The S2 and S4 sites map
rounding loops, named L1, L2, L3, and L4 (Figure 2B). mainly to the L3 and L4 loops; thus the P2 and P4
The L1 loop constitutes one side of the groove whereas residues exhibit greater sequence variation. For exam-
L4 represents the other side (Figure 2C). Loop L3 and ple, the L4 loop in caspase-1, -8, or -9 is considerably
the following ␤ hairpin, collectively referred to as L3, shorter than that in caspase-3 or -7, resulting in a shal-
are located at the base of the groove. Loop L2, which lower substrate-binding groove (Figure 2D). This obser-
harbors the catalytic residue Cys, is positioned at one vation is consistent with a bulky hydrophobic residue
end of the groove with the side chain of Cys pointing as the preferred P4 residue for caspase-1, -8, or -9.
along the groove, poised for binding and catalysis. The conformational similarity at the active site is ex-
Among these four loops, L1 and L3 exhibit a relatively tended to surrounding structural elements. Most nota-
conserved length as well as composition among all bly, the loops L4 and L2 from one catalytic subunit are
mammalian caspases (Figure 1). In contrast, L2 and L4 stabilized by the N terminus (loop L2⬘) of the small sub-
are highly divergent. These four loops determine the unit of the other catalytic subunit (Figure 2), forming the
sequence specificity of the substrates. The binding so-called “loop bundle” (Chai et al., 2001a).
pockets for the P4-P3-P2-P1 residues of the substrate
are named S4-S3-S2-S1 subsites, respectively. Struc- Activation Mechanisms of the Effector Procaspases
tural studies using covalent peptide inhibitors reveal Except procaspase-9, all other procaspase zymogens
that these pockets are located mostly between the base are inactive and require proteolytic activation. Given the
(L3) and the two sides (L1 and L4). presence of the four active site loops in the zymogen,
Review
461

Figure 2. Structural Features of Caspases


(A) Representative structure of an inhibitor-
bound caspase-3 (PDB code 1DD1). The
small and large subunits are colored orange
and blue, respectively. The bound peptide in-
hibitor is shown in pink. The four surface
loops that constitute the catalytic groove of
one heterodimer are labeled. The apostrophe
denotes the other heterodimer. Note that L2⬘
stabilizes the active site of the adjacent het-
erodimer.
(B) The active site conformation of all known
caspases is conserved. Of the four loops, L1
and L3 are relatively constant while L2 and
L4 exhibit greater variability. The catalytic
residue Cys is highlighted in red.
(C) Schematic diagram of the substrate-bind-
ing groove. L1 and L4 constitute two parallel
sides of the groove while L3 serves as the
base. L2, harboring the catalytic residue Cys,
is positioned at one end of the groove, poised
for catalysis. L2⬘ plays a critical role by stabi-
lizing the conformation of the L2 and L4
loops.
(D) Conformation of the L4 loop in five known
caspase structures. The L4 loop dictates the
P4 specificity of the substrate. Short L4 loops
(caspase-1, -8, and -9) allow bulkier and hy-
drophobic residue in the P4 position whereas
extended L4 loops (caspase-3 and -7) prefer
Asp. Coloring scheme is the same as (B). Fig-
ures were prepared using MOLSCRIPT
(Klaulis, 1991) and GRASP (Nicholls et al.,
1991).

it was intuitively unclear why this is the case. The recent deviation for all aligned C␣ atoms. However, three of
crystal structure of procaspase-7 (Figure 3A) provides the four active site loops adopt drastically different con-
an explanation to this puzzle (Chai et al., 2001a; Riedl formations. L3, which forms the base of the catalytic
et al., 2001a). groove in the active caspase-7, is unraveled and loos-
Compared to the inhibitor-bound caspase-7, the core ened above the base (Figure 3C). Loop L4, which forms
structural elements of the procaspase-7 zymogen are one side of the catalytic groove in the active caspase-7,
nearly identical, with less than 0.8 Å root-mean-square- is located farther away from L3, flattening the active site

Figure 3. Mechanisms of Procaspase-7 Activation and Substrate Binding


(A) Structure of a procaspase-7 zymogen (PDB code 1K86). Compared to that of the inhibitor-bound caspase-7, the conformation of the active
site loops does not support substrate binding or catalysis. The L2⬘ loop, locked in a closed conformation by covalent linkage, is occluded
from adopting its productive and open conformation.
(B) Structure of an active and free caspase-7 (PDB code 1K88). The active site loops are still flexible. Despite an interdomain cleavage, the
L2⬘ loop still exists in the closed conformation, indicating an induced-fit mechanism for binding to inhibitors/substrates.
(C) Comparison of the conformation of the active site loops. Compared to the procaspase-7 zymogen or the free caspase-7, the L2⬘ loop is
flipped 180o in the inhibitor-bound caspase-7 to stabilize loops L2 and L4.
Molecular Cell
462

