Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Critical Reviews in Oncology/Hematology 66 (2008) 52–64

Concepts of activated T cell death


Dirk Brenner, Peter H. Krammer, Rüdiger Arnold ∗
Tumor Immunology Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
Accepted 16 January 2008

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
2. Shut-down of the immune response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
3. Regulation of AICD by cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
4. NF-␬B is essential for T cell survival . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
5. HPK1 controls the onset of AICD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
6. Suppression of NF-␬B induces AICD. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
7. AICD can involve death receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
8. AICD and CD95 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
9. AICD independent of death receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
10. Activated T cell death without TCR restimulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
11. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63

Abstract
Lymphocytes of the adaptive immune system play a crucial role in defending the organism against pathogens. Initial stimulation via antigen
receptors induces activation and proliferation of lymphocytes to generate effector cells that clear the pathogen from the body. During the
shut-down of the immune response activated lymphocytes are removed by two mechanisms. T cells that are restimulated during the end of the
immune response die by activation-induced cell death (AICD), whereas activated lymphocytes which are not restimulated die by activated cell
autonomous death (ACAD). Here, we discuss the regulation of AICD and ACAD in T cells and review the role of cytokines, T cell receptor
(TCR) proximal signaling mediators like hematopoietic progenitor kinase 1 (HPK1) and the NF-␬B pathway. We distinguish between AICD
dependent on or independent of death receptor ligation, and discuss caspase-independent death of T cells.
© 2008 Elsevier Ireland Ltd. All rights reserved.

Keywords: Apoptosis; T cells; AICD; ACAD; Immune response; HPK1; NF-␬B

1. Introduction
Abbreviations: AICD, activation-induced cell death; ACAD, activated
Apoptosis is involved in T cell development, T cell recep-
T cell autonomous death; Bcl-2, B cell leukemia-2; BCR, B cell recep-
tor; CD95L, CD95-ligand; DISC, death-inducing signaling complex; FLIP, tor (TCR) repertoire selection and T cell homeostasis. T
FLICE-inhibitory protein; HPK1, hematopoietic progenitor kinase 1; HPK1- cells develop from common lymphocyte progenitors in the
C, C-terminal fragment of HPK1; IL-2, interleukin-2; IKK, I␬B kinase bone marrow and migrate to the thymus. In the thymus
complex; MAPK, mitogen-activated protein kinase; NF-AT, nuclear factor developing T cells pass through a series of distinct phases
of activated T cells; NF-␬B, nuclear factor regulating ␬-chain expression in
which are characterized by differential expression of cell
B cells; PKB/PKC, protein kinase B/C; TCR, T cell receptor; tg, transgenic.
∗ Corresponding author. Tel.: +49 6221 423769; fax: +49 6221 411715. surface receptors, like TCR, CD4 and CD8. The thymus
E-mail address: r.arnold@dkfz.de (R. Arnold). fosters the development of functional competent, but not

1040-8428/$ – see front matter © 2008 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.critrevonc.2008.01.002
D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64 53

