Você está na página 1de 2

Developmental Cell

Previews

Krebs Cycle Moonlights in Caspase Regulation


Adi Minis1 and Hermann Steller1,*
1Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York,

NY 10065, USA
*Correspondence: steller@mail.rockefeller.edu
http://dx.doi.org/10.1016/j.devcel.2016.03.016

In this issue of Developmental Cell, Aram et al. (2016) identify a mechanism that uses a Krebs cycle protein to
control local activation of a ubiquitin ligase complex at the mitochondrial outer membrane for temporally and
spatially restricted caspase activation during Drosophila sperm differentiation.

Caspases, a conserved family of cysteine and depends on apoptotic proteins, cells, it is not sufficient for caspase activa-
proteases, have been intensely studied as including caspase-3-type effector cas- tion in spermatids. Furthermore, although
key executioners of apoptosis, the most pases (Arama et al., 2003). Earlier studies the authors suggest that the binding of
common form of natural cell death. In had already revealed the critical impor- A-Sb to the CRL3 complex is important
addition, caspases also play important tance of the caspase inhibitor dBruce, the for its activation by neddylation, their
non-apoptotic roles in development and apoptosome activator Cytochrome C, RNAi experiments show a more promi-
differentiation (Yi and Yuan, 2009). A strik- and a Cullin-3-based ubiquitin ligase com- nent effect on the localization to the
ing example is the non-lethal use of plex (CRL3) for regulating caspase activity mitochondria than on the levels of neddy-
apoptotic effector caspases for cellular in a non-lethal manner (Arama et al., lation. Collectively, these results imply
remodeling (Fuchs and Steller, 2011; 2007). Furthermore, the Arama lab previ- that Cul3-neddylation and caspase acti-
Schuldiner and Yaron, 2015). Apoptotic ously identified a pseudosubstrate inhibi- vation are uncoupled in spermatids and
proteins are required for the regulated tor for CRL3, termed Soti, which inhibits that another layer of regulation operates
destruction of axons and dendrites to the activity of this complex in a spatially at the MOM. This could be mediated
sculpt the nervous system, the degrading graded manner and thereby restricts cas- through an interaction with other mito-
of the nucleus and membrane-bound or- pase activity in time and space (Kaplan chondrial proteins or lipids, post-transla-
ganelles during lens fiber differentiation, et al., 2010). This fine-tuning of caspase tional modification, or the proximity to
and terminal differentiation of spermatids. levels is crucial for male fertility, as either other apoptosis-related proteins that are
This raises the intriguing question of how too low or too high caspase activity has necessary for caspase activation, such
the activation of apoptotic caspases can disastrous consequences. as Cytochrome C.
be restricted in time and space to avoid In the current study, Aram et al. (2016) Irrespective of the exact underlying
death of the entire cell. In this issue of used a yeast two-hybrid approach fol- biochemical mechanism, the current
Developmental Cell, Aram et al. (2016) lowed by co-immunoprecipitation experi- work by Aram et al. (2016) reveals an
report a major advance that sheds new ments to identify an ATP-specific form of unexpected ‘‘moonlighting’’ function of
light onto this question. Using Drosophila the Succinyl-CoA synthetase b subunit A-Sb for the spatio-temporal regulation
melanogaster spermatogenesis as a (A-Sb) as another key regulator of CRL3 of a Cullin-3-based ubiquitin ligase com-
model system, the authors uncover an un- activity. Typically, A-Sb is found in the plex and caspase activity. At the same
expected role for a Krebs cycle protein, mitochondrial matrix and functions within time, these findings raise many new
Succinyl-CoA synthetase b (A-Sb), in pro- the Krebs cycle. However, in Drosophila and intriguing questions. Foremost, why
moting temporally and spatially restricted spermatids, a significant portion of A-Sb would a Krebs cycle protein involve a
caspase activation at the mitochondrial is localized on the MOM where it can function at the surface of mitochondria
outer membrane (MOM) (Figure 1). bind to CRL3. In a series of elegant exper- to regulate protein degradation? The
During terminal sperm differentiation in iments that combine cell fractionation, im- important role of mitochondria in the regu-
Drosophila, the vast majority of proteins muno-EM, biochemical analyses, domain lation of apoptosis has long been recog-
are degraded to generate highly special- swapping experiments, and genetic and nized, as a source of regulatory proteins,
ized and minimalistic cells devoid of major structure-function studies, Aram et al. but also as a platform for the sequestra-
organelles. This process occurs in cysts (2016) now demonstrate that A-Sb pro- tion of apoptotic proteins (Fuchs and
containing 64 haploid cells that remain motes caspase activation by antago- Steller, 2011). Furthermore, sperm differ-
interconnected due to incomplete cytoki- nizing Soti to activate the CRL3 complex entiation in insects involves very dramatic
nesis (Fuller, 1993). The terminal differen- at the MOM. They also show that the changes in the structure of mitochondria.
tiation of spermatids involves dramatic localization and function of A-Sb in devel- Therefore, it is possible that A-Sb helps
cell elongation and separation into indi- oping sperm depends on its N-terminal to sense the appropriate metabolic state
vidual spermatids; hence, this process is domain. This is interesting because for the initiation of caspase activity.
termed ‘‘individualization.’’ Importantly, although the CRL3-binding domain of Intriguingly, two additional non-canonical
this process involves the degradation of A-Sb, located at its C terminus, is suffi- functions of A-Sb outside mitochondria
the majority of proteins and organelles cient for caspase activation in S2 culture have also been suggested, namely in

