Sei sulla pagina 1di 10

Critical Reviews in Oncology/Hematology 82 (2012) 249–258

Necroptosis: An emerging form of programmed cell death


Wei Wu, Peng Liu ∗ , Jianyong Li ∗
Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing 210029, PR China
Accepted 10 August 2011

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
2. Signaling pathway of necroptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
2.1. Initiators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
2.2. Formation of the complex I . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
2.3. The formation of complex II . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
2.4. The formation of necrosome complex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
3. Execution of necroptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
3.1. Reactive oxygen species (ROS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
3.2. ANT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
3.3. RNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
3.4. PLA2 and LOXs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
4. Necroptosis and other types of programmed cell death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
4.1. Necroptosis and apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
4.2. Necroptosis and autophagy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
4.3. Necroptosis and PARP-induced programmed necrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
5. Necroptosis in physiological and pathological processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
6. Necroptosis and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 258

Abstract
Necrosis plays an important role in multiple physiological and pathological processes. Recently, a relatively new form of necrosis has been
characterized as “necroptosis”. Morphologically, necroptosis exhibits the features of necrosis; however, necroptosis exhibits a unique signaling
pathway that requires the involvement of receptor interaction protein kinase 1 and 3 (RIP1 and RIP3) and can be specifically inhibited by
necrostatins. Necroptosis has been found to contribute to the regulation of immune system, cancer development as well as cellular responses
to multiple stresses. In this review, we will summarize the signaling pathway, biological effects and pathological significance of this specific
form of programmed cell death.
© 2011 Elsevier Ireland Ltd. All rights reserved.

Keywords: Necroptosis; Apoptosis; Cell death; Cancer

∗ Corresponding authors. Tel.: +86 25 83781120; fax: +86 25 83781120.


E-mail addresses: liupeng8888@yahoo.com.cn (P. Liu), lijianyonglm@medmail.com.cn (J. Li).

1040-8428/$ – see front matter © 2011 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.critrevonc.2011.08.004
250 W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258

1. Introduction Since there is more than one kind of necroptotic initiator, it


is not clear whether or not there is a common downstream sig-
Cell survival has been the subject of active research for naling pathway. Among initiators, the TNF-␣/TNFR-induced
several decades, and over time research studies have evolved pathway has received the most intensive scrutiny. The molec-
to include a further investigation of cell death. There are ular mechanisms of TNFR ligation induced necroptotic
two principal types of cell death according to morphological signaling are described below.
appearance: apoptosis and necrosis [1]. Apoptosis is charac-
terized by the rounding up of cells, pseudopod retraction, 2.2. Formation of the complex I
pyknosis, chromatin condensation, nuclear fragmentation,
and the appearance of apoptotic bodies [2]. Apoptosis is TNF-␣ is produced by activated macrophages [13] and is a
executed in intrinsic or extrinsic pathways in response to var- homotrimer protein with each subunit containing 157 amino
ious cell death stimuli and is regulated by a specific class acids. Although currently TNF-␣ is commonly considered an
of compounds, such as the caspase cascade [3]. Necrosis is apoptotic inducer, it was first identified as an agent capable of
morphologically identified by the swelling of organelles, an causing tumor cell necrosis [14,15]. New evidence demon-
increased cell volume, disruption of the plasma membrane, strating that TNF-␣ is an inducer of programmed necrosis
and loss of intracellular contents. It has been previously has recently emerged.
demonstrated that necrosis can be triggered not only by death TNFR1 or TNFR2, located on the cell surface, are specific
domain receptors, but also through AIF-mediated chromati- receptors of TNF-␣. As TNFR2 does not have a death domain,
nolysis or alkylation-induced DNA damage [4–6]. In contrast TNFR1 plays the principal role in TNF-␣-mediated activation
to apoptosis, which has been demonstrated to involve pro- and triggers a series of intracellular events.
grammed cell death, necrosis has been commonly viewed as Initially, TNF-␣ binds to the extracellular portion of
an accidental and unregulated event. However, an increasing TNFR1, creating allosteric changes in the intracellular por-
body of evidence indicates that necrosis can also be exe- tion of TNFR1 [16]. There are four cysteine-rich domains
cuted by regulated mechanisms [7]. The recently coined term (CRDs) in TNFR1. The first CRD, known as pre-ligand
“necroptosis” refers to one particular form of programmed assembly domain (PLAD), is essential for the formation
necrosis induced by stimulating death receptors with ago- of pre-assembled receptors which can bind to TNF-␣ with
nists such as TNF␣, FasL, and TRAIL. Necroptosis has its higher affinity [17]. After binding to TNF-␣, the silencer
own unique signaling pathway which requires the involve- of death domains (SODD) is released from the intracellu-
ment of receptor interaction protein kinase 1 and 3 (RIP1 lar domain of TNFR1 with the help of various proteins and
and RIP3), and can be specifically inhibited by necrostatins enzymes. TNFR1 and TNFR2 trigger the downstream signal-
[4,8,9]. Compared to apoptosis, of which signaling path- ing by forming complex I with proteins containing a death
ways that have been studied for many years, the underlying domain, such as TNF-␣ receptor-associated death domain
mechanisms of necroptosis remain poorly understood. In (TRADD), RIP1, Fas-associated death domain (FADD), and
this review, we will provide a detailed description of the several E3 ubiquitin ligases, such as TNF-␣ receptor asso-
mechanisms of necroptosis and briefly discuss the potential ciated factor 2/5 (TRAF2/5), inhibitor of apoptosis proteins
impact of necroptosis on physiological processes and tumor (IAPs) cIAP1 and cIAP2. Ubiquitination of these proteins is
pathogenesis. important for the regulation of the activity of complex I and
impacts the outcome of the cell [18].
RIP1 is a member of the RIP family exhibiting a homol-
2. Signaling pathway of necroptosis ogous N-terminal kinase domain. The ubiquitination state
of RIP1 determines whether it functions as a pro-survival
2.1. Initiators molecule or a kinase promoting cell death. RIP1 is initially
recruited to complex I by TNFR1, and is polyubiquitinated
Necroptosis is induced by a class of death receptors that by TRAF2/5, cIAP1, and cIAP2 at the position of lysine 63
include TNFR1, TNFR2, and Fas. Upon binding with their [19,20]. The ubiquitination of RIP1 initiates the recruitment
agonists, these death receptors induce cells toward either sur- and activation of the IKK complex and NEMO and promotes
vival or death depending on the circumstances. Until recently, the activation of the NF-␬B pathway, inducing a pro-survival
death receptors were considered to induce apoptosis only. cellular mode. Deubiquitination of RIP1 could inhibit NF-
However, it has been recently demonstrated that death recep- ␬B pathway and this leads to an inclination of cell death
tors potentially leads to necroptosis with the involvement of pathways. Two proteins have been shown to exhibit the activ-
RIP1 when the apoptotic pathway is blocked [4,10]. It has ity of RIP1 deubiquitination in the regulation of the NF-␬B
also been established that the agonists of the Toll-like receptor pathway. Cylindromatosis (CYLD) is encoded by a tumor-
(TLR) induce caspase-independent necrosis [11,12]. Consis- suppressor gene Cyld. It blocks the activation of NF-␬B by
tently, multiple genes involved in the TLR pathway are found cleaving Lys63 -linked polyubiquitin chains from several pro-
in necroptotic signaling, indicating that the TLR pathway teins [21]. Tumor cells carrying inactive CYLD exhibit an
may be involved in necroptosis. elevated proliferation activity and a decreased apoptosis rate
W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258 251

[22,23]. A20 removes Lys63 -linked ubiquitin by trigging the


proteasome-dependent degradation of E3 ligases, such as
TRAF2 and cIAPs, and negatively regulates NF-␬B pathway
[24,25]. It has been previously demonstrated that ubiquitina-
tion of RIP1 is necessary for activation of the NF-␬B pathway
[26]. However, the kinase activity of RIP1 is not a requirement
for this process [27,28]. Thus, the key element in the regula-
tion of the TNF-induced NF-␬B pathway is the ubiquitination
status of RIP1, regardless of whether the RIP1 kinase is acti-
vated. Complex I is situated at the crossroads of cell survival
and death, switching between various signaling pathways in
response to an array of stimuli and microenvironments.