self-reactive mature T cells by positive and negative selec-


tion. Positive selection ensures that the TCR is capable
to recognize its cognate ligand, the peptide binding major
histocompatibility complex (MHC). T cells whose TCR is
unable to bind to MHC do not gain survival signals and
undergo apoptosis in a process called ‘death by neglect’. Dur-
ing negative selection T cells are eliminated which respond
to MHC bound self-antigens presented within the thymus.
Therefore, T cell apoptosis during negative selection ensures
central tolerance. Maturation of T cells within the thymus
leads to the development of CD4+ and CD8+ single posi-
tive T cells which have diverse functions in the peripheral
immune system. CD4+ T cells develop into T helper cells
(Th) which can be further subdivided into Th1 and Th2
cells. The Th1 cells initiate cellular immune responses and
secrete, e.g. IFN-␥. The Th2 cells are important for humoral
immunity and secrete, e.g. IL-4, IL-5, IL-10 and TGF-␤.
Besides Th1 and Th2 cells several other subpopulations with
distinct cytokine profiles and functions have been identi- Fig. 1. Scheme of a T cell immune response. Upon antigen challenge, T cells
fied. For example CD4+ CD25+ regulatory T cells (Treg), are activated and proliferate. In the early expansion phase T cells are resistant
which express high levels of IL-10 and TGF-␤, but little IL- towards activation-induced cell death (AICD) or activated cell autonomous
death (ACAD). After reaching the peak of the immune response T cells
2 and IL-4 and suppress T cell functions in order to avoid become sensitive towards cell death and T cell numbers decline during the
chronic activation or autoreactivity of T cells in the periph- contraction phase. While ACAD is independent of secondary stimulation,
ery. Another immunosuppressive group of CD4+ T cells are induction of AICD depends on restimulation via the T cell receptor.
Th3 cells which seem to act via TGF-␤ as the dominant
cytokine. 2. Shut-down of the immune response
In contrast to CD4+ T cells whose effector mechanisms
are mainly based on cytokine secretion, CD8+ T cells serve The immune system defends the organism against infec-
as cytotoxic T cells which directly kill target cells. CD8+ T tions. Serving as the first line of defense the innate immune
cells are critical in controlling viral infections and infections system recognizes evolutionary conserved structures on dif-
by intracellular bacteria. In addition CD8+ T cells are cru- ferent pathogens. Defenses by the adaptive immune system,
cial in host defense against some protozoa. Once a foreign in contrast, are based on clonal selection of a diverse lym-
antigen is detected by CD8+ T cells on the surface MHC I phocyte repertoire. After initiation of the immune response
of an infected cell this cell is primed for death. Cytotoxic lymphocytes proliferate and differentiate into effector cells.
T cells kill their target cells by inducing apoptosis mainly This process is called expansion phase and is needed to gen-
by the release of lytic granules like perforin and granzymes erate sufficient numbers of effector T cells specific for the
or in some cases by the ligation of the death receptor CD95 pathogen (Fig. 1). Following the immune response activated
(APO1/Fas). lymphocytes have to be eliminated. The high proliferative
Naı̈ve T cells are found mainly in the peripheral lym- capacity of antigen-activated lymphocytes requires an effec-
phoid organs until they are activated by foreign antigens tive control of their life span in order to maintain lymphocyte
presented by antigen presenting cells (APC). Upon activa- homeostasis. Programmed cell death (apoptosis) allows for
tion, T cells enter a clonal proliferation phase and invade the the decline of lymphocyte numbers during the contraction
sites of infection or inflammation. After antigen clearance phase of an immune response. Two concepts for the induction
the clonally expanded activated T cell population is removed of T cell death during the shut-down of an immune response
by apoptosis, which is essential for T cell homeostasis and have been described: activation-induced cell death (AICD)
prevents the development of autoimmunity and lymphomas and activated T cell autonomous death (ACAD). In this con-
[1]. Only a few T cells that have previously been exposed text AICD describes the induction of cell death in already
to antigen survive and develop into memory T cells which activated T cells by restimulation of their T cell receptors
respond more rapidly to the same antigen than resting naive (TCR) (Fig. 1, lower panel). During the expansion phase
T cells. of the immune response T cells possess an AICD-resistant
This review focuses on the mechanisms of apoptosis phenotype, whereas they gradually shift towards AICD sen-
induction which are critical for the removal of activated T sitivity during the contraction phase of the immune response
cells. We discuss the concepts of activation-induced cell death [2]. AICD can depend on or be independent of death recep-
(AICD) and activated T cell autonomous death (ACAD) and tor engagement and therefore involves extrinsic or intrinsic
summarize the involvement of various apoptosis pathways cell death pathways (see Sections 7–9). In contrast to AICD,
and their molecular regulators. activated T cell death by ACAD during the contraction phase
54 D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64

