Vous êtes sur la page 1sur 19

Apoptosis (2010) 15:331349

DOI 10.1007/s10495-009-0432-9

UNUSUAL MODEL SYSTEMS FOR CELL DEATH RESEARCH

The zebrafish as a model organism for the study of apoptosis


Peter M. Eimon Avi Ashkenazi

Published online: 24 December 2009


Springer Science+Business Media, LLC 2009

Abstract Apoptosis plays important roles in embryo- Introduction


genesis, tissue homeostasis, and immune system regula-
tion. The zebrafish (Danio rerio) is a powerful vertebrate There are two key apoptotic signaling mechanisms in
model organism that has been extensively used to study mammals and other vertebrates: the cell-intrinsic (or
apoptotic cell death during normal development and under mitochondrial) pathway and the cell-extrinsic (or death
conditions of cellular stress. In the past 5 years, a detailed receptor) pathway. The intrinsic pathway can be triggered
picture has begun to emerge of the molecular underpin- by a variety of cues including cellular damage, growth
nings of the cell-intrinsic and the cell-extrinsic apoptosis factor withdrawal, and chemotherapeutic drugs. It is reg-
signaling pathways in zebrafish. We begin this review with ulated by the Bcl-2 gene family and functions through the
an introduction to the techniques and experimental initiator protease caspase-9 [1]. The extrinsic pathway
approaches that are used to study apoptosis in zebrafish. plays important roles in immune system operation and
We follow with a general overview of developmental homeostasis. It is triggered by death receptor ligation and
apoptosis during zebrafish embryogenesis. Finally, we functions through a death-inducing signaling complex
present a comprehensive review of the intrinsic and (DISC) that includes the initiator proteases caspase-8 and
extrinsic apoptosis pathways in zebrafish, focusing on the -10 [2]. The extrinsic pathway is linked to the intrinsic
high degree of conservation with humans and other mam- pathway through Bid, a Bcl-2 family protein that can be
mals. Recent publications that draw upon the unique cleaved and activated by caspase-8. Additional pathways,
advantages of the zebrafish system to study novel aspects such as the granzyme B and endoplasmic reticulum path-
of apoptosis regulation and function are highlighted ways, may also mediate aspects of apoptosis under certain
throughout. conditions. All pathways converge at the level of the
effector caspases (caspase-3, -6 and -7), which carry out
Keywords Apoptosis  Zebrafish  Intrinsic pathway  the molecular mechanics of programmed cell death [3].
Extrinsic pathway  Bcl-2  IAP  Death receptor  The intrinsic apoptosis pathway appears to have evolved
Apo2L/TRAIL  Caspase around the same time as multicellular organisms, and Bcl-2
family orthologs have been identified in all metazoans
analyzed to date [1]. In contrast, the extrinsic pathway is a
more recent evolutionary development. No tumor necrosis
factor (TNF) or TNF receptor (TNFR) superfamily mem-
bers have been found in Caenorhabditis elegans (C. ele-
P. M. Eimon gans). Death receptors (TNFRs possessing a cytoplasmic
Zygogen, LLC, Atlanta, GA, USA death domain motif) have been reported almost exclusively
in vertebrates, although recently a death domain-containing
A. Ashkenazi (&)
TNFR has been identified in the Zhikong scallop (Chlamys
Department of Molecular Oncology, Genentech Inc.,
1 DNA Way, MS 42, South San Francisco, CA 94080, USA farreri) [4]. In the following review we will summarize the
e-mail: aa@gene.com current understanding of the intrinsic and extrinsic

123
332 Apoptosis (2010) 15:331349

apoptotic pathways in zebrafish. We will highlight simi- (http://www.ensembl.org/Danio_rerio/Info/Index). More com-


larities and differences between these pathways in zebra- plete manual annotation is being carried out in collabora-
fish, mammals, and more divergent model organisms such tion with ZFIN and can be accessed through the Vega
as fruit flies and nematodes. In order to promote consis- database (http://vega.sanger.ac.uk/Danio_rerio/index.html).
tency in zebrafish nomenclature, we have used the official As a result of these and other approaches, an exceptionally
gene names and symbols endorsed by Entrez and the large number of genes and molecular pathways have been
Zebrafish Information Network (ZFIN) throughout this well characterized in zebrafish and help to validate this
review. In some cases, official zebrafish gene names are model organism as highly relevant for the study of verte-
paired with their more familiar human designation (e.g. brate biology.
tnfrsf21/dr6). LocusLink (LOC) designations are used for Several experimental protocols have been developed
zebrafish genes without an official full name. that allow apoptotic cells to be visualized in vivo in zeb-
rafish embryos. One of the most commonly used tech-
niques utilizes the vital dye acridine orange (AO), a nucleic
Approaches to studying apoptosis in zebrafish acid intercalating dye that emits green fluorescence when
bound to dsDNA and selectively stains apoptotic rather
The zebrafish is a small freshwater teleost of the cyprinid than necrotic cells [17, 18]. Analysis of AO staining in
family (carps and minnows) in the class of ray-finned fishes zebrafish embryos is complicated by autofluorescence or
(Actinopterygii). Zebrafish have served as an important non-specific uptake in the yolk, iridophores, hatching
research organism for decades, particularly in the fields of gland, and neuromasts [19]. A recent publication describes
genetics and developmental biology [5]. A unique combi- an imaging protocol that enables rapid quantification of AO
nation of features makes them particularly attractive as a staining in large groups of embryos [20]. Approaches such
vertebrate model. These include their small size (embryos as this are crucial, since AO staining exhibits considerable
are *0.7 mm in diameter at fertilization and grow to embryo-to-embryo variability and large numbers of
3.5 mm within 3 days), high fecundity (a single zebrafish embryos must be analyzed to ensure statistical rigor.
pair can produce hundreds of embryos per week), short Hoechst 33342, a DNA-binding vital dye, has also been
generation time (adults reach sexual maturity used in live zebrafish embryos to visualize the formation of
in *3 months), and genetic tractability. Embryos are fer- chromatin clumps characteristic of apoptotic nuclei [21].
tilized externally, undergo rapid and synchronous devel- Finally, a transgenic zebrafish line expressing mitochond-
opment, and are optically transparent. Most organs become rially-targeted green fluorescent protein (GFP) has been
functional between 3 and 5 days post fertilization (dpf) [6]. developed to monitor mitochondrial fragmentation in
The popularity of zebrafish increased substantially fol- apoptotic cells in vivo [22].
lowing several publications in 1996 demonstrating that Many assays used to detect apoptosis in mammalian
they are amenable to large-scale forward genetic screens tissue samples and cell culture have been adapted to fixed
and describing over 1000 mutants with visible phenotypes zebrafish embryos and embryonic lysates. Fixed embryos
during early development [7, 8]. Previously, large-scale can be stained by the terminal dUTP nick end-labeling
screening approaches had been limited to invertebrates (TUNEL) method, which detects fragmented DNA char-
such as C. elegans and Drosophila melanogaster. acteristic of apoptotic cells [18, 23]. According to some
Many additional tools further enhance the utility of reports, use of the TUNEL method on zebrafish embryos
zebrafish as a model organism. Reverse genetic approaches older than 48 h post fertilization (hpf) is hampered by
include using antisense morpholino oligonucleotides to unacceptable background staining and nonspecific labeling
knock down gene function transiently in embryos [9], tar- of periderm cells [19]. Fixed embryos can also be immu-
geting induced local lesions in genomes (TILLING) to stained using a monoclonal antibody that recognizes the
identify stable mutations in genes of interest [10], and zinc- cleaved, active form of caspase-3 [24]. Fluorogenic and
finger nucleases to generate locus-specific mutations for bioluminescent substrates have been used to monitor the
targeted gene inactivation [11]. Techniques have been activity of caspase-3, caspase-8, and caspase-9 in zebrafish
devised for generating stable transgenic lines that express embryo lysates and in dissociated cells [21, 25, 26].
fluorescent reporter proteins or other genes of interest Negron and Lockshin [21] have used a number of
under the control of tissue-specific promoters [1214]. additional techniques to characterize the biochemical
Commercial zebrafish DNA microarrays are available for modifications associated with apoptotic cell death in zeb-
expression profiling, and protocols have been developed rafish embryos and report broad conservation of apoptotic
for proteomic and metabolomic analysis [15, 16]. The hallmarks between fish and mammals. Extensive intranu-
Sanger Institute has sequenced the zebrafish genome cleosomal DNA fragmentation was confirmed by agarose
and an annotated assembly is available in Ensembl gel electrophoresis and poly(ADP-ribose) polymerase

123
Apoptosis (2010) 15:331349 333

(PARP) cleavage was detected by Western blot. Apoptotic the apoptotic machinery in pre-gastrula stage embryos has
cells translocated phosphatidylserinea component of the been documented in Xenopus laevis [37]. In contrast,
apoptotic cell-surface eat-me signalto the outer leaflet apoptotic cell death has been reported in mammalian
of the plasma membrane. Transmission electron micros- embryos as early as the 16-cell stage [34, 38].
copy was used to confirm that apoptotic cells in zebrafish Following gastrulation, apoptotic cells are detected in
exhibit chromatin condensation and convolution of the various organs and tissues of zebrafish embryos [19]. Cole
nuclear membrane. Engulfment of apoptotic cell corpses by and Ross [39] have used the TUNEL method to map
neighboring cells has also been observed in zebrafish developmental apoptosis in zebrafish between gastrulation
embryos using electron microscopy; engulfment can be and 4 dpf. The authors describe transient high levels of
inhibited by knockdown of the phosphatidylserine receptor developmental apoptosis in a number of structures
jmjd6 [27]. In zebrafish, like mammals, apoptosis and including the olfactory placodes (2272 hpf), eyes (peak-
necrosis share a number of common features and it is ing at 36 hpf in the retina and 2048 hpf in the lens), lat-
important to use two or more distinct assays to confirm that eral line neuromasts (beginning at 2430 hpf), Rohon-
cell death is truly apoptotic. Beard neurons (peaking between 3648 hpf), trigeminal
Many experimental methods for inducing apoptosis in neurons (peaking between 3648 hpf), urogenital system
mammalian cells are also effective in zebrafish. Chemical (peaking between 3036 hpf), and tail bud (peaking at
reagents that have been evaluated include aphidicolin [28], 29 hpf). Similar patterns of developmental apoptosis have
camptothecin [28, 29], cycloheximide [21], hydroxyurea been observed in other fish species such as medaka
[28], and staurosporine [22]. Apoptotic cell death has also (Oryzias latipes) [40].
been demonstrated in response to heat shock [30], ultra- Developmental apoptosis has been studied in detail in
violet irradiation [29], and c-irradiation [3032]. Similar to several zebrafish neuronal populations. Cell death is
mammals, stress-induced apoptosis in zebrafish can be widespread in the central and peripheral nervous systems
blocked by the pan-caspase inhibitor Z-VAD-FMK and by of most vertebrates during development. In many regions,
the cytoprotective agent amifostine [26]. more than 50% of newly formed neurons are eliminated
[41]. In motoneurons, one well-characterized period of
apoptosis occurs when axons contact their postsynaptic
Developmental apoptosis in zebrafish targets. Competition between neurons for limiting amounts
of muscle-derived neurotrophic factors is thought to be a
In multicellular organisms, normal development depends major driver of apoptosis in this context [42]. Eisen and
upon a carefully orchestrated balance between cell prolif- Melancon [43] have used zebrafish embryos to study the
eration, differentiation, and death. Apoptosis is the most regulation of motoneuron apoptosis in greater detail. In
common mechanism employed during development to adult zebrafish, each ventral myotome is innervated by a
remove damaged, misplaced, or otherwise unwanted cells single motoneuron designated CaP. During early develop-
[33, 34]. Additional forms of programmed cell death ment, however, a second motoneuron (designated VaP) is
particularly autophagy (or type II cell death)are utilized often present alongside CaP in the spinal cord. Both CaP
in certain developmental contexts but fall outside the scope and VaP are morphologically equivalent and send out
of this review. Although over-production of cells that will axons that pause briefly at an intermediate target known as
ultimately be eliminated by apoptosis may appear ener- the horizontal myoseptum (HM). The cell whose axon first
getically wasteful, it is a common feature of metazoan reaches the HM adopts the CaP fate and goes on to
development and occurs in species ranging from nema- innervate the ventral myotome. The remaining cell adopts
todes to humans [34, 35]. the VaP fate, its axon remains paused at the HM, and it
Zebrafish embryos are resistant to apoptosis before they undergoes apoptotic death by 36 hpf. Through a series of
reach the maternal-to-zygotic transition (MZT, occurring cell transplant and cell ablation studies, the authors show
between 4 and 7 hpf), the stage during early development that VaP motoneuron death is governed by a death-pro-
when the zygotic genome becomes the primary source of moting signal originating from HM cells and triggered by
mRNA in the embryo [21, 28, 36]. Embryos treated with the CaP axon [43, 44]. Developmental apoptosis is also
camptothecin, cycloheximide, nocodazole, or staurosporine widespread among neurons of the mammalian retina,
prior to the end of MZT exhibit an arrest of cell prolifer- where two consecutive phases of apoptosis have been
ation but survive until the mid-gastrula stage (*7 hpf), at described. The earlier phase occurs during neurogenesis
which point they undergo rapid and synchronous cell death. and cell migration, while the later phase occurs during
Acquisition of apoptotic competence appears to be under target innervation [45, 46]. A similar pattern of apoptosis is
post-translational control, as it is not impacted by contin- seen in the developing zebrafish retina, with an initial
uous treatment with cycloheximide. A similar inhibition of phase peaking at 34 dpf and a second phase at 67 dpf