pocket. Most importantly, loop L2, which contains the observation argues that the substrate-bound state is
catalytic residue Cys186, is rotated 90⬚, making this resi- transient and can be trapped by the covalent peptide
due inaccessible to solvent. Among the four loops, only inhibitors. It remains to be seen whether this mechanism
L1 retains its active site conformation. is generally applicable to other caspases.
These rearrangements in the procaspase-7 zymogen
do not allow formation of a substrate-binding groove. Inhibition of Caspases by IAPs
In particular, the loop bundle seen in the inhibitor-bound Caspases are regulated by many cellular processes.
caspases is missing in the procaspase-7 zymogen as For example, caspases are subject to transcriptional
the L2⬘ loop is flipped by 180⬚, existing in a “closed” regulation and posttranslational modification (Earnshaw
conformation (Figure 3C). These structural rearrange- et al., 1999). In addition, active caspases can be perma-
ments explain why the procaspase-7 zymogen does not nently eliminated by the ubiquitination-mediated protea-
possess detectable catalytic activity. some degradation pathway (Huang et al., 2000; Suzuki
What makes the active site of the procaspase-7 zymo- et al., 2001b).
gen so different from that of the inhibitor-bound cas- The enzymatic activity of caspases is subject to inhibi-
pase-7? One essential reason is that the unprocessed tion by the conserved IAP (inhibitor of apoptosis) family
nature of the procaspase-7 zymogen locks the L2⬘ loop of proteins (Deveraux and Reed, 1999; Hay, 2000). The
in the closed conformation and occludes it from forming IAPs, originally identified in the genome of baculovirus
a loop bundle with the L2 and L4 loops. This inability based on their ability to suppress apoptosis in infected
can only be removed by a proteolytic cleavage after host cells, antagonize cell death by interacting with and
Asp198, which allows L2⬘ to switch to its open conforma- inhibiting the enzymatic activity of mature caspases.
tion as observed in the inhibitor-bound caspase-7. As Eight distinct mammalian IAPs, including XIAP, c-IAP1,
the interactions that stabilize the loop bundle are gener- c-IAP2, and ML-IAP/Livin (Ashhab et al., 2001; Kasof
ally conserved among caspases, this mechanism is and Gomes, 2001; Vucic et al., 2000), have been identi-
likely to be true for other caspases. fied, and they target the initiator caspase, caspase-9,
In this mechanism, the ability of L2⬘ to move freely and the effector caspases, caspase-3 and -7 (Deveraux
in response to inhibitor/substrate binding is a decisive and Reed, 1999). These IAP proteins do not inhibit other
feature. In caspase-7, this ability is acquired through caspases, such as caspase-6 or -8.
proteolytic cleavage after Asp198. But, because L2⬘ is The functional unit in IAPs is the baculoviral IAP repeat
at the N terminus of the small subunit, inverting the order (BIR), which contains ⵑ80 amino acids folded around a
of primary sequences of the large and small subunits zinc atom. XIAP, c-IAP1, and c-IAP2 contain three BIR
could free L2⬘ and hence constitutively activate cas- domains each, with the different BIR domains exhibiting
pases. Indeed, this prediction was confirmed for cas- distinct functions. In these IAPs, the third BIR domain
pase-3 and -6 (Srinivasula et al., 1998b) as well as for (BIR3) potently inhibits the activity of processed cas-
the Drosophila caspase drICE (Wang et al., 1999). pase-9 whereas the linker region between BIR1 and
Why does procaspase-9 exhibit a basal level of activ- BIR2 selectively targets caspase-3 and -7 (Fesik and
ity prior to proteolytic activation (Stennicke et al., 1999)? Shi, 2001; Shi, 2001). This inhibition is highly specific;
The answer may lie in the fact that caspase-9 contains for example, BIR3 exhibits no inhibition of caspase-3
an expanded L2 loop, which could allow enough confor- even at elevated concentrations whereas the linker re-
mational flexibility for the L2⬘ loop to move to its produc- gion between BIR1 and BIR2 has no impact on caspase-
tive conformation without an interdomain cleavage. 9. Mutational analysis revealed that, in the XIAP-linker-
BIR2 fragment, substitution of Asp148 or Leu141 to Ala
Mechanisms of Substrate Binding nearly abolished caspase-3 inhibition (Sun et al., 1999).
Because the active sites of all inhibitor-bound caspases The corresponding BIR domains in XIAP, c-IAP1, and
are highly similar (Figure 2B), it was once thought that c-IAP2 are highly conserved in both sequence and func-
the substrate-binding grooves on caspases are pre- tion. For example, BIR3 of XIAP is more similar to BIR3
formed. However, in the structure of the free caspase-7 of c-IAP1 or c-IAP2 than to BIR2 or BIR1 of XIAP. In
(Chai et al., 2001a), these loops are flexible and quite contrast to these IAPs, ML-IAP/Livin, which is highly
different from those in the inhibitor-bound caspase-7 expressed in melanoma and lymphoma, contains only
(Figure 3B). Surprisingly, despite activation cleavage, a single BIR domain but was reported to inhibit both
the L2’ loop in the isolated active caspase-7 still exists caspase-3 and -9 (Ashhab et al., 2001; Kasof and
in the closed conformation, mimicking the procaspase-7 Gomes, 2001; Vucic et al., 2000). Another single BIR-
zymogen (Figure 3C). Consequently, the loop bundle is containing IAP, survivin, does not inhibit caspase activ-
not properly assembled and the catalytic cleft is not well ity in vitro. In contrast to the relatively stable expression
defined. levels of other IAPs, expression of survivin oscillates
This unanticipated finding indicates that the isolated with cell cycle and peaks at the G2/M phase (Li et al.,
active caspase-7 adopts a conformation intermediate 1998). Survivin appears to play an important role in mito-
between that of the zymogen and that of the inhibitor- sis (Altieri, 2001; Li et al., 1998).
bound caspase-7. Thus inhibitor/substrate binding trig-
gers a 180⬚ flipping of the L2⬘ loop in the active Mechanisms of IAP-Mediated Inhibition
caspase-7, changing its conformation from a closed to of Effector Caspases
an open form (Figure 3C). Each cycle of substrate bind- Recent structural analyses reveal the precise mecha-
ing and catalysis is a process of induced fit, accompa- nism of IAP-mediated inhibition of caspase-3 and -7
nied by the back-and-forth flipping of the L2⬘ loop. This (Chai et al., 2001b; Huang et al., 2001; Riedl et al., 2001b).
Review
463

Figure 4. Mechanisms of IAP-Mediated Inhi-


bition of Effector Caspases and p35-Medi-
ated Pan-Caspase Inhibition
(A) Superposition of the structures of cas-
pase-3 (brown) and -7 (green) together with
their bound XIAP fragments colored blue and
pink, respectively. The interactions primarily
occur between a linker segment N-terminal
to the BIR2 domain of XIAP and the active
site of caspase-3 or -7 (boxed region). The
small red circle on BIR2 marks where the
Smac N-terminal tetrapeptide binds and the
large red circle indicates a likely second inter-
face with Smac.
(B) Close-up view of the active sites of cas-
pase-3 and -7 bound to their respective XIAP
fragments. Two hydrophobic residues of
XIAP, Leu141 and Val146, make multiple van
der Waals interactions with a conserved hy-
drophobic pocket on caspase-3 or -7. Asp148
of XIAP, occupying the S4 pocket, hydrogen
bonds to neighboring residues in caspase-3
or -7.
(C) Close-up view of the covalent inhibition
of caspase-8 by p35. The thioester intermedi-
ate is shown between Asp87 of p35 and
Cys360 (active site residue). The N terminus
of p35 restricts solvent access to this inter-
mediate.

In the structures, the linker peptide N-terminal to XIAP- Waals contacts to surrounding caspase residues as
BIR2 forms highly similar interactions with both cas- does the P2 residue. In contrast to the covalent peptide
pase-3 and -7 (Figure 4A). Compared to the covalent inhibitors, Gly144 is located close to the S1 pocket,
peptide inhibitors, the linker segment of XIAP occupies within van der Waals contact distance of the catalytic
the active site of caspases in a reverse orientation, re- Cys in the caspases.
sulting in a blockade of substrate entry (Figure 4B). Interestingly, although the linker sequence between
Asp148 of XIAP, which was shown to be critical for the BIR1 and BIR2 of XIAP plays a dominant role in inhibiting
inhibition of caspase-3, binds the S4 pocket in the same caspase-3 and -7, this fragment in isolation is insufficient
manner as the P4 residue of the covalent peptide inhibi- (Chai et al., 2001b; Sun et al., 1999). However, an engi-
tors. In addition, Val146 makes a similar set of van der neered protein with the linker peptide fused either N-
Molecular Cell
464