does not require restimulation of the TCR (Fig. 1, upper Ca2+ mobilization as well as a reduced expression of CD95L
panel). ACAD is not mediated by death receptors, depends [22]. In addition, Itk-deficient T cells lack Egr2 and Egr3
on the intrinsic cell death pathway and involves the mito- expression. Egr2 and Egr3 are two transcription factors which
chondria (see Sections 10 and 11) [3]. Most likely AICD have been implicated in TCR-induced CD95L expression
and ACAD cooperate during the shut-down of an immune [23]. It is not clear how exactly IL-2 and TCR-stimulation
response. While AICD seems to be most important for elimi- cooperate in CD95L expression as both stimuli use differ-
nation of chronically activated and potentially autoreactive T ent pathways to induce the CD95L gene [24]. Sensitization
cells [4], ACAD most prominently contributes to the down- towards AICD is accompanied by T cell proliferation [25]
regulation of T cell numbers during the contraction phase which is dependent on IL-2, and it is conceivable that other
[5]. mechanisms than expression of CD95L contribute to IL-2-
driven sensitization towards apoptosis.
The cytokine IL-15 shares several functions with IL-2. The
3. Regulation of AICD by cytokines receptors for both cytokines share the beta (IL-2R␤) chain
and the common gamma (␥c ) chain and differ only in their
Upon initial TCR stimulation T cells are activated to elicit alpha chains (IL-2R␣ vs. IL-15R␣). Decreased IL-2 levels
an immune response. TCR restimulation of already acti- in IL-15 transgenic mice were reported to interfere with sur-
vated T cells triggers AICD [6,7]. It has become evident that vival and death during AICD resulting in elevated levels of
AICD is an important mechanism to eliminate autoreactive CD8+ , CD44+ memory T cells [26]. Increased survival of
T cells and to ensure tolerance in the periphery [8]. Sensi- CD8+ memory T cells was also found in wild type mice upon
tivity towards AICD is strictly controlled according to the repeated IL-15 injection [27,28]. Therefore, both cytokines
activation status of T cells [9–12]. stimulate T cell proliferation and act as a survival factor, but
One essential factor for the proliferation of T cells and sen- IL-15 can inhibit the IL-2 dependent sensitization towards
sitization towards AICD is the cytokine interleukin-2 (IL-2) AICD.
[13]. Activation of naive AICD-resistant T cells triggers the Two other cytokines which positively influence AICD by
expression of IL-2 and its high affinity receptor subunit IL- different mechanisms are IL-4 and interferon-␥ (IFN-␥). In T
2R␣ (CD25) [14,15]. IL-2 is essential for T cell proliferation, cells derived from IL-4 deficient mice, AICD is significantly
serves as survival factor and ensures T cell effector functions reduced [29]. Interestingly, IL-4 sensitizes towards AICD via
to allow the development of a protective immune response an IL-2-dependent mechanism which involves transcriptional
[16]. T cell survival mediated by IL-2 involves various sig- upregulation of the IL-2 receptor. As a consequence, AICD
naling pathways including proteins like signal transducer and resistance of IL-4 deficient T cells could be overcome by
activator of transcription (STAT), mitogen-activated protein exogenous addition of IL-2. On the molecular level IL-4 and
kinase (MAPK) and protein kinase B (PKB/AKT). Signal- IL-2 seem to be necessary for degradation of the apoptosis
ing via the IL-2R␤ chain and the SH2 domain-containing inhibitor cellular FLICE-inhibitory protein (c-FLIP) which
transforming protein (SHC) induces PKB/AKT-mediated correlates with AICD sensitivity [29]. T cells from IFN-
expression of the anti-apoptotic factor Bcl-2, which protects ␥-deficient mice are also resistant to AICD [30]. In CD4+
the cell against the induction of apoptosis [16]. PKB/AKT T cells, IFN-␥ controls caspase-8 expression in a STAT-
and MAPK have been implicated in phosphorylation of the dependent manner. Consequently, rescue of IFN-␥-deficient
Bcl-2 family members, either supporting their anti-apoptotic T cells with exogenous IFN-␥ restores caspase-8 levels and
function or inhibiting their pro-apoptotic function [17]. susceptibility to AICD.
Besides its function as a survival factor, IL-2 is also
required for the sensitization towards AICD upon expansion
of T cells [18]. This sensitization is not found in T cells of 4. NF-␬B is essential for T cell survival
IL-2 deficient or IL-2R␣ deficient mice [19]. The molecu-
lar mechanisms by which IL-2 sensitizes T cells to AICD It is presently still unsolved how the TCR signal is inte-
are not completely understood. Increased expression of the grated to provide activation, proliferation and survival upon
death receptor ligand CD95L (Fas-L/Apo-1L) (see Section initial stimulation and cell death by AICD upon restimula-
6) and decreased expression of the anti-apoptotic molecule tion. TCR stimulation can trigger gene expression of proteins
c-FLIP (see Section 7) are suggested to be IL-2 dependent with anti-apoptotic functions, like c-FLIP, Bcl-2, Bcl-XL ,
[19]. The IL-2 inducible T cell kinase (Itk) which belongs Bcl-2A1, IAP/XIAP [31] or GADD45␤ [31–33] and proteins
to the Tec family of tyrosine kinases may play an important with pro-apoptotic functions like CD95L [34] and Nur77
role in this process [20]. Members of this kinase family have [35] (Fig. 2). The balance between expression of pro- and
an impact on TCR-mediated calcium (Ca2+ ) mobilization and anti-apoptotic proteins determines T cell fate and is largely
are known to activate phospholipase C␥1 (PLC␥1) [21]. Ca2+ dependent on the type of signal received by the cell, e.g.
signaling, however, is a prerequisite for expression of CD95L, strength of TCR stimulation and costimulation and the pres-
another molecule involved in AICD [8]. In accordance with ence of intracellular signaling modulators downstream of the
these studies Itk-deficient mice show reduced TCR-induces TCR, e.g. adapter molecules and kinases [36].
D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64 55