123
334 Apoptosis (2010) 15:331349

[47]. The first phase is restricted to the ganglion cell layer cell death in the central nervous system (CNS; for exam-
(GCL) and the inner nuclear layer (INL), while the second ples, see Abdelilah et al. [58], Furutani-Seiki et al. [18],
phase encompasses cells of outer nuclear layer (ONL) in Fadool et al. [59], and Rodriguez and Driever [19]).
addition to the GCL and INL. Although the onset and Mutations that affect cell death in many other organs and
progression of retinal apoptosis in zebrafish is reminiscent tissues have also been described (reviewed in Pyati et al.
of mammals, the total number of apoptotic cells detected in [60]). It is probable that aberrant cell death in most of these
zebrafish is significantly less. mutants does not stem from defects in the core apoptotic
Developmental apoptosis in non-neuronal cell types has machinery, but rather serves to eliminate cells that are
also been characterized in zebrafish. Elevated levels of unhealthy, misplaced, or otherwise deficient due to dis-
apoptotic cells are detected in the zebrafish lens around ruptions in the tightly regulated process of embryogenesis.
24 hpf as the developing lens is detaching from the surface In addition to forward genetic screens, knockdown exper-
ectoderm. It has been suggested that apoptosis aids in iments have suggested that many zebrafish genes have
separation of the lens from the ectoderm [48], similar to a loss-of-function phenotypes characterized by excessive
mechanism reported during development of the rat lens apoptotic cell death (reviewed in Pyati et al. [60]).
vesicle [49]. Two more recent studies have failed to con- Unfortunately, interpretation of these results is greatly
firm that apoptosis is a significant factor in separation of complicated by recent findings that morpholino antisense
the zebrafish lens from the surface ectoderm [50, 51]. oligonucleotides and other knockdown technologies often
Apoptosis may instead be used to clear undifferentiated or induce nonspecific off-target apoptosis in zebrafish [61].
lens-foreign cells that could interfere with subsequent Off-target apoptosis is most pronounced in the CNS and
development [48, 51]. TUNEL-positive cells in the lens appears to be mediated through tp53 activation. For a more
may also arise from programmed removal of nuclei and detailed discussion of mutants and morpholino knockdown
other organelles from lens fiber cells, a process that shares embryos in which developmental cell death is perturbed,
many hallmarks of apoptosis including chromatin frag- readers are encouraged to consult Pyati et al. [60]. The
mentation [52]. remainder of this review will focus specifically on the
In some cases, developmental apoptosis underlies pro- molecular mechanism governing the intrinsic and extrinsic
cesses that have not been evolutionarily conserved between apoptosis pathways in zebrafish.
zebrafish and other vertebrates. Rohon-Beard (RB) cells
are an interesting example of this phenomenon. These
primary sensory neurons are found in anamniote verte- The cell-intrinsic apoptosis pathway
brates such as teleosts and amphibians but are not present
in birds or mammals. RB cells exist only transiently during The zebrafish Bcl-2 family
early development and their sensory functions are soon
replaced by neurons of the dorsal root ganglia. In zebrafish, The Bcl-2 gene family serves as the chief regulator of
RB cells are eliminated during the first 4 days of devel- the intrinsic apoptosis pathway in mammals [6264]. The
opment in a caspase-dependent manner. Apoptosis appears Bcl-2 family can be divided into three subfamilies based
to be driven in part by loss of the neurotrophic factor ntf3 on apoptotic function and the presence of up to four distinct
and may also be regulated by voltage-gated Na? current a-helical motifs known as Bcl-2 homology (BH) domains
[5355]. The inner ear is another region where the role of (designated BH1, BH2, BH3, and BH4). Anti-apoptotic
developmental apoptosis has not been conserved through- family members (e.g. Bcl-2, Bcl-xL, Bcl-w, and Mcl-1)
out vertebrate evolution. In both birds and mammals an possess all four BH domains. Pro-apoptotic family mem-
apoptotic hot spot has been characterized in the ven- bers are categorized as multidomain proteins (containing
tromedial wall of the developing inner ear adjacent to the BH1, BH2, and BH3 domains; e.g. Bax, Bak, and Bok) and
auditory ganglion [56]. A thorough examination of apop- BH3-only proteins (e.g. Bid, Bad, Bik, Bim, Bmf, Noxa,
totic cells in the developing ears of both zebrafish and Puma, Hrk, and the Bnip proteins). Members of the BH3-
Xenopus laevis failed to detect a similar hot spot, although only subfamily serve as apical sensors for various apoptotic
apoptosis was observed in the statoacoustic ganglion in stimuli and are capable of promoting apoptosis through
agreement with previous reports in birds and mammals activation of the multidomain pro-apoptotic subfamily and/
[57]. or suppression of the anti-apoptotic subfamily. Interactions
Large-scale mutagenesis screens in zebrafish have between subfamily members occur when the BH3 domains
identified numerous genes with loss-of-function pheno- on pro-apoptotic proteins bind to a hydrophobic groove
types that affect developmental apoptosis. By far the formed by the BH1, BH2, and BH3 domains on anti-
largest class of these mutants is associated with elevated apoptotic proteins [65].

123
Apoptosis (2010) 15:331349 335

Table 1 Apoptosis associated genes in zebrafish


Gene family Mammalian gene Zebrafish Other names Reference Zebrafish Synteny
homologa sequence chromosome w/human

TNF LTa (TNFSF1)


TNF (TNFSF2) tnfa TNF1, tnf-alpha, NM_212859 19 Yes [172]
TNF-a_v1, TNF-a_v3
TNF (TNFSF2) tnfb TNF2, tnf, Tnf-alpha, NM_001024447 15 Yes [24]
TNF-a_v2
CD95L/FasL (TNFSF6) faslg FasL NM_001042701 16
Apo2L/TRAIL (TNFSF10) tnfsf10l DL1a, TRAIL-like NM_131843 7
Apo2L/TRAIL (TNFSF10) tnfsf10l2 DL2 NM_001002593 24 Yes [24]
Apo2L/TRAIL (TNFSF10) tnfsf10l3 DL1b NM_001042713 21
Apo2L/TRAIL (TNFSF10) tnfsf10l4 DL3 NM_001013283 5
TL1A (TNFSF15) LOC560966 similar to TL1A NM_001123259 8
Unknown lta TNF-N NM_001024821 15
TNFR TNFR1 (TNFRSF1A) tnfrsf1a TNFR1 NM_213190 16 Yes [24]
CD95/Fas (TNFRSF6) fas XM_685355 17 Yes [24]
DR4 & DR5 hdr ZH-DR NM_194391 5
(TNFRSF10A & B)
DR4 & DR5 tnfrsfa OTR, tnf NM_131840 21
(TNFRSF10A & B)
DR6 (TNFRSF21) tnfrsf21 DR6 NM_001042688 20 Yes [24]
DR3 (TNFRSF25)
Adaptors CRADD/RAIDD cradd NM_001006066 4 Yes (ZFIN)
DEDD zgc:92202 NM_001002639 8
FADD fadd XM_001923858 7 Yes [24]
EDARADD LOC566761 similar to EDRADD XM_690046 17
MADD LOC100150917 similar to MADD XM_001923955 7
PIDD/LRDD LOC571033 similar to LRDD XM_694585 25
RIP1/RIPK1 ripk1l rip1 NM_001043350 2
TRADD tradd NM_131607 7 Yes (ZFIN)
Caspase Unknown caspase-a caspy NM_131505 16
Unknown caspase-b caspy2 NM_152884 1
Caspase-2 caspase-2 NM_001042695 16 Yes [24]
Caspase-3 caspase-3a casp3 NM_131877 1 Yes [24]
Caspase-3 caspase-3b casp3l NM_001048066 7 Yes [24]
Caspase-6 caspase-6 casp6a NM_001020497 3
Caspase-6 caspase-6l1 casp6b NM_001005973 3
Caspase-6 caspase-6l2 casp6c NM_001039980 3
Caspase-7 caspase-7 casp7a NM_001020607 23 Yes [24]
Caspase-7 LOC100000522 casp7b, similar to Casp7 XM_001340668 20
Caspase-7 LOC798445 similar to Casp7 XM_001338854 10
Caspase-8 caspase-8 casp8a NM_131510 6 Yes [146]
Caspase-8 caspase-8l1 casp8a-like NM_001098619 16
Caspase-8 caspase-8l2 casp8b, casp10-like XM_680338 9
Caspase-9 caspase-9 NM_001007404 23 Yes [24]
Caspase-10 LOC795066 caspxb, similar to Casp10 XM_001335127 16
Caspase-10 caspase-xa NM_001083862 6
zgc:194469 card-casp8 NM_001136252 6
c-FLIP/CFLAR cflar FLIP, Clarp1 NM_194399 11

123
336 Apoptosis (2010) 15:331349

Table 1 continued
Gene family Mammalian gene Zebrafish Other names Reference Zebrafish Synteny
homologa sequence chromosome w/human

Bcl2 Bcl-2 bcl2 zBlp2 NM_001030253 24


(Pro-Survival) Bcl-xL (BCL2L1) bcl2l zBlp1, zfBLP1, zfBcl-xL NM_131807 23 Yes [31]
Bcl-w (BCL2L2)
Mcl-1 mcl1a zfMLP1, zfMcl-1a NM_131599 19 Yes [31]
Mcl-1 mcl1b NM_194394 16 Yes [31]
BCL2L10 bcl2l10 zNR13, Nrz, NR-13, mcl1l NM_194398 18 Yes [31]
Bfl-1 (BCL2A1)
BCL2L12
Bcl2 Bax baxa zBax1, bax NM_131562 3 Yes (ZFIN)
(Pro-Apoptotic) Bax baxb zBax2 NM_001013296 3 Yes [31]
Bak
Bok boka zBok1, bok NM_001003612 6 Yes [31]
Bok bokb zBok2 NM_201185 2 Yes [31]
Bcl-G (BCL2L14)
Bcl-rambo (BCL2L13) bcl2l13 NM_001044891 18 Yes (ZFIN)
Bfk (BCL2L15)
Bid bida zBid NM_001079826 18
Bad bad NM_131579 7 Yes [31]
Bik bik NM_001045038 4 Yes [31]
Bim (BCL2L11) bcl2l11 BIM NM_001135791 13
Bmf bmf1 NM_001045224 3
Bmf bmf2 NM_001045473 Yes [31]
Bnip1 LOC795732 similar to Bnip1 XM_001333689 21
Bnip1 LOC565822 similar to SEC20 XM_684156 14
Bnip2 bnip2 NM_201218 Unmapped Yes (ZFIN)
Bnip2 LOC100149386 similar to Bnip2 NM_001128394 16
Bnip3 zgc:123120 nip3b NM_212693 17
Bnip3 bnip3l nip3a NM_205571 10 Yes (ZFIN)
Noxa/PMAIP1 pmaip1 zNoxa NM_001045474 14
Puma/BBC3 bbc3 zPuma NM_001045472 15 Yes [31]
Hrk
IAP NAIP
cIAP1 (BIRC2) birc2 IAP1, birc3 NM_194395 21 Yes (ZFIN)
cIAP2 (BIRC3)
XIAP (BIRC4) xiap birc4 NM_194396 14
Survivin (BIRC5) birc5a bir5a, survivin, survivin 1 NM_194397 12 Yes [136]
Survivin (BIRC5) birc5b survivin 2 NM_145195 23
Apollon (BIRC6) LOC796547 similar to birc6 XM_001336866 17
MLIAP (BIRC7)
ILP2 (BIRC8)
Other A20/TNFAIP3 LOC564497 similar to TNFAIP3 XM_687830 13
Apaf-1 apaf1 NM_001045243 4 Yes (ZFIN)
APP appa app NM_131564 1 Yes [136]
ATM atm XM_001921710 12
ATR DKEY- atr XM_691071 2
231J24.1

