Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Current Biology

Review

Just So Stories about the Evolution of Apoptosis


Douglas R. Green* and Patrick Fitzgerald
Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
*Correspondence: douglas.green@stjude.org
http://dx.doi.org/10.1016/j.cub.2016.05.023

Apoptosis is a form of active cell death engaged by developmental cues as well as many different cellular
stresses in which the dying cell essentially ‘packages’ itself for removal. The process of apoptotic cell death,
as defined at the molecular level, is unique to the Metazoa (animals). Yet active cell death exists in non-animal
organisms, and in some cases molecules involved in such death show some sequence similarities to those
involved in apoptosis, leading to extensive speculation regarding the evolution of apoptosis. Here, we
examine such speculation from the perspective of the functional properties of molecules of the mitochondrial
apoptotic cell death pathway. We suggest scenarios for the evolution of one pathway of apoptosis, the mito-
chondrial pathway, and consider how they might be tested. We conclude with a ‘Just So Story’ of how the
mitochondrial pathway of apoptosis might have evolved during eukaryotic evolution.

Introduction: Just So Stories First Principles


‘‘‘Explain! Explain! Explain!’’’ How the First Letter Was Written1 ‘‘‘Ah!’ said Tegumai. ‘Will that do to begin with?’’’ How the Alpha-
Since Darwin, evolutionary biologists have engaged in bet Was Made
rigorous, and often not-so-rigorous, speculation into the selec- Much of the speculation into the evolution of molecular path-
tive events that favored the emergence of characteristics associ- ways is based on the principle of homology and the relationships
ated with living things. More recently, these have been extended between the sequences of genes or proteins. As discussed
to molecular events that function at the cellular level. With the below, there are caveats to drawing conclusions from such
elucidation of genome sequences and the development of ana- limited information without more detailed knowledge of how
lytic tools to compare these sequences, many biologists with the proteins encoded by these genes interact and function.
only a passing knowledge of evolutionary theory have felt free This is the position taken herein: a consideration of protein func-
to elaborate on how molecular pathways and processes evolved, tion beyond sequence similarity can alter our views (and our Just
often with minimal information beyond protein sequence. In the So Stories) about how a pathway may have evolved. Indeed, by
discussion presented herein, we make no claims regarding the such exploration of protein function, we can begin to test our
robustness of the analysis or the rigor of the speculation, as we stories.
are firmly seated among those with, at best, only a superficial An important (and, in retrospect, obvious) precept in any
appreciation of evolutionary concepts. Those with a deeper un- discussion of evolution is that all currently existing organisms
derstanding should feel free to criticize or disregard these ideas, and, by extension, biological molecules have ‘evolved’ for pre-
but for those who, like us, have an interest in cell death mecha- cisely the same period of time. In considering genes from two
nisms and wonder about their origins, the following discussion different organisms, we do not state that ‘X evolved from Y’,
might be viewed as a joint exploration into possibilities. but rather can speculate that ‘X and Y had a common ancestor’.
A ‘Just So Story’ is an untestable and thus unfalsifiable As discussed below, this is a critical point that allows us to
idea, such as a scenario for the evolution of a biological speculate that a function of a particular protein present in one or-
trait or process. It is just that, a story. But, as suggested else- ganism may have been lost in another, despite similarity in their
where (http://epjournal.net/blog/2012/09/just-so-stories-are-bad- sequences. It is also important to point out that sequence
explanations-functions-are-much-better-explanations/), ‘‘the similarity is not proof of homology, and that similar genes may
goal should not be to expel stories from science, but rather to be homologs, orthologs, or paralogs, with different evolutionary
identify the stories that are also good explanations’’. Here, we relationships.
will explore some stories specifically relating to the evolution of One further principle relates specifically to our consideration of
one particular type of cell death — apoptosis — and how they apoptosis. This cell death process has been broadly defined by
might be investigated (if not falsified). In the process, we discuss morphology; animal (metazoan) cells that die by apoptosis lose
at least one bewildering paradox that lies at the heart of the evo- volume, display condensation of nuclear chromatin, sustain
lution of a major pathway of apoptosis in animals and consider a structures of intracellular organelles (such as mitochondria and
possible resolution. endoplasmic reticulum), often show protrusions from the plasma
It is important to note that this is not the first foray into the evo- membrane (blebbing), and frequently fragment the nucleus and/
lution of cell death, and several excellent reviews should be or cell body. This has led to a search for similar morphologies
examined by the interested reader [1–3]. in dying non-metazoan cells, with subsequent evolutionary
speculation. More recently, however, a molecular definition of
apoptosis has been proposed [4], and it is this definition (elabo-
1All quotes are from Rudyard Kipling, Just So Stories. rated below) that is employed herein.

R620 Current Biology 26, R620–R627, July 11, 2016 ª 2016 Elsevier Ltd.
Current Biology

Review

A Chordates, echinoderms, platyhelminths B Nematodes C Drosophila

Mitochondrion Mitochondrion Mitochondrion

BCL-2 Apoptosome
APAF1
family BH3
MOMP BCL-2
APAF1
BH3 Cyt c family
Apoptosome
Apoptosome
IAP
IAP IAPi
Initiator caspase
IAPi

Initiator Caspase
caspase
* *
* * *
Executioner
caspase Executioner
caspase

Current Biology

Figure 1. Variations on a theme: the mitochondrial pathway of apoptosis.


(A) In chordates, echinoderms, platyhelminths, and possibly other phyla, apoptotic conditions engage the members of the Bcl-2 family, which either promote
(green) or inhibit (red) MOMP in response to activation of BH3-only proteins. MOMP results in the release of cytochrome c (cyt c) and other proteins of the
intermembrane space. Cytochrome c binds to APAF1, which oligomerizes to create the apoptosome that in turn binds to and activates an initiator caspase. The
initiator caspase cleaves and thereby activates executioner caspases. Both the initiator and executioner caspases are inhibited by an inhibitor of apoptosis
protein (IAP). MOMP releases antagonists of the IAP (IAPi), permitting the executioner caspases to orchestrate apoptosis. Asterisks (*) refer to active caspases.
(B) In nematodes, the anti-apoptotic Bcl-2 protein sequesters the APAF1 homolog. Apoptotic conditions induce expression of BH3-only proteins, which bind to
the Bcl-2 protein, releasing the APAF1 homolog to form an apoptosome. This binds and thereby activates the caspase to promote apoptosis. (C) In Drosophila,
the APAF1 homolog appears to spontaneously form an apoptosome, but the initiator caspase is inhibited by an IAP. Apoptotic conditions promote expression of
IAPi, permitting the apoptosome to now activate the initiator caspase, which in turn cleaves and thereby activates executioner caspases, promoting apoptosis.
Note that MOMP, upstream of caspase activation, occurs only in the pathway shown in (A).