Contrary to activation of NF-␬B, inhibition of NF-␬B


is considered to fulfill an important pro-apoptotic function
[51,52]. It has been shown that the onset of AICD correlates
with reduced NF-␬B activity in the nucleus [33]. One molecu-
lar regulator of NF-␬B involved in AICD is the hematopoietic
progenitor kinase 1 (HPK1) which is discussed in the next
section.

5. HPK1 controls the onset of AICD

Recently it was shown that the TCR-proximal kinase


hematopoietic progenitor kinase 1 (HPK1) is involved in the
regulation of cell death in primary T cells [37,53]. HPK1
is a serine–threonine kinase which is exclusively expressed
in hematopoietic tissues and activates the c-Jun N-terminal
kinase (JNK) pathway [54] and the NF-␬B pathway [55].
Fig. 2. Pro- and anti-apoptotic genes are induced by TCR stimulation. Stim- HPK1 is activated upon stimulation of immunoreceptors
ulation of the T cell receptor (TCR) by its cognate ligand induces the
in T and B cells [56,57] and is recruited to the immune
expression of pro-apoptotic as well as anti-apoptotic genes. A shift of the
equilibrium induces activation and survival on the one side or death by synapse [58,59]. During the expansion phase of an immune
AICD on the other side. TCR stimulation can induce gene expression of response, AICD-resistant T cells contain full-length HPK1,
the anti-apoptotic molecules c-FLIP, Bcl-2, Bcl-XL , Bcl-2A1, IAP/XIAP, which is an essential mediator of TCR-induced NF-␬B sig-
GADD45␤ and the pro-apoptotic molecules CD95L, and Nur77. naling (Fig. 3). Transient knockdown of HPK1 in T cell lines
blocks activation of IKK and NF-␬B [37]. In highly prolif-
Naı̈ve and activated T cells interpret the same signal erating and viable T cells during expansion HPK1 is cleaved
in two different ways. Recent studies emphasize that dif- into its N- and C-terminal fragments [37]. Despite of HPK1
ferential regulation of NF-␬B influences the transition of being a caspase target [55,60] this cleavage is not associ-
AICD-resistant to AICD-sensitive T cells [33,37,38]. Regu- ated with apoptosis induction [61], but it is crucially needed
lation of lymphocyte fate and execution of immune functions for the induction of AICD sensitivity during T cell expan-
are connected with transcription factors of the NF-␬B fam- sion [62]. The presence of the C-terminal cleavage product
ily [31,39,40]. NF-␬B determines life and death decisions of HPK1 (HPK1-C) sensitizes T cells towards AICD [37,53]
in developing lymphocytes and provides important signals
in T cells to ensure cell survival [41,42]. NF-␬B activa-
tion in response to antigen receptor ligation is mediated
by a high molecular weight I␬B kinase (IKK) complex
[43]. The IKK complex comprises two enzymatic subunits,
IKK␣ and IKK␤, and the regulatory subunit IKK␥ (NEMO)
[44]. Activation of the IKK complex upon stimulation of
the T cell receptor (TCR) depends on the caspase recruit-
ment domain (CARD)-containing signaling adapter protein
Carma1 (CARD11) [45]. TCR stimulation leads activation
of protein kinase C (PKC)␪ which in turn phosphorylates
Carma1 within its linker region between the coiled-coil and
the PDZ domains [46–48]. Subsequently, Carma1 interacts
with the CARD domain of Bcl10, which recruits the adaptor
protein Malt1 [49]. The Malt1-interacting E3 ubiquitin ligase
TRAF6 triggers Lys63-linked polyubiquitinylation of IKK␥
and induces activation of the IKK complex in T cells [50]. Fig. 3. HPK1-C sensitizes T cells to AICD by suppression of NF-␬B activity.
Role of hematopoietic progenitor kinase 1 (HPK1) in T cell receptor (TCR)-
NF-␬B/Rel proteins are dimeric, sequence-specific tran-
mediated AICD. HPK1 is cleaved into HPK1-N and HPK1-C in T cells.
scription factors which are rendered inactive within the HPK1-C renders T cells AICD-sensitive. HPK1-mediated I␬B Kinase (IKK)
cytoplasm by interaction with inhibitory I␬B proteins [43]. activation in AICD-resistant T cells leads to NF-␬B activation by liberation
Activation of the IKK complex results in phosphorylation and of the NF-␬B heterodimer (p65/p50) from its inhibitor I␬B. The presence of
subsequent ubiquitination and degradation of the inhibitor HPK1-C blocks IKK activation downstream of the TCR, thereby blunting
nuclear translocation of NF-␬B and suppressing NF-␬B target gene expres-
of NF-␬B (I␬B). Liberated NF-␬B dimers translocate to the
sion. Therefore, full length HPK1 confers NF-␬B activation and resistance
nucleus where they bind and transactivate promoter regions towards AICD while HPK1-C is blocking NF-␬B and sensitizes towards
of NF-␬B-regulated genes [44]. AICD.
56 D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64