123
Apoptosis (2010) 15:331349 337

Table 1 continued
Gene family Mammalian gene Zebrafish Other names Reference Zebrafish Synteny
homologa sequence chromosome w/human

CHK1/CHEK1 chek1 chk1 NM_200193 21


CHK2/CHEK2 chek2 chk2 NM_200045 5
COP1/RFWD2 zgc:163067 NM_001089542 2
Cullin 3 cullin 3a cullin 3 NM_199691 2
Cytochrome c zgc:86706 NM_001002068 6
Mdm2 mdm2 NM_131364 4 Yes [136]
Omi/HtrA2 LOC799006 XM_001339375 13
Omi/HtrA2 LOC100003308 NM_001109735 13
Omi/HtrA2 LOC560031 XM_702479 10
Omi/HtrA2 LOC797876 XM_001338303 13
Omi/HtrA2 LOC560704 XM_001921275 13
Omi/HtrA2 LOC560164 XM_001921286 13
Omi/HtrA2 LOC562245 XM_001921346 13
Omi/HtrA2 LOC797799 NM_001110528 13
Omi/HtrA2 LOC100148101 XM_001921090 21
Omi/HtrA2 LOC793110 XM_001332768 10
Omi/HtrA2 LOC793521 XM_001333269 10
Omi/HtrA2 zgc:174193 NM_001114706 13
Omi/HtrA2 LOC799537 NM_001110170 13
Omi/HtrA2 LOC799634 XM_001335165 21
Omi/HtrA2 LOC100150281 XM_001923368 21
Omi/HtrA2 LOC799293 XM_001922056 21
Omi/HtrA2 LOC100149563 NM_001128820 13
p21/CDKN1A cdkn1a p21, cip1, waf1 XM_001923789 22 Yes (ZFIN)
p53 tp53 p53, brp53, drp53 NM_131327 5 Yes [136]
p62/SQSTM1 sequestosome 1 NM_213173 14 Yes (ZFIN)b
p62/SQSTM1 LOC100148880 similar to Sequestosome 1 XM_001922985 14
Phosphatidylserine receptor jmjd6 psr, ptdsr, zfpsr NM_170761 3
Pirh2/RCHY1 rchy1 NM_212600 5
SMAC/DIABLO zgc:63938 NM_200346 15
SMAC/DIABLO zgc:158776 XM_693904 21
a
LocusLink (LOC) designations are used for genes without an official full name
b
Conserved synteny reported with non-human mammalian species

Orthologs of nearly every member of the Bcl-2 family zebrafish Bcl-2 proteins remain to be described, particu-
have been described in zebrafish (Table 1) [31, 62, 6671]. larly among members of the divergent BH3-only subfam-
The only exceptions are three members of the anti-apop- ily. The wide range of Bcl-2 family members identified to
totic subfamily (Bcl-w, Bfl-1, and BCL2L12), one member date suggests that zebrafish may be a better system for
of the multidomain pro-apoptotic subfamily (Bak), and modeling the mammalian intrinsic apoptosis pathway than
three members of the BH3-only subfamily (Bcl-G, Bfk, and simple metazoans such as Drosophila, where only two
Hrk). Zebrafish multidomain Bcl-2 family members show multidomain Bcl-2 proteins have been described (buffy and
the highest degree of conservation with their mammalian debcl) and C. elegans, where one multidomain Bcl-2 pro-
counterparts. Members of the BH3-only subfamily display tein (ced-9) and two BH3-only protein (ced-13 and egl-1)
greater sequence divergence, making conserved synteny have been described (Fig. 1) [62, 72, 73].
within the genome an important criterion for establishing There is a great deal of functional conservation between
orthology [31, 66, 67]. It is probable that additional pro-apoptotic Bcl-2 proteins in zebrafish and mammals.

123
338 Apoptosis (2010) 15:331349

Fig. 1 Comparison of Apoptotic Pathways in C. elegans, Drosophila, all been shown to trigger apoptosis through cell surface death
Fish, and Mammals. The diagram illustrates core components of the receptors but this activity has not yet been formally demonstrated for
intrinsic and extrinsic apoptosis pathways in evolutionarily diverse faslg. Similarly, anti-apoptotic activity has been demonstrated for
species. In zebrafish, red arrows indicate interactions with strong birc2, birc5a, and birc5b but not the other zebrafish IAPs. A link
supporting evidence based on co-immunoprecipitation studies and/or between the extrinsic and intrinsic pathways in zebrafish is considered
antisense morpholino oligonucleotide knockdown experiments. In confirmed based on Eimon et al. [24] although it remains unclear if
some instances, interactions may not be established for all members bida is responsible for mediating this interaction
of a given family. For example, tnfsf10l, tnfsf10l2, and tnfsf10l3 have

Overexpression of zebrafish BH3-only subfamily members embryos [66]. Among the zebrafish pro-apoptotic Bcl-2
bida, bik, bcl2l11/bim, bmf1, bmf2, pmaip1/noxa, bbc3/ proteins, bad and bida are notably weak inducers of apop-
puma, and bnip3l and the multidomain pro-apoptotic sub- tosis. Interestingly, although bad does not strongly induce
family members baxa and baxb in early stage embryos apoptosis on its own, it is capable of sensitizing embryos to
induces caspase-3 activation and apoptosis [31, 66, 74]. c-irradiation [66]. Knockdown of bad also prevents apop-
Conversely, inhibition of bbc3/puma, baxa, or baxb using totic cell death in the CNS of embryos that are defective for
antisense morpholino oligonucleotides protects embryos glucose uptake due to inhibition of the slc2a1 membrane
from apoptosis in response to c-irradiation [31]. As in transporter [75]. Zebrafish bida does not share conserved
mammals, the pro-apoptotic function of zebrafish BH3-only synteny with mammalian Bid, leading to speculation that it
proteins requires an intact BH3 domain. Mutation of this is either functionally diverged or not a true ortholog [66].
domain blocks the ability of zebrafish bida, bcl2l11/bim, Zebrafish BH3-only proteins are capable of triggering
pmaip1/noxa, and bbc3/puma to induce apoptosis in apoptosis in mammalian cells, confirming a high degree of

123
Apoptosis (2010) 15:331349 339

functional conservation. Transfection of COS-1 cells with anti-apoptotic Bcl-2 subfamily members retain sufficient
zebrafish bad induces DNA fragmentation and chromatin conservation to function in mammalian cells. Zebrafish
condensation [76]. Zebrafish BH3 domain peptides from bcl2l10 promotes survival following serum withdrawal in
bcl2l11/bim, pmaip1/noxa, and bbc3/puma induce cyto- COS-7 cells [85] and zebrafish bcl2 promotes survival
chrome c release in mitochondria isolated from mouse following growth factor withdrawal in murine FL5.12 cells
embryonic fibroblasts (MEFs). Significantly, none of these [86]. In the latter example, the zebrafish bcl2 protein was
peptides show activity in mitochondria isolated from Bax-/-, shown to bind directly to human Bim, Bax, and Puma by
Bak-/- double knockout MEFs, indicating that the zebrafish immunoprecipitation.
BH3 peptides are capable of interacting with endogenous In mammals, anti-apoptotic Bcl-2 proteins associate
murine multidomain pro-apoptotic proteins [66]. with pro-apoptotic family members through their BH3
Important regulatory sequences are conserved between domains. It is assumed that specific patterns of interaction
pro-apoptotic Bcl-2 proteins in zebrafish and mammals. exist between individual Bcl-2 family members, but the
Mammalian Bad is regulated by phosphorylation of three exact hierarchy of these interactions is controversial [87,
conserved serine residues, which facilitates binding of 88]. Initial studies in zebrafish show that bcl2 can inhibit
14-3-3 proteins and promotes cell survival [77]. Two of apoptosis induced by overexpression of the multidomain
these residues (serines 136 and 155 in mice) are conserved pro-apoptotic protein baxa and the BH3-only proteins bida,
in zebrafish bad and appear to play a similar regulatory bmf1, bmf2, pmaip1/noxa, and bbc3/puma. In contrast, bcl2
role. When they are changed to aspartate to mimic phos- fails to inhibit baxb and only weakly inhibits bik [31]. In
phorylation, zebrafish bad is no longer capable of sensi- GST pull-down experiments, full-length zebrafish bcl2
tizing embryos to radiation-induced apoptosis. Conversely, protein binds strongly to the BH3 domain peptide from
when they are changed to alanine to prevent phosphory- zebrafish bim, binds weakly to bida, bad, and bbc3/puma,
lation, overexpression of bad alone strongly induces and does not bind at all to pmaip1/noxa [66]. Additional
apoptosis in embryos [66]. This observation indicates that work is required to reconcile these observations and
the proteins pro-apoptotic activity is normally inhibited by determine if individual Bcl-2 family members exhibit
serine phosphorylation during early development. Mam- similar protein-protein interaction patterns in zebrafish and
malian Bim is regulated by ERK1/2-dependent phosphor- mammals.
ylation of serine 69 (leading to proteasomal degradation)
[78, 79], AKT-dependent phosphorylation of serine 87
[80], and localization to microtubules [81]. Zebrafish The zebrafish apoptosome and effector caspases
bcl2l11/bim shows conservation of the serine 69 and 87
residues, but appears to lack an obvious dynein light chain- In response to apoptotic stimuli, pro-apoptotic Bcl-2 family
binding site [66]. In mammals, the BH3-only protein Bid members induce mitochondrial outer-membrane perme-
plays a unique role as a bridge between the extrinsic and abilization and trigger release of pro-apoptotic factors into
intrinsic apoptosis pathways [82]. While it remains the cytosol. One such factor is cytochrome c, which binds
unknown if bida fulfills a similar role in zebrafish, the adaptor Apaf-1 and the initiator caspase-9 to form a
sequence analysis reveals a putative caspase-8 cleavage complex known as the apoptosome. Once assembled, the
site that would ostensibly allow for proteolytic activation apoptosome activates caspase-9, which in turn activates the
by the extrinsic apoptosis pathway [31]. effector caspases (e.g caspase-3, -6, and -7) through pro-
Zebrafish anti-apoptotic Bcl-2 subfamily members also teolytic processing. Activated effector caspases cleave
show structural and functional conservation with their multiple major cellular substrates and are responsible for
mammalian counterparts. Overexpression of zebrafish bcl2, amplifying pro-apoptotic signals from both intrinsic and
bcl2l/bcl-xL, mcl1a and mcl1b inhibits apoptosis induced extrinsic pathways. In addition to cytochrome c, two other
by ectopic expression of BH3-only subfamily members and mitochondrial factorsSmac/Diablo and the serine prote-
by c-irradiation [31, 66]. Simultaneous knockdown of both ase Omi/HtrA2are released following membrane per-
mcl1a and mcl1b significantly decreases viability in zeb- meabilization and indirectly promote apoptosis by
rafish embryos, demonstrating that anti-apoptotic Bcl-2 inhibiting a family of anti-apoptotic proteins known as
proteins play an important role in promoting survival inhibitor of apoptosis proteins (IAPs) [89].
during early development. In contrast, knockdown of bcl2 Although the zebrafish apoptosome has not been char-
has no effect on embryo viability [31]. These loss-of- acterized in detail, it appears highly homologous to its
function phenotypes in zebrafish are similar to those mammalian counterpart. Orthologs of all major apoptosome
observed in knockout mice. Mcl-1-/- mice die around components have been identified in the zebrafish genome
embryonic day E4.0 [83] while Bcl-2-/- mice are viable (Table 1) [71]. Mitochondrial fragmentation is observed
and show no embryonic defects [84]. Several zebrafish in zebrafish embryos undergoing staurosporine-induced