Developmental Cell 37, April 4, 2016 ª2016 Elsevier Inc. 1


Developmental Cell

Previews

plexes. In addition, although the CRL3


complex is clearly important for caspase
activation during sperm differentiation,
there may be other substrates with rele-
vant biological functions. Another impor-
tant unresolved question is how exactly
A-Sb overcomes the inhibition of CRL3
by Soti. Furthermore, it will be interesting
to investigate the precise mechanism
that determines when and how A-Sb be-
comes located to the MOM, since this
appears to be the rate-limiting step in acti-
vating CRL3 in this system. Finally, it will
be important to explore whether these
findings are restricted to Drosophila
sperm differentiation or represent a more
general regulatory mechanism. Given
the tremendous importance of under-
standing both mechanisms that restrict
caspase activity and the regulation of
CRL3 complexes, it is likely that the
answers to many of these questions will
emerge soon.

REFERENCES

Aram, L., Braun, T., Braverman, C., Kaplan, Y.,


Ravid, L., Levin-Zaidman, S., and Arama, E.
(2016). Dev. Cell 37, this issue, 15–33.

Arama, E., Agapite, J., and Steller, H. (2003). Dev.


Cell 4, 687–697.

Arama, E., Bader, M., Rieckhof, G.E., and Steller,


H. (2007). PLoS Biol. 5, e251.

Fuchs, Y., and Steller, H. (2011). Cell 147, 742–758.


Figure 1. Spatially Restricted Caspase Activation at the Mitochondrial Membrane during
Sperm Differentiation Fuller, M.T. (1993). In The Development of
(A) The non-lethal activity apoptotic effector caspases is regulated by multiple pathways at the mito- Drosophila melanogaster, M. Bate and A.M. Arias,
chondrial surface. First, Cytochrome C is necessary to promote caspase activation. Second, the pseu- eds. (Cold Spring Harbor Laboratory Press),
dosubstrate Soti and A-Sbt, a testis-specific isoform of the Krebs cycle enzyme Succinyl-CoA synthase b, pp. 71–147.
have antagonistic effects on the activity of a Culin-3-based ubiquitin ligase (CRL3) complex. Activation of
CRL3 promotes degradation of the caspase-inhibitor dBruce, which in turn restricts the potentially lethal Gao, L., Fei, H., Connors, N.C., Zhang, J., and Lev-
activity of caspases in time and space. The interplay between Soti and A-Sbt achieves spatial and tem- itan, I.B. (2008). J. Neurophysiol. 99, 2736–2740.
poral restriction of caspase activity that is necessary for the selective destruction of subcellular structures
during sperm terminal differentiation. Hughes, J.R., Meireles, A.M., Fisher, K.H., Garcia,
(B) Soti and A-Sbt regulate CRL3 to promote caspase activation. Soti inhibits CRL3 by blocking substrate A., Antrobus, P.R., Wainman, A., Zitzmann, N.,
binding. Upon the onset of sperm differentiation, A-Sbt levels increase on the mitochondrial outer Deane, C., Ohkura, H., and Wakefield, J.G.
membrane (MOM) and compete with Soti for binding to CRL3. This allows for spatially restricted activation (2008). PLoS Biol. 6, e98.
of CRL3 at the MOM, which in turn promotes degradation of dBruce and caspase activation.
Kaplan, Y., Gibbs-Bar, L., Kalifa, Y., Feinstein-Rot-
kopf, Y., and Arama, E. (2010). Dev. Cell 19,
160–173.
modulating the activity of a voltage-gated possible that A-Sb has multiple moonlight
potassium channel and Skp-1-mediated jobs, or perhaps all these observations Schuldiner, O., and Yaron, A. (2015). Cell. Mol. Life
Sci. 72, 101–119.
centrosome regulation (Gao et al., 2008; reflect a more general role of this protein
Hughes et al., 2008). Therefore, it is in Cullin-based ubiquitin ligase com- Yi, C.H., and Yuan, J. (2009). Dev. Cell 16, 21–34.

2 Developmental Cell 37, April 4, 2016

Você também pode gostar