HPK1-C inhibits NF-␬B activation [55] and sequesters the Recently it has been reported that HIV-1-expressed
inactive IKK complex by binding to IKK␣ and IKK␤ (Fig. 3) trans-activator of transcription (HIV-1 Tat) can enhance or
[37]. substitute for ROS signaling in T cells [34]. This might
Presence of HPK1 as a full-length or a truncated molecule explain strongly enhanced AICD of T cells from AIDS
leads to different results upon TCR stimulation. Full-length patients [71,72]. During progression of AIDS, HIV-1 Tat
HPK1 supports activation of NF-␬B, resistance towards and CD4+ -dependent Ca2+ mobilization induce expression
AICD and T cell proliferation during the expansion phase. of CD95L and lead to T cell depletion by AICD.
Presence of HPK1-C results in inhibition of NF-␬B acti-
vation, sensitization towards AICD and shut-down of an
immune response. As TCR stimulation results in expres- 7. AICD can involve death receptors
sion of proteins with pro- and anti-apoptotic functions,
HPK1-C-mediated blocking of the anti-apoptotic NF-␬B It is commonly believed that AICD involves the engage-
pathway shifts the equilibrium towards apoptosis and sen- ment of death receptors, like CD95 (Apo-1/Fas) and the
sitizes towards AICD (Fig. 3). TNF receptor. Apoptosis mediated by CD95 is essential
It is tempting to speculate that conversion of HPK1 to for T cell homeostasis since defects within this pathway
HPK1-C shifts the outcome of the very same TCR signal cause abnormal T cell accumulation, lymphadenopathy and
simply by modulation of gene expression. A similar conclu- autoimmunity [73]. Three naturally occurring mutations have
sion was drawn by a recent report correlating loss of nuclear been widely used to study the impact of the CD95/CD95L-
location of the NF-␬B proteins p65/RelA with induction of system in T cell AICD. In lpr mice a retrotransposon is
AICD [33]. Therefore, the HPK1/HPK1-C ratio might serve inserted into intron 2 of the CD95 gene leading to a dramatic
as a molecular timer for the T cell to sense age and limit its decrease in CD95 surface expression [74]. An additional
life span. mutation which results in a phenotype congenic to lpr is
lprcg . The lprcg phenotype is caused by a single point
mutation within the death domain of CD95 which abro-
6. Suppression of NF-␬B induces AICD gates apoptotic signaling [75]. The gld mutation results in
the generation of a defective CD95L [76]. T cells from
Several other groups have linked NF-␬B-suppression mice with these mutations show reduced but clearly evi-
to susceptibility towards cell death [31,63,64]. The NF- dent AICD in the presence of IL-2 [77]. Until now it is
␬B-dependent inhibition of the expression of the tumor unclear which CD95/CD95L-independent mechanism medi-
suppressor p73 is required for the survival of antigen- ates AICD under these conditions.
stimulated T cells [38], while expression of p73 has been Although it has been suggested that tumor necrosis factor
shown to be essential for AICD [65]. Furthermore, transgenic (TNF) might be involved, its role in AICD is controversial.
expression of the IKK-inhibitory protein I␬B␣ increases While TNF and TNF receptor deficient mice show normal
TCR-induced AICD [38,66]. AICD, blocking of TNF reduces TCR-induced cell death in
Another mechanism depending on NF-␬B-inhibition and vitro and in vivo [78,79]. TCR stimulation can induce expres-
considered to exhibit important pro-apoptotic function is the sion of TNF and its role might be comparable to the role
generation of reactive oxygen species (ROS). TCR stimula- of the CD95/CD95L system in the early phase of AICD.
tion leads to fast generation of ROS [34]. A recent study has TNF, however, might be important in a late phase of AICD
shown that the core components of TCR signaling includ- [80].
ing ZAP70, LAT, SLP76 and PLC␥1 are crucial for ROS Another death receptor which has been described to medi-
induction [67]. ROS production is mediated by the mito- ate AICD of CD8+ T cells depending on CD4+ T cell help
chondrial respiratory chain complex I involving NADPH is TNF-related apoptosis-inducing ligand (TRAIL) recep-
oxidase II [67,68]. Interestingly, PKC␪ which is activated tor [81]. CD8+ T cells which have been primed in the
by TCR stimulation translocates to the mitochondria and presence of CD4+ T cells acquire the ability to enter a sec-
is required for activation-induced generation of ROS [67]. ond, restimulation-dependent round of clonal proliferation.
Increasing ROS levels suppress Bcl-2 expression and thereby In contrast to that, CD8+ T cells which have been initially
initiate the mitochondrial death pathway [69]. Furthermore primed without CD4+ T cell help (helpless T cells) do not
ROS has been shown to be essential for CD95L-induction respond with a second round of clonal proliferation, but
in response to stimulation of the TCR [34]. Recent studies undergo AICD mediated by TRAIL. Therefore it is thought
have shown that ROS production alone is not sufficient to that regulation of TRAIL expression by CD4+ T cells is
efficiently induce expression of CD95L, but requires Ca2+ an important mechanism for CD8+ memory generation and
mobilization in addition [34]. Stimulation of the TCR results adaptive immunity.
in increased intracellular Ca2+ levels depending on PLC-␥1 It has also been shown that redistribution of death
and possibly also on ROS itself [70]. Therefore, the com- receptors into lipid rafts enhances the susceptibility to
bined action of Ca2+ and ROS leads to efficient induction of apoptosis. TCR stimulation of CD4+ T cells results in
CD95L expression and sensitizes towards AICD. a redistribution of CD95 into lipid rafts [82]. Interest-
D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64 57