123
340 Apoptosis (2010) 15:331349

apoptosis, suggesting that activation of the intrinsic apop- apoptosis to exert anti-apoptotic functions, although not all
tosis pathway triggers disruption of the mitochondrial outer- family members are dedicated inhibitors of apoptosis [94].
membrane [22]. Zebrafish caspase-9 can rapidly trigger Eight IAPs have been identified in humans and at least four
caspase-3 activation and apoptosis when overexpressed of these have orthologs in zebrafish (Table 1) [71, 95, 96].
in zebrafish embryos [24]. Activity assays utilizing The closely related mammalian c-IAP1 and c-IAP2 genes
caspase-specific bioluminescent substrates confirm that are represented by a single ancestral birc2/ciap1 gene in
both c-irradiation and staurosporine cause strong activation zebrafish and other lower vertebrates [96]. Zebrafish
of caspase-9 in zebrafish [26]. orthologs of the IAP inhibitors Smac/Diablo and Omi/
Multiple effector caspases have been identified in the HtrA2 have also been identified but remain to be func-
zebrafish genome (Table 1) [24, 71]. Zebrafish effector tionally characterized (Table 1).
caspases cleave many of the same protein substrates known Several recent publications show that the IAP family
in mammals, including PARP and 14 novel human cas- plays a critical role in endothelial cell survival and vascular
pase-3 substrates [21, 90]. These observations suggest that homeostasis in zebrafish. A forward genetic screen for
effector caspase targets are broadly conserved between fish zebrafish vascular defects identified the tomato (tom)
and mammals. There are two zebrafish orthologs of cas- mutation, which produces a hemorrhagic phenotype, vas-
pase-3 and at least one (caspase-3a) is capable of inducing cular regression, and endothelial cell apoptosis [96]. The
apoptosis following overexpression in cultured cells and tom mutation is located in the zebrafish birc2/ciap1 gene
zebrafish embryos [24, 25]. Recombinant caspase-3a and produces a nonfunctional protein product. The anti-
shows strong activity toward the mammalian caspase-3 and apoptotic function of birc2/ciap1 in endothelial cells is
-7 substrate Ac-DEVD-MCA and weak activity toward dependent upon 1) binding to TNFR-associated factor 2
caspase-1, -6, -8, and -9 substrates [25]. The finding that (TRAF2) and 2) the E3-ubiquitin ligase activity of the
zebrafish caspase-3a has low but significant activity RING domain. Inhibitors of caspase-8 can rescue the birc2/
against a mammalian caspase-6-specific substrate has led ciap-/- phenotype. These observations are consistent with
to the suggestion that it plays dual roles, partitioned mammalian c-IAPs, which interact with TNF receptors
between two different effector caspases in mammals [90]. through TRAF2 and regulate TNFa-mediated activation of
Caspase-3 enzyme activity is detected in zebrafish embryos the prosurvival NF-jB pathway through polyubiquitination
following overexpression of caspase-3a and treatment by of receptor-interacting protein 1 (RIP1) [2, 97101]. A
cycloheximide [21, 25]. Activation of caspase-3 in zebra- second member of the zebrafish IAP family, birc5a/survi-
fish following c-irradiation and overexpression of caspase- vin, has also been reported to play a role in endothelial cell
3a, caspase-9, and pro-apoptotic Bcl-2 family members has survival [95, 102]. Knockdown of birc5a/survivin results in
been confirmed by immunohistochemistry using a cross- disorganized angiogenesis, reduced vasculature, and
reactive monoclonal antibody against the cleaved, active increased numbers of apoptotic cells. Surprisingly, knock-
form of caspase-3 [24, 31]. Structural information for down of the closely related birc5b/survivin has a much less
caspase-3a has been obtained by X-ray crystallography, dramatic effect on angiogenesis although its expression
providing a unique level of detail for this important com- pattern during early embryogenesis is similar to that of
ponent of the zebrafish apoptotic machinery [91]. In birc5a/survivin [102] Knockdown of either birc5a or birc5b
addition to the canonical effector caspases, two novel also triggers apoptosis in several other tissues including
zebrafish caspases (caspase a and caspase b) containing hematopoietic cells and the neural tube [102, 103].
N-terminal pyrin domains and sharing greatest homology Several lines of evidence indicate that IAPs, and more
with mammalian caspase-1 have been reported to induce generally endothelial cell apoptosis, are important for
apoptosis following overexpression [92]. This may in fact normal vascular development in mammals as well as zeb-
be more akin to pyroposis, a novel form of proinflamma- rafish [104]. Endothelial cell apoptosis has been observed
tory cell death mediated by caspase-1 in mammals [93]. in mouse embryos beginning at E14.5 and inhibition of this
process by the anti-apoptotic Bcl-2 protein results in
abnormally dense and disorganized networks of small
Zebrafish IAP proteins and IAP antagonists vessels [105]. Survivin-/- knockout mice die by E3.5 [106],
but selective inactivation of Survivin in endothelial cells
IAPs have been described in both invertebrates and using the cre-lox system results in embryos that survive to
vertebrates and are characterized by the presence of a E12.5 and exhibit extensive hemorrhaging throughout the
C-terminal RING domain and one to three N-terminal body, diffuse vascular endothelial apoptosis, and increased
baculoviral IAP repeat (BIR) domains (hence the alternate pericyte-endothelial gaps [107]. Although there are no
designation BIRCBIR-containing proteins). IAPs can reports of vascular defects in mice deficient for c-IAP1 or
interact directly with caspases and other effectors of c-IAP2 [108, 109], this may be due to the high degree of

123
Apoptosis (2010) 15:331349 341

conservation between these two family members, which baxa [113], perp [122], and bbc3/puma [31]. Puma has
allows for functional redundancy in some contexts [99, been identified as a key mediator of p53-dependent apop-
100]. This hypothesis is supported by the finding that tosis in mammals based on data from knockout mice [123]
simultaneous depletion of c-IAP1 and c-IAP2 in human and bbc3/puma appears to play a similarly essential role in
umbilical cord vascular endothelial cells (HUVECs) sen- zebrafish [31]. Upstream regulation of tp53 has also been
sitizes these cells to killing by TNFa, the TNFR1 ligand characterized in zebafish. As in mammals, the E3 ubiquitin
[96]. ligase mdm2 serves as a primary negative regulator of tp53
activation. Knockdown of zebrafish mdm2 results in
Zebrafish p53 and the intrinsic apoptosis pathway extensive apoptosis, which can be rescued by simultaneous
knockdown of tp53 [29]. Other ubiquitin ligases known to
The tumor suppressor protein p53 is a multifunctional negatively regulate p53 in mammalssuch as COP1 [124]
transcription factor that plays a central role in determining and Pirh2 [125]are present in the zebrafish genome but
how cells respond to stresses such as DNA damage, have not been functionally characterized. Epistasis analysis
hypoxia, and oncogenic signaling [110]. In stressed cells, in piy mutant embryos shows that two genes known to
p53 is stabilized and converted from a latent to an active mediate the DNA damage response upstream of p53 in
form. Activation of p53 can lead to a variety of outcomes, mammalsataxia telangiectasia mutated (atm) and
including cell cycle arrest (allowing for DNA repair) and checkpoint kinase 2 (chek2)are also essential for tp53-
apoptosis (to eliminate cells damaged beyond repair). In dependent apoptosis in zebrafish [116]. Seven novel acti-
the latter case, p53-dependent apoptosis is primarily med- vators of p53 (Hey1, Hes1, TFAP4, Osr1, NR2F2, SFRS10,
iated by stimulation of the intrinsic apoptosis pathway. and FLJ11339), initially identified in a high-throughput
Stabilized p53 accumulates in the nucleus and can directly in vitro screen using a human cell line, are capable of
regulate the expression of many pro-apoptotic genes (e.g. inducing tp53 expression in zebrafish embryos [126].
Bid, Noxa, Puma, Apaf1). Mounting evidence indicates Several recent studies have used zebrafish to identify and
that transcription-independent activities also play an validate novel mechanisms of p53 regulation that are
important role in p53-induced apoptosis. Specifically, p53 broadly conserved throughout vertebrate evolution. Le et al.
can translocate to the mitochondrial outer membrane fol- [127] identified miR-125b, a brain-enriched microRNA
lowing induction and directly interact with both pro- and (miRNA), as a potential negative regulator of p53, by
anti-apoptotic members of the Bcl-2 family [111, 112]. searching for miRNA response elements that have been
The tp53 gene is a critical mediator of apoptosis in conserved between zebrafish and humans. Overexpression
response to DNA damage in zebrafish. A variety of DNA of miR-125b reduces p53 levels and inhibits p53-dependent
damaging reagents have been shown to upregulate zebra- apoptosis in primary human lung fibroblasts and the neu-
fish tp53 transcription [29, 113] and increase tp53 protein roblastoma cell line SHSY5Y. In zebrafish, knockdown of
levels [114]. Knockdown of tp53 blocks apoptosis arising miR-125b leads to increased apoptosis in wild-type
from a wide range of stressors, including c-irradiation [32], embryos, but not in embryos from the tp53M214K mutant
ultraviolet irradiation, camptothecin treatment [29], inhi- line. Injection of synthetic miR-125b duplex into zebrafish
bition of the DNA crosslink repair gene fancd2 [115], and embryos rescues the knockdown phenotype and inhibits
compromised DNA replication in mutant lines such as apoptosis following DNA damage. Chen et al. [128] show
flathead, caf1b, and pinball eye [116118]. A similar that D113p53, the zebrafish counterpart of the human p53
degree of protection is observed when embryos are pre- isoform D133p53, is activated in a p53-dependent manner
treated with Pifithrin-a, a pharmacological inhibitor of p53 following DNA damage and functions to antagonize
[119]. Resistance to DNA damage-induced apoptosis is apoptosis, in part through upregulation of bcl2l/bcl-xL.
also seen in tp53M214K (sometimes referred to as tp53e7), a Knockdown of endogenous D113p53, using a morpholino
zebrafish line with a dominant negative mutation in the targeting the D113p53 5-UTR, increases the sensitivity of
tp53 locus that destroys transcriptional activity [113]. In embryos to c-irradiation-induced apoptosis.
addition to reduced apoptosis, the tp53M214K line begins to Sidi et al. [86] have used the zebrafish tp53M214K mutant
develop malignant peripheral nerve sheath tumors at to better understand p53-independent apoptotic responses
approximately 8.5 months of age, confirming tp53 is a and gain important insights into the function of caspase-2.
critical tumor suppressor protein in fish. The related tran- Knockdown of checkpoint kinase 1 (chek1) was found to
scription factors tp63 and tp73 do not appear to play sig- restore sensitivity to c-irradiation-induced apoptosis in
nificant roles in apoptosis in zebrafish [120, 121]. tp53M214K embryos. Surprisingly, although dying cells in
As in mammals, zebrafish tp53 autoregulates its own chek1-knockdown embryos are TUNEL positive, caspase-3
expression [29] and promotes transcription of a diverse activation is not observed and cell death cannot be blocked
array of target genes, including cdkn1a/p21 [29], mdm2 and by bcl2l/bcl-xL overexpression or bbc3/puma knockdown.