Apoptosis, with a Focus on the Mitochondrial Pathway (‘induced proximity’ [9,11]), rather than by cleavage. (Although
‘‘Hear and attend and listen; for this befell and behappened and initiator caspases undergo auto-cleavage upon activation, this
became and was.’’ The Cat that Walked by Himself is not the activating event, and cleavage of an initiator caspase
Apoptosis, as a defined molecular process, is a type of cell is not proof of its activation [12,13].)
death unique to metazoans. The morphological characteristics The enforced dimerization of initiator caspases, leading to
of apoptosis are brought about by the actions of a set of cysteine their activation, occurs by the action of adapter proteins, which
proteases, the executioner caspases, which are only found in an- form an ‘activation platform’ that binds to interaction regions in
imals [5]. When activated, the executioner caspases cleave the prodomain of the caspase. An apoptotic pathway is defined
approximately a thousand substrates, including several that by the mode of formation of the caspase activation platform and
have been identified as mediating the characteristic changes in the identity of the initiator caspase recruited by the adapter.
the dying cell, including membrane blebbing, DNA fragmenta- Here, we focus on the so-called intrinsic or mitochondrial
tion, phosphatidylserine externalization (which promotes clear- pathway of apoptosis.
ance by phagocytes), loss of electron transport function in In the mitochondrial pathway (Figure 1), the initiator caspase,
mitochondria, and other events [6–8]. caspase-9, is activated when it binds to its activation platform
Executioner caspases, in their inactive form, are dimers that (the apoptosome), composed of an oligomer of the adapter
become activated upon cleavage at a site located between the protein APAF1. Inactive APAF1 is a monomer that oligomerizes
resulting large and small subunits of the active protease [9]. In upon interaction with cytochrome c, which is normally seq-
the majority of cases (the exception being cleavage by the serine uestered in the mitochondrial intermembrane space. During
protease granzyme B, which is introduced into cells by cyto- apoptosis, the proteins of the intermembrane space are released
toxic lymphocytes [10]), this activating cleavage is mediated by to the cytosol upon mitochondrial outer membrane permeabili-
initiator caspases. Unlike the executioner caspases, initiator cas- zation (MOMP). These include cytochrome c, as well as pro-
pases are monomeric and are activated by enforced dimerization teins (including Smac/Diablo and Omi/Htra2) that function to

Current Biology 26, R620–R627, July 11, 2016 R621


Current Biology

Review

inactivate an inhibitor of active executioner caspases, X-IAP [14]. identified that is cleaved by either protease and has been sug-
The latter role for MOMP links the mitochondrial pathway to gested to participate in cell death [26]. Such findings have led
other apoptotic pathways. It will be important to note that, while to the idea that apoptosis and these death processes in non-
animal cytochrome c activates APAF1 (indeed, it appears that metazoan organisms have a common ancestor [27].
any animal cytochrome c will activate mammalian APAF1 [15]), There is a difficulty with this idea, however. Despite some
apocytochrome c, which lacks the heme group, does not acti- sequence and structural similarity between caspases and meta-
vate APAF1, nor does cytochrome c from non-animal sources, caspases, their biochemistry is fundamentally different. The prin-
such as yeast. ciple of induced proximity, discussed above, is central to the way
The process of MOMP is mediated by the action of members in which initiator caspases are activated by the platforms
of the BCL-2 protein family. These include the effectors of composed of their adapters, but metacaspases are monomers
MOMP (e.g. BAX and BAK), which upon activation cause the and are not activated by dimerization [5]. Therefore, recruitment
permeabilization of the mitochondrial outer membrane. Anti- to an adapter protein, even one similar to APAF1, should not acti-
apoptotic BCL-2 proteins (e.g., BCL-2, BCL-xL, and MCL-1) pre- vate a metacaspase. The functional pathways of apoptosis,
vent MOMP, binding to the activated effectors or to the proteins involving the interactions between adapters, initiator caspases,
that activate them. A third sub-family, the BH3-only proteins, and executioner caspases, are therefore unlikely to be paralleled
either activate the effectors (e.g., BID, BIM, and PUMA) and/or by the putative homologs in plants or yeast. If caspases and
neutralize the anti-apoptotic proteins (e.g., BAD, BMF, HRK, metacaspases have a common molecular ancestor, there is no
PUMA, and NOXA) [16]. During cellular stress (or other pro- evidence that their mechanisms of activation are the same. Of
apoptotic conditions) changes in the expression and/or function course this position could change if we were to gain an under-
of the BH3-only proteins initiate the process of MOMP via such standing of whether and how molecular relatives of APAF1 might
interactions. Even if caspase activation downstream of MOMP activate metacaspases in plant cell death (an idea for which there
is blocked or disrupted, the process of MOMP can condemn is currently no supporting evidence) [19].
cells to death (albeit non-apoptotic death) [17], although cells
can survive MOMP in some situations [14,18]. Functional Similarities in the Mitochondrial Pathway of
All of the components of the mitochondrial pathway of Apoptosis
apoptosis have homologs throughout the animals, but, to date, ‘‘Make me different from all other animals by five this afternoon.’’
functional homologs of these proteins have not been convinc- The Sing-Song of Old Man Kangaroo
ingly demonstrated outside of metazoans. Before engaging The key molecular components of the mitochondrial pathway
in a discussion of these homologs and their functions, however, of apoptosis in mammals, as outlined above, are proteins of the
it may be useful to examine claims regarding apoptosis in BCL-2 family, cytochrome c, APAF1, an initiator caspase (cas-
non-metazoans. pase-9), and executioner caspases (Figure 1). Attempts to
identify homologs of these proteins have met with success
Caspases versus Metacaspases: Non-Animal throughout many animal phyla. Homologs of chordate BCL-2
Apoptosis? proteins have been identified in poriferans (sponges) [28], cnidar-
‘‘Before the High and Far-Off Times, O my Best Beloved, came ians (Hydra) [29], platyhelminths (planaria) [15], arthropods (flies)
the Time of the Very Beginnings.’’ The Crab That Played with [30], and nematodes [31], among others [1]. Cytochrome c is
the Sea among the most highly conserved proteins, found in all animals
Active cell death processes are not unique to animals and (and eukaryotes, as well as bacteria). APAF1 homologs have
have been described in plants [19], yeast [20], protozoa [21], been described for representatives of several animal phyla,
and bacteria [22]. In some cases, attempts have been made to and can even be found in the placozoans (Trichoplax) (see Fig-
relate these events to apoptosis, sometimes implying an evolu- ures S1–S3 in the Supplemental Information), which may repre-
tionary relationship. While that view will be challenged here, we sent the most basal animal phylum (although this is debatable
contend that the study of such non-animal cell death has value [32]). And a great many caspases, including caspases with pro-
in its own right, both as a way to understand the life and death domains containing protein interaction motifs similar to the pro-
of those organisms (and cells in the case of plants) and perhaps domain of caspase-9, have been widely identified in animal
to elucidate general principles of active cell death. phyla. Based on these considerations, it is widely held that the
Caspase proteases are not found outside of the animals; mitochondrial pathway of apoptosis, or at least the proteins
therefore, if apoptosis is strictly defined as involving the activa- that comprise it, arose with the animals.
tion and function of caspases, apoptosis does not exist outside It is when we consider the biochemical functions of these pro-
the animals. However, a family of proteases that are related to teins in apoptosis that the picture becomes much more murky.
caspases — the metacaspases — have been implicated in cell One example is the relationships between cytochrome c,
death in plants and yeast [23]. (Another caspase-like protease APAF1, and caspase-9. In those cases that have been examined
type — the paracaspases, present in animals as well as in functionally, the interaction between APAF1-like molecules and
Dictyostelium — is not involved in cell death in any organism, caspase-9-like molecules results in activation of the caspase,
including Dictyostelium, which lacks an apoptosis-like cell death as observed in arthropods [33], nematodes [34], and chordates
[24].) Further, in plants, some cell death processes appear to [35] (Figure 1). The role of cytochrome c is more complex. While
involve proteins related to the adapter APAF1 [25]. While meta- in chordates cytochrome c is required to activate APAF1 to form
caspases cleave substrates after an arginine residue, unlike the activation platform for caspase-9, this is not the case for
caspases, which cleave after aspartate, a substrate has been nematodes, where the APAF1 homolog, CED-4, lacks the region