2.3. The formation of complex II

Following deubiquitination, RIP1 is released from com-


plex I to the cytoplasm, and subsequently recruited to
complex II. TRADD is also released from complex I after
the internalization of ligand-bound TNFR1. Internalization
of ligand-bound TNFR1 is essential for the formation of
complex II, as it has been previously demonstrated that Fig. 1. Formation of necrosome After binding to TNF-␣, the intracel-
the inhibition of TNFR1 internalization leads to a resis- lular domain of TNFR1 summons a series of proteins, including TNF
receptor-associated death domain (TRADD), receptor-associated death pro-
tance to apoptosis in cells [29]. Complex II, also known as tein1 (RIP1), and Fas-associated death domain (FADD). Meanwhile, several
the death-inducing signaling complex (DISC), is composed E3 ligases, such as TNF receptor associated factor 2/5 (TRAF2/5), inhibitor
of TRADD, FADD, RIP1, and caspase-8. Knock-down of of apoptosis proteins (IAPs) cIAP1 and cIAP2 are also recruited to TNFR1.
CYLD inhibits TNF-induced necroptosis [12], which indi- Those proteins mentioned above form “complex I”. RIP1 is ubiquitilatized
cates that the deubiquitination of RIP1 is an important step in complex I, and has the ability to activate of the I␬B kinase (IKK) complex
and NF-␬B essential modulator (NEMO), leading to the activation of pro-
in TNF-induced necroptosis. However, there is no other evi- survival NF-␬B pathway in consequence. When RIP1 is deubiquitilitized by
dence regarding whether other deubiquitinating enzymes, cylindromatosis (CYLD), it is released to the cytoplams, and forms complex
such as A20, are as essential as CYLD to necroptosis. Inhibi- II with TRADD, FADD, RIP3 and caspase-8. While caspase-8 is activated, it
tion of cIAPs accelerates complex II formation by decreasing would cleave RIP3 and induce apoptosis through caspase cascade. However,
the ubiquitination of RIP1 [30]. TRAF2, another E3 ligase, when caspase-8 is blocked, phosphorylated RIP1 and RIP3 form necrosome
and initiate necroptosis.
has been demonstrated to be essential for TNF-␣ induced
necrosis because TRAF2−/− cells show resistance to TNF-
␣ induced necrosis [31]. This effect is potentially due to necroptosis [35]. Additionally, RIP3 is cleaved at Asp328 by
the fact that TRAF2 is required for the formation of com- caspase-8 under apoptotic stimuli, suppressing the ability of
plex I. FADD is one of the domains recruited to complex RIP3 to induce caspase-independent cell death [37]. When
II and its effect on necroptosis varies depending upon cell the apoptotic pathway is blocked, necroptosis prevails.
type. FADD is essential for necroptosis induced by TNF-
␣ in mouse embryonic fibroblasts (MEFs) [18], but it is not 2.4. The formation of necrosome complex
required for necroptotic process in leukemic Jurkat cells [32].
For T cells in a proliferative phase, FADD acts as a negative When the apoptosis pathway is inhibited, a complex
regulator in necroptosis [33]. The mechanism underlying the named “necrosome” is formed (Fig. 1). Necrosome is primar-
various roles of FADD remains unclear. It has been reported ily composed of RIP1 and RIP3, and it distinctly enhances
that TRADD is generally necessary for necroptosis with the necroptosis [38]. RIP3 accelerates recruitment of RIP1 to the
exception of necroptosis induced by Smac mimetics [34,35]. necrosome, and the kinase activity of both proteins is required
Thus, whether TRADD is required for necroptosis poten- in this process [39]. The RIP1 kinase inhibitor Nec-1 inhibits
tially depends upon the type of stimulus involved. Complex complex II formation, and RIP1 recruitment to complex II
II may activate two downstream signaling pathways: apopto- is crucial for the induction of pronecrotic complex II kinase
sis and necroptosis. When capase-8 is activated, it deprives activity. However, RIP1 kinase activity is not required for the
RIP1 of its activity by cleaving it, driving complex II into a formation of complex I. It has been previously reported that
pro-apoptosis mode. Cleavage of RIP1 by caspase-8 not only RIP3 is required in RIP1 phosphorylation in TNF-␣ induced
abrogates the stimulatory role of RIP1 in the activation of necroptosis. However, the phosphorylation mediated by RIP3
NF-␬B pathway [36], but also exhibits a negative effect on is very weak and is similar to the level of RIP1 autophos-
necroptosis because the kinase activity of RIP1 is crucial in phorylation [40]. Additionally, only ubiquitinated RIP1 is
252 W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258