ingly, unlike antigen receptors which accumulate in lipid procaspase-8a (FLICE, MACH-␣1, Mch5␤), procaspase-8b
rafts upon triggering, reorganisation of CD95 is indepen- (MACH-␣2), procaspase-10 and procaspase-2 [85–87]. The
dent of CD95L-ligation. The TCR-induced translocation of precise molecular events of CD95-induced caspase activa-
CD95 into lipid rafts increases sensitivity to CD95-induced tion according to the ‘induced proximity model’ are reviewed
apoptosis and therefore may account for the clonotypic elsewhere [87,85].
specificity of restimulated T cell death during immune The cellular FLICE-inhibitory protein (c-FLIP) is a potent
responses. inhibitor of caspase activation at the DISC [88]. c-FLIP
The complexity is further increased by the fact that resembles a catalytic inactive procaspase with a tandem DED.
blocking of death receptors alone does not completely res- The ratio between c-FLIP and the procaspases at the DISC
cue from TCR-induced T cell death. Caspase-independent determine whether apoptosis is initiated or not [85]. Several
mechanisms have been described as well as a granzyme different c-FLIP-isoforms are described on the protein level
B-dependent pathway [83]. It is important that T cells use including a long and a short isoform, c-FLIPL and c-FLIPS
various mechanisms to execute AICD. The different path- [89,90], and an isoform which was recently found in Raji
ways allow a fine-tuned regulation of AICD and might enable cells, c-FLIPR [91,92].
different T cell subtypes to respond differently to TCR res- c-FLIPL and c-FLIPS have been implicated in regulation
timulation. of AICD [19,93]. Upon initial T cell activation c-FLIPS is
strongly upregulated [94,95]. This increase in expression of
c-FLIP correlates with the resistance of freshly activated T
8. AICD and CD95 cells towards CD95-induced apoptosis. In expanded AICD-
and CD95-sensitive T cells c-FLIPS levels decline, suggest-
Stimulation of CD95 by its cognate ligand CD95L induces ing that c-FLIPS might be the switch that renders T cells
the formation of the death-inducing signaling complex susceptible to CD95-dependent AICD [96]. Downregulation
(DISC) [84] (Fig. 4). The CD95 DISC contains the oligomer- of c-FLIP seems to depend on T cell proliferation and it is
ized, most likely trimerized CD95 and a death domain (DD) declining in an IL-2 dependent manner most likely due to
containing adaptor molecule Fas-associated death domain the G1/S transition of c-FLIP protein expression [13,95,97].
(FADD). FADD is recruited to CD95 by homotypic DD The important role of c-FLIPS in AICD is underlined by
interactions and mediates interaction and dimerization of the observation that costimulation via CD28 rescues T cells