or C-terminal to BIR1 was fully able to bind and inhibit p35, which occludes access by water molecules (Figure
caspase-3 while neither BIR1 nor BIR2 in isolation exhib- 4C). Another protein, the serpin CrmA derived from the
ited any effect (Sun et al., 1999). These observations cowpox virus, can also inhibit several caspases, likely
suggest that the linker peptide needs to be presented through similar covalent modification. This unique cova-
in a “productive” conformation by a surrounding BIR lent mechanism adds to the complexity of caspase inhi-
domain. In support of this hypothesis, the linker peptide bition by natural proteins. It is important to note, how-
fused to glutathione S-transferase (GST) was able to ever, that an equivalent of p35 in mammals has not been
inhibit caspase-3 and -7 (Chai et al., 2001b; Huang et identified.
al., 2001). Nevertheless, the BIR domains also contribute
to the inhibition of caspases, as XIAP exhibits about 20- Function of Smac/DIABLO
fold higher potency than the GST-linker peptide fusion During apoptosis, the IAP-mediated caspase inhibition
(Chai et al., 2001b; Huang et al., 2001). Consistent with is removed by a mitochondrial protein named Smac
this observation, XIAP-BIR2 also makes direct contacts (second mitochondria-derived activator of caspases)
to caspase-3 in the crystal structure (Riedl et al., 2001b). (Du et al., 2000) or DIABLO (direct IAP-binding protein
More importantly, the BIR domains provide a scaffold for with low pI) (Verhagen et al., 2000). Upon stimulation of
Smac/DIABLO binding in order to relieve IAP-mediated apoptosis, Smac/DIABLO is released from the inter-
inhibition of the effector caspases (see below). membrane space of mitochondria into the cytosol, to-
gether with Cyt. c. Whereas Cyt. c directly activates
Mechanisms of IAP-Mediated Inhibition Apaf-1 and caspase-9, Smac/DIABLO interacts with
of Caspase-9 multiple IAPs and removes IAP-mediated inhibition of
Only processed caspase-9 is subject to inhibition by both initiator and effector caspases (Chai et al., 2000;
IAPs. Although both BIR2 and BIR3 of XIAP can inhibit Srinivasula et al., 2000).
caspase-9, BIR3 displays tighter binding affinity and Smac functions as an elongated dimer, spanning over
higher potency. Mutation of Trp310 or Glu314 in BIR3 130 Å in length (Chai et al., 2000) (Figure 5A). The wild-
completely abrogates XIAP-mediated inhibition of cas- type Smac protein binds to both the BIR2 and the BIR3
pase-9, while amino acids outside of BIR3 are unneces- domains of XIAP but not the BIR1 domain. In contrast,
sary for this inhibition (Sun et al., 2000). In the structure, the monomeric Smac mutants interact strongly with
these two residues are located close to each other, BIR3 but do not form a stable complex with BIR2 (Chai
suggesting that this region is involved in binding and et al., 2000). Because the linker sequence immediately
inhibiting caspase-9. Indeed, BIR3 of XIAP binds to the preceding BIR2 is involved in binding and inhibiting cas-
N terminus of the small subunit of caspase-9 that be- pase-3, Smac monomers cannot relieve the IAP-medi-
comes exposed after proteolytic processing (Srinivasula ated caspase-3 inhibition. Despite the ability to interact
et al., 2001). This binding presumably brings BIR3 close with BIR3, the monomeric Smac mutants are less able
to the active site of caspase-9, which may block sub- to relieve IAP-mediated caspase-9 inhibition (Chai et al.,
strate entry and subsequent catalysis. In support of this 2000).
hypothesis, when the N-terminal four residues of the The N-terminal mitochondria-targeting sequence of
caspase-3 small subunit were replaced with those from Smac is proteolytically removed upon import (Du et al.,
caspase-9, the resulting protein could be inhibited by 2000). These freshly exposed N-terminal residues play
BIR3 (Srinivasula et al., 2001). an indispensable role in Smac function; a seven residue
The interaction between the N-terminal four residues peptide including these residues can remove the XIAP-
of the caspase-9 small subunit and XIAP-BIR3 is neces- mediated caspase-9 inhibition. Strikingly, a missense
sary but may not be sufficient for the XIAP-mediated mutation of the N-terminal residue Ala to Met in Smac
inhibition. Mutation of His343 in XIAP-BIR3, which is leads to a complete loss of interactions with XIAP and
located in a different area than Trp310 and Glu314, re- the concomitant loss of Smac function (Chai et al., 2000).
sults in a loss of caspase-9 inhibition, suggesting that
His343 also makes important contacts to caspase-9 A Tetrapeptide IAP-Binding Motif
(Sun et al., 2000). The molecular explanation for the indispensable role of
the Smac N-terminal sequences is provided by struc-
Covalent Inhibition of Caspases by p35 tures of XIAP-BIR3 bound to either a monomeric Smac
IAPs are not the only natural inhibitors to caspases. In protein (Wu et al., 2000) or a nine residue Smac peptide
contrast to XIAP, which only affects caspase-3, -7, and (Liu et al., 2000). The Smac N-terminal tetrapeptide (Ala-
-9, the baculoviral p35 protein is a pan-caspase inhibitor, Val-Pro-Ile) recognizes an acidic surface groove on
and it potently targets most caspases both in vivo and in BIR3, with the first residue Ala binding a hydrophobic
vitro (Miller, 1999). Caspase inhibition by p35 correlates pocket and making hydrogen bonds to neighboring
with the cleavage of its reactive site loop after Asp87, XIAP residues (Figure 5B). The next three residues also
which leads to the translocation of the N terminus of interact with surrounding hydrophobic residues in BIR3.
p35 into the active site of caspases (Bump et al., 1995; To accommodate these interactions, the N terminus of
Zhou et al., 1998). The crystal structure of caspase-8 Smac must be free, thus explaining why only the mature
in complex with p35 reveals that the catalytic residue Smac can bind to the IAPs. Modeling studies indicate
Cys360 of caspase-8 is covalently linked to the Asp87 of that replacement of Ala by a bulkier residue will cause
p35 through a thioester bond (Xu et al., 2001). Although a steric hindrance while Gly substitution may result in an
thioester bond is generally susceptible to hydrolysis, entropic penalty as well as loss of binding in the hy-
this bond is protected by the neighboring N terminus of drophobic pocket. This analysis explains why mutation
Review
465

Figure 5. A Conserved IAP-Binding Tetra-


peptide Motif
(A) Structure of the mature Smac. The disor-
dered N-terminal residues are shown as dot-
ted lines.
(B) Close-up view of the Smac N-terminal tet-
rapeptide binding to the BIR3 surface groove.
The BIR3 domain is shown either by electro-
static potential (left panel) or in ribbon dia-
gram (right panel) to highlight the interac-
tions. The amino and carbonyl groups of the
N-terminal Ala make several hydrogen bonds
to conserved residues in XIAP.
(C) A conserved motif of IAP-binding tetra-
peptides. The tetrapeptide motif has the con-
sensus sequence A-(V/T/I)-(P/A)-(F/Y/I/V/S).
The Drosophila proteins have an additional
binding component (conserved 6th–8th res-
idues).
(D) A conserved IAP-binding mode from
mammals to fruit flies. The structure of
DIAP1-BIR2 is superimposed with that of the
XIAP-BIR3 domain, with their corresponding
bound peptides Hid (pink), Grim (blue), and
Smac/DIABLO (green).