R622 Current Biology 26, R620–R627, July 11, 2016


Current Biology

Review

of the molecule required for cytochrome c interaction [34,36]. In to interact with cytochrome c may have been ‘lost’. This may
arthropods (e.g. Drosophila), the APAF1 homolog has this re- also be the case in arthropods, some of which maintain the cyto-
gion, but the evidence that cytochrome c activates this APAF1 chrome c-binding (WD) domain (Figures S1–S3) but have an
homolog is contentious; some studies suggest that cytochrome APAF1-like molecule that does not appear to interact with cyto-
c mutants can be identified that are defective in apoptosis [37], chrome c. This idea only has validity if the common ancestor of
while in vitro studies in Drosophila cells failed to find a role for platyhelminths, nematodes, and arthropods possessed an
cytochrome c in caspase activation [38–41]. In contrast, cyto- APAF1 that was activated by holocytochrome c; the alternative
solic extracts from platyhelminths (planaria) and echinoderms proposal is that cytochrome c-mediated APAF1 activation arose
(sea urchins and sand dollars) displayed robust caspase activa- only after such a ‘split’.
tion in response to cytochrome c addition [15], suggesting that Can we reconcile these differences, which, despite sequence
APAF1 in such animals is activated by cytochrome c. similarity and the same ultimate effect, suggest very different
In chordates, the BCL-2 proteins regulate and effect MOMP, molecular pathways? That is, can we tell a Just So Story to
which is required for the release of cytochrome c into the cytosol explain how apoptosis evolved in the animals? And can it be
to activate APAF1 (as discussed above). Although BCL-2 pro- tested? To approach this, it may first help to explore a form of
teins have been found throughout the animals, as noted, the cell death that is related to apoptosis, but different. This is the
involvement of MOMP is less clear. During apoptosis in platyhel- process of pyroptosis.
minths, cytochrome c release (indicative of MOMP) can be
observed [15], but no MOMP is seen during apoptosis in nema- Pyroptosis, a Caspase-Dependent Cell Death Distinct
todes [42]. Consistent with these observations, a platyhelminth from Apoptosis
BCL-2 family protein from planaria has MOMP activity [15], while ‘‘It was all his ’satiable curiosity.’’ The Elephant’s Child
the single nematode BCL-2 protein in Caenorhabditis elegans Cell death by pyroptosis is a non-apoptotic cell death, more
does not [12], and instead has an anti-apoptotic function unre- closely allied to necrosis. Like apoptosis, however, it is caspase-
lated to MOMP (see below). In Drosophila, cytochrome c release dependent, involving the activation of either caspase-1 or a related
is observed during apoptosis, but this release depends upon caspase (caspase-11 in rodents, caspase-4 or -5 in primates)
the function of caspases [43], unlike most cases of MOMP in (Figure 2). In general, pyroptosis occurs in response to infection.
chordate cells. The activation of caspases-4, -5, and -11 is distinct from that
Arthropod BCL-2 proteins, to date, have not been observed to of other monomeric caspases in that the adapter responsible for
regulate apoptosis directly [44], although they may have indirect activation by induced proximity is not encoded by the host
effects [45,46]. Instead, it appears that apoptosis in the arthro- genome, but rather by intracellular bacteria. Lipopolysaccharide
pods takes a different path. The Drosophila APAF1 homolog, derived from the outer membrane of Gram-negative bacteria
ARK, appears to be constitutively active, but the activation of directly engages the prodomains of these caspases, causing
the initiator caspase, DRONC, is held in check by an inhibitor, their activation [49]. The caspase then cleaves a protein called
DIAP1 (related to X-IAP in mammals, see above). Upon Gasdermin d, which in its cleaved form kills the cell (by a mech-
apoptosis induction, proteins that antagonize DIAP1 are ex- anism that is yet to be elucidated) [50,51].
pressed, releasing active DRONC to proceed with executioner Intracellular DNA viruses can trigger pyroptosis via another
caspase activation and cell death [47]. These proteins share a mechanism, involving the engagement of a cytosolic DNA
functional motif with mammalian antagonists of X-IAP, but, un- sensor, AIM2, which oligomerizes to create an activation plat-
like the mammalian proteins, they are not sequestered in the form for caspase-1. Such activation platforms for caspase-1
mitochondrial intermembrane space. are termed inflammasomes because caspase-1 cleaves the
A paradox emerges when we compare the functions of BCL-2 proforms of the inflammatory cytokines interleukin-1 and inter-
proteins in nematodes and chordates. In C. elegans, the anti- leukin-18, as well as Gasdermin d. While the cleavage of Gasder-
apoptotic BCL-2 protein, CED-9, directly binds to the APAF1 min d by caspase-1 contributes to rapid cell death, cells lacking
homolog, CED-4, preventing its function [48]. The BH3-only pro- Gasdermin d nevertheless are still able to die in response to cas-
tein Egl-1 disrupts this interaction, permitting CED-4 to bind and pase-1 activation [50,51].
activate the caspase CED-3. In contrast, anti-apoptotic BCL-2 Unlike other monomeric caspases, caspase-1 has several
proteins in chordates do not inhibit APAF1 or the activation of activation platforms. Most of these involve proteins of the Nod-
caspase-9, but instead prevent MOMP, which is needed for cy- like receptor (NLR) family, which includes several involved in
tochrome c and APAF1 activation. Despite the similarities be- caspase-1 activation, such as NLRP1, NLRP3, and NLRC4.
tween the proteins and the overall effect (i.e., in both cases These proteins are activated to oligomerize in response to
anti-apoptotic BCL-2 proteins prevent APAF1 activation), the different bacterial products and form a complex with another
biochemistry is distinct. It is important to note that there is adapter, ASC, which then binds to and activates caspase-1
nothing paradoxical about a protein assuming different functions (Figure 2). In addition, extracellular bacterial products, and other
in different organisms; what we suggest is surprising is that a conditions produced by bacterial infection, can trigger the acti-
superficial function (inhibition of caspase activation by BCL-2 vation of NLRP3 via other mechanisms, leading to caspase-1
proteins) appears to be achieved via fundamentally different activation and pyroptosis.
biochemical mechanisms (inhibition of MOMP versus direct inhi- Many NLR proteins are described in mammals, but most have
bition of APAF1). unknown functions, or known functions that do not include activa-
Comparing APAF1 homologs from different animal phyla, it tion of caspase-1. Strikingly, APAF1 is often considered a member
seems likely that in nematodes the ability of the APAF1 protein of the NLR family, on the basis of sequence similarity and domain