123
342 Apoptosis (2010) 15:331349

Epistasis analysis demonstrates that zebrafish orthologs The cell-extrinsic apoptosis pathway
of ATM, ATR, and the initiator caspase-2 are critical for
radiation-induced apoptosis in tp53M214K/chek1-knock- Zebrafish death receptors and ligands
down embryos. Experiments in HeLa cells suggest that a
similar Chek1-suppressed caspase-2 pathway exists in The extrinsic apoptosis pathway is engaged when certain
mammalian cells defective for p53 activity. ligands of the TNF superfamily bind to specialized mem-
ATM and ATR are well-characterized apical kinases bers of the TNFR superfamily known as death receptors.
that respond to DNA damage by activating a number of Death receptors are characterized by the presence of a
downstream proteins including p53 [129]. The precise role conserved *80-amino-acid death domain motif in the
of caspase-2 in apoptosis, however, is a matter of long- cytoplasmic tail that plays a central role in formation of the
standing debate [3]. In mammalian systems caspase-2 has DISC [132]. The human genome contains six death
been linked to both the intrinsic and extrinsic pathways, receptors and five cognate TNF ligands (Fig. 2). An
although it does not appear to be required for cell death in extracellular fragment of b-amyloid precursor protein
most contexts [130]. The mechanism of caspase-2 acti- (APP) has recently been shown to act as a ligand for DR6
vation has not been fully elucidated, although it is thought and drive axonal degeneration and neuronal death in mice
to involve a protein complex similar to the apoptosome [133]. This arrangement of receptors and ligands is well
that incorporates the adaptor proteins RAIDD and PIDD conserved in mammals, although many species possess
(designated the PIDDosome) [131]. Orthologs of both only a single ortholog of the closely related death receptors
RAIDD (cradd) and PIDD (LOC571033) have been designated DR4 and DR5 in humans and other primates
identified in the zebrafish genome (Table 1) and overex- [134].
pression of casp2 on its own is sufficient to induce Recent studies have shed considerable light on death
apoptosis in embryos [24]. The roles of the RAIDD and receptor ligands in zebrafish and other teleosts. Orthologs
PIDD orthologs in zebrafish have yet to be studied and of TNF (tnfa and tnfb), CD95/FasL (faslg), Apo2L/TRAIL
their requirement in the novel Chek1-suppressed caspase-2 (tnfsf10l, tnfsf10l2, tnfsf10l3, and tnfsf10l4), and APP
pathway is unclear. (appa) have been identified based on phylogenetic analysis

Fig. 2 Zebrafish Death Receptors and Ligands. The diagram illus- co-immunoprecipitation studies and/or antisense morpholino oligo-
trates all TNF and TNFR superfamily members that have been nucleotide knockdown experiments. Gray arrows indicate interac-
reported to date in zebrafish. Official zebrafish gene designations are tions that are hypothesized to occur based on studies in other
given in lower case italic type and corresponding mammalian vertebrate species but remain to be experimentally verified in
homologs are given in upper case bold. Red arrows indicate ligand- zebrafish. The zebrafish homolog of APP is included as a potential
receptor interactions that have been confirmed in zebrafish by ligand for tnfrsf21/dr6 based on recent evidence in mammals

123
Apoptosis (2010) 15:331349 343

[24, 135139]. Co-immunoprecipitation studies confirm and cellular FLICE-inhibitory protein (c-FLIP), an enzy-
that tnfa binds selectively to tnfrsf1a, the zebrafish ortholog matically inactive relative of caspase-8 and -10 [144]. In
of TNFR1, as expected [24]. A probable zebrafish TL1A contrast, engagement of the adaptor protein TRADD leads
ortholog (LOC560966) has also been described based on to assembly of Complex I, which induces transcription of
phylogenetic analysis and the presence of conserved cys- both proinflammatory cytokines and antiapoptotic factors
teine residues [135]. The status of LTa in zebrafish and through the IKK/NF-jB, JNK, and p38 MAPK pathways
other teleosts remains unclear. Savan et al. [137] describe a [145]. In mammals, only DR4, DR5, and CD95/Fas
novel TNF gene (TNF-N; subsequently designated lta) that which signal primarily through FADDare direct activa-
appears similar to mammalian LTa based on its proximity tors of the extrinsic apoptosis pathway. TNFR1 and DR3
to the zebrafish tnf locus. However, the intronic structure of bind TRADD and mediate immune-stimulatory activity.
lta is distinct from human LTa and it clusters with OX40L, Cross talk is possible between these two distinct death
GITRL, and CD30L in an unrooted phylogenetic tree receptor signaling pathways through distal signaling com-
[135]. plexes that are nucleated by FADD and TRADD [2].
Of the four zebrafish Apo2L/TRAIL homologs, Zebrafish orthologs have been identified for most com-
tnfsf10l2 exhibits the greatest structural similarity with ponents of the DISC and Complex I, including the pivotal
mammalian Apo2L/TRAIL [24]. Both tnfsf10l and adaptor proteins FADD and TRADD (Table 1) [24, 71].
tnfsf10l3 are more structurally related to Apo2L-likea Reverse genetic analysis has been used to interrogate the
second copy of Apo2L/TRAIL found in avian species but requirement for fadd and tradd in the zebrafish DISC.
not mammalsdue to additional cysteine residues in their Inhibition of fadd, but not tradd, is sufficient to block
extracellular domains [140]. Zebrafish tnfsf10l4 shows the apoptosis induced by overexpression of the Apo2L/TRAIL
greatest sequence divergence and only groups weakly with orthologs tnfsf10l and tnfsf10l3 in zebrafish embryos [24].
other Apo2L/TRAIL and RANKL sequences [135]. Transient overexpression of zebrafish death receptors
Death receptors are also well conserved between zeb- (tnfrsf1a, fas, hdr, and tnfrsfa) and fadd in human cells
rafish and mammals. Clear orthologs of TNFR1 (tnfrsf1a), triggers apoptosis, indicating that the death domain motif
CD95/Fas (fas), and DR6 (tnfrsf21) have been identified has been sufficiently well conserved between teleosts and
based on the arrangement of their cysteine-rich extracel- mammals to enable cross-species protein interactions
lular domains, phylogenetic analysis of their death within the DISC [24].
domains, and conserved synteny with mammalian genes A single zebrafish ortholog of caspase-8 (casp8a), con-
(Table 1) [24]. In addition, two death domain-containing taining N-terminal death effector domains (DEDs) and a
receptors designated hdr [141, 142] and tnfrsfa (previously QACQG active-site motif characteristic of mammalian
known as Ovarian TNF Receptor or otr) [139] have been caspase-8 and -10, has been described [24, 146]. Several
described in zebrafish. These receptors have less clear-cut lines of evidence show that zebrafish casp8a is functionally
mammalian orthologs. They contain extracellular domains conserved with its mammalian counterpart. FADD directly
that are most similar to CD95/Fas and death domains that associates with casp8a through homophilic DED interac-
are most similar to DR4 and DR5. The finding that both hdr tions [146]. Zebrafish casp8a can mediate apoptosis fol-
and tnfrsfa bind Apo2L/TRAIL orthologs and are essential lowing death receptor activation in mouse embryonic
for Apo2L/TRAIL-induced apoptosis in zebrafish argues fibroblast (MEF) cells that lack endogenous caspase-8
strongly for categorizing them as homologs of DR4 and [146]. The cowpox virus serpin cytokine response modifier
DR5 (Fig. 2) [24]. DR3 remains the only mammalian death A (CrmA)which inhibits caspase-8, -10, and -1 in
receptor without a zebrafish counterpart, although the mammalsblocks Apo2L/TRAIL-induced apoptosis in
identification of a zebrafish ligand with homology to TL1A zebrafish embryos [24].
[135] suggests such an ortholog may yet be found. Additional caspase-8 related genes are present in zeb-
rafish (Table 1) [24, 146]. These include casp8l1, casp8l2,
Components of the zebrafish DISC and three genes containing an N-terminal caspase recruit-
ment domain (CARD) in place of the DED motifs (caspxa,
Activated death receptors mediate downstream activity LOC795066, and zgc:194469). Casp8l1 contains the dis-
through one of two distinct signaling complexes. Engage- tinctive QACQG active-site motif but lacks N-terminal
ment of the adaptor protein FADD leads to DISC recruit- DEDs. In contrast, casp8l2 possesses DEDs but contains a
ment and drives proapoptotic signaling through caspase-8 QACRG active site that is more reminiscent of caspase-2.
(as well as caspase-10 in human cells) [132]. Activation of In addition, zebrafish possess a clear ortholog of c-FLIP
caspase-8 is positively controlled by Cullin-3-mediated (cflar) that contains N-terminal DEDs and an enzymati-
polyubiquitination and p62-dependent aggregation [143] cally inactive caspase cataltic domain. Overexpression of
and negatively controlled by the deubiquitinase A20 [143] cflar in zebrafish embryos prevents induction of apoptosis

123
344 Apoptosis (2010) 15:331349

by the Apo2L/TRAIL orthologs tnfsf10l and tnfsf10l3 [24], (particularly CD95/Fas and DR6) in the CNS is thought
consistent with its roles as an inhibitor of death receptor- to influence a variety of neuropathological processes
mediated apoptosis in mammals. [133, 153].

Biological roles of the extrinsic apoptosis pathway


in zebrafish Concluding remarks

Although the extrinsic pathway is critical for normal Invertebrate model organisms such as C. elegans and
functioning of the mammalian immune system, it remains Drosophila have been used widely and successfully to
unclear if it plays a similar role in zebrafish. In humans, study apoptosis [154, 155]. The ease with which these
mutations in CD95/Fas, CD95L/FasL, and caspase-10 are systems can be genetically manipulated has been instru-
responsible for autoimmune lymphoproliferative syn- mental in identifying and characterizing core components
drome, while mutations in caspase-8 cause deficiencies in of the apoptotic machinery and understanding how these
both lymphocyte proliferation and activation [2, 147]. molecules are regulated under both normal and pathologi-
Knockout studies in mice confirm that the extrinsic path- cal conditions. In spite of their advantages, invertebrate
way has widely conserved roles in immune system regu- models cannot replicate many aspects of apoptosis known
lation [3, 148, 149]. In zebrafish, T and B cells do not to be important in mammals. Apoptosis that occurs as a
develop until 3 dpf or later [150], making analysis of the normal part of embryogenesis and adult tissue homeostasis
extrinsic pathway in these cell types by morpholino is often specific to organs and tissues that lack analogs in
knockdown techniques impractical. Langenau et al. [151] invertebrate species. In other cases, apoptosis arises from
have used a stable transgenic line expressing bcl2 under the disease pathologies that cannot be modeled in nematodes
control of the rag2 promoter to show that apoptosis plays and flies. Additionally, apoptotic pathways have evolved to
an integral role in zebrafish T- and B-cell homeostasis, but a far greater degree of complexity and diversity in mam-
additional transgenic or mutant lines will be required to mals and other vertebrates (Fig. 1) [156, 157]. For exam-
verify a specific role for the extrinsic pathway in these cell ple, more than 25 Bcl-2 superfamily members have been
types. Two studies demonstrate that the zebrafish death identified in humans [62] versus three in C. elegans (egl-1,
receptor hdr plays a homeostatic role in at least one non- ced-9, and ced-13) [158] and two in Drosophila (buffy and
lymphoid cell type. Long et al. [141] found that hdr is debcl) [159]. An even more extreme case is the TNFR
specifically expressed in the hematopoietic lineage and superfamily, comprising approximately 30 members in
used transgenic zebrafish expressing a dominant negative humans including six death domain-containing receptors
hdr version to demonstrate that receptor inhibition leads to responsible for engaging the extrinsic pathway [160]. This
an accumulation of red blood cells. Kwan et al. [142] superfamily is not present at all in C. elegans and consists
reported similar findings in zebrafish embryos following of only a single member lacking a death domain (eiger) in
morpholino knockdown of hdr. In the context of these Drosophila [161]. Finally, there are fundamental differ-
observations, it is interesting to note that overexpression of ences in the way apoptotic pathways function between
tnfsf10l3, the hdr ligand, selectively induces apoptosis in invertebrates and mammals. Drosophila Bcl-2 proteins
the erythroid lineage [24]. appear to be peripheral players responsible for fine-tuning
Embryonic gene expression patterns suggest that death stress-induced apoptosis [162, 163], while in mammals
receptor signaling may play a role in numerous zebrafish they serve as the chief regulators of the intrinsic pathway.
tissues [24]. In addition to the hematopoietic lineage, hdr is This difference may be linked to the increased importance
strongly expressed in the developing notochord during of mitochondria as activators of the intrinsic pathway in
early development. The closely related death receptor mammals versus C. elegans and Drosophila. In mammals,
tnfrsfa is also expressed in the notochord during this per- Bcl-2 proteins regulate the release of pro-apoptotic mole-
iod, as is fadd. Tnfrsfa and the Apo2L/TRAIL homologs cules such as cytochrome c, Smac/Diablo, and Omi/HtrA2
tnfsf10l2 and tnfsf10l3 are detected in neuromasts of the from the mitochondria. Unlike mammals, cytochrome c
lateral line. These mechanosensory organs contain hair does not play a role in apoptosome formation in C. elegans
cells similar to those of the mammalian inner ear and are and its role in Drosophila appears to be extremely limited
known to undergo constant, apoptosis-mediated turnover in [159, 164]. The Reaper, Grim, and Hid (RGH) proteins
zebrafish [152]. Finally, several death receptors (tnfrsf1a, functional counterparts of Smac/Diabloplay a central
tnfrsfa, and tnfrsf21/dr6) and ligands (tnfsf10l, tnfsf10l2, role in Drosophila apoptosis but are regulated primarily via
and tnfsf10l3) are expressed in regions of the developing increased expression rather than mitochondrial sequestra-
CNS. Their role in zebrafish remains unknown, but in tion [162, 165]. Fundamental differences are also apparent
mammals the expression of death receptors and ligands in the extrinsic pathway. Eiger triggers cell death through a