of Ala to Met or Gly abrogated interactions with the BIR tides from Grim, Hid, and Sickle reveal that the binding
domains. Since most of the important interface residues of these N-terminal tetrapeptides precisely match that
of BIR3 are conserved in BIR2, the isolated N-terminal of the Smac-XIAP interactions (Srinivasula et al., 2002;
Smac peptides can also interact with the BIR2 domain Wu et al., 2001) (Figure 5D). For Grim and Hid, the next
of XIAP, though with a lower binding affinity (Liu et al., three conserved residues also contribute to DIAP1 bind-
2000). ing through hydrophobic interactions.
In fruit flies, the anti-death IAP, DIAP1, binds and Using a direct XIAP-binding assay, another mamma-
inhibits several Drosophila caspases (Hay, 2000). This lian protein, HtrA2/Omi, was recently identified by five
function is countered by the proapoptotic proteins, independent groups (Hedge et al., 2002; Martin et al.,
Reaper, Grim, Hid, and more recently, Sickle (Christich 2002; Suzuki et al., 2001a; van Loo et al., 2002; Verhagen
et al., 2002; Srinivasula et al., 2002; Wing et al., 2002), et al., 2002). Similar to Smac, HtrA2/Omi is believed to
which physically interact with the BIR2 domain of DIAP1 be a resident protein of the intermembrane space of
and remove its inhibitory effect on caspases. Thus mitochondria and is released to the cytosol during apo-
Reaper, Grim, Hid, and Sickle are functional homologs ptosis. The mature form of HtrA2/Omi contains three
of the mammalian protein Smac/DIABLO. Supporting functional segments, an IAP-binding tetrapeptide motif
this notion, the N-terminal tetrapeptides of Reaper, at its N terminus, a central serine protease domain, and a
Grim, Hid, and Sickle closely resemble that of the Smac/ PDZ domain at its C terminus. Interestingly, the catalytic
DIABLO (Figure 5C). The Smac-binding surface groove activity of the serine protease domain is absolutely es-
on XIAP-BIR3 comprises highly conserved residues sential to the HtrA2/Omi-mediated cell killing activity
among the BIR3 domains of c-IAP1 and c-IAP2 and the whereas the N-terminal IAP-binding motif appears less
BIR2 domain of DIAP1, suggesting a conserved mode critical. HtrA2/Omi is unlikely to induce cell death
of interactions. Indeed, the crystal structures of DIAP1- through a nonspecific protease activity as it appears to
BIR2 by itself and in complex with the N-terminal pep- exhibit stringent substrate specificity (Gray et al., 2000).
Molecular Cell
466

Figure 6. Schematic Diagram of the Mechanisms of Caspase Activation and Inhibition


(Left box) The tetrapeptide motif (red arrows), present in the N termini of Smac and the small subunit of caspase-9, binds to a surface groove
on the BIR domains of IAPs (XIAP, c-IAP1, c-IAP2, and Livin/ML-IAP). This motif is responsible for both caspase-9 inhibition through binding
BIR3 and the relief of inhibition through displacement of the caspase-9 tetrapeptide with a Smac tetrapeptide. Positive feedback cleavage
by caspase-3 or -7 permanently removes IAP-mediated inhibition of caspase-9. The released caspase-9 linker peptide can antagonize IAP-
mediated inhibition of other caspases.
(Right box) On the other hand, a linker segment N-terminal to the BIR2 domain of XIAP is primarily responsible for caspase-3 inhibition. Smac
binding to the BIR2 domain may make this linker segment unavailable for binding to caspase-3. For clarity, only BIR domains but not the full-
length IAPs are shown in this figure.

Since PDZ domains typically interact with the C-terminal During apoptosis, caspase-9 can be further cleaved
hydrophobic residues of membrane receptors and ion after Asp330 by downstream caspases such as cas-
channels (Harris and Lim, 2001), the PDZ domain in pase-3 (Figure 6). This positive feedback not only perma-
HtrA2/Omi may further regulate its protease activity. The nently removes XIAP-mediated caspase-9 inhibition but
precise mechanism of HtrA2/Omi-mediated apoptosis also releases a 15 residue peptide that is able to relieve
remains elusive. IAP-mediated inhibition of other caspases. This mecha-
nism ensures that less than a stoichiometric amount of
Removal of Caspase Inhibition by Smac/DIABLO Smac can remove the IAP-mediated caspase-9 inhibi-
During apoptosis, Smac/DIABLO reactivates the pro- tion as transient activation of caspase-9 may lead to the
cessed initiator as well as effector caspases through activation of caspase-3 and ensuing positive feedback.
distinct mechanisms, although both require physical in- Although the Smac tetrapeptide in isolation can re-
teractions with IAPs. The N terminus (Ala-Thr-Pro-Phe) move IAP-mediated caspase-9 inhibition, it plays a less
of the small subunit of caspase-9 was found to contain direct role in the removal of IAP-mediated inhibition of
an IAP-binding tetrapeptide motif (Srinivasula et al., effector caspases (Figure 6). The binding site for this
2001). Subsequent experiments confirmed that this se- tetrapeptide motif maps to the surface of BIR2 or BIR3,
quence is indeed primarily responsible for the interac- whereas the fragment responsible for inhibiting cas-
tions between the processed caspase-9 and XIAP (Srini- pase-3 or -7 is located between BIR1 and BIR2 of XIAP.
vasula et al., 2001). In the absence of proteolytic Although a conclusive mechanism remains elusive,
processing, procaspase-9 is unable to interact with modeling studies of a Smac/BIR2/caspase-3 complex
IAPs. Proteolytic processing of procaspase-9 at Asp315 suggest that steric clashes preclude XIAP-BIR2 from
leads to the exposure of an internal tetrapeptide motif, simultaneously binding to caspase-3 and Smac (Chai et
which recruits IAPs to inhibit caspase-9 (Figure 6). The al., 2001b). In this model, binding to the BIR2 domain
mature Smac binds IAPs, again using a similar N-ter- requires not only the N-terminal tetrapeptide of Smac
minal tetrapeptide. Thus, a conserved IAP-binding motif but also an extensive surface available only in the wild-
in caspase-9 and Smac mediates opposing effects on type dimeric Smac protein. This model is consistent
caspase activity (Figure 6). with the observation that monomeric Smac mutants only
Review
467

Figure 7. Structure of Caspase-9 and the


Apoptosome
(A) Two views of the heptameric apoptosome
comprising Apaf-1, Cyt. c, and dATP. The
CARD domains are located in the central hub
while the WD40 repeats were mapped to the
end of the spoke.
(B) Structure of the inhibitor-bound caspase-
9. Only half of the caspase-9 dimer contains
a well-formed active site (shown in green),
covalently bound by an inhibitor. The active
site loops in the other half, shown in pink, do
not form a functional catalytic groove.
(C) A proposed model for the activation of
caspase-9. The apoptosome-bound cas-
pase-9 exists in an inactive conformation.
The high local concentrations of caspase-9
within the apoptosome drive the recruitment
of additional caspase-9 monomers, which are
activated upon dimerization.