detectable in necroptosis-resistant cells that exhibit a low L929 cells, and L929 cells could commit necroptosis treated
RIP3 expression level, indicating that RIP3 may facilitate with TNF-␣ or z-VAD alone, or both simultaneously [49]. An
the deubiquitination of RIP1. experiment performed in Harvard Cell Biology Department
Similar to other RIPs, RIP3 has an N-terminal kinase [12] showed that necroptosis might be controlled at tran-
domain. However, its C-terminus lacks a death domain or scriptional level, indicating a possible genetically explanation
CARD motif. The biological function of RIP3 remains con- about the cellular choice of necroptosis.
troversial. It has been reported that RIP3 is able to inhibit Necrostatin-1 (Nec-1) was initially defined as an inhibitor
RIP1 from activating the NK-␬B pathway [40]. Alternatively, of necroptosis because of the specific inhibitory effect of
RIP3 could activate the NK-␬B pathway when it is over- RIP1 [9,50]. It provides neuron-protection regarding brain
expressed [41]. However, the absence of RIP3 does not inhibit trauma, hypoxia, and ischemia/reperfusion injuries [51,52].
NK-␬B pathway activation [42]. Recent evidence demon- Further studies have revealed the underlying mechanism with
strates that RIP3 is critical in necroptosis induced by various greater detail: the inactivation of RIP1 blocks the recruitment
stimuli [39,43]. It has been reported that RIP3 knockdown and interaction of RIP3 and RIP1, thus inhibits the formation
leads to a notable inhibition of necroptosis in HT-29 cells of necrosome, which is essential for necroptosis [40]. It is
[43]. Studies have demonstrated a low RIP3 expression level well established that RIP1 is the only active site of Nec-
in cells resistant to necroptosis, and transfection of these cells 1 [50]. A recent report indicated that Nec-1 protected cells
with the RIP3 gene enables them to undergo necroptosis when from necrosis by blocking the reduction of mitochondrial
the apoptotic pathway is blocked [44]. Phosphorylation of membrane potential (MMP) and activating the NF-␬B path-
RIP3 is essential in necroptosis, but the exact mechanism way, supporting mitochondrion as a potential active site of
remains unclear [39]. A recent report identified RIP3 as an Nec-1 [53]. Further investigations are needed to confirm the
essential mediator in TNF-␣ induced necroptosis using a new characteristics of Nec-1, as those characteristics may
screening technique involving RNA interference [44]. The only emerge in certain cell types or under certain stimulus.
interaction between RIP1 and RIP3 is based upon RIP homo- Aside from Nec-1, other members of the Necrostatin family,
typic interaction motif (RHIM). Mutations of RHIMs in RIP1 such as Nec5 [54] and Nec7 [55] have also been found to be
or RIP3 may block the formation of necrosomes and protect capable of inhibiting necroptosis too, although their targets
cells from necroptosis [45]. Moreover, RIP3 kinase activity remain to be elucidated.
is also required for the interaction between RIP1 and RIP3
[39].
Although RIP1 and RIP3 have been reported to be 3. Execution of necroptosis
necessary for necroptosis in most experimental models,
there are some contradictory reports. The necroptosis Execution necroptosis is equally important, but much
induced by TCR in FADD−/− T cells has been demon- less clarified compared to the upstream signaling pathway.
strated to be RIP1-dependent only [36]. Studies on murine It is unlikely that necrosomes could lead to cell death by
cytomegalovirus infection have identified a RIP3-dependent, destroying cellular organelles directly, as no obvious co-
RIP1-independent necroptosis induced in infected cells [46]. localization of necrosome or RIP3 has been found in any
Briefly, kinase activity of RIP1, RIP3, and the interaction cellular organelles [43]. Thus, necrosome may function
of RIP1 and RIP3 is required to guarantee the activation as an upstream signal and potentially triggers the execu-
of necroptotic pathway. Aside from the molecules discussed tion of cell death through various mechanisms. It has been
above, other necroptotic regulatory factors exist. demonstrated that necroptosis shares identical sub-cellular
As mention above, apoptosis suppression may drive cells events with necrosis, such as oxidative burst, mitochondrial
to use necroptosis as an alternative cell death mechanism. membrane hyperpolarization, lysosomal membrane perme-
Some caspase inhibitors, such as zVAD.fmk and BocD.fmk, abilization and plasma membrane permeabilization, but the
could induce necroptosis through autocrine production of mechanisms underlying those processes might be different
TNF-␣ [47]. However, treatment with Smac mimetic, which from necrosis [56]. We will discuss these subcellular events
mimics the function of Smac protein, could only result in in detail in the following sections.
apoptotic cell death, even though it can also induce autocrine
of TNF-␣ [48]. Most apoptotic inhibitors require the pres- 3.1. Reactive oxygen species (ROS)
ence of exogenous TNF-␣ to induce necroptosis. It has been
demonstrated that only certain cell types are able to execute ROS potentially leads to cell death by directly oxidizing or
necroptosis in response to TNF-␣ stimulus when the apopto- triggering various downstream pathways [57]. It has been pre-
sis pathway is inhibited or inactive. These cells include mouse viously demonstrated that TNF-␣ induced necrosis requires
fibrosarcoma L929 cells, human T cell leukemia Jurkat cells, the participation of ROS [58], although the exact mechanism
human monocytic leukemia U937 cells, MEFs, and human that triggers ROS production remains poorly understood.
colon cancer HT-29 cells. Among these cell lines, L929 cells Mitochondria are the principal producers of ROS in cells
are most extensively utilized in experiments related to necrop- [56,59,60]. RIP3 accelerates mitochondrial ROS production
tosis, as TNF-␣ induces necrosis prominently to apoptosis in and mitochondrial metabolism through the activation of a
W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258 253

series of metabolism-related enzymes. In T293 cells, RIP3 RIP1 and TNF-␣ have both been demonstrated to be essential
enhances the activity of glycogen phosphorylase (PYLG), for the binding of zVAD.fmk to ANT. This study also reported
glutamate-ammonia ligase (GLUL), and glutamate dehydro- that CYPD exhibits a protective role against cell death by sup-
genase 1 (GLUD1) during TNF-␣ induced necroptosis [61]. pressing zVAD.fmk binding to ANT. It has been shown that
All of these enzymes are essential for ROS production. PYLG CYPD is consistently up-regulated in several human tumor
is the rate-limiting enzyme in glycogen degradation and the tissues including breast, ovary, and uterus [75]. However,
glucose-1-phosphate released by PYLG is important in gly- another study reported that CYPD was required in oxidative
colysis. GLUL and GLUD1 are key enzymes in the process damage-induced cell death [76].
of oxidative phosphorylation. Moreover, by enhancing these
metobolism-related enzymes, RIP3 may also participate in 3.3. RNS
the decision regarding the choice of cell death because varia-
tion in energy metabolism status potentially leads to differing Nitric oxide (NO) is produced in endothelial cells by
mechanisms of cell-death [62]. endothelial nitric oxide synthase (eNOS). It affects many
NADPH oxidases are a family of enzymes specifically physical and pathological processes, such as vascular relax-
important in ROS production [63]. Several of the oxidases, ation, inflammation, cell proliferation, and cell death [77–80].
such as Nox1, Nox2, Nox3, Nox4, and p47phox, have been NO interacts with mitochondria [81], and influences cellular
shown to be upregulated in the presence of TNF-␣ [64–67]. bioenergetics as well as oxygen consumption [82,83]. NO can
Nox1 is activated by TNF-␣ and thus generates superoxides lead to programmed cell death in endothelial cells in a manner
in MEFs [66]. In this process, Nox1 forms a complex with similar to the necroptosis induced by TNF-␣ [84], and RIP1,
TRADD, RIP1, and a small GTPase called Rac1 [64]. Hence, RIP3, and ROS are all involved in the process. However, no
for necrosis induced by TNF-␣, RIP1 is required in ROS pro- receptor is required here. Since necrotic cell death is inhib-
duction [31]. However, in HT-29 cells, ROS is not required for ited by Nec-1 and is dependant upon RIP3, and possibly RIP1
necrosis induced by TNF-␣, Smac mimetic, and zVAD.fmk in addition, it may represent a type of necroptosis. However,
[43]. its mechanism is very different from the necroptosis induced
The c-Jun NH2-terminal kinase (JNK) plays a duel role by TNF-␣. Further investigation is warranted to clarify the
in TNF-␣-induced necrosis. On one hand, JNK plays a pro- underlying mechanisms in NO-induced necroptosis.
survival role and suppresses apoptosis induced by TNF-␣;
on the other hand, JNK is a pro-necrotic signal and initi- 3.4. PLA2 and LOXs
ates TNF-␣-induced necrotic cell death in fibroblasts [68].
JNK is activated downstream of RIP1 and TRAF2 in necro- PLA2 is a family of enzymes that liberate and cleave
sis [69,70], and TRAF2 is essential for JNK activation [71]. free fatty acids and lysophospholipids at the sn-2 posi-
JNK leads to necrosis by producing ROS, potentially by tion of glycerophospholipids. cPLA2 (calcium-dependent
impacting mitochondrial function [72], whereas intracellu- cytosolic type) is a member of PLA2 primarily responsi-
lar production of ROS activates the pro-necrotic effect of ble for initiating arachidonic acid (AA) metabolism [85].
JNK [57]. Therefore, JNK and ROS form a positive feedback The transmission and activation of cPLA2 requires calcium
loop and enhance necrosis. Recently, it has been reported that and phosphorylation [86,87]. cPLA2 plays an important role
JNK potentially promotes autocrine production of TNF-␣ by in TNF-␣-induced necrotic cell death in L929 cells and
mediating activating protein-1 (AP-1) activation in L929 cells MEFs, as well as in chemical or oxidant-induced necrosis in
treated with zVAD.fmk, consequently accelerating necropto- renal epithelial cells [88–90]. Lipoxygenase (LOXs) acts as a
sis [47]. downstream effector of PLA2 and is activated by an increased
concentration of calcium resulting from the generation of
3.2. ANT free fatty acids. LOXs contributes to lipid hydroperoxidation
and leads to the disruption of organelle and plasma mem-
Beyond the production of ROS, mitochondria also con- branes [91]. It has been reported that LOXs is involved in
tribute to necrotic cell death through an ADP/ATP-related both apoptosis and necrosis induced by TNF-␣ [92,93].
pathway. Synthesis of ATP in mitochondria requires the
normal activity of adenine nucleotide translocase (ANT),
an ADP/ATP carrier located in the inner mitochondrial 4. Necroptosis and other types of programmed cell
membrane. The activity of ANT is potentially affected by death
interaction with VDAC and cyclophilin D (CYPD). CYPD is
an important regulator of mitochondrial permeability transi- 4.1. Necroptosis and apoptosis
tion pore (MPTP) [73]. RIP1-dependent inhibition of ANT
has been reported in the programmed necrosis induced by Necroptosis and apoptosis induced by death receptor liga-
TNF-␣ and zVAD.fmk in U937 cells [74]; zVAD.fmk poten- tion share the same pathway from initiation to the formation
tially blocks the ability of ANT to transport cytoplasmic ADP, of complex II. Complex II is composed of FADD, RIP1,
thereby inducing a massive ATP depletion in mitochondria. RIP3, and caspase-8. When caspase-8 is inhibited, both RIP1
254 W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258