Current Biology 26, R620–R627, July 11, 2016 R623


Current Biology

Review
Figure 2. Variations on a theme: caspase
A LPS-mediated B Inflammasome- C Apoptosome- mediated activation platforms.
activation mediated activation activation (A) Some caspases (caspase-4, caspase-5, cas-
pase-11) bind directly to intracellular lipopolysac-
charides (LPS), causing caspase dimerization
Cyt c and, hence, activation. (B) An example of an
NLRP3 inflammasome is shown. NLRP3 is activated to
LPS APAF1 oligomerize under conditions, for example, in
response to a decrease in intracellular K+ con-
Caspase
Apoptosome centrations. NLRP3 binds to an adaptor protein,
K+ efflux which in turn binds to caspase-1, resulting in its
activation. (C) The activation of APAF1 by cyto-
chrome c, as described in Figure 1A. Asterisks (*)
refer to active caspases.

* Caspase
Initiator
caspase
Adapter leading to the death of the cell. Given the
* * nature of the pyroptotic caspases-4, -5,
and -11, we can envision that the mecha-
nism was a simple one, based on the ‘in-
vention’ of a monomeric protease that
was activated upon binding to a bacterial
product not found in the Archaea. If in-
Inflammasome
fected, this cell died, thereby sparing its
clone mates the subsequent infection by
Current Biology the replicating pathogen. This would lead
to a Red Queen scenario, in which the in-
structure. While it is possible that APAF1 arose independently of fecting organism evolves mechanisms to avoid the Archaea’s
the NLRs found in inflammasomes (e.g., by convergent evolution), cell death mechanism and, in turn, the Archaea evolves strategies
for the purposes of our Just So Story below we will consider to counter these evasions. These counter-strategies included the
APAF1 and NLRs to be evolutionarily related molecules. emergence of adapters that would recognize a range of bacterial
NLR proteins are found not only throughout the animals, but also products, lending increased surveillance to the mechanism. This
in plants: these are the plant APAF1-related proteins mentioned might have been an ancestor of the inflammasomes. We will return
above. In many animals, there is a bewildering number of NLR pro- to this escalation, below.
teins, far beyond the large numbers identified in chordates. Among the unique bacterial products recognized by these
A cell death response to intracellular infection is an obvious adapters was cytochrome c and the proteins needed for its
way in which obligate intracellular parasites can be controlled maturation, present in bacteria and absent in many of the
by removing the infected cell before the invader can replicate. Archaea [53]. Here it may be important to note that, while a cyto-
Indeed, this can help to explain active cell death in single-celled chrome c-like protein may have been present, the adapter in our
organisms. We have previously argued [52] that an active cell story was specialized to recognize the mature form of cyto-
death mechanism that might be envisioned to allow single-celled chrome c, complete with heme. Most Archaea lack the machin-
organisms, as a group, to survive limited nutrient availability ery to construct this holocytochrome c [53]. (As an aside, it is
(a scenario that is often invoked in discussions of the evolution worth remembering that APAF1 in animals recognizes only holo-
of cell death) is not an evolutionarily stable strategy, as ‘cheaters’ cytochrome c.)
that lose the cell death mechanism will inevitably be favored by At some point, an a-purple bacterium resembling Rickettsia
selection. However, if we consider a pathogen that has adapted [54] invaded a member of the Archaea, possibly of the ‘TACK’
to infect a single-celled organism and therefore, by extension, its superphylum [55], and, rather than the latter dying, a symbiosis
clone mates, death of the infected cell is now stable; an individ- arose that led to the formation of mitochondria. This symbiosis
ual that loses the cell death mechanism is no longer favored. hypothesis for the evolution of mitochondria has remarkable
These considerations and the observations above lead us to support and is widely accepted [56].
our Just So Story. But for our purposes we might suppose that it was not a ‘per-
fect union’. While under some circumstances the proto-mito-
How the Animal’s Cells Learned to Die chondria and the Archaea that housed it might both gain tremen-
‘‘‘I don’t think it was at all like that,’ said Painted Jaguar, but he dous selective advantages, in other situations dissolution of the
felt a little puzzled; ‘but, please, say it again more distinctly.’’’ The symbiosis might favor the proto-mitochondria, at least those that
Beginning of the Armadillos had sustained their ability to replicate outside the host. In that
While it is tempting to begin our story with ‘once upon a very long setting, the proto-mitochondria might engage a strategy com-
time ago’, we will spare the reader this indignity. Nevertheless, mon to bacteria — the production of pore-forming toxins. These
what follows is only a story, by necessity. We begin with an would target the nearest membrane, i.e. that surrounding the
assumption that, at some early point in the evolution of eukaryotes, bacteria, and induce lysis. Proteins that exist between the inner
a mechanism existed for the recognition of intracellular infection, (bacterial) and outer (host) membrane would then be released to