Fig. 4. Extrinsic CD95L-dependent AICD-pathways. Freshly activated T cells (left) form low amounts of death-inducing signaling complex (DISC). Under
this condition CD95 engagement by CD95L activates low amounts of caspase-8. Caspase-8 cleaves Bid to tBid which migrates to the mitochondria. In freshly
activated T cells the pro-apoptotic ability of tBid is blocked by elevated levels of anti-apoptotic Bcl-XL . The presence of c-FLIP blocks activation of caspase-8
at the DISC. As a consequence these T cells are resistant to CD95-dependent AICD. Long-term activated T cells form large amounts of DISC. This results
in robust activation of caspase-8 which subsequently leads to activation of caspase-3 and the execution of apoptosis. Additionally, long-term activated T cells
exhibit low Bcl-XL levels which allow activation of the mitochondrial death pathway by tBid.
58 D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64

from TCR-induced apoptosis [42,94]. This costimulation is antigen-activated T cells, but is also required for expression
followed by upregulation of c-FLIPS which abrogates CD95- of CD95L during AICD.
induced procaspase-8 activation at the DISC.
Another mechanism which influences CD95-induced
apoptosis sensitivity and which can be linked to AICD is 9. AICD independent of death receptors
the capacity of differential DISC formation [95,98]. Freshly
activated T cells show low activation of caspase-8 and depend The initiation of an adaptive immune response is a strictly
on amplification of the apoptotic signal by the mitochondria. regulated process and it is largely dependent on the nature of
In these cells low amounts of caspase-8 are activated which the antigen. Besides Th1, Th2 and Th3 cells, recent stud-
cleave the cytosolic Bcl-2 family member Bid (Fig. 4). Trun- ies have identified a new T helper population which was
cated Bid (tBid) migrates to the mitochondria, where it fulfils named Th17. Production of IL-17 and also of IL-22, was
its pro-apoptotic role and induces the release of cytochrome found to be characteristic for this subpopulation [108]. How-
c (cyt c) into the cytosol. This is followed by formation of the ever in this review we will focus on Th1 and Th2 cells. The
apoptosome, caspase-9 activation and subsequent activation decision between Th1 and Th2 depends on cytokines and
of caspase-3 and execution of cell death. affinity of the antigen to the TCR. Th1 vs. Th2 cells direct
The resistance of freshly activated T cells against CD95- the outcome of the adaptive immune system towards cell-
induced apoptosis is caused by c-FLIP and Bcl-XL which based vs. antibody-based immunity, respectively. While Th1
are upregulated upon initial T cell activation. While c-FLIP cells enable macrophages to efficiently destroy intracellu-
lowers the amount of caspase-8-activation Bcl-XL blocks the lar microorganisms, Th2 cells activate the antibody-based
mitochondria-dependent cell death pathway (Fig. 4) [95,96]. defense by driving B cell differentiation and eosinophil/mast
During the expansion phase of the immune response T cells cell immune responses. In a physiological situation one of
shift towards AICD-sensitivity. In these T cells triggering the two subsets prevails. In a pathologic situation prevalence
of CD95 activates caspase-8 in sufficient amounts to exe- of one of the subsets might induce autoimmunity, allergic
cute apoptosis independently of the mitochondrial pathway. reactions or chronic infections.
In addition, T cells switch from a Bcl-XL high to a Bcl-XL AICD of Th1 and AICD of Th2 cells differs in several
low state allowing activation of the mitochondrial cell death points. It has been reported that Th1 cells are more suscepti-
