Download as pdf or txt
Download as pdf or txt
You are on page 1of 3

Methods 61 (2013) 87–89

Contents lists available at SciVerse ScienceDirect

Methods
journal homepage: www.elsevier.com/locate/ymeth

Guest Editor’s Introduction

Distinguishing between apoptosis, necrosis, necroptosis and other cell death


modalities

This issue of Methods contains a series of articles that cover the pyroptosis. Necrotic cell death bears none of the striking features
major techniques used to measure apoptosis, necrosis and varia- that characterize apoptotic cells, such as extensive membrane
tions thereof, at the population, single cell, organelle and molecular blebbing and hypercondensation and fragmentation of the nucleus.
level. Although we used to think of apoptosis as programmed or Instead, necrotic cells undergo extensive organelle and cell swell-
regulated cell death and necrosis as strictly non-programmed or ing, leading to decondensation of nuclei which become markedly
unregulated cell death [1,2], data have emerged in recent years less stained with fluorescent dyes such as DAPI or propidium io-
which suggest that the boundaries between these two modes of dide. Thus, this mode of cell death is relatively easy to distinguish
cell death can become blurred in certain situations [3,4]. from apoptosis on the basis of morphological criteria.
To summarize, for those that are new to the field, apoptosis is a Necroptosis, which is a form of necrosis that appears to be dri-
mode of cell death that is characterized by a series of morpholog- ven as a consequence of the deregulated activity of specific mole-
ical and biochemical alterations to the cell architecture that pack- cules, is under intensive investigation at present. This mode of cell
age a cell up for removal by cells with phagocytic capacity [5]. death is typically seen in response to engagement of certain mem-
Crucially, apoptotic cells are recognized by phagocytes and are en- bers of the ‘death receptor’ subset of the TNF superfamily, particu-
gulfed before they leak their contents. Thus, apoptosis ensures that larly of the TNF receptor itself, when caspase activity is inhibited
when a cell needs to be removed from a tissue, this occurs in an or- [7]. The caveat is that death receptor-induced necroptosis is only
derly fashion that minimizes disruption to neighboring cells. The observed in cells that express the kinase, RIPK3. In cells lacking
major consideration during apoptosis is that intracellular contents RIPK3, inhibition of caspase activity completely blocks TNF-in-
do not leak into the extracellular space because this could (a) dam- duced cell death, but in RIPK3-expressing cells results in a necro-
age surrounding cells, and (b) trigger inflammation through release tic-like mode of cell death [4,8]. Necroptosis results from
of molecules with immune-activating activity, the so-called ‘alar- excessive recruitment of RIPK3 onto RIPK1, thereby promoting
mins’ [5]. Thus, apoptosis is largely concerned with avoiding dis- excessive RIPK1 kinase activity, which is cytotoxic. Thus, necropto-
turbance to tissues in which there is ongoing homeostatic cell sis represents somewhat of a kinase bomb that is set off within the
death. Most of the biochemical and morphological changes that cell if the normal caspase wiring is interfered with. Under normal
typify apoptosis are the consequence of activation of a subset of circumstances, caspase-8 restrains recruitment of RIPK3 onto
the caspase family of proteases (caspases 3, 6, 7, 8 and 9) RIPK1 in two ways, first by cleaving RIPK1 and second by cleaving
[1,5]. The latter caspases operate similar to a controlled demolition the deubiquitinating enzyme CYLD. CYLD negatively regulates the
squad, coordinating the packaging and disposal of cells in a manner degree of RIPK1 ubiquitination that occurs in response to death
that minimizes damage to neighbors and the initiation of inflam- receptor engagement, an event that is involved in assembling sig-
mation [5]. Methods for measuring apoptosis typically rely on naling complexes on RIPK1 to propagate death receptor-induced
the detection of caspase-dependent events, such as exposure of NFjB activation. However, untrammeled CYLD activity can lead
plasma membrane phosphatidylserine [6], that precede uptake of to deubiquitination of RIPK1 and excessive recruitment of RIPK3
vital dyes such as trypan blue or propidium iodide. Alternatively, as a consequence. Thus, by cleaving RIPK1 and CYLD, caspase-8
the striking morphological features of apoptotic cells (such as com- fine-tunes the composition of the RIPK1/RIPK3 complex, which, if
paction and fragmentation of the cell nucleus), which are also ef- perturbed, deregulates the RIPK1/RIPK3 kinase complex, resulting
fected through caspase activation, are still highly relevant for in necroptosis. What is not clear at present is why. Moreover, the
methods for detecting this mode of cell death. physiological relevance of this mode of cell death is currently de-
In contrast to apoptosis, necrosis is generally uncontrolled and bated but may occur upon infection with certain viruses that en-
involves the sudden loss of membrane integrity, release of extra- code caspase inhibitors. Irrespective of the precise physiological
cellular contents, leading to activation of the immune system and relevance of necroptosis, which is under investigation at present,
extensive inflammation [2,4]. Necrosis is typically not associated the major means of distinguishing conventional necrosis from nec-
with caspase activation, although the exception to this is where roptosis is through probing for the involvement of RIPK1, or RIPK3,
cell death follows aggressive activation of the inflammatory subset where cell death occurs in the absence of caspase involvement.
of caspases (caspases 1, 4 and 5), a mode of cell death termed This can be done either through ‘knocking down’ expression of