R624 Current Biology 26, R620–R627, July 11, 2016


Current Biology

Review

the cytosol. Some of these (perhaps including holocytochrome Once we examine animals, however, we see not only caspases
c) might then trigger the proto-inflammasome response in the and APAF1-like proteins, but often a radiation of the genes encod-
host, resulting in its death. ing them (so that an organism may have many). An example is in
A component of the symbiosis hypothesis is that genes pre- the basal Placozoa (Figure S4), or in the echinoderm Strongylo-
sent in the symbiont somehow ‘transfer’ to the nucleus of the centrotus purpuratus, where a cursory search revealed 76
host, coming under the host’s control and thus enforcing the possible caspases (not shown). Based on our considerations
symbiosis [56]. Among these might be the toxins, normally pro- above, it is tempting to suggest that many of these are involved
duced by the bacteria. In our story, these would now be host pro- in the recognition of intracellular invaders, although at this
teins, and would function as the BCL-2 protein family effector time we do not know if any caspases (beyond mammalian cas-
proteins, capable of permeabilizing the mitochondrial outer pases-4, -5, and -11) are activated upon binding to bacterial
membranes to effect MOMP, release cytochrome c, and activate products.
a specialized ‘inflammasome’ (the APAF1 apoptosome) that en- At this point, we have only our Just So Story, and the presence
gages caspase-9. of the pathway we have described throughout the animals. In the
The BCL-2 family effectors oligomerize via their BH3 regions to relatively basal platyhelminths, different BCL-2 proteins promote
cause MOMP [57]. Anti-apoptotic BCL-2 proteins bind to this re- [15] and prevent [61] MOMP and cell death, and cytochrome c
gion in activated effectors, thereby blocking MOMP. The BH3- promotes caspase activation [15]. Again, though, we have a
only proteins possess just the BH3 domain (hence the name) problem. If anti-apoptotic BCL-2 proteins function by blocking
and bind to the BH3-binding pockets of the effectors to activate effector oligomerization and MOMP, how did the situation in
them [57,58] or to the anti-apoptotic proteins to neutralize them. C. elegans arise, where the anti-apoptotic BCL-2 protein acts
Therefore, for our Just So Story we propose that the common by binding the APAF1 protein, which does not itself interact
ancestor of the BCL-2 proteins was a toxin resembling a pro- with cytochrome c (and no MOMP is required for its activation)?
apoptotic BCL-2 effector of MOMP. Like the pro-apoptotic effector proteins, where binding to an
Before moving on to discuss how this imaginary primordial activating BH3-only protein induces a conformational change
pathway of apoptosis diverged in the animals, we can ask if there in the former [57,58], the binding of the nematode BH3-only pro-
is any evidence in support of our story, whether there are funda- tein to the anti-apoptotic BCL-2 protein also induces a confor-
mental problems with it, and/or propose ways to test it. The first mational change, in this case releasing the APAF1-like protein
structural studies of anti-apoptotic BCL-2 proteins noted similar- [48]. How can these different functions for a similar molecular
ities in structure with bacterial pore-forming toxins [59], raising mechanism be reconciled?
the possibility that the proto-BCL-2 effector may have been One possibility is that a common ancestor of a BCL-2 pro-
such a toxin. If so, we have not found it in existing bacteria nor tein had both activities and could directly inhibit APAF1 as well
in the exceptionally large mitochondrial genomes of the Jakobid as regulate MOMP. If so, we can posit that one or the other func-
protozoans (which most closely resemble proteobacterial ge- tion might have been lost in the lineage leading to nematodes.
nomes [60]). The BCL-2 proteins of basal phyla, such as Cnidaria, Porifera,
Our story also features an APAF1-like sensor that creates the and Placozoa (Figure S5), have been identified, and it would
proto-caspase activation platform. Although NLR proteins are be interesting to determine whether any of these actually bind
found in non-metazoans (especially plants), the non-metazoan to and inhibit the corresponding APAF1-like proteins. Of course,
caspase-like proteins do not have the property of being acti- this is not a simple analysis, although it might be possible
vated on such platforms (as discussed above). Therefore, if our to determine the likelihood of such interactions using in silico
story is to have any relationship to reality, we must postulate approaches, examining whether the binding interactions deter-
that our proto-apoptotic pathway arose with a common ancestor mined structurally in the nematode proteins [48] might be
of the Metazoa. Choanoflagellates are thought to be the closest possible in the proteins from other organisms. The same ana-
living relatives of the Metazoa and, while their genomes encode lyses might be carried out for interactions between holocyto-
several proteins with similarities to caspases (https://www. chrome c and APAF1, which have also been elucidated at the
broadinstitute.org/annotation/genome/multicellularity_project/ structural level [36].
MultiHome.html), to date we do not know if any functional cas- Intriguingly, the BCL-2 protein of nematodes [62], and those of
pases exist in these organisms, nor whether these might have arthropods [43] and chordates [63], regulate mitochondrial dy-
the property of activation by induced proximity. namics, i.e., the fission and fusion of mitochondria. Further, mito-
Our Just So Story, however, posits that our primordial chondrial shape has been implicated in the induction of MOMP
pathway would have existed well before the origins of the Choa- by mammalian BCL-2 effectors [64]. A common mechanism
noflagellates and, indeed, during the emergence of eurkaryotes. may thus exist for the function of these proteins, whether to
If so, why do multicellular kingdoms other than the Metazoa cause MOMP or to sequester (and release) APAF1. At this point,
appear to lack a mitochondrial pathway of apoptosis? At this however, such a unified model is lacking.
point, compelling evidence for such a pathway does not exist
outside the animals, as we discussed. This may be a fatal flaw Conclusions and Perspectives
for our story. But then again, it is only a story. It may be that ves- ‘‘They are quite contented as they are.’’ How the Leopard Got His
tiges of such a pathway do exist outside the Metazoa, and Spots
remain to be found. Notwithstanding our previously discussed Our discussion of the evolution of cell death in animals focuses
issues with metacaspases, further investigation of their functions on the mitochondrial pathway of apoptosis, how its presence
in cell death may elaborate such vestiges. throughout the Metazoa takes different forms, and how it might