and RIP3 are phosphorylated and the “necrosome” is acti- ily form mitochondria [103]. Catalase is a scavenger enzyme
vated, inducing necroptosis in consequence. However, when of ROS. It has been reported that catalase is discomposed
caspase-8 remains activated, it attenuates the activity of RIP3 by autophagy, leading to an imbalance of intracellular ROS
through cleavage and forms the complex IIa with FADD and [104]. Secondly, PARP1 is over-activated by autophagy and
RIP1, leading to apoptosis. Among the factors that affect the leads to necrotic cell death. However, whether PARP1 acti-
switch between apoptosis and necroptosis are: the nature of vation occurs upstream or downstream of ROS production
the stimulus, type of cell, and activity of caspases. As previ- remains controversial [105]. In contrast to the results men-
ously mentioned, only certain types of stimulus are capable tioned above, it has been reported that autophagy protects
of inducing necroptosis, and not all cell types are able to L929 cells from necroptosis induced by zVAD.fmk [98].
die through necroptosis. In concordance with this finding, a Rapamycin, a molecule that increases the flux of autophagy,
recent study demonstrated that shikonin, previously known as was demonstrated to inhibit zVAD-induced cell necrosis.
an apoptosis inducer [94], was able to induce necroptosis in Moreover, suppression of autophagy enhanced the necropto-
MCF-7 and HEK293 cells, but only apoptosis in HL60 cells sis induced by zVAD.fmk. Given that zVAD.fmk are capable
[95]. However, necroptosis in HL-60 cells can be induced of inhibiting the activity of cathepsins and calpains and
by DT-GMCSF (diphtheria toxin granulocyte-macrophage that the two proteases proved to be essential for autophagy,
colony-stimulating factor), a novel fusion toxin treatment it is possible that zVAD.fmk suppresses autophagy by
for AML [96]. These results underscore the importance blocking cathepsins and calpains directly, and that the sup-
of proper stimuli to trigger necroptosis. The exact mecha- pression of pro-survival autophagy leads to necroptosis in
nism of cell type-dependence and stimulus-dependence of L929 cells.
necroptosis remains unclear. The cells capable of exhibiting
necroptosis appear to have nothing in common. Conversion
between necroptosis and apoptosis has been identified in the 4.3. Necroptosis and PARP-induced programmed
same cell line, in accordance to differing stimuli and cir- necrosis
cumstance [97]. The alternation of mitochondrial membrane
potential may partially contribute to the conversion. In cer- Poly (ADP-ribose) polymerase-1 (PARP-1) is a nuclear
tain circumstances, necroptosis and apoptosis could occur enzyme activated by DNA-strand breaks, and plays a key
simultaneously in cells [3]. role in repairing DNA damage. Over-activation of PARP-1
potentially leads to NAD + reduction, ATP depletion, cellu-
4.2. Necroptosis and autophagy lar energy failure, and release of apoptosis-inducing factor
(AIF) from mitochondria to nucleus, which result in mas-
Autophagic cell death is characterized by massive sive DNA damage and necrotic cell death. PARP-1-mediated
autophagosomes, a two-membrane vesicle containing cyto- necrosis contributes to several pathological processes, such
plasmic organelles or degenerating cytosol. Autophagy has as ischemia-reperfusion, inflammation, ROS-induced injury,
proven to be protective to cells, as knock-down of autophagy- and glutamate excitotoxicity. Dysfunction of mitochondria
related genes appears to promote cell death [98]. However, is a key step in PARP-1-induced necrosis. The mecha-
as some investigations have recently discovered, autophagy nism of PARP-1 induced mitochondria dysfunction remains
may also contribute to necroptotic cell death [50,52,99,100] unknown. It is potentially associated with the modification
even though it is not indispensable for necroptosis [101]. of Bcl-2 families, essential regulators of membrane perme-
The mechanism underlying autophagy activation remains ability in mitochondria. BNip3 is a member of the BH3-only
controversial. It has been reported that a signaling pathway Bcl-2 family. A recent study reported that BNip3 mediated
involving caspase-8, RIP1, JNK, and c-JUN may contribute TNF-␣-induced necroptosis in alveolar lung cells [106]. In
to autophagy activation in L929 cells [102]. c-Src was PARP-1 induced necrosis, calpains are required for translo-
recently reported to be associated with caspase-8 in L929 cation of AIF from mitochondrial membrane to nucleus
cells at resting state. zVAD.fmk Treatment releases the acti- [107]. Calpain is a protease regulated by intracellular calcium
vated c-Src, and triggered c-Src-dependent JNK and ERK level. The released AIF binds to DNA, leading to chromatin
signaling pathways [103]. In proliferative T cells, FADD condensation and DNA fragmentation, and finally induces
and caspase-8 work in concert to limit autophagy and pro- necrosis [108]. Inhibition of PARP fails to protect Jurkat cells
tect T cells from necroptosis [99]. Recently, it has also from necroptosis, indicating that PARP activation is poten-
been reported that necroptosis promotes autophagy, as Nec- tially not be involved in necroptosis [50]. However, it has
1 reduced LC3 levels in FADD−/− cells and rescued the been previously reported that Nec-1 blocks the transloca-
cycling and proliferation of FADD−/− T cells [99] simul- tion of AIF from mitochondria to nucleus and suppresses the
taneously. This phenomenon indicates the possibility that activation of PARP-1 [109,110]. These findings suggest the
autophagy and necroptosis may potentially form a posi- possibility that PARP-1 induced necrosis acts downstream
tive feedback loop and reinforce the process of cell death. of necroptosis. Moreover, PARP-2, the closest homologue to
Autophagy might accelerate cell necrosis in two ways. Firstly, PARP-1, has been identified as one of the essential regulators
autophagy induces an increased level of ROS released primar- of necroptosis by a genome-wide siRNA screen study [12].
W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258 255