weakly interacted with BIR2 and were unable to remove closely resemble a large family of disease-resistant
the IAP-mediated caspase-3 inhibition (Chai et al., 2000). genes in plants (Dangl and Jones, 2001).
The induced proximity model appears to be consis-
tent with a number of observations. For example, bacte-
Induced Proximity Model for the Initiator Procaspases rially expressed caspases are almost always processed
The initiator caspases invariably contain one of two pro- to their mature forms, presumably due to high local
tein-protein interaction motifs, the CARD (caspase re- concentrations. In mammalian cells, forced oligomeriza-
cruitment domain) or the DED (death effector domain). tion of procaspase-8 through the use of FKBP (FK506
These motifs interact with similar motifs present on binding protein) and FK1012 (a dimeric form of FK506)
oligomerized adapter proteins, thus bringing multiple led to activation of procaspase-8 and subsequent apo-
initiator caspase molecules into close proximity with one ptosis (Muzio et al., 1998; Yang et al., 1998a). Similar
another and presumably facilitating their autoactivation. results were reported for the mammalian caspase-9
This hypothesis is summarized as the induced-proximity (Srinivasula et al., 1998a) and the C. elegans CED3 (Yang
model (Salvesen and Dixit, 1999). For example, procas- et al., 1998b).
pase-8 contains two copies of the DED, which interact Although induced proximity undoubtedly leads to cas-
with the DED of the adapter protein FADD (Fas-associ- pase activation, it remains unclear whether this is indeed
ated death domain). FADD, in turn, is a component of how the initiator caspases are activated under physio-
the multimeric death-inducing signaling complex (DISC) logical conditions. For example, effector caspases can
at the cell membrane (Ashkenazi and Dixit, 1998). Through also be autoactivated through induced proximity; yet
association with FADD, procaspase-8 is brought into in vivo they are activated by the initiator caspases. In
the DISC, resulting in its autocleavage and activation. addition, unprocessed procaspase-9 is nearly as active
Another well-characterized paradigm involves pro- as mature caspase-9 (Srinivasula et al., 2001) and the
caspase-9 activation. During apoptosis, Cyt. c is re- primary function of the apoptosome is to allosterically
leased from mitochondria into the cytoplasm, where it enhance caspase-9 activity rather than to facilitate its
binds to Apaf-1 and, in the presence of dATP or ATP, autoactivation (Rodriguez and Lazebnik, 1999). Further-
assembles into a complex called the apoptosome (Saleh more, forced oligomerization of the initiator caspases
et al., 1999; Zou et al., 1999). The primary function of may not recapitulate the physiological context, in terms
the apoptosome is to recruit procaspase-9 through a of protein expression levels, and more importantly, the
CARD-CARD interaction between caspase-9 and Apaf-1 specific protein-protein interactions that are required
and facilitate autoactivation of caspase-9. Interestingly, for the precise positioning and activation of the initiator
Apaf-1 is a member of a conserved family of adaptor caspases.
proteins that contain an N-terminal CARD, a central nu-
cleotide-binding oligomerization domain (NOD), and a
C-terminal sensing domain for intracellular ligands (Ino- Caspase-9 and the Apoptosome
hara and Nunez, 2001). Members of the NOD family also It was a rather startling discovery that the processed
include Ipaf (Poyet et al., 2001), Nod1 (Inohara et al., caspase-9 is only marginally active in the absence of the
1999), and Nod2 (Ogura et al., 2001). The caspase-acti- apoptosome, prompting the concept of a holo-enzyme
vating mechanisms by these NODs are likely to be con- (Rodriguez and Lazebnik, 1999). Through association
served as well. Interestingly, the NOD proteins in animals with the apoptosome, the catalytic activity of caspase-9
Molecular Cell
468

is enhanced more than 1000-fold (Rodriguez and Lazeb- of procaspase zymogen, the mechanisms of IAP-medi-
nik, 1999). Surprisingly, the unprocessed caspase-9 can ated caspase inhibition, and the Smac-mediated re-
be similarly maintained in this “hyperactive” state, dem- moval of caspase inhibition by IAPs. The identification of
onstrating that the proteolytic processing is unneces- a conserved tetrapeptide IAP-binding motif helps unify
sary for the activation of procaspase-9 (Srinivasula et mechanisms of apoptosis regulation from fruit flies to
al., 2001). mammals. Despite these advances, many important
The primary component of the apoptosome, Apaf-1, questions remain unanswered. Our understanding of
comprises an N-terminal CARD domain, a CED4 homol- how the initiator caspases are activated is far from com-
ogy domain, and 13 repeats of WD40 at its C-terminal plete. Although the functions of the BIR2 and BIR3 do-
half. The three-dimensional structure of the apoptosome mains are well understood in XIAP, c-IAP1 and c-IAP2,
at 27 Å resolution reveals a wheel-shaped heptameric it is unclear what role BIR1 plays in apoptosis and how
complex, with the CARD domains of Apaf-1 located at the different BIR domains cooperate in the full-length
the central hub and the WD40 repeats at the extended IAP to regulate cell death. In addition, it remains to be
spokes (Acehan et al., 2002) (Figure 7A). Docking of examined whether the principles derived from mamma-
caspase-9 to this apoptosome resulted in a dome- lian caspases are generally applicable to other species
shaped structure in the center; however, the bulk of the in which no structural information is yet available. Future
caspase-9 was not visible in these EM studies. Although structural and biochemical studies should clarify these
the concept of apoptosome had been biochemically es- issues. Furthermore, several of the known caspases
tablished, the visualization of the apoptosome allows function primarily in the inflammatory response rather
the identification of the relevant components and proves than in programmed cell death. Thus, activation of a
the structural involvement of Cyt. c in this functional caspase cascade does not always lead to apoptosis.
assembly. This structure should also remove remaining Elucidation of the pathways involving these caspases
doubts about the integrity of the apoptosome as a single will reveal important insights into how different cas-
structural entity. Nevertheless, because of the low reso- pases regulate distinct biological processes.
lution, the identified domain organization remains to be
validated. Acknowledgments
Although most other caspases exist exclusively as
I would like to thank members of my laboratory for discussion, C.
homodimers in solution, under physiological conditions,
Akey for help with the preparation of Figure 7, and F. Hughson
caspase-9 was found to exist in an equilibrium between for helpful comments. This research is supported by the National
predominantly monomers and a small amount of dimers Institutes of Health (R01-CA90269), the Searle Scholar Foundation,
(Renatus et al., 2001). Fractions corresponding to dimers and the Rita Allen Foundation.
and monomers from gel filtration were separately ana-
lyzed for their catalytic activity, and only the dimer frac- References
tions were found to be active. Interestingly, biochemical
Acehan, D., Jiang, X., Morgan, D.G., Heuser, J.E., Wang, X., and
as well as structural analyses revealed that dimerization
Akey, C.W. (2002). Three-dimensional structure of the apoptosome:
resulted in the formation of only one functional active implications for assembly, procaspase-9 binding, and activation.
site (Renatus et al., 2001) (Figure 7B). Based on these Mol. Cell 9, 423–432.
observations, it was proposed that dimer formation may Alnemri, E.S., Livingston, D.J., Nicholson, D.W., Salvesen, G.,
drive the activation of caspase-9. While these studies Thornberry, N.A., Wong, W.W., and Yuan, J. (1996). Human ICE/
provide solid leads on the mechanisms of caspase-9 CED-3 protease nomenclature. Cell 87, 171.
activation, they also raise many questions. For example, Altieri, D.C. (2001). Cytokenesis, apoptosis and survivin: three for
why didn’t the diluted dimers dissociate into the inactive tango? Cell Death Differential 8, 4–5.
monomers after gel filtration? And why couldn’t the far Ashhab, Y., Alian, A., Polliack, A., Panet, A., and Yehuda, D.B. (2001).
more concentrated monomers form active dimers? It Two splicing variants of a new inhibitor of apoptosis gene with
different biological properties and tissue distribution pattern. FEBS
also remains to be seen how the extremely hydrophobic
Lett. 495, 56–60.
dimerization interface is shielded from solvent in mono-
Ashkenazi, A., and Dixit, V.M. (1998). Death receptors: signaling and
meric caspase-9.
modulation. Science 281, 1305–1308.
Based on structural information on caspase-9 and
Blanchard, H., Kodandapani, L., Mittl, P.R.E., Di Marco, S., Krebs,
the apoptosome, a model was proposed to explain the J.K., Wu, J.C., Tomaselli, K.J., and Grutter, M.G. (1999). The three
activation of procaspase-9 (Acehan et al., 2002) (Figure dimersional structure of caspase-8: an initiator enzyme in apoptosis.
7C). In this model, a heptameric apoptosome binds Structure 7, 1125–1133.
seven monomers of inactive caspase-9. The high local Budihardjo, I., Oliver, H., Lutter, M., Luo, X., and Wang, X. (1999).
concentrations of caspase-9 within this apoptosome Biochemical pathways of caspase activation during apoptosis.
drive the efficient recruitment of additional inactive cas- Annu. Rev. Cell Dev. Biol. 15, 269–290.
pase-9 monomers, which become activated upon bind- Bump, N.J., Hackett, M., Hugunin, M., Seshagiri, S., Brady, K., Chen,
ing. This interesting model contains a strong assumption P., Ferenz, C., Franklin, S., Ghayur, T., and Li, P. (1995). Inhibition
that caspase-9 activity in the dimers is identical to that of ICE family proteases by baculovirus antiapoptotic protein p35.
Science 269, 1885–1888.
in the apoptosome holoenzyme, which remains to be
Chai, J., Du, C., Wu, J.-W., Kyin, S., Wang, X., and Shi, Y. (2000).
experimentally examined.
Structural and biochemical basis of apoptotic activation by Smac/
DIABLO. Nature 406, 855–862.
Future Perspectives Chai, J., Shiozaki, E., Srinivasula, S.M., Alnemri, E.S., and Shi, Y.
Recent investigation on caspase regulation has led to (2001a). Crystal structure of a procaspase-7 zymogen: mechanisms
a significantly improved understanding of the activation of activation and substrate binding. Cell 107, 399–407.
Review
469