Current Biology 26, R620–R627, July 11, 2016 R625


Current Biology

Review

have evolved. In doing so, we have promoted one set of sce- 7. Green, D.R. (2011). Means to an End: Apoptosis and other Cell Death
Mechanisms (Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory
narios that does not take into account possible alternatives. Press).
For example, the apparent conservation of the death receptor
pathway of apoptosis in Cnidaria [65] raises the possibility that 8. Toda, S., Nishi, C., Yanagihashi, Y., Segawa, K., and Nagata, S. (2015).
Clearance of apoptotic cells and pyrenocytes. Curr. Top. Dev. Biol. 114,
the evolution of this pathway predated that of the mitochondrial 267–295.
pathway, which was then co-opted during the evolution of the
mitochondrial pathway. Further, other forms of cell death, such 9. Boatright, K.M., Renatus, M., Scott, F.L., Sperandio, S., Shin, H., Pedersen,
I.M., Ricci, J.E., Edris, W.A., Sutherlin, D.P., Green, D.R., et al. (2003). A uni-
as forms of programmed necrosis (active molecular mechanisms fied model for apical caspase activation. Mol. Cell 11, 529–541.
leading to necrotic cell death), which include mitochondrial
10. Ewen, C.L., Kane, K.P., and Bleackley, R.C. (2012). A quarter century of
mechanisms, are not considered in our discussion and may granzymes. Cell Death Differ. 19, 28–35.
well have had a prominent role in the emergence of apoptosis.
Most investigations into the evolution of cell death in the ani- 11. Salvesen, G.S., and Dixit, V.M. (1999). Caspase activation: the induced-
proximity model. Proc. Natl. Acad. Sci. USA 96, 10964–10967.
mals rely on sequence similarities at the protein level, with the
assumption that form specifies function. As we have discussed, 12. Delivani, P., Adrain, C., Taylor, R.C., Duriez, P.J., and Martin, S.J. (2006).
however, the biochemical mechanisms whereby a molecule may Role for CED-9 and Egl-1 as regulators of mitochondrial fission and fusion
dynamics. Mol. Cell 21, 761–773.
promote or inhibit cell death can be distinct in different animal
phyla. In some cases we have proposed that this represents a 13. McStay, G.P., Salvesen, G.S., and Green, D.R. (2008). Overlapping cleav-
age motif selectivity of caspases: implications for analysis of apoptotic
paradox, such that the superficial function of a protein (e.g., pathways. Cell Death Differ. 15, 322–331.
the function of anti-apoptotic BCL-2 proteins to prevent
apoptosis) can take different forms (e.g., inhibition of MOMP 14. Tait, S.W., and Green, D.R. (2010). Mitochondria and cell death: outer
membrane permeabilization and beyond. Nat. Rev. Mol. Cell Biol. 11,
versus direct inhibition of APAF1). While this might be ‘explained’ 621–632.
by gene duplication and divergence of biochemical function, the
source of the paradox is not why two similar proteins may func- 15. Bender, C.E., Fitzgerald, P., Tait, S.W., Llambi, F., McStay, G.P., Tupper,
D.O., Pellettieri, J., Sanchez Alvarado, A., Salvesen, G.S., and Green, D.R.
tion differently (which is not unexpected), but why the differing (2012). Mitochondrial pathway of apoptosis is ancestral in metazoans.
functions have the same superficial effect. Ultimately, the resolu- Proc. Natl. Acad. Sci. USA 109, 4904–4909.
tion of such paradoxes will reside in the detailed analysis of 16. Chipuk, J.E., Moldoveanu, T., Llambi, F., Parsons, M.J., and Green, D.R.
similar proteins from many phyla. While intellectually satisfying, (2010). The BCL-2 family reunion. Mol. Cell 37, 299–310.
such studies face challenges, not least of all in how they will be
17. Chipuk, J.E., and Green, D.R. (2005). Do inducers of apoptosis trigger cas-
regarded by funding agencies. pase-independent cell death? Nat. Rev. Mol. Cell. Biol. 6, 268–275.
The reader should not take our scenario for the evolution of the
18. Lopez, J., and Tait, S.W. (2015). Mitochondrial apoptosis: killing cancer
mitochondrial pathway of apoptosis too seriously. It is proposed
using the enemy within. Br. J. Cancer 112, 957–962.
as an exercise in how one might frame a story to raise questions,
and where the answers to those questions may lie. Ultimately, 19. Coll, N.S., Epple, P., and Dangl, J.L. (2011). Programmed cell death in the
plant immune system. Cell Death Differ. 18, 1247–1256.
our story will likely remain a Just So Story. But in the meantime,
we hope that the tale has been thought provoking, or at least 20. Strich, R. (2015). Programmed cell death initiation and execution in
entertaining. In the end, that is what stories are for. budding yeast. Genetics 200, 1003–1014.

21. Reece, S.E., Pollitt, L.C., Colegrave, N., and Gardner, A. (2011). The mean-
SUPPLEMENTAL INFORMATION ing of death: evolution and ecology of apoptosis in protozoan parasites.
PLoS Pathog. 7, e1002320.
Supplemental Information includes five supporting figures and can be found
22. Engelberg-Kulka, H., Amitai, S., Kolodkin-Gal, I., and Hazan, R. (2006).
with this article online at http://dx.doi.org/10.1016/j.cub.2016.05.023.
Bacterial programmed cell death and multicellular behavior in bacteria.
PLoS Genet. 2, e135.
REFERENCES
23. Salvesen, G.S., Hempel, A., and Coll, N.S. (2015). Protease signaling in an-
imal and plant regulated cell death. FEBS J. http://dx.doi.org/10.1111/
1. Zmasek, C.M., and Godzik, A. (2013). Evolution of the animal apoptosis
febs.13616.
network. Cold Spring Harb. Perspect. Biol. 5, a008649.