5. Necroptosis in physiological and pathological Necroptosis may accelerate cancer cell death or enhance
processes the sensitivity of tumor cells to anti-cancer treatment
[96,100,119,120]. A study on pediatric acute lymphoblastic
Necroptosis may play a regulatory role in the development leukemia (ALL) revealed the important role of necroptosis
of the immune system. It is related to the regulation of T in overcoming the glucocorticoid resistance of ALL cells
cell proliferation [33,99] and macrophage survival [12]. An [100]. Additionally, research has demonstrated that necropto-
increased expression of necroptosis related genes have been sis participates in acute myeloid leukemia (AML) cell death
found in mouse immune cells [12]. RIP3 expression is found induced by a novel fusion toxin treatment [96]. Other research
to be elevated in immunologic organs, such as thymus and has demonstrated that necroptosis is able to overcome resis-
spleen in mice [43]. tance to cancer drugs mediated by P-glycoprotein, Bcl-2,
Programmed necrosis plays an important role in the and Bcl-xL in cancer cell lines [95]. All these results indi-
response to viral infection by serving as a backup mechanism cate necroptosis may be a potent therapeutic target for the
of cell death other than apoptosis. Virus-infected cells, which treatment of cancer.
are resistant to apoptosis, are found to be highly sensitive to
necroptosis [111]. Knock-down of RIP3 in vaccinia virus
(VV) infected cells apparently protects the cells from cell 7. Conclusions
death induced by activated T cells or TNF-␣. Additionally,
TNF-␣ potentially induces necroptosis without the presence As a newly identified form of cell death, necroptosis plays
of zVAD.fmk in VV infected cells [39], indicating that viral an important role in immune system regulation, tissue injury,
infection may serve to sensitize infected cells to necroptosis. and cancer development. Necroptosis is might be triggered by
Regarding the innate immune response following viral infec- TNF-␣ and caspase inhibitors. RIP1 and RIP3 are critical reg-
tion, it has been demonstrated that the inflammation caused ulators of necroptosis, and necrostatin-1, an inhibitor of RIP1,
by VV infection is evidently reduced in RIP3−/− mice [112], specifically blocks necroptosis. Necroptosis is a potential
as well as TNFR1−/− and TNFR2−/− cells [32,112], sug- therapeutic target for many diseases. Inhibition of necrop-
gesting the necroptotic pathway plays a key role in triggering tosis potentially protects cells from severe injury induced
inflammation. by trauma or inflammation. Hence, the necrostatin family
Recently, a viral inhibitor of RIP3-dependent necroptosis may serve as a potential therapeutic tool for diseases such
has been reported [46]. The findings indicated that murine as ischemic/reperfusion of brain and myocardiac infarction.
cytomegalovirus (MCMV) M45, which encodes the viral Acceleration of necroptosis may provide a backup route to
inhibitor of RIP activation (vIRA), potentially inhibits RIP cell death for tumor cells, particularly for those resistant to
induced necrotic cell death and accelerates viral replication. apoptosis-inducing cancer therapy.
It has been demonstrated that M45 inhibits TNFR/FasL/TNF-
␣-induced necroptosis by either inhibiting RIP1, RIP1–RIP3
complex formation, or RIP3 alone [113,114]. Reviewers
Necroptosis is also involved in several types of tissue
damage. Enhanced expression of RIP3 was detected in pan- Junying Yuan, Ph.D., Harvard Medical School, Depart-
creatitis tissue induced by cerulein, and deletion of RIP3 ment of Cell Biology, 250 Longwood Avenue, Boston, MA
was shown to prevent the tissue damage [39,43]. Moreover, 02115, United States.
necroptosis participates in hypoxia injury, ischemia reperfu- Tao Cheng, M.D., University of Pittsburgh School of
sion damage of brain and retina, cortical impact, myocardial Medicine, Department of Radiation Oncology, Pittsburgh,
infarction, and excitotoxicity in neurons [51,52,115–117]. PA, United States.

6. Necroptosis and cancer Conflict of interest

Necroptosis serves as a “backup” pathway of cell death No conflict of interest is declared.


differing from apoptosis. Defects in necroptosis or variations
of necroptosis-related genes may contribute to the patho-
logical process of human malignancies. Our research group Acknowledgements
has discovered necroptotic defects in chronic lymphocytic
leukemia (CLL) cells (unpublished results). RIP3 SNPs have This work was supported by National Natural Science
been identified in non-Hodgkin lymphoma patients. Genetic Foundation of China (30500603, 30871104, 30971295,
variation of TNF and TLR signaling pathways have also been 81170486, 81170485), International Cooperation Program
detected [118]. Inactive mutation of CYLD was found to of Jiangsu Province (BZ2010041), the Program for Devel-
accelerate proliferation and migration in epidermal cancer opment of Innovative Research Team in the First Affiliated
cells [22]. Hospital of NJMU, and A Project Funded by the Priority Aca-
256 W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258