Chai, J., Shiozaki, E., Srinivasula, S.M., Wu, Q., Datta, P., Alnemri, Alnemri, E.S., and Wamg, X. (1997). Cytochrome c and dATP-depen-
E.S., and Shi, Y. (2001b). Structural basis of caspase-7 inhibiton by dent formation of Apaf-1/caspase-9 complex initiates an apoptotic
XIAP. Cell 104, 769–780. protease cascade. Cell 91, 479–489.
Christich, A., Kauppila, S., Chen, P., Sogame, N., and Abrams, J.M. Li, F., Ambrosini, G., Chu, E.Y., Plescia, J., Tognin, S., Marchisio,
(2002). The damage-responsive Drosophila gene Sickle encodes a P.C., and Altieri, D.C. (1998). Control of apoptosis and mitotic spindle
novel IAP binding protein similar to but distinct from Reaper, Grim checkpoint by survivin. Nature 396, 580–584.
and Hid. Curr. Biol. 12, 137–140. Liu, X., Zou, H., Slaughter, C., and Wang, X. (1997). DFF, a heterodi-
Dangl, J.L., and Jones, J.D.G. (2001). Plant pathogens and inte- meric protein that functions downstream of caspase-3 to trigger
grated defense responses to infection. Nature 411, 826–833. DNA fragmentation during apoptosis. Cell 89, 175–184.
Deveraux, Q.L., and Reed, J.C. (1999). IAP family proteins— Liu, X., Li, P., Widlak, P., Zou, H., Luo, X., Garrard, W.T., and Wang,
suppressors of apoptosis. Genes Dev. 13, 239–252. X. (1998). The 40-kDa subunit of DNA fragmentation factor induces
Du, C., Fang, M., Li, Y., and Wang, X. (2000). Smac, a mitochondrial DNA fragmentation and chromatin condensation during apoptosis.
protein that promotes cytochrome c-dependent caspase activation Proc. Natl. Acad. Sci. USA 95, 8461–8466.
during apoptosis. Cell 102, 33–42. Liu, Z., Sun, C., Olejniczak, E.T., Meadows, R.P., Betz, S.F., Oost,
Earnshaw, W.C., Martins, L.M., and Kaufmann, S.H. (1999). Mamma- T., Herrmann, J., Wu, J.C., and Fesik, S.W. (2000). Structural basis
lian caspases: structure, activation, substrates, and functions during for binding of Smac/DIABLO to the XIAP BIR3 domain. Nature 408,
apoptosis. Annu. Rev. Biochem. 68, 383–424. 1004–1008.
Enari, M., Sakahira, H., Yokoyama, H., Okawa, K., Iwamatsu, A., and Martin, L.M., Iaccarino, I., Tenev, T., Gschmeissner, S., Totty, N.F.,
Nagata, S. (1998). A caspase-activated DNase that degrades DNA Lemoine, N.R., Savopoulos, J., Gray, C.W., Creasy, C.L., Dingwall,
during apoptosis, and its inhibitor ICAD. Nature 391, 43–50. C., et al. (2002). The serine protease Omi/HtrA2 regulates apoptosis
Fesik, S.W. (2000). Insights into programmed cell death through by binding XIAP through a Reaper-like motif. J. Biol. Chem. 277,
structural biology. Cell 103, 273–282. 439–444.
Fesik, S.W., and Shi, Y. (2001). Controlling caspases. Science 294, Miller, L.K. (1999). An exegesis of IAPs:salvation and surprises from
1477–1478. BIR motifs. Trends Cell Biol. 9, 323–328.
Gray, C.W., Ward, R.V., Karran, E., Turconi, S., Rowles, A., Viglienghi, Mittl, P.R., Di Marco, S., Krebs, J.F., Bai, X., Karanewsky, D.S.,
D., Southan, C., Barton, A., Fantom, K.G., West, A., et al. (2000). Priestle, J.P., Tomaselli, K.J., and Grutter, M.G. (1997). Structure of
Characterization of human HtrA2, a novel serine protease involved recombinant human CPP32 in complex with the tetrapeptide acetyl-
in the mammalian cellular stress response. Eur. J. Biochem. 267, Asp-Val-Ala-Asp fluoromethyl ketone. J. Biol. Chem. 272, 6539–
5699–5710. 6547.
Hanahan, D., and Weinberg, R.A. (2000). The hallmarks of cancer. Muzio, M., Stockwell, B.R., Stennicke, H.R., Salvesen, G.S., and
Cell 100, 57–70. Dixit, V.M. (1998). An induced proximity model for caspase-8 activa-
Harris, B.Z., and Lim, W.A. (2001). Mechanism and role of PDZ tion. J. Biol. Chem. 273, 2926–2930.
domains in signaling complex assembly. J. Cell Sci. 114, 3219–3231. Nicholls, A., Sharp, K.A., and Honig, B. (1991). Protein folding and
Hawkins, C.J., Yoo, S.J., Peterson, E.P., Wang, S.L., Vernooy, S.Y., association: insights from the interfacial and thermodynamic prop-