2. Munoz-Pinedo, C. (2012). Signaling pathways that regulate life and cell 24. Giusti, C., Luciani, M.F., and Golstein, P. (2010). A second signal for auto-
death: evolution of apoptosis in the context of self-defense. Adv. Exp. phagic cell death? Autophagy 6, 823–824.
Med. Biol. 738, 124–143.
25. Jacob, F., Vernaldi, S., and Maekawa, T. (2013). Evolution and conserva-
3. Ameisen, J.C. (2002). On the origin, evolution, and nature of programmed tion of plant NLR functions. Front. Immunol. 4, 297.
cell death: a timeline of four billion years. Cell Death Differ. 9, 367–393.
26. Sundstrom, J.F., Vaculova, A., Smertenko, A.P., Savenkov, E.I., Golovko,
4. Galluzzi, L., Vitale, I., Abrams, J.M., Alnemri, E.S., Baehrecke, E.H., Bla- A., Minina, E., Tiwari, B.S., Rodriguez-Nieto, S., Zamyatnin, A.A., Jr., Vali-
gosklonny, M.V., Dawson, T.M., Dawson, V.L., El-Deiry, W.S., Fulda, S., neva, T., et al. (2009). Tudor staphylococcal nuclease is an evolutionarily
et al. (2012). Molecular definitions of cell death subroutines: recommenda- conserved component of the programmed cell death degradome. Nat.
tions of the Nomenclature Committee on Cell Death 2012. Cell Death Cell Biol. 11, 1347–1354.
Differ. 19, 107–120.
27. Wilkinson, D., and Ramsdale, M. (2011). Proteases and caspase-like activ-
5. McLuskey, K., and Mottram, J.C. (2015). Comparative structural analysis ity in the yeast Saccharomyces cerevisiae. Biochem. Soc. Trans. 39,
of the caspase family with other clan CD cysteine peptidases. Biochem. J. 1502–1508.
466, 219–232.
28. Wiens, M., Diehl-Seifert, B., and Muller, W.E. (2001). Sponge Bcl-2 homol-
6. Crawford, E.D., and Wells, J.A. (2011). Caspase substrates and cellular re- ogous protein (BHP2-GC) confers distinct stress resistance to human
modeling. Annu. Rev. Biochem. 80, 1055–1087. HEK-293 cells. Cell Death Differ. 8, 887–898.

R626 Current Biology 26, R620–R627, July 11, 2016


Current Biology

Review
29. Lasi, M., Pauly, B., Schmidt, N., Cikala, M., Stiening, B., Kasbauer, T., Zen- 48. Yan, N., Chai, J., Lee, E.S., Gu, L., Liu, Q., He, J., Wu, J.W., Kokel, D., Li,
ner, G., Popp, T., Wagner, A., Knapp, R.T., et al. (2010). The molecular cell H., Hao, Q., et al. (2005). Structure of the CED-4-CED-9 complex provides
death machinery in the simple cnidarian Hydra includes an expanded cas- insights into programmed cell death in Caenorhabditis elegans. Nature
pase family and pro- and anti-apoptotic Bcl-2 proteins. Cell Res. 20, 437, 831–837.
812–825.
49. Shi, J., Zhao, Y., Wang, Y., Gao, W., Ding, J., Li, P., Hu, L., and Shao, F.
30. Doumanis, J., Dorstyn, L., and Kumar, S. (2007). Molecular determinants (2014). Inflammatory caspases are innate immune receptors for intracel-
of the subcellular localization of the Drosophila Bcl-2 homologues DEBCL lular LPS. Nature 514, 187–192.
and BUFFY. Cell Death Differ. 14, 907–915.
50. Shi, J., Zhao, Y., Wang, K., Shi, X., Wang, Y., Huang, H., Zhuang, Y., Cai,
31. Hengartner, M.O., and Horvitz, H.R. (1994). C. elegans cell survival gene T., Wang, F., and Shao, F. (2015). Cleavage of GSDMD by inflammatory
ced-9 encodes a functional homolog of the mammalian proto-oncogene caspases determines pyroptotic cell death. Nature 526, 660–665.
bcl-2. Cell 76, 665–676.
51. Kayagaki, N., Stowe, I.B., Lee, B.L., O’Rourke, K., Anderson, K., Warming,
32. Dohrmann, M., and Worheide, G. (2013). Novel scenarios of early animal S., Cuellar, T., Haley, B., Roose-Girma, M., Phung, Q.T., et al. (2015). Cas-
evolution–is it time to rewrite textbooks? Integr. Comp. Biol. 53, 503–511. pase-11 cleaves gasdermin D for non-canonical inflammasome signalling.
Nature 526, 666–671.
33. Pang, Y., Bai, X.C., Yan, C., Hao, Q., Chen, Z., Wang, J.W., Scheres, S.H.,
and Shi, Y. (2015). Structure of the apoptosome: mechanistic insights into 52. James, E.R., and Green, D.R. (2002). Infection and the origins of
activation of an initiator caspase from Drosophila. Genes Dev. 29, apoptosis. Cell Death Differ. 9, 355–357.
277–287.
53. Kletzin, A., Heimerl, T., Flechsler, J., van Niftrik, L., Rachel, R., and Klingl,
34. Huang, W., Jiang, T., Choi, W., Qi, S., Pang, Y., Hu, Q., Xu, Y., Gong, X., A. (2015). Cytochromes c in Archaea: distribution, maturation, cell archi-
Jeffrey, P.D., Wang, J., et al. (2013). Mechanistic insights into CED-4- tecture, and the special case of Ignicoccus hospitalis. Front. Microbiol.
mediated activation of CED-3. Genes Dev. 27, 2039–2048. 6, 439.