demic Program Development of Jiangsu Higher Education enhanced proliferative, survival and angiogenic properties to epider-
Institutions. mal cancer cells. Oncogene 2010;29:6522–32.
[23] Urbanik T, Kohler BC, Boger RJ, et al. Down-regulation of CYLD as
a trigger for NF-␬ activation and a mechanism of apoptotic resistance
in hepatocellular carcinoma cells. Int J Oncol 2011;38:121–31.
References [24] Wertz IE, O’Rourke KM, Zhou H, et al. De-ubiquitination and ubiq-
uitin ligase domains of A20 downregulate NF-␬B signalling. Nature
[1] Kroemer G, Galluzzi L, Vandenabeele P, et al. Classification of cell 2004;430:694–9.
death: recommendations of the Nomenclature Committee on Cell [25] Shembade N, Ma A, Harhaj EW. Inhibition of NF-␬B signaling
Death. Cell Death Differ 2009;16:3–11. by A20 through disruption of ubiquitin enzyme complexes. Science
[2] Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phe- 2010;327:1135–9.
nomenon with wide-ranging implications in tissue kinetics. Br J [26] Ea CK, Deng L, Xia ZP, Pineda G, Chen ZJ. Activation of IKK by
Cancer 1972;26:239–57. TNF␣ requires site-specific ubiquitination of RIP1 and polyubiquitin
[3] Galluzzi L, Joza N, Tasdemir E, et al. No death without life:vital by NEMO. Mol Cell 2006;22:245–57.
function of apoptotic effectors. Cell Death Differ 2009;16:1093–107. [27] Wong WW, Gentle IE, Nachbur U, Anderton H, Vaux DL, Silke J.
[4] Holler N, Zaru R, Micheau O, et al. Fas triggers an alternative, RIPK1 is not essential for TNFR1-induced activation of NF-␬B. Cell
caspase-8-independent cell death pathway using the kinase RIP as Death Differ 2010;17:482–7.
effector molecule. Nat Immunol 2000;1:489–95. [28] Lee TH, Shank J, Cusson N, Kelliher MA. The kinase activity of Rip1
[5] Artus C, Boujrad H, Bouharrour A, et al. AIF promotes chromatinoly- is not required for tumor necrosis factor-␣-induced I␬B kinase or p38
sis and caspase-independent programmed necrosis by interacting with MAP kinase activation or for the ubiquitination of Rip1 by Traf2. J
histone H2AX. EMBO J 2010;29:1585–99. Biol Chem 2004;279:33185–91.
[6] Zong WX, Ditsworth D, Bauer DE, Wang ZQ, Thompson CB. Alky- [29] Schneider-Brachert W, Tchikov V, Merkel O, et al. Inhibition of TNF
lating DNA damage stimulates a regulated form of necrotic cell death. receptor 1 internalization by adenovirus 14.7K as a novel immune
Genes Dev 2004;18:1223–6. escape mechanism. J Clin Invest 2006;116:2901–13.
[7] Edinger AL, Thompson CB. Death by design:apoptosis, necrosis or [30] Bertrand MJ, Milutinovic S, Dickson KM, et al. cIAP1 and cIAP2
autophagy. Curr Opin Cell Biol 2004;16:663–9. facilitate cancer cell survival by functioning as E3 ligases that pro-
[8] Galluzzi L, Kepp O, Kroemer G. RIP kinases initiate programmed motes RIP1 ubiquitination. Mol Cell 2008;30:689–700.
necrosis. J Mol Cell Biol 2009;1:8–10. [31] Lin Y, Choksi S, Shen HM, et al. Tumor necrosis factor-induced non-
[9] Degterev A, Hitomi J, Germscheid M, et al. Identification of RIP1 apoptotic cell death requires receptor-interacting protein-mediated
kinase as a specific cellular target of necrostations. Nat Chem Biol cellular reactive oxygen species accumulation. J Biol Chem
2008;4:313–21. 2004;279:10822–8.
[10] Fiers W, Beyaert R, Boone E, et al. TNF-induced intracellular sig- [32] Chan FK, Shisler J, Bixby JG, et al. A role for tumor necrosis factor
naling leading to gene induction or to cytotoxicity by necrosis or by receptor-2 and receptor-interacting protein in programmed necrosis
apoptosis. J Inflamm 1995;47:67–75. and antiviral responses. J Biol Chem 2003;278:51613–21.
[11] Kalai M, Van Loo G, Vanden Berghe T, et al. Tipping the balance [33] Osborn SL, Diehl G, Han SJ, et al. Fas-associated death domain
between necrosis and apoptosis in human and murine cells treated (FADD) is a negative regulator of T-cell receptor-mediated necropto-
with interferon and dsDNA. Cell Death Differ 2002;9:981–94. sis. Proc Natl Acad Sci USA 2010;107:13034–9.
[12] Hitomi J, Christofferson DE, Ng A, et al. Identification of a molecular [34] Ermolaeva MA, Michallet MC, Papadopoulou N, et al. Function of
signaling network that regulates a cellular necrotic cell death pathway. TRADD in tumor necrosis factor 1 signaling and in TRIF-dependent
Cell 2008;135:1311–23. inflammatory responses. Nat Immunol 2008;9:1037–46.
[13] Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science [35] Wang L, Du F, Wang X. TNF-alpha induced two distinct caspase-8
2002;296:1634–5. activation pathways. Cell 2008;133:693–703.
[14] Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. [36] Kim JW, Choi EJ, Joe CO. Activation of death-inducing signaling
An endotoxin-induced serum factor that causes necrosis of tumors. complex (DISC) by pro-apoptotic C-terminal fragment of RIP. Onco-
Proc Natl Acad Sci USA 1975;72:3666–70. gene 2000;19:4491–9.
[15] Laster SM, Wood JG, Gooding LR. Tumor necrosis factor can [37] Feng S, Yang Y, Mei Y, et al. Cleavage of RIP3 inactivates its caspase-
induce both apoptotic and necrotic forms of cell lysis. J Immunol independent apoptosis pathway by removal of kinase domain. Cell
1988;141:2629–34. Signal 2007;19:2056–67.
[16] Andera L. Signaling activated by the death receptors of the TNFR [38] Declercq W, Vanden Berghe T, Vandenabeele P. RIP kinases at the
family. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub crossroads of cell death and survival. Cell 2009;138:229–32.
2009;153:173–80. [39] Cho YS, Challa S, Moquin D, et al. Phosphorylation-driven assembly
[17] Chan FK, Chun HJ, Zheng L, Siegel RM, Bui KL, Lenardo MJ. A of the RIP1–RIP3 complex regulated programmed necrosis and virus-
domain in TNF receptors that mediates ligand-independent receptor induced inflammation. Cell 2009;137:1112–23.
assembly and signaling. Science 2000;288:2351–4. [40] Sun X, Lee J, Navas T, Baldwin DT, Stewart TA, Dixit VM. RIP3, a
[18] Wertz IE, Dixit VM. Ubiquitin-mediated regulation of TNFR1 sig- novel apoptosis-induing kinase. J Biol Chem 1999;274:16871–5.
naling. Cytokine Growth Factor Rev 2008;19:313–24. [41] Zhao L, Wang G, Lu D, et al. Homocysteine, hRIP3 and congenital
[19] Mahoney DJ, Cheung HH, Mrad RL, et al. Both cIAP1 and cIAP2 cardiovascular malformations. Anat Embryol 2006;211:203–12.
regulate TNFalpha-mediated NF-kappaB activation. Proc Natl Acad [42] Newton K, Sun X, Dixit VM. Kinase RIP3 is dispensable for normal
Sci USA 2008;105:11778–83. NF-␬B, signaling by the B-cell and T-cell receptors, tumor necro-
[20] Varfolomeev E, Goncharov T, Fedorova AV, et al. c-IAP1 and c-IAP2 sis factor receptor 1, and Toll-like receptor 2 and 4. Mol Cell Biol
are critical mediators of tumor necrosis factor alpha(TNFalpha)- 2004;24:1464–9.
induced NF-kappaB activation. J Biol Chem 2008;283:24295–9. [43] He S, Wang L, Miao L, et al. Receptor interacting protein
[21] Kovalenko A, Chable-Bessia C, Cantarella G, Israel A, Wallach D, kinase-3 determines cellular necrotic response to TNF-␣. Cell
Courtois G. The tumor suppressor CYLD negatively regulates NF-␬B 2009;137:1100–11.
signaling pathway by deubiquitination. Nature 2003;424:801–5. [44] Cho Y, Challa S, Chan FK. A RNA interderence screen identifies RIP3
[22] Alameda JP, Moreno-Maldonado R, Navarro M, et al. An inactivat- as an essential inducer of TNF-induced programmed necrosis. Adv
ing CYLD mutation promotes skin tumor progression by conferring Exp Med Biol 2011;691:589–93.
W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258 257