123
Apoptosis (2010) 15:331349 345

JNK-dependent mechanism using the adaptor protein zebrafish embryos following inhibition of IAPs [96] or
TNF-receptor-associated factor 6 (TRAF6, formerly known ectopic expression of Apo2L/TRAIL homologs [24] could
as dTRAF2) [161, 166]. Drosophila homologs of FADD serve as the basis for in vivo screens to identify small
(dFADD) and caspase-8 (Dredd), have not been linked to molecules with similar pro-apoptotic activities. It is likely
the extrinsic pathway and instead appear to promote tran- that future apoptosis research in zebrafish will continue to
scription of antimicrobial peptide genes following bacterial exploit the strengths of this model system, in particular its
infection [167]. genetic tractability. To date, loss-of-function mutants have
Due to these limitations, a vertebrate model of apoptosis been reported only for a handful of the apoptosis-related
that is genetically tractable, amenable to analysis during genes listed in Table 1. Therefore, it will be important to
early development, and suitable for experimental approa- identify and characterize additional mutant lines that are
ches with relatively high throughput is desirable. The deficient in core components of the intrinsic and extrinsic
zebrafish fulfills all of these criteria and, as this review apoptosis pathways. The failure of large-scale forward
shows, has a diversity of apoptosis-related genes compa- genetic screens to identify such mutants so far suggests that
rable to mammals (Fig. 1). In addition to preserving the reverse genetic techniques, such at TILLING and gene
complexity of mammalian systems, zebrafish apoptosis targeting through zinc-finger nucleases, may be the pre-
pathways are functionally more analogous to mammals ferred approaches. The value of such genetic tools is nicely
than invertebrates on a number of levels. The Bcl-2 family demonstrated by recent publications using birc2/ciap1 and
appears to play a far more crucial role in regulating the tp53 mutant lines to elucidate novel apoptosis pathways,
intrinsic pathway in zebrafish than Drosophila, as demon- regulatory mechanisms, and physiological functions
strated by the rapid and extensive cell death triggered by [86, 96, 127, 128].
knockdown of mcl1a and mcl1b, the requirement for bbc3/
puma in DNA damage-induced apoptosis, and the ability of
anti-apoptotic family members to inhibit apoptosis in References
response to both stress stimuli and extrinsic pathway acti-
vation [24, 31, 151]. In contrast to Drosophila, the zebra- 1. Youle RJ, Strasser A (2008) The BCL-2 protein family:
fish extrinsic pathway requires both FADD and caspase-8 opposing activities that mediate cell death. Nat Rev Mol Cell
Biol 9:4759
to trigger apoptosis and undergoes amplification through 2. Wilson NS, Dixit V, Ashkenazi A (2009) Death receptor signal
the intrinsic pathway, similar to type II cells in mammals transducers: nodes of coordination in immune signaling net-
[24]. These findings suggest the zebrafish has the potential works. Nat Immunol 10:348355
to become a powerful in vivo model for studying cell death. 3. Chowdhury I, Tharakan B, Bhat GK (2008) Caspases - an
update. Comp Biochem Physiol B Biochem Mol Biol 151:1027
It remains important to verify a number of features of 4. Li L, Qiu L, Song L et al (2009) First molluscan TNFR
zebrafish apoptosis. In particular, the roles of the mito- homologue in Zhikong scallop: molecular characterization and
chondria and mitochondrial factors such as cytochrome c, expression analysis. Fish Shellfish Immunol 27:625632
Smac/Diablo, and Omi/HtrA2 need to be clarified, as these 5. Streisinger G, Walker C, Dower N, Knauber D, Singer F (1981)
Production of clones of homozygous diploid zebra fish (Brac-
are among the most striking points of divergence between hydanio rerio). Nature 291:293296
invertebrates and mammals. Demonstrating an essential 6. Westerfield M (1995) The zebrafish book: a guide for the lab-
role for cytochrome c (zgc:86706) in formation of the oratory use of zebrafish (Danio rerio). University of Oregon
zebrafish apoptosome may require a three-dimensional Press, Eugene
7. Driever W, Solnica-Krezel L, Schier AF et al (1996) A genetic
structure of the complex, but it should be possible to screen for mutations affecting embryogenesis in zebrafish.
address changes in the subcellular localization of Smac/ Development 123:3746
Diablo and Omi/HtrA2 homologs with far less difficulty. 8. Haffter P, Nusslein-Volhard C (1996) Large scale genetics in a
Fish models may provide unique advantages for the small vertebrate, the zebrafish. Int J Dev Biol 40:221227
9. Nasevicius A, Ekker SC (2000) Effective targeted gene
study of apoptosis [168, 169]. Zebrafish and other teleost knockdown in zebrafish. Nat Genet 26:216220
species are useful for studying apoptotic cell death occur- 10. Wienholds E, Schulte-Merker S, Walderich B, Plasterk RH
ring not only during embryonic development but also in (2002) Target-selected inactivation of the zebrafish rag1 gene.
response to a variety of external stimuli including changes Science 297:99102
11. Amacher SL (2008) Emerging gene knockout technology in
in oxygen availability, osmotic stress, high and low tem- zebrafish: zinc-finger nucleases. Brief Funct Genomic Proteomic
peratures, and exposure to toxins. Many small molecule 7:460464
drugs exhibit conserved activities between zebrafish and 12. Kurita K, Burgess SM, Sakai N (2004) Transgenic zebrafish
humans [170, 171], raising the possibility that zebrafish produced by retroviral infection of in vitro-cultured sperm. Proc
Natl Acad Sci U S A 101:12631267
assays may also prove useful for identifying novel drugs 13. Davidson AE, Balciunas D, Mohn D et al (2003) Efficient gene
designed to induce apoptosis for the treatment of cancer. delivery and gene expression in zebrafish using the sleeping
For example, the highly specific phenotypes observed in beauty transposon. Dev Biol 263:191202

123
346 Apoptosis (2010) 15:331349

14. Meng A, Jessen JR, Lin S (1999) Transgenesis. Methods Cell 36. Ikegami R, Zhang J, Rivera-Bennetts AK, Yager TD (1997)
Biol 60:133148 Activation of the metaphase checkpoint and an apoptosis pro-
15. Ong ES, Chor CF, Zou L, Ong CN (2009) A multi-analytical gramme in the early zebrafish embryo, by treatment with the
approach for metabolomic profiling of zebrafish (Danio rerio) spindle-destabilising agent nocodazole. Zygote 5:329350
livers. Mol Biosyst 5:288298 37. Hensey C, Gautier J (1997) A developmental timer that regulates
16. Link V, Shevchenko A, Heisenberg CP (2006) Proteomics of apoptosis at the onset of gastrulation. Mech Dev 69:183195
early zebrafish embryos. BMC Dev Biol 6:1 38. Spanos S, Rice S, Karagiannis P et al (2002) Caspase activity
17. Abrams JM, White K, Fessler LI, Steller H (1993) Programmed and expression of cell death genes during development of
cell death during Drosophila embryogenesis. Development human preimplantation embryos. Reproduction 124:353363
117:2943 39. Cole LK, Ross LS (2001) Apoptosis in the developing zebrafish
18. Furutani-Seiki M, Jiang YJ, Brand M et al (1996) Neural embryo. Dev Biol 240:123142
degeneration mutants in the zebrafish, Danio rerio. Development 40. Iijima N, Yokoyama T (2007) Apoptosis in the medaka embryo
123:229239 in the early developmental stage. Acta Histochem Cytochem
19. Rodriguez M, Driever W (1997) Mutations resulting in transient 40:17
and localized degeneration in the developing zebrafish brain. 41. Oppenheim RW (1991) Cell death during development of the
Biochem Cell Biol 75:579600 nervous system. Annu Rev Neurosci 14:453501
20. Tucker B, Lardelli M (2007) A rapid apoptosis assay measuring 42. Pettmann B, Henderson CE (1998) Neuronal cell death. Neuron
relative acridine orange fluorescence in zebrafish embryos. 20:633647
Zebrafish 4:113116 43. Eisen JS, Melancon E (2001) Interactions with identified muscle
21. Negron JF, Lockshin RA (2004) Activation of apoptosis and cells break motoneuron equivalence in embryonic zebrafish. Nat
caspase-3 in zebrafish early gastrulae. Dev Dyn 231:161170 Neurosci 4:10651070
22. Kim MJ, Kang KH, Kim CH, Choi SY (2008) Real-time 44. Gould TW, Oppenheim RW (2001) Stepping stone to death. Nat
imaging of mitochondria in transgenic zebrafish expressing Neurosci 4:10531054
mitochondrially targeted GFP. Biotechniques 45:331334 45. Guerin MB, McKernan DP, OBrien CJ, Cotter TG (2006) Ret-
23. Gavrieli Y, Sherman Y, Ben-Sasson SA (1992) Identification of inal ganglion cells: dying to survive. Int J Dev Biol 50:665674
programmed cell death in situ via specific labeling of nuclear 46. Vecino E, Hernandez M, Garcia M (2004) Cell death in the
DNA fragmentation. J Cell Biol 119:493501 developing vertebrate retina. Int J Dev Biol 48:965974
24. Eimon PM, Kratz E, Varfolomeev E et al (2006) Delineation of 47. Biehlmaier O, Neuhauss SC, Kohler K (2001) Onset and time
the cell-extrinsic apoptosis pathway in the zebrafish. Cell Death course of apoptosis in the developing zebrafish retina. Cell
Differ 13:16191630 Tissue Res 306:199207
25. Yabu T, Kishi S, Okazaki T, Yamashita M (2001) Character- 48. Dahm R, Schonthaler HB, Soehn AS, van Marle J, Vrensen GF
ization of zebrafish caspase-3 and induction of apoptosis through (2007) Development and adult morphology of the eye lens in the
ceramide generation in fish fathead minnow tailbud cells and zebrafish. Exp Eye Res 85:7489
zebrafish embryo. Biochem J 360:3947 49. Mohamed YH, Amemiya T (2003) Apoptosis and lens vesicle
26. Geiger GA, Parker SE, Beothy AP, Tucker JA, Mullins MC, development. Eur J Ophthalmol 13:110
Kao GD (2006) Zebrafish as a biosensor? Effects of ionizing 50. Greiling TM, Aose M, Clark JI (2009) Cell fate and differ-
radiation and amifostine on embryonic viability and develop- entiation of the developing ocular lens. Invest Ophthalmol Vis
ment. Cancer Res 66:81728181 Sci. doi:10.1167/iovs.09-4388
27. Hong JR, Lin GH, Lin CJ et al (2004) Phosphatidylserine 51. Greiling TM, Clark JI (2009) Early lens development in the
receptor is required for the engulfment of dead apoptotic cells zebrafish: a three-dimensional time-lapse analysis. Dev Dyn
and for normal embryonic development in zebrafish. Develop- 238:22542265
ment 131:54175427 52. Bassnett S (2009) On the mechanism of organelle degradation in
28. Ikegami R, Hunter P, Yager TD (1999) Developmental activa- the vertebrate lens. Exp Eye Res 88:133139
tion of the capability to undergo checkpoint-induced apoptosis 53. Reyes R, Haendel M, Grant D, Melancon E, Eisen JS (2004)
in the early zebrafish embryo. Dev Biol 209:409433 Slow degeneration of zebrafish Rohon-Beard neurons during
29. Langheinrich U, Hennen E, Stott G, Vacun G (2002) Zebrafish programmed cell death. Dev Dyn 229:3041
as a model organism for the identification and characterization 54. Svoboda KR, Linares AE, Ribera AB (2001) Activity regulates
of drugs and genes affecting p53 signaling. Curr Biol 12:2023 programmed cell death of zebrafish Rohon-Beard neurons.
2028 Development 128:35113520
30. Yamashita M (2003) Apoptosis in zebrafish development. Comp 55. Williams JA, Barrios A, Gatchalian C, Rubin L, Wilson SW,
Biochem Physiol B Biochem Mol Biol 136:731742 Holder N (2000) Programmed cell death in zebrafish rohon
31. Kratz E, Eimon PM, Mukhyala K et al (2006) Functional beard neurons is influenced by TrkC1/NT-3 signaling. Dev Biol
characterization of the Bcl-2 gene family in the zebrafish. Cell 226:220230
Death Differ 13:16311640 56. Lang H, Bever MM, Fekete DM (2000) Cell proliferation and
32. Bladen CL, Lam WK, Dynan WS, Kozlowski DJ (2005) DNA cell death in the developing chick inner ear: spatial and temporal
damage response and Ku80 function in the vertebrate embryo. patterns. J Comp Neurol 417:205220
Nucleic Acids Res 33:30023010 57. Bever MM, Fekete DM (1999) Ventromedial focus of cell death
33. Greenwood J, Gautier J (2005) From oogenesis through gas- is absent during development of Xenopus and zebrafish inner
trulation: developmental regulation of apoptosis. Semin Cell ears. J Neurocytol 28:781793
Dev Biol 16:215224 58. Abdelilah S, Mountcastle-Shah E, Harvey M et al (1996)
34. Penaloza C, Lin L, Lockshin RA, Zakeri Z (2006) Cell death in Mutations affecting neural survival in the zebrafish Danio rerio.
development: shaping the embryo. Histochem Cell Biol Development 123:217227
126:149158 59. Fadool JM, Brockerhoff SE, Hyatt GA, Dowling JE (1997)
35. Meier P, Finch A, Evan G (2000) Apoptosis in development. Mutations affecting eye morphology in the developing zebrafish
Nature 407:796801 (Danio rerio). Dev Genet 20:288295