1046-2023/$ - see front matter Ó 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.ymeth.2013.06.001
88 Guest Editor’s Introduction / Methods 61 (2013) 87–89

RIPK1 or RIPK3 using siRNA or shRNA or by means of the RIPK1 death pathway are known, useful predictions may be made con-
inhibitor necrostatin [7]. cerning how best to engage or manipulate such pathways for ther-
The issue begins with an overview of the morphological fea- apeutic purposes.
tures of apoptosis by Henry et al. [9]. Morphological features of cell The assembly of large multi-protein complexes mediate many
death still remain a key means of distinguishing apoptosis from of the signaling events within a cell. Several of these complexes
necrosis, with the former typically associated with extensive cell have been identified as key mediators of apoptotic and necrotic cell
shrinkage and plasma membrane blebbing and the latter typified death including, the Death Inducing Signaling Complex (DISC) and
by cell swelling and gross membrane rupture. Henry et al., also de- the apoptosome, with the necrosome and RIPoptosome identified
scribe flow cytometry-based methods for quantitating apoptosis and extensively characterized in recent years. The precise compo-
based upon DNA fragmentation, caspase activation and phosphati- sition of these complexes is currently an area of intense research
dylserine externalization, which are very well established methods due to the increasing evidence that cell death outcomes are often
in the field [9]. determined by subtle compositional and stoichiometric changes
This is followed by a comprehensive discussion of the differ- within these complexes [4]. In this issue, MacFarlene and col-
ences between apoptosis and necrosis by Vandenabeele and col- leagues provide a detailed range of methodologies to characterize
leagues [10], who also describe a variety of assays that can be the assembly of these complexes in vitro including affinity purifica-
used to discriminate between these different modes of cell death tion, sucrose fractionation, size exclusion chromatography and
with particular emphasis on the L929 cell line that is widely used reconstitution of the death-inducing complexes [16].
to study TNF-induced necroptosis. Bortoluci and colleagues de- Moving onto methods used to evaluate cell death in vivo, Amar-
scribe a novel form of cell death, called pyroptosis, that has fea- ante-Mendes and colleagues detail a method for the assessment of
tures of apoptosis as well as necrosis and is induced through cytotoxic T-lymphocyte (CTL) killing within the complex microen-
delivery of bacterial flagellin into the cytoplasm of macrophages vironment in which immune responses take place [17]. This is par-
[11]. This form of cell death, which appears to be dependent on ticularly important given the conflicting data published on CTL-
activation of inflammatory caspases, such as caspase-1, is rapid mediated cytotoxicity arising from studies performed in vitro ver-
and associated with the release of pro-inflammatory cytokines sus in vivo.
[11]. This is followed by an article by Derry and colleagues on the
Mitochondria have a central role in the initiation of apoptosis evaluation of cell death in the nematode worm C. elegans [18], a
through releasing cytochrome c, which acts as a co-factor for the model system that has been exploited very successfully for the
assembly of a key caspase-activating complex, called the apopto- identification of many conserved components of cell death path-
some [1]. The Bcl-2 family of proteins regulate this cytochrome c ways. Derry and colleagues utilize fluorescent markers in conjunc-
release step in a positive and negative way, with opposing factions tion with RNAi screening to identify new genes and pathways that
within this family fine-tuning the balance between life and death regulate apoptosis in the worm germline [18].
[1]. In response to diverse cytotoxic insults, members of the BH3- To complete this issue of Methods, we turn our attention to the
only subset of the Bcl-2 family can be mobilized to facilitate cyto- use of genetically engineered mice in the study of cell death. There
chrome c release by triggering opening of the Bax/Bak channel is no doubt that gene-targeted mice have become an indispensible
within the mitochondrial outer membrane. However, Bax/Bak resource in every area of biological research. The cell death field is
channel assembly can be countered by the pro-survival subset of no exception to this and such models have provided key insights
the Bcl-2 family, such as Bcl-2 and Bcl-xL. Mitochondrial outer into the physiological roles and importance of many pro-apoptotic
membrane permeabilization (MOMP), as a result of Bax/Bak activa- and anti-apoptotic proteins in vivo. Gain-of-function as well as
tion represents a defining event where a cell commits to apoptosis loss-of-function gene mutations have helped to delineate cell
[1]. Waterhouse and colleagues detail high-throughput flow death pathways, as well as tease out redundant and non-redun-
cytometry-based methods to measure uptake of cationic dyes, as dant functions of many apoptosis-related proteins. However, while
well as cytochrome c translocation, to distinguish healthy cells much has been learned from studies using ‘knockout’ mice and de-
with intact mitochondria from dying cells where permeabilisation rived cell lines, Villunger and colleagues highlight the potential pit-
of mitochondria occurs [12]. Biochemical analysis of apoptosis- falls of interpreting data generated from different genetic
associated mitochondrial events, such as Bax/Bak activation, can backgrounds [19]. While the use of ‘knockouts’ clearly have their
also be conveniently studied using purified populations of isolated place, it is often meaningless to simply compare cell death re-
mitochondria, a topic which is covered by Chipuk and colleagues sponses from cell lines derived from one genotype versus another.
[13]. The proximity of a tumor cell to undergo MOMP, due to the Such crude comparisons, while superficially sophisticated, are of-
interplay between members of the Bcl-2 family, may be one of ten as crude as comparing one tumor cell line to another on the ba-
the reasons why chemotherapeutic drugs are effective on some sis that they differ by only one expressed gene.
but not all cancers and is the topic of the article by Letai and col- In conclusion, for those that wonder why it matters which way
leagues [14]. They describe an assay developed by the Letai lab, a cell dies, it is useful to consider why multicellular organisms have
called ‘BH3-profiling’, which utilizes peptides based on BH3 do- developed such complex strategies to regulate cell death. The ma-
mains of BH3-only proteins to measure the proximity of mitochon- jor driving forces appear to be the avoidance of collateral damage
dria within specific tumor cell populations to MOMP and, by to surrounding healthy neighbors, as well as the avoidance of
extension, apoptosis. Profiling of tumor cell mitochondria in this inflammation. Or conversely, where it is desirable that the immune
way, prior to chemotherapy, may help to guide oncologists to- system becomes activated, the exact opposite of this is true. Much
wards drugs or drug combinations that elicit favorable cell death like in daily life, it is important to know whether a death has oc-
responses, thereby sparing patients needless treatment cycles with curred as a result of natural causes or foul play. Each scenario war-
drugs that are ineffective for their particular tumor. rants a very different response by those close to the recently
Along similar lines, it is also possible that responses to pertur- deceased, as well as the authorities.
bations in the cell death machinery may be subject to computa-
tional modeling in silico. Prehn and colleagues outline the use of References
signal transduction models to quantify and temporally analyze
changes to particular proteins in response to apoptotic stimuli [1] R.C. Taylor, S.P. Cullen, S.J. Martin, Nat. Rev. Mol. Cell Biol. 9 (2008) 231–241.
[2] G. Kroemer, S.J. Martin, Nat. Med. 11 (2005) 725–730.
[15]. In situations where the concentrations of all members of a cell
Guest Editor’s Introduction / Methods 61 (2013) 87–89 89