[45] Sun X, Yin J, Starovasnik MA, Fairbrother WJ, Dixit VM. Iden- [67] Moe KT, Aulia S, Jiang F, et al. Differential upregulation of Nox
tification of a novel homotypic interaction motif required for the homologues of NADPH oxidase by tumor necrosis factor-alpha in
phosphorylation of receptor-interacting protein (RIP) by RIP3. J Biol human aortic smooth muscle and embryonic kidney cells. J Cell Mol
Chem 2002;277:9505–11. Med 2007;10:231–9.
[46] Upton JW, Kaiser WJ, Mocarski ES. Virus inhibition of RIP3- [68] Ventura JJ, Cogswell P, Flavell RA, Baldwin Jr AS, Davis RJ. JNK
dependent necrosis. Cell Host Microbe 2010;7:321–413. potentiates TNF-stimulated necrosis by increasing the production of
[47] Wu YT, Tan HL, Huang Q, Sun XJ, Zhu X, Shen HM. zVAD-induced cytotoxic reactive oxygen species. Genes Dev 2004;18:2905–15.
necroptosis in L929 cells depends on autocrine production of TNF- [69] Shen HM, Lin Y, Choksi S, et al. Essential role of receptor-interacting
␣ mediated by the PKC-MAPKs-AP-1 pathway. Cell Death Differ protein and TRAF2 in oxidative stress-induced cell death. Mol Cell
2011;18:26–37. Biol 2004;24:5914–22.
[48] Petersen SL, Wang L, Yalcin-Chin A, et al. Autocrine TNFalpha [70] Xu Y, Huang S, Liu ZG, Han J. Poly(ADP-ribose) polymerase-1 sig-
signaling renders human cancer cells susceptible to Smac-mimetic- naling to mitochondria in necrotic cell death requires RIP1/TRAF2-
induced apoptosis. Cancer Cell 2007;12:445–56. medited JNK1 activation. J Biol Chem 2006;281:8788–95.
[49] Vercammen D, Brouckaert G, Denecker G, et al. Dual signaling of [71] Lee SY, Reichlin A, Santana A, Sokol KA, Nussenzweig MC,
the Fas receptor:initiation of both apoptotic and necrotic cell death Choi Y. TRAF2 is essential for JNK but not NF-kappaB activa-
pathways. J Exp Med 1998;188:919–30. tion and regulates lymphocyte proliferation and survival. Immunity
[50] Degterev A, Huang Z, Boyce M, et al. Chemical inhibitor of nonapop- 1997;7:703–13.
totic cell death with therapeutic potential for ischemic brain injury. [72] Davis RJ. Signal transduction by the JNK group of MAP kinases. Cell
Nat Chem Biol 2005;1:112–9. 2000;103:239–52.
[51] Xu X, Chua KW, Chua CC, Liu CF, Hamdy RC, Chua BH. [73] Nakagawa T, Shimizu S, Watanabe T, et al. Cyclophilin D-dependent
Synergistic protective effects of humanin and necrostatin-1 on mitochondrial permeability transition regulates some necrotic but not
hypoxia and ischemia/reperfusion injury. Brain Res 2010;1355: apoptotic cell death. Nature 2005;434:652–8.
189–94. [74] Temkin V, Huang Q, Liu H, Osada H, Pope RM. Inhibition of
[52] Rosenbaum DM, Degterev A, David J, et al. Necroptosis, a novel ADP/ATP exchange in receptor-interacting protein-mediated necro-
form of caspase-independent cell death, contributes to neuronal dam- sis. Mol Cell Biol 2006;26:2215–25.
age in a retinal ischemia-reperfusion injury model. J Neurosci Res [75] Schubert A, Grimm S, Cyclophilin D. A component of the per-
2010;88:1569–76. meability transition-pore, is an apoptosis repressor. Cancer Res
[53] Hsu TS, Yang PM, Tsai JS, Lin LY. Attenuation of cadmium-induced 2004;64:85–93.
necrotic cell death by necrostatin-1: potential necrostatin-1 acting [76] Baines CP, Kaiser RA, Purcell NH, et al. Loss of cyclophilin D reveals
sites. Toxicol Appl Pharmacol 2009;235:153–62. a critical role for mitochondrial permeability transition in cell death.
[54] Wang K, Li J, Degterev A, Hsu E, Yuan J, Yuan C. Structure–activity Nature 2005;434:658–62.
relationship of a novel necroptosis inhibitor, necrostatin-5. Bioorg [77] Bredt DS. Endogenous nitric oxide synthesis:biological functions and
Med Chem Lett 2007;17:1455–65. pathophysiology. Free Radic Res 1999;31:577–96.
[55] Zheng W, Degterev A, Hsu E, Yuan J, Yuan C. Structure–activity [78] van der Vliet A, Eiserich JP, Cross CE. Nitric oxide: a pro-
relationship of a novel necroptosis inhibitor, necrostatin-7. Bioorg inflammatory mediator in lung disease? Respir Res 2000;1:67–72.
Med Chem Lett 2008;18:4923–35. [79] Zheng J, Wen Y, Austin JL, Chen DB. Exogenous nitric oxide stim-
[56] Berghe TV, Vanlangenakker N, Parghoens E, et al. Necroptosis, necro- ulates cell proliferation via activation of a mitogen-activated protein
sis and secondary necrosis converge on similar cellular disintegration kinase pathway in ovine fetoplacental artery endothelial cells. Biol
features. Cell Death Differ 2010;17:922–30. Reprod 2006;74:375–82.
[57] Morgan MJ, Kim YS, Liu Z. Lipid rafts and oxidative stress-induced [80] Uehara T, Nakamura T, Yao D, et al. S-nitrosylated protein-disulphide
cell death. Antioxid Redox Signal 2007;9:1471–83. isomerase links protein misfolding to neurodegeneration. Nature
[58] Sakon S, Xue X, Takekawa M, et al. NF-kappaB inhibits TNF-induced 2006;441:513–7.
accumulation of ROS that mediate prolonged MAPK activation and [81] Burwell LS, Brookes PS. Mitochondria as a target for the cardiopro-
necrotic cell death. EMBO J 2003;22:3898–909. tective effects of nitric oxide in ischemia-reperfusion injury. Antioxid
[59] Jezek P, Hlavata L. Mitochondria in homeostasis of reactive oxy- Redox Signal 2008;10:579–99.
gen species in cell, tissues, and organism. Int J Biochem Cell Biol [82] Cooper CE, Giulivi C. Nitric oxide regulation of mitochondrial oxy-
2005;37:2478–503. gen consumption II: molecular mechanism and tissue physiology. Am
[60] Chen Q, Vazquez EJ, Moghaddas S, Hoppel CL, Lesnefsky EJ. Pro- J Physiol Cell Physiol 2007;292:C1993–2003.
duction of reactive oxygen species by mitochondria: central role of [83] Moncada S, Erusalimsky JD. Does nitric oxide modulate mito-
complex III. J Biol Chem 2003;278:36027–31. chondrial energy generation and apoptosis? Nat Rev Mol Cell Biol
[61] Zhang DW, Shao J, Lin J, et al. RIP3, an energy metabolism regula- 2002;3:214–20.
tor that switches TNF-induced cell death from apoptosis to necrosis. [84] Davis CW, Hawkins BJ, Ramasamy S, et al. Nitration of the mitochon-
Science 2009;325:332–6. drial complex I subunit NDUFB8 elicits RIP1-and-RIP3-mediated
[62] Nicotera P, Melino G. Regulation of the apoptosis-necrosis switch. necrosis. Free Radic Biol Med 2010;48:306–17.
Oncogene 2004;23:2757–65. [85] Akiba S, Sato T. Cellular function of calcium-independent phospho-
[63] Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat lipase A2 . Biol Pharm Bull 2004;27:1174–8.
Rev Immunol 2004;4:181–9. [86] Evans JH, Spencer DM, Zweifach A, Leslie CC. Intracellular calcium
[64] Kim YS, Morgan MJ, Choksi S, Liu ZG. TNF-induced activation of signals regulating cytoslic phospholipase A2 translocation to internal
the Nox1 NADPH oxidase and its role in the induction of necrotic membranes. J Biol Chem 2001;276:30150–60.
cell death. Mol Cell 2007;26:675–87. [87] Hefner Y, Borsch-Haubold AG, Murakami M, et al. Serine 727
[65] Dang PM, Stensballe A, Boussetta T, et al. A specific p47phox- phosphorylation and activation of cytosolic phospholipase A2 by
serine phosphorylated by convergent MAPKs mediates neutrophil MNK1-related protein kinases. J Biol Chem 2000;275:37542–51.
NADPH oxidase priming at inflammatory sites. J Clin Invest [88] Hayakawa M, Ishida N, Takeuchi K, et al. Arachidonic acid-selective
2006;116:2033–43. cytosolic phospholipase A2 is crucial in the cytotoxic action of tumor
[66] Li L, He Q, Huang X, et al. Nox3-derived reactive oxygen species necrosis factor. J Biol Chem 1993;26:11290–5.
promote TNF-alpha-induced reductions in hepatocyte glycogen levels [89] El Mahdani NE, Ameyar M, Cai Z, Colard O, Masliah J,
via a JNK pathway. FEBS Lett 2010;584:995–1000. Chouaib S. Resistance to TNF-induced cytotoxicity correlates with
258 W. Wu et al. / Critical Reviews in Oncology/Hematology 82 (2012) 249–258