123
Apoptosis (2010) 15:331349 347

60. Pyati UJ, Look AT, Hammerschmidt M (2007) Zebrafish as a phosphorylation and proteasome-dependent degradation of the
powerful vertebrate model system for in vivo studies of cell BH3-only protein, Bim. J Biol Chem 278:1881118816
death. Semin Cancer Biol 17:154165 80. Qi XJ, Wildey GM, Howe PH (2006) Evidence that Ser87 of
61. Robu ME, Larson JD, Nasevicius A et al (2007) p53 activation BimEL is phosphorylated by Akt and regulates BimEL apop-
by knockdown technologies. PLoS Genet 3:e78 totic function. J Biol Chem 281:813823
62. Aouacheria A, Brunet F, Gouy M (2005) Phylogenomics of life- 81. Puthalakath H, Huang DC, OReilly LA, King SM, Strasser A
or-death switches in multicellular animals: Bcl-2, BH3-Only, (1999) The proapoptotic activity of the Bcl-2 family member
and BNip families of apoptotic regulators. Mol Biol Evol Bim is regulated by interaction with the dynein motor complex.
22:23952416 Mol Cell 3:287296
63. Cory S, Adams JM (2002) The Bcl2 family: regulators of the 82. Li H, Zhu H, Xu C-J, Yuan J (1998) Cleavage of BID by caspase
cellular life-or-death switch. Nat Rev Cancer 2:647656 8 mediates the mitochondrial damage in the Fas pathway to
64. Puthalakath H, Strasser A (2002) Keeping killers on a tight apoptosis. Cell 94:491501
leash: transcriptional and post-translational control of the pro- 83. Rinkenberger JL, Horning S, Klocke B, Roth K, Korsmeyer SJ
apoptotic activity of BH3-only proteins. Cell Death Differ (2000) Mcl-1 deficiency results in peri-implantation embryonic
9:505512 lethality. Genes Dev 14:2327
65. Strasser A (2005) The role of BH3-only proteins in the immune 84. Nakayama K, Negishi I, Kuida K, Sawa H, Loh DY (1994)
system. Nat Rev Immunol 5:189200 Targeted disruption of Bcl-2 alpha beta in mice: occurrence of
66. Jette CA, Flanagan AM, Ryan J et al (2008) BIM and other gray hair, polycystic kidney disease, and lymphocytopenia. Proc
BCL-2 family proteins exhibit cross-species conservation of Natl Acad Sci U S A 91:37003704
function between zebrafish and mammals. Cell Death Differ 85. Arnaud E, Ferri KF, Thibaut J et al (2006) The zebrafish bcl-2
15:10631072 homologue Nrz controls development during somitogenesis and
67. Coultas L, Huang DC, Adams JM, Strasser A (2002) Pro- gastrulation via apoptosis-dependent and -independent mecha-
apoptotic BH3-only Bcl-2 family members in vertebrate model nisms. Cell Death Differ 13:11281137
organisms suitable for genetic experimentation. Cell Death 86. Sidi S, Sanda T, Kennedy RD et al (2008) Chk1 suppresses a
Differ 9:11631166 caspase-2 apoptotic response to DNA damage that bypasses p53,
68. Chen MC, Gong HY, Cheng CY, Wang JP, Hong JR, Wu JL Bcl-2, and caspase-3. Cell 133:864877
(2001) Cloning and characterization of zfBLP1, a Bcl-XL 87. Certo M, Del Gaizo Moore V, Nishino M et al (2006) Mito-
homologue from the zebrafish, Danio rerio. Biochim Biophys chondria primed by death signals determine cellular addiction to
Acta 1519:127133 antiapoptotic BCL-2 family members. Cancer Cell 9:351365
69. Chen MC, Gong HY, Cheng CY, Wang JP, Hong JR, Wu JL 88. Chen L, Willis SN, Wei A et al (2005) Differential targeting of
(2000) Cloning and characterization of a novel nuclear Bcl-2 prosurvival Bcl-2 proteins by their BH3-only ligands allows
family protein, zfMcl-1a, in zebrafish embryo. Biochem Bio- complementary apoptotic function. Mol Cell 17:393403
phys Res Commun 279:725731 89. van Loo G, Saelens X, van Gurp M, MacFarlane M, Martin SJ,
70. Bae J, Hsu SY, Leo CP, Zell K, Hsueh AJ (2001) Under- Vandenabeele P (2002) The role of mitochondrial factors in
phosphorylated BAD interacts with diverse antiapoptotic Bcl-2 apoptosis: a Russian roulette with more than one bullet. Cell
family proteins to regulate apoptosis. Apoptosis 6:319330 Death Differ 9:10311042
71. Inohara N, Nunez G (2000) Genes with homology to mamma- 90. Valencia CA, Bailey C, Liu R (2007) Novel zebrafish caspase-3
lian apoptosis regulators identified in zebrafish. Cell Death substrates. Biochem Biophys Res Commun 361:311316
Differ 7:509510 91. Chakraborty C, Nandi SS, Sinha S, Gera VK (2006) Zebrafish
72. Doumanis J, Dorstyn L, Kumar S (2007) Molecular determinants caspase-3: molecular cloning, characterization, crystallization
of the subcellular localization of the Drosophila Bcl-2 homo- and phylogenetic analysis. Protein Pept Lett 13:633640
logues DEBCL and BUFFY. Cell Death Differ 14:907915 92. Masumoto J, Zhou W, Chen FF et al (2003) Caspy, a zebrafish
73. Schumacher B, Schertel C, Wittenburg N et al (2005) C. elegans caspase, activated by ASC oligomerization is required for pha-
ced-13 can promote apoptosis and is induced in response to ryngeal arch development. J Biol Chem 278:42684276
DNA damage. Cell Death Differ 12:153161 93. Fink SL, Cookson BT (2005) Apoptosis, pyroptosis, and
74. Wang WD, Huang CJ, Lu YF et al (2006) Heart-targeted necrosis: mechanistic description of dead and dying eukaryotic
overexpression of Nip3a in zebrafish embryos causes abnormal cells. Infect Immun 73:19071916
heart development and cardiac dysfunction. Biochem Biophys 94. Dubrez-Daloz L, Dupoux A, Cartier J (2008) IAPs: more than
Res Commun 347:979987 just inhibitors of apoptosis proteins. Cell Cycle 7:10361046
75. Jensen PJ, Gitlin JD, Carayannopoulos MO (2006) GLUT1 95. Ma A, Lin R, Chan PK et al (2007) The role of survivin in
deficiency links nutrient availability and apoptosis during angiogenesis during zebrafish embryonic development. BMC
embryonic development. J Biol Chem 281:1338213387 Dev Biol 7:50
76. Hsieh YC, Chang MS, Chen JY et al (2003) Cloning of zebrafish 96. Santoro MM, Samuel T, Mitchell T, Reed JC, Stainier DY
BAD, a BH3-only proapoptotic protein, whose overexpression (2007) Birc2 (cIap1) regulates endothelial cell integrity and
leads to apoptosis in COS-1 cells and zebrafish embryos. Bio- blood vessel homeostasis. Nat Genet 39:13971402
chem Biophys Res Commun 304:667675 97. Vince JE, Wong WW, Khan N et al (2007) IAP antagonists
77. Datta SR, Katsov A, Hu L et al (2000) 14-3-3 proteins and target cIAP1 to induce TNFalpha-dependent apoptosis. Cell
survival kinases cooperate to inactivate BAD by BH3 domain 131:682693
phosphorylation. Mol Cell 6:4151 98. Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS
78. Luciano F, Jacquel A, Colosetti P et al (2003) Phosphorylation Jr (1998) NF-kappaB antiapoptosis: induction of TRAF1 and
of Bim-EL by Erk1/2 on serine 69 promotes its degradation via TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation.
the proteasome pathway and regulates its proapoptotic function. Science 281:16801683
Oncogene 22:67856793 99. Mahoney DJ, Cheung HH, Mrad RL et al (2008) Both cIAP1
79. Ley R, Balmanno K, Hadfield K, Weston C, Cook SJ (2003) and cIAP2 regulate TNFalpha-mediated NF-kappaB activation.
Activation of the ERK1/2 signaling pathway promotes Proc Natl Acad Sci U S A 105:1177811783