35. Zou, H., Henzel, W.J., Liu, X., Lutschg, A., and Wang, X. (1997). Apaf-1, a 54. Andersson, S.G., Zomorodipour, A., Andersson, J.O., Sicheritz-Ponten,
human protein homologous to C. elegans CED-4, participates in cyto- T., Alsmark, U.C., Podowski, R.M., Naslund, A.K., Eriksson, A.S., Winkler,
chrome c-dependent activation of caspase-3. Cell 90, 405–413. H.H., and Kurland, C.G. (1998). The genome sequence of Rickettsia pro-
wazekii and the origin of mitochondria. Nature 396, 133–140.
36. Zhou, M., Li, Y., Hu, Q., Bai, X.C., Huang, W., Yan, C., Scheres, S.H., and
Shi, Y. (2015). Atomic structure of the apoptosome: mechanism of cyto- 55. Guy, L., and Ettema, T.J. (2011). The archaeal ‘TACK’ superphylum and
chrome c- and dATP-mediated activation of Apaf-1. Genes Dev. 29, the origin of eukaryotes. Trends Microbiol. 19, 580–587.
2349–2361.
56. Ku, C., Nelson-Sathi, S., Roettger, M., Sousa, F.L., Lockhart, P.J., Bryant,
D., Hazkani-Covo, E., McInerney, J.O., Landan, G., and Martin, W.F.
37. Arama, E., Bader, M., Srivastava, M., Bergmann, A., and Steller, H. (2006).
(2015). Endosymbiotic origin and differential loss of eukaryotic genes. Na-
The two Drosophila cytochrome C proteins can function in both respiration
ture 524, 427–432.
and caspase activation. EMBO J. 25, 232–243.
57. Czabotar, P.E., Westphal, D., Dewson, G., Ma, S., Hockings, C., Fairlie,
38. Means, J.C., Muro, I., and Clem, R.J. (2006). Lack of involvement of mito-
W.D., Lee, E.F., Yao, S., Robin, A.Y., Smith, B.J., et al. (2013). Bax crystal
chondrial factors in caspase activation in a Drosophila cell-free system.
structures reveal how BH3 domains activate Bax and nucleate its oligo-
Cell Death Differ. 13, 1222–1234.
merization to induce apoptosis. Cell 152, 519–531.
39. Dorstyn, L., Mills, K., Lazebnik, Y., and Kumar, S. (2004). The two cyto- 58. Moldoveanu, T., Grace, C.R., Llambi, F., Nourse, A., Fitzgerald, P., Gehr-
chrome c species, DC3 and DC4, are not required for caspase activation ing, K., Kriwacki, R.W., and Green, D.R. (2013). BID-induced structural
and apoptosis in Drosophila cells. J. Cell Biol. 167, 405–410. changes in BAK promote apoptosis. Nat. Struct. Mol. Biol. 20, 589–597.
40. Dorstyn, L., Read, S., Cakouros, D., Huh, J.R., Hay, B.A., and Kumar, S. 59. Muchmore, S.W., Sattler, M., Liang, H., Meadows, R.P., Harlan, J.E.,
(2002). The role of cytochrome c in caspase activation in Drosophila mel- Yoon, H.S., Nettesheim, D., Chang, B.S., Thompson, C.B., Wong, S.L.,
anogaster cells. J. Cell Biol. 156, 1089–1098. et al. (1996). X-ray and NMR structure of human Bcl-xL, an inhibitor of pro-
grammed cell death. Nature 381, 335–341.
41. Zimmermann, K.C., Ricci, J.E., Droin, N.M., and Green, D.R. (2002). The
role of ARK in stress-induced apoptosis in Drosophila cells. J. Cell Biol. 60. Burger, G., Gray, M.W., Forget, L., and Lang, B.F. (2013). Strikingly bacte-
156, 1077–1087. ria-like and gene-rich mitochondrial genomes throughout jakobid protists.
Genome Biol. Evol. 5, 418–438.
42. Jagasia, R., Grote, P., Westermann, B., and Conradt, B. (2005). DRP-1-
mediated mitochondrial fragmentation during EGL-1-induced cell death 61. Pellettieri, J., Fitzgerald, P., Watanabe, S., Mancuso, J., Green, D.R., and
in C. elegans. Nature 433, 754–760. Sanchez Alvarado, A. (2010). Cell death and tissue remodeling in planarian
regeneration. Dev. Biol. 338, 76–85.
43. Abdelwahid, E., Yokokura, T., Krieser, R.J., Balasundaram, S., Fowle,
W.H., and White, K. (2007). Mitochondrial disruption in Drosophila 62. Lu, Y., Rolland, S.G., and Conradt, B. (2011). A molecular switch that gov-
apoptosis. Dev. Cell 12, 793–806. erns mitochondrial fusion and fission mediated by the BCL2-like protein
CED-9 of Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 108,
44. Galindo, K.A., Lu, W.J., Park, J.H., and Abrams, J.M. (2009). The Bax/Bak E813–E822.
ortholog in Drosophila, Debcl, exerts limited control over programmed cell
death. Development 136, 275–283. 63. Hoppins, S., Edlich, F., Cleland, M.M., Banerjee, S., McCaffery, J.M.,
Youle, R.J., and Nunnari, J. (2011). The soluble form of Bax regulates mito-
45. Tanner, E.A., Blute, T.A., Brachmann, C.B., and McCall, K. (2011). Bcl-2 chondrial fusion via MFN2 homotypic complexes. Mol. Cell 41, 150–160.
proteins and autophagy regulate mitochondrial dynamics during pro-
grammed cell death in the Drosophila ovary. Development 138, 327–338. 64. Renault, T.T., Floros, K.V., Elkholi, R., Corrigan, K.A., Kushnareva, Y.,
Wieder, S.Y., Lindtner, C., Serasinghe, M.N., Asciolla, J.J., Buettner, C.,
46. Monserrate, J.P., Chen, M.Y., and Brachmann, C.B. (2012). Drosophila et al. (2015). Mitochondrial shape governs BAX-induced membrane per-
larvae lacking the bcl-2 gene, buffy, are sensitive to nutrient stress, main- meabilization and apoptosis. Mol. Cell 57, 69–82.
tain increased basal target of rapamycin (Tor) signaling and exhibit charac-
teristics of altered basal energy metabolism. BMC Biol. 10, 63. 65. Quistad, S.D., Stotland, A., Barott, K.L., Smurthwaite, C.A., Hilton, B.J.,
Grasis, J.A., Wolkowicz, R., and Rohwer, F.L. (2014). Evolution of TNF-
47. Steller, H. (2008). Regulation of apoptosis in Drosophila. Cell Death Differ. induced apoptosis reveals 550 My of functional conservation. Proc. Natl.
15, 1132–1138. Acad. Sci. USA 111, 9567–9572.

Current Biology 26, R620–R627, July 11, 2016 R627

You might also like