and Hay, B.A. (2000). The Drosophila caspase DRONC cleaves fol- erties of hydrocarbons. Proteins: Struct. Funct. Genet. 11, 281–296.
lowing glutamate or aspartate and is regulated by DIAP1, HID, and Ogura, Y., Inohara, N., Benito, A., Chen, F.F., Yamaoka, S., and
GRIM. J. Biol. Chem. 275, 27084–27093. Nunez, G. (2001). Nod2, a Nod1/Apaf-1 family member that is re-
Hay, B.A. (2000). Understanding IAP function and regulation: a view stricted to monocytes and activates NF-kB. J. Biol. Chem. 276,
from Drosophila. Cell Death Differential 7, 1045–1056. 4812–4818.
Hedge, R., Srinivasula, S.M., Wassell, R., Mukattash, R., Cilenti, L., Poyet, J.-L., Srinivasula, S.M., Tnani, M., Razmara, M., Fernandes-
Zhang, Z., DuBois, G., Lazebnik, Y., Zervos, A.S., Fernandes- Alnemri, T., and Alnemri, E.S. (2001). Identification of Ipaf, a human
Alnemri, T., et al. (2002). Identification of Omi/HtrA2 as a mitochon- caspase-1-activating protein related to Apaf-1. J. Biol. Chem. 276,
drial apoptotic serine protease that disrupts IAP-caspase interac- 28309–28313.
tion. J. Biol. Chem. 277, 432–438. Renatus, M., Stennicke, H.R., Scott, F.L., Liddington, R.C., and
Horvitz, H.R. (1999). Genetic control of programmed cell death in Salvesen, G.S. (2001). Dimer formation drives the activation of the
the nematode Caenorhabditis elegans. Cancer Res. 59, 1701–1706. cell death protease caspase 9. Proc. Natl. Acad. Sci. USA 98, 14250–
Huang, H.-K., Joazeiro, C.A.P., Bonfoco, E., Kamada, S., Leverson, 14255.
J.D., and Hunter, T. (2000). The inhibitor of apoptosis, cIAP2, func- Riedl, S.J., Fuentes-Prior, P., Renatus, M., Kairies, N., Krapp, S.,
tions as a ubiquitin-protein ligase and promotes in vitro monoubiqui- Huber, R., Savesen, G.S., and Bode, W. (2001a). Structural basis for
tination of caspase 3 and 7. J. Biol. Chem. 275, 26661–26664. the activation of human procaspase-7. Proc. Natl. Acad. Sci. USA
Huang, Y., Park, Y.C., Rich, R.L., Segal, D., Myszka, D.G., and Wu, 98, 14790–14795.
H. (2001). Structural basis of caspase inhibition by XIAP: differential Riedl, S.J., Renatus, M., Schwarzenbacher, R., Zhou, Q., Sun, C.,
roles of the Linker versus the BIR domain. Cell 104, 781–790. Fesik, S.W., Liddington, R.C., and Salvesen, G.S. (2001b). Structural
Inohara, N., Koseki, T., del Peso, L., Hu, Y., Yee, C., Chen, S., Carrio, basis for the inhibition of caspase-3 by XIAP. Cell 104, 791–800.
R., Merino, J., Liu, D., Ni, J., et al. (1999). Nod1, an Apaf-1-like Rodriguez, J., and Lazebnik, Y. (1999). Caspase-9 and Apaf-1 form
activator of caspase-9 and nuclear factor-kB. J. Biol. Chem. 274, an active holoenzyme. Genes Dev. 13, 3179–3184.
14560–14567. Rotonda, J., Nicholson, D.W., Fazil, K.M., Gallant, M., Gareau, Y.,
Inohara, N., and Nunez, G. (2001). The NOD: a signaling module that Labelle, M., Peterson, E.P., Rasper, D.M., Ruel, R., Vaillancourt, J.P.,
regulates apoptosis and host defense against pathogens. Oncogene et al. (1996). The three-dimensional structure of apopain/CPP32, a
20, 6473–6481. key mediator of apoptosis. Nat. Struct. Biol. 3, 619–625.
Jacobson, M.D., Weil, M., and Raff, M.C. (1997). Programmed cell Saleh, A., Srinivasula, S.M., Acharya, S., Fishel, R., and Alnemri, E.S.
death in animal development. Cell 88, 347–354. (1999). Cytochrome c and dATP-mediated oligomerization of Apaf-1
Kasof, G.M., and Gomes, B.C. (2001). Livin, a novel inhibitor of is a prerequisite for procaspase-9 activation. J. Biol. Chem. 274,
apoptosis protein family member. J. Biol. Chem. 276, 3238–3246. 17941–17945.
Klaulis, P.J. (1991). Molscript: a program to produce both detailed Salvesen, G.S., and Dixit, V.M. (1997). Caspases: intracellular signal-
and schematic plots of protein structures. J. Appl. Crystallogr. 24, ing by proteolysis. Cell 91, 443–446.
946–950. Salvesen, G.S., and Dixit, V.M. (1999). Caspase activation: the in-
Li, P., Nijhawan, D., Budihardjo, I., Srinivasula, S.M., Ahmad, M., duced-proximity model. Proc. Natl. Acad. Sci. USA 96, 10964–10967.
Molecular Cell
470