an abnormal cleavage of cytosolic phopholipase A2 . J Immunol [109] Xu X, Chua CC, Kong J, et al. Necrostatin-1 protects against
2000;165:6756–61. glutamate-induced glutathione depletion and caspase-independent
[90] Kohjimoto Y, Kennington L, Scheid CR, Honeyman TW. Role of cell death in HT-22 cells. J Neurochem 2007;103:2004–14.
phospholipase A2 in the cytotoxic effects of oxalate in cultured renal [110] Xu X, Chua CC, Zhang M, et al. The role of PARP activa-
epithelial cells. Kidney Int 1999;56:1432–41. tion in glutamate-induced necroptosis in HT-22 cells. Brain Res
[91] van Leyen K, Duvoisin RM, Engelhardt H, Wiedmann M. A func- 2010;1343:206–12.
tion for lipoxygenase in programmed organelle degradation. Nature [111] Li M, Beg AA. Induction of necrotic-like cell death by tumor necrosis
1998;395:392–5. factor alpha and caspase inhibitors: novel mechanism for killing virus-
[92] Maccarrone M, Melino G, Finazzi-Agro A. Lipoxygenases and infected cells. J Virol 2000;74:7470–7.
their involvement in programmed cell death. Cell Death Differ [112] Ruby J, Bluethmann H, Peschon JJ. Antiviral activity of tumor necro-
2001;8:776–84. sis factor (TNF) is mediated via p55 and p57 TNF receptors. J Exp
[93] Festjens N, Kalai M, Smet J, et al. Butylated hydroxyanisole is Med 1997;186:1591–6.
more than a reactive oxygen species scavenger. Cell Death Differ [113] Mack C, Sickmann A, Lembo D, Brune W. Inhibition of proinflam-
2006;13:166–9. matory and innate immune signaling pathways by a cytomegalovirus
[94] Wu Z, Wu LJ, Li LH, Tashiro S, Onodera S, Ikejima T. Shikonin RIP1-interacting protein. Proc Natl Acad Sci USA 2008;105:3094–9.
regulates HeLa cell death via caspase-3 activation and blockage of [114] Upton JW, Kaiser WJ, Mocarski ES. Cytomegalovirus M45 cell death
DNA synthesis. J Asian Nat Prod Res 2004;6:155–66. suppression requires receptor-interacting protein (RIP) homotypic
[95] Han W, Li L, Qiu S, et al. Shikonin circumvents cancer drug resistance interaction motif (RHIM)-dependent interaction with RIP1. J Biol
by induction of a necroptotic death. Mol Cancer Ther 2007;6:1641–9. Chem 2008;283:16966–70.
[96] Horita H, Frankel AE, Thorburn A. Acute myeloid leukemia-targeted [115] Smith CC, Davidson SM, Lim SY, Simpkin JC, Hothersall JS, Yellon
toxin activates both apoptotic and necroptotic death mechanisms. DM. Necrostatin: a potential novel cardioprotective agent? Cardio-
PLoS One 2008;12:e3909. vasc Drugs Ther 2007;21:227–33.
[97] Han W, Xie J, Li L, Liu Z, Hu X. Necrostatin-1 reverts shikonin- [116] Li Y, Yang X, Ma C, Qiao J, Zhang C. Necroptosis contributes to
induced necroptosis to apoptosis. Apoptosis 2009;14:674–86. the NMDA-induced excitotoxicity in rat’s cultured cortical neurons.
[98] Wu YT, Tan HL, Huang Q, Kim YS, Pan N, Ong WY. Autophagy plays Neurosci Lett 2008;447:120–3.
a protective role during zVAD-induced necrotic cell death. Autophagy [117] You Z, Savitz SI, Yang J, et al. Necrostatin-1 reduces histopathology
2008;4:457–66. and improves functional outcome after controlled cortical impact in
[99] Bell BD, Leverrier S, Weist BM, et al. FADD and caspase-8 control mice. J Cereb Blood Flow Metab 2008;28:1564–73.
the outcome of autophagic signaling in proliferation T cells. Proc Natl [118] Cerhan JR, Ansell SM, Fredericksen ZS, et al. Genetic variation
Acad Sci USA 2008;105:16677–82. in 1253 immune and inflammation genes and risk of non-Hodgkin
[100] Bonapace L, Bornhauser BC, Schmitz M, et al. Induction of lymphoma. Blood 2007;110:4455–63.
autophagy-dependent necroptosis is required for childhood acute lym- [119] Mantel F, Frey B, Haslinger S, et al. Combination of lonising irradi-
phoblastic leukemia cells to overcome glucocorticoid resistance. J ation and hyperthermia activates programmed apoptotic and necrotic
Clin Invest 2010;120:1310–23. cell death pathways in human colorectal carcinoma cells. Strahlenther
[101] Shiraishi H, Okamoto H, Hara H, Yoshida H. Alternative cell death of Onkol 2010;186:587–99.
Apaf1-deficient neural progenitor cells withdrawal of EGF or insulin. [120] Zhang H, Zhong C, Shi L, Guo Y, Fan Z. Granulysin induces cathepsin
Biochim Biophys Acta 2010;1800:405–15. B release from lysosomes of target tumor cells to attack mitochon-
[102] Yu L, Alva A, Su H, et al. Regulation of an ATG7-belin 1 program of dria through processing of bid leading to necroptosis. J Immunol
autophagic cell death by caspase-8. Science 2004;304:1500–2. 2009;182:6993–7000.
[103] Chen SY, Chiu LY, Maa MC, Wang JS, Chien CL, Lin WW. zVAD-
induced autophagic cell death requires c-Src-dependent ERK and
JNK activation and reactive oxygen species generation. Autophagy
2011;7:217–28. Biographies
[104] Yu L, Wan F, Dutta S, et al. Autophagic programmed cell
death by selective catalase degradation. Proc Natl Acad Sci USA
2006;103:4952–7. Dr. Peng Liu is a professor of Department of Hematology
[105] Xu Y, Kim SO, Li Y, Han J. Autophagy contributes to caspase- at the First Affiliated Hospital of Nanjing Medical University.
independent macrophage cell death. J Biol Chem 2006;281: He specializes in clinical hematology with a research focus in
19179–87. molecular biology of leukemia. He also has a strong interest in
[106] Kim JY, Kim YJ, Lee S, Park JH. BNip3 is a mediator of TNF-induced
developing novel strategies to improve the efficacy of cancer
necrotic cell death. Apoptosis 2011;16:114–26.
[107] Cao G, Xing J, Xiao X, et al. Critical role of calpain I in mitochondrial chemotherapy.
release of apoptosis-inducing factor in ischemic neuronal injury. J
Dr. Jianyong Li is a professor and Director of Department
Neurosci 2007;27:9278–93.
[108] Ye H, Cande C, Stephanou NC, et al. DNA binding is required for of Hematology at the First Affiliated Hospital of Nanjing
the apoptogenic action of apoptosis inducing factor. Nat Struct Biol Medical University. He has a strong interest in clinical and
2002;9:680–4. basic study of lymphoid malignacies.

Potrebbero piacerti anche