123
348 Apoptosis (2010) 15:331349

100. Varfolomeev E, Goncharov T, Fedorova AV et al (2008) c-IAP1 121. Lee H, Kimelman D (2002) A dominant-negative form of p63 is
and c-IAP2 are critical mediators of tumor necrosis factor alpha required for epidermal proliferation in zebrafish. Dev Cell
(TNFalpha)-induced NF-kappaB activation. J Biol Chem 283: 2:607616
2429524299 122. Nowak M, Koster C, Hammerschmidt M (2005) Perp is required
101. Varfolomeev E, Blankenship JW, Wayson SM et al (2007) IAP for tissue-specific cell survival during zebrafish development.
antagonists induce autoubiquitination of c-IAPs, NF-kappaB Cell Death Differ 12:5264
activation, and TNFalpha-dependent apoptosis. Cell 131:669681 123. Yu J, Zhang L (2003) No PUMA, no death: implications for
102. Delvaeye M, De Vriese A, Zwerts F et al (2009) Role of the 2 p53-dependent apoptosis. Cancer Cell 4:248249
zebrafish survivin genes in vasculo-angiogenesis, neurogenesis, 124. Dornan D, Wertz I, Shimizu H et al (2004) The ubiquitin ligase
cardiogenesis and hematopoiesis. BMC Dev Biol 9:25 COP1 is a critical negative regulator of p53. Nature 429:8692
103. Ma AC, Chung MI, Liang R, Leung AY (2009) The role of 125. Leng RP, Lin Y, Ma W et al (2003) Pirh2, a p53-induced
survivin2 in primitive hematopoiesis during zebrafish develop- ubiquitin-protein ligase, promotes p53 degradation. Cell 112:
ment. Leukemia 23:712720 779791
104. Luttun A, Verhamme P (2008) Keeping your vascular integrity: 126. Huang Q, Raya A, DeJesus P et al (2004) Identification of p53
what can we learn from fish? Bioessays 30:418422 regulators by genome-wide functional analysis. Proc Natl Acad
105. Duval H, Johnson N, Li J et al (2007) Vascular development is Sci U S A 101:34563461
disrupted by endothelial cell-specific expression of the anti- 127. Le MT, Teh C, Shyh-Chang N et al (2009) MicroRNA-125b is a
apoptotic protein Bcl-2. Angiogenesis 10:5568 novel negative regulator of p53. Genes Dev 23:862876
106. Conway EM, Pollefeyt S, Steiner-Mosonyi M et al (2002) 128. Chen J, Ng SM, Chang C et al (2009) p53 isoform delta113p53
Deficiency of survivin in transgenic mice exacerbates Fas- is a p53 target gene that antagonizes p53 apoptotic activity via
induced apoptosis via mitochondrial pathways. Gastroenterol- BclxL activation in zebrafish. Genes Dev 23:278290
ogy 123:619631 129. Abraham RT (2001) Cell cycle checkpoint signaling through the
107. Zwerts F, Lupu F, De Vriese A et al (2007) Lack of endothelial ATM and ATR kinases. Genes Dev 15:21772196
cell survivin causes embryonic defects in angiogenesis, cardio- 130. Kumar S (2007) Caspase function in programmed cell death.
genesis, and neural tube closure. Blood 109:47424752 Cell Death Differ 14:3243
108. Conte D, Holcik M, Lefebvre CA et al (2006) Inhibitor of 131. Tinel A, Tschopp J (2004) The PIDDosome, a protein complex
apoptosis protein cIAP2 is essential for lipopolysaccharide- implicated in activation of caspase-2 in response to genotoxic
induced macrophage survival. Mol Cell Biol 26:699708 stress. Science 304:843846
109. Conze DB, Albert L, Ferrick DA et al (2005) Posttranscriptional 132. Ashkenazi A, Dixit VM (1998) Death receptors: signaling and
downregulation of c-IAP2 by the ubiquitin protein ligase c-IAP1 modulation. Science 281:13051308
in vivo. Mol Cell Biol 25:33483356 133. Nikolaev A, McLaughlin T, OLeary DD, Tessier-Lavigne M
110. Vousden KH, Prives C (2009) Blinded by the light: the growing (2009) APP binds DR6 to trigger axon pruning and neuron death
complexity of p53. Cell 137:413431 via distinct caspases. Nature 457:981989
111. Fuster JJ, Sanz-Gonzalez SM, Moll UM, Andres V (2007) 134. Wu G, Burns T, Zhan Y, Alnemri E, El-Deiry W (1999)
Classic and novel roles of p53: prospects for anticancer therapy. Molecular cloning and functional analysis of the mouse homo-
Trends Mol Med 13:192199 logue of the KILLER/DR5 tumor necrosis factor-related apop-
112. Chipuk JE, Green DR (2006) Dissecting p53-dependent apop- tosis-inducing ligand (TRAIL) death receptor. Cancer Res
tosis. Cell Death Differ 13:9941002 59:27702775
113. Berghmans S, Murphey RD, Wienholds E et al (2005) tp53 135. Glenney GW, Wiens GD (2007) Early diversification of the
mutant zebrafish develop malignant peripheral nerve sheath TNF superfamily in teleosts: genomic characterization and
tumors. Proc Natl Acad Sci U S A 102:407412 expression analysis. J Immunol 178:79557973
114. Lee KC, Goh WL, Xu M et al (2008) Detection of the p53 136. Woods IG, Wilson C, Friedlander B et al (2005) The zebrafish
response in zebrafish embryos using new monoclonal antibodies. gene map defines ancestral vertebrate chromosomes. Genome
Oncogene 27:629640 Res 15:13071314
115. Liu TX, Howlett NG, Deng M et al (2003) Knockdown of 137. Savan R, Kono T, Igawa D, Sakai M (2005) A novel tumor
zebrafish Fancd2 causes developmental abnormalities via p53- necrosis factor (TNF) gene present in tandem with the TNF-
dependent apoptosis. Dev Cell 5:903914 alpha gene on the same chromosome in teleosts. Immunoge-
116. Yamaguchi M, Fujimori-Tonou N, Yoshimura Y, Kishi T, netics 57:140150
Okamoto H, Masai I (2008) Mutation of DNA primase causes 138. Pressley ME, Phelan PE 3rd, Witten PE, Mellon MT, Kim CH
extensive apoptosis of retinal neurons through the activation of (2005) Pathogenesis and inflammatory response to Edwardsiella
DNA damage checkpoint and tumor suppressor p53. Develop- tarda infection in the zebrafish. Dev Comp Immunol 29:501513
ment 135:12471257 139. Bobe J, Goetz FW (2001) Molecular cloning and expression of a
117. Fischer S, Prykhozhij S, Rau MJ, Neumann CJ (2007) Mutation TNF receptor and two TNF ligands in the fish ovary. Comp
of zebrafish caf-1b results in S phase arrest, defective differen- Biochem Physiol B Biochem Mol Biol 129:475481
tiation, and p53-mediated apoptosis during organogenesis. Cell 140. Hymowitz SG, OConnel MP, Ultsch MH et al (2000) A unique
Cycle 6:29622969 zinc-binding site revealed by a high-resolution X-ray structure
118. Plaster N, Sonntag C, Busse CE, Hammerschmidt M (2006) p53 of homotrimeric Apo2L/TRAIL. Biochemistry 39:633640
deficiency rescues apoptosis and differentiation of multiple cell 141. Long Q, Huang H, Shafizadeh E, Liu N, Lin S (2000) Stimu-
types in zebrafish flathead mutants deficient for zygotic DNA lation of erythropoiesis by inhibiting a new hematopoietic death
polymerase delta1. Cell Death Differ 13:223235 receptor in transgenic zebrafish. Nat Cell Biol 2:549552
119. Davidson W, Ren Q, Kari G, Kashi O, Dicker AP, Rodeck U 142. Kwan TT, Liang R, Verfaillie CM et al (2006) Regulation of
(2008) Inhibition of p73 function by Pifithrin-alpha as revealed primitive hematopoiesis in zebrafish embryos by the death
by studies in zebrafish embryos. Cell Cycle 7:12241230 receptor gene. Exp Hematol 34:2734
120. Rentzsch F, Kramer C, Hammerschmidt M (2003) Specific and 143. Jin Z, Li Y, Pitti R et al (2009) Cullin3-based polyubiquitination
conserved roles of TAp73 during zebrafish development. Gene and p62-dependent aggregation of caspase-8 mediate extrinsic
323:1930 apoptosis signaling. Cell 137:721735

123
Apoptosis (2010) 15:331349 349

144. Krueger A, Schmitz I, Baumann S, Krammer PH, Kirchhoff S 158. Nehme R, Conradt B (2008) Egl-1: a key activator of apoptotic
(2001) Cellular FLICE-inhibitory protein splice variants inhibit cell death in C. elegans. Oncogene 27(Suppl 1):S3040
different steps of caspase-8 activation at the CD95 death- 159. Krieser RJ, White K (2009) Inside an enigma: do mitochondria
inducing signaling complex. J Biol Chem 276:2063320640 contribute to cell death in Drosophila? Apoptosis 14:961968
145. Pobezinskaya YL, Kim YS, Choksi S et al (2008) The function 160. Hehlgans T, Pfeffer K (2005) The intriguing biology of the
of TRADD in signaling through tumor necrosis factor receptor 1 tumour necrosis factor/tumour necrosis factor receptor super-
and TRIF-dependent Toll-like receptors. Nat Immunol 9:1047 family: players, rules and the games. Immunology 115:120
1054 161. Kauppila S, Maaty WS, Chen P et al (2003) Eiger and its
146. Sakata S, Yan Y, Satou Y et al (2007) Conserved function of receptor, Wengen, comprise a TNF-like system in Drosophila.
caspase-8 in apoptosis during bony fish evolution. Gene Oncogene 22:48604867
396:134148 162. Hay BA, Guo M (2006) Caspase-dependent cell death in Dro-
147. Chun HJ, Zheng L, Ahmad M et al (2002) Pleiotropic defects in sophila. Annu Rev Cell Dev Biol 22:623650
lymphocyte activation caused by caspase-8 mutations lead to 163. Sevrioukov EA, Burr J, Huang EW et al (2007) Drosophila Bcl-
human immunodeficiency. Nature 419:395399 2 proteins participate in stress-induced apoptosis, but are not
148. Strasser A, Jost PJ, Nagata S (2009) The many roles of FAS required for normal development. Genesis 45:184193
receptor signaling in the immune system. Immunity 30:180192 164. Yu X, Wang L, Acehan D, Wang X, Akey CW (2006) Three-
149. Falschlehner C, Schaefer U, Walczak H (2009) Following dimensional structure of a double apoptosome formed by the
TRAILs path in the immune system. Immunology 127:145154 Drosophila Apaf-1 related killer. J Mol Biol 355:577589
150. Trede NS, Langenau DM, Traver D, Look AT, Zon LI (2004) 165. Bilak A, Su TT (2009) Regulation of Drosophila melanogaster
The use of zebrafish to understand immunity. Immunity 20:367 pro-apoptotic gene hid. Apoptosis 14:943949
379 166. Igaki T, Pastor-Pareja JC, Aonuma H, Miura M, Xu T (2009)
151. Langenau DM, Jette C, Berghmans S et al (2005) Suppression of Intrinsic tumor suppression and epithelial maintenance by
apoptosis by bcl-2 overexpression in lymphoid cells of trans- endocytic activation of Eiger/TNF signaling in Drosophila. Dev
genic zebrafish. Blood 105:32783285 Cell 16:458465
152. Williams JA, Holder N (2000) Cell turnover in neuromasts of 167. Kuranaga E, Miura M (2007) Nonapoptotic functions of casp-
zebrafish larvae. Hear Res 143:171181 ases: caspases as regulatory molecules for immunity and cell-
153. Reich A, Spering C, Schulz JB (2008) Death receptor Fas fate determination. Trends Cell Biol 17:135144
(CD95) signaling in the central nervous system: tuning neuro- 168. Krumschnabel G, Podrabsky JE (2009) Fish as model systems
plasticity? Trends Neurosci 31:478486 for the study of vertebrate apoptosis. Apoptosis 14:121
154. Peden E, Killian DJ, Xue D (2008) Cell death specification in C. 169. dos Santos NM, do Vale A, Reis MI, Silva MT (2008) Fish and
elegans. Cell Cycle 7:24792484 apoptosis: molecules and pathways. Curr Pharm Des 14:148169
155. Mollereau B (2009) Cell death: what can we learn from flies? 170. Zon LI, Peterson RT (2005) In vivo drug discovery in the
editorial for the special review issue on Drosophila apoptosis. zebrafish. Nat Rev Drug Discov 4:3544
Apoptosis 14:929934 171. Eimon PM, Rubinstein AL (2009) The use of in vivo zebrafish
156. Wang C, Youle RJ (2009) The role of mitochondria in apoptosis. assays in drug toxicity screening. Expert Opin Drug Metab
Annu Rev Genet 43:95118 Toxicol 5:393401
157. Oberst A, Bender C, Green DR (2008) Living with death: the 172. Sambrook JG, Figueroa F, Beck S (2005) A genome-wide sur-
evolution of the mitochondrial pathway of apoptosis in animals. vey of Major Histocompatibility Complex (MHC) genes and
Cell Death Differ 15:11391146 their paralogues in zebrafish. BMC Genomics 6:152

123

Vous aimerez peut-être aussi