Shi, Y. (2001). A structural view of mitochondria-mediated apoptosis. (2002). HtrA2 promotes cell death through its serine protease activity
Nat. Struct. Biol. 8, 394–401. and its ability to antagonize inhibitor of apoptosis proteins. J. Biol.
Soengas, M.S., Capodieci, P., Polsky, D., Mora, J., Esteller, M., Chem. 277, 445–454.
Opitz-Araya, X., McCombie, R., Herman, J.G., Gerald, W.L., Lazeb- Vucic, D., Stennicke, H.R., Pisabarro, M.T., Salvesen, G.S., and Dixit,
nik, Y.A., et al. (2001). Inactivation of the apoptosis effector Apaf-1 V.M. (2000). ML-IAP, a novel inhibitor of apoptosis that is preferen-
in malignant melanoma. Nature 409, 207–211. tially expressed in human melanomas. Curr. Biol. 10, 1359–1366.
Srinivasula, S.M., Ahmad, M., Fernandes-Alnemri, T., and Alnemri, Walker, N.P., Talanian, R.V., Brady, K.D., Dang, L.C., Bump, N.J.,
E.S. (1998a). Autoactivation of procaspase-9 by Apaf-1-mediated Ferenz, C.R., Franklin, S., Ghayur, T., Hackett, M.C., and Hammill,
oligomerization. Mol. Cell 1, 949–957. L.D. (1994). Crystal structure of the cysteine protease interleukin-
Srinivasula, S.M., Ahmad, M., MacFarlane, M., Luo, Z., Huang, Z., 1b-converting enzyme: a (p20/p10)2 homodimer. Cell 78, 343–352.
Fernandes-Alnemri, T., and Alnemri, E.S. (1998b). Generation of con- Wang, S., Hawkins, C., Yoo, S., Muller, H.-A., and Hay, B. (1999).
stitutively active recombinant caspase-3 and -6 by rearrangement The Drosophila caspase inhibitor DIAP1 is essential for cell survival
of their subunits. J. Biol. Chem. 273, 10107–10111. and is negatively regulated by HID. Cell 98, 453–463.
Srinivasula, S.M., Datta, P., Fan, X.J., Fernandes-Alnemri, T., Huang, Watt, W., Koeplinger, K.A., Mildner, A.M., Heinrikson, R.L., Tomas-
Z., and Alnemri, E.S. (2000). Molecular determinants of the caspase- selli, A.G., and Watenpaugh, K.D. (1999). The atomic-resolution
promoting activity of Smac/DIABLO and its role in the death receptor structure of human caspase-8, a key activator of apoptosis. Struc-
pathway. J. Biol. Chem. 275, 36152–36157. ture 7, 1135–1143.
Srinivasula, S.M., Saleh, A., Hedge, R., Datta, P., Shiozaki, E., Chai, Wei, Y., Fox, T., Chambers, S.P., Sintchak, J.-A., Coll, J.T., Golec,
J., Robbins, P.D., Fernandes-Alnemri, T., Shi, Y., and Alnemri, E.S. J.M.C., Swenson, L., Wilson, K.P., and Charifson, P.S. (2000). The
(2001). A conserved XIAP-interaction motif in caspase-9 and Smac/ structures of caspases-1, -3, -7 and -8 reveal the basis for substrate
DIABLO mediates opposing effects on caspase activity and apopto- and inhibitor selectivity. Chem. Biol. 7, 423–432.
sis. Nature 409, 112–116. Wilson, K.P., Black, J.-A., Thomson, J.A., Kim, E.E., Griffith, J.P.,
Srinivasula, S.M., Datta, P., Kobayashi, M., Fujioka, M., Wu, J.-W., Navia, M.A., Murcko, M.A., Chambers, S.P., Aldape, R.A., Raybuck,
Hedge, R., Zhang, Z., Mukattash, R., Fernandes-Alnemri, T., Shi, Y., S.A., and Livingston, D.J. (1994). Structure and mechanism of in-
et al. (2002). Sickle, a novel Drosophila death gene in the reaper/ terleukin-1b converting enzyme. Nature 370, 270–275.
hid/grim region encodes an IAP-inhibitory protein. Curr. Biol. 12, Wing, J.P., Karres, J., Ogdahl, J.L., Zhou, L., Schwartz, L.M., and
125–130. Nambu, J.R. (2002). Drosophila sickle is a novel grim-reaper cell
Stennicke, H.R., Deveraux, Q.L., Humke, E.W., Reed, J.C., Dixit, death activator. Curr. Biol. 12, 131–135.
V.M., and Salvesen, G.S. (1999). Caspase-9 can be activated without Wu, G., Chai, J., Suber, T.L., Wu, J.-W., Du, C., Wang, X., and Shi, Y.
proteolytic processing. J. Biol. Chem. 274, 8359–8362. (2000). Structural basis of IAP recognition by Smac/DIABLO. Nature
408, 1008–1012.
Sun, C., Cai, M., Gunasekera, A.H., Meadows, R.P., Wang, H., Chen,
J., Zhang, H., Wu, W., Xu, N., Ng, S.-C., et al. (1999). NMR structure Wu, J.-W., Cocina, A.E., Chai, J., Hay, B.A., and Shi, Y. (2001).
and mutagenesis of the inhibitor-of-apoptosis protein XIAP. Nature Structural analysis of a functional DIAP1 fragment bound to Grim
401, 818–822. and Hid peptides. Mol. Cell 8, 95–104.
Sun, C., Cai, M., Meadows, R.P., Xu, N., Gunasekera, A.H., Herr- Xu, G., Cirilli, M., Huang, Y., Rich, R.L., Myszka, D.G., and Wu, H.
mann, J., Wu, J.C., and Fesik, S.W. (2000). NMR structure and muta- (2001). Covalent inhibition revealed by the crystal structure of the
genesis of the third BIR domain of the inhibitor of apoptosis protein caspase-8/p35 complex. Nature 410, 494–497.
XIAP. J. Biol. Chem. 275, 33777–33781. Yang, X., Chang, H.Y., and Baltimore, D. (1998a). Autoproteolytic
Suzuki, Y., Imai, Y., Nakayama, H., Takahashi, K., Takio, K., and activation of pro-caspases by oligomerization. Mol. Cell 1, 319–325.
Takahashi, R. (2001a). A serine protease, HtrA2, is released from Yang, X., Chang, H.Y., and Baltimore, D. (1998b). Essential role of
the mitochondria and interacts with XIAP, inducing cell death. Mol. CED-4 oligomerization in CED-3 activation and apoptosis. Science
Cell 8, 613–621. 281, 1355–1357.
Suzuki, Y., Nakabayashi, Y., and Takahashi, R. (2001b). Ubiquitin- Yuan, J., Shaham, S., Ledoux, S., Ellis, H.M., and Horvitz, H.R. (1993).
protein ligase activity of X-linked inhibitor of apoptosis protein pro- The C. elegans cell death gene Ced-3 encodes a protein similar to
motes proteasomal degradation of caspase-3 and enhances its anti- mammalian interleukin-1 beta-converting enzyme. Cell 75, 641–652.
apoptotic effect in Fas-induced cell death. Proc. Natl. Acad. Sci. Yuan, J., and Yankner, B.A. (2000). Apoptosis in the nervous system.
USA 98, 8662–8667. Nature 407, 802–809.
Thompson, C.B. (1995). Apoptosis in the pathogenesis and treat- Zhou, Q., Krebs, J.F., Snipas, S.J., Price, A., Alnemri, E.S., Tomaselli,
ment of disease. Science 267, 1456–1462. K.J., and Salvesen, G.S. (1998). Interaction of the baculovirus anti-
Thornberry, N.A., and Lazebnik, Y. (1998). Caspases: enemies within. apoptotic protein p35 with caspases. Specificity, kinetics, and char-
Science 281, 1312–1316. acterization of the caspase/p35 complex. Biochemistry 37, 10757–
10765.
Thornberry, N.A., Rano, T.A., Peterson, E.P., Rasper, D.M., Timkey,
T., Garcia-Calvo, M., Houtzager, V.M., Nordstrom, P.A., Roy, S., Zou, H., Li, Y., Liu, X., and Wang, X. (1999). An APAF-1-cytochrome
Vaillancourt, J.P., et al. (1997). A combinatorial approach defines c multimeric complex is a functional apoptosome that activates
specificities of members of the caspase family and granzyme B. procaspase-9. J. Biol. Chem. 274, 11549–11556.
Functional relationships established for key mediators of apoptosis.
J. Biol. Chem. 272, 17907–17911.
van Loo, G., van Gurp, M., Depuydt, B., Srinivasula, S.M., Rodriguez,
I., Alnemri, E.S., Gevaert, K., Vandekerckhove, J., Declercq, W., and
Vandenabeele, P. (2002). The serine protease Omi/HtrA2 is released
from mitochondria during apoptosis. Omi interacts with caspase-
inhibitor XIAP and induces enhanced caspase activity. Cell Death
Differential 9, 20–26.
Verhagen, A.M., Ekert, P.G., Pakusch, M., Silke, J., Connolly, L.M.,
Reid, G.E., Moritz, R.L., Simpson, R.J., and Vaux, D.L. (2000). Identifi-
cation of DIABLO, a mammalian protein that promotes apoptosis
by binding to and antagonizing IAP proteins. Cell 102, 43–53.
Verhagen, A.M., Silke, J., Ekert, P.G., Pakusch, M., Kaufmann, H.,
Connolly, L.M., Day, C.L., Tikoo, A., Burke, R., Wrobel, C., et al.

You might also like