[3] A. Degterev, J. Yuan, Nat. Rev. Mol. Cell Biol. 9 (2008) 378–390. [17] T. Clemente, M.R. Dominguez, N.J. Vieira, M.M. Rodrigues, G.P. Amarante-
[4] A. Kaczmarek, P. Vandenabeele, D.V. Krysko, Immunity 38 (2013) 209–223. Mendes, Methods 61 (2013) 103–107.
[5] S.J. Martin, C.M. Henry, S.P. Cullen, Mol. Cell. 46 (2012) 387–397. [18] B. Lant, W.B. Derry, Methods 61 (2013) 174–182.
[6] S.J. Martin, C.P.M. Reutelingsperger, J. Exp. Med. 182 (1995) 1545–1556. [19] C. Manzl, F. Baumgartner, Methods 61 (2013) 128–135.
[7] A. Degterev, Z. Huang, M. Boyce, Y. Li, P. Jagtap, N. Mizushima, T.J. Mitchison,
M.A. Mizushima, J. Yuan, Nat. Chem. Biol. 1 (2005) 112–119.
[8] P. Vandenabeele, L. Galluzzi, T. Vanden Berghe, G. Kroemer, Nat. Rev. Mol. Cell
Seamus J. Martin
Biol. 11 (2010) 700–714. Conor M. Henry
[9] C.M. Henry, E. Hollville, S.J. Martin, Methods 61 (2013) 88–95. Molecular Cell Biology Laboratory,
[10] T. Vanden Berghe, S. Grootjans, V. Goossens, Y. Dondelinger, D.V. Krysko, N.
Dept. of Genetics, Trinity College, Dublin 2, Ireland
Takahashi, Methods 61 (2013) 115–127.
[11] S.L. Lage, G.P. Amarante-Mendes, K.R. Bortoluci, Methods 61 (2013) 108–114. Fax: +353 1 679 8558. (S.J. Martin)
[12] M.E. Christensen, E.S. Jansen, W. Sanchez, N.J. Waterhouse, Methods 61 (2013) E-mail address: martinsj@tcd.ie (S.J. Martin)
136–143.
[13] T.T. Renault, F.V. Floros, J.E. Chipuk, Methods 61 (2013) 144–153.
[14] J.A. Ryan, Methods 61 (2013) 154–162.
[15] M. Rehm, J.H.M. Prehn, Methods 61 (2013) 163–171.
[16] M.A. Hughes, C. Langlais, K. Cain, Methods 61 (2013) 98–104.

You might also like