pathway [95]. ble to AICD compared to Th2 cells [109]. This is consistent
T cell activation itself influences the susceptibility to with various reports showing an increased CD95L sensitiv-
CD95-induced apoptosis by two mechanisms: First, initial ity in Th1 cells. Consequently Th1 or Th2 cells can kill Th1
T cell activation leads to upregulation of CD95 and second, but not Th2 target cells [110–112]. A study showed that in
TCR-stimulation leads to CD95L-independent relocalization contrast to Th1, Th2 cells undergo AICD independently of
of CD95 into lipids rafts and thereby increases the sensitivity CD95/CD95L engagement, but dependent on granzyme B
to clonotypic deletion [82]. In summary, CD95-dependent [83]. The serine protease granzyme B, which is present in
cell death is an important mechanism in AICD. The regu- intracellular lytic granules, was first defined to play a role
lation of this pathway is complex and involves alterations in the effector function of cytotoxic T lymphocytes [113].
in signal transduction, caspase-8 activation at the DISC and Upregulation of granzyme B expression in response to TCR
localization of CD95. stimulation was shown in Th2 cells and could be abrogated
Several studies highlight the role of PKC␪ in CD95- by the Th2 survival factor vasocative interstine peptide [114].
dependent AICD. PKC␪ can synergize with calcineurin to In the presence of granzyme B AICD of Th2 cells could not
provide robust CD95L expression upon TCR stimulation be prevented by blocking of CD95L and AICD of Th2 cells
[99]. This was further supported by a recent study show- from granzyme B-deficient mice was completely abrogated
ing that T cells from PKC␪ knock out mice fail to induce [82,83]. This indicates that AICD of Th2 cells is completely
CD95L [100]. TCR-mediated induction of CD95L is depen- dependent on granzyme B. Furthermore, accumulation of
dent on the transcription factors NF-AT, AP-1 and NF-␬B Th2 cytokines in granzyme B deficient mice increased the
[23,101–104]. In this scenario PKC␪ is crucial for full susceptibility to allergen-induced asthma [83]. This under-
NF-AT activation as mutations of the NF-AT binding sites lines the physiological significance of TCR-induced AICD
within the CD95L promoter abrogate the ability of PKC␪ to in controlling a balanced immune response.
induce CD95L. In addition, PKC␪ seems to influence CD95- Recently, we have shown that T and B lymphocytes from
dependent cell death directly. CD4+ T cells from PKC␪ HPK1-C transgenic mice undergo AICD independently of
deficient mice are less sensitive towards CD95-induced apop- the CD95/CD95L system, but involve caspase-9 [62]. Knock
tosis, which is supported by a weaker activation of caspase-8 down of HPK1/HPK1-C or Bim by siRNA showed that
and Bid compared to wild type T cells [105]. Deletion CD95L-dependent and HPK1/HPK1-C-dependent cell death
of PKC␪ differentially affects CD8+ and CD4+ T cells by pathways complement each other in AICD of primary T
impairing BclXL expression upon activation [106]. PKC␪ cells. These results define HPK1-C as a mediator of CD95L-
might also mediate cell survival by phosphorylation of Bad independent AICD, which contributes to the removal of
[107]. In summary, PKC␪ confers activation and survival of activated lymphocytes.
D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64 59

10. Activated T cell death without TCR restimulation only proteins are more regulators than executioners of death
and are localized in various organelles or in the cytoplasm
Recently, a novel concept of activated T cell death has [17]. Under survival conditions they are held in an inactive
been established which has been named activated T cell state. The pro-apoptotic BH3-only protein Bid for example
autonomous death (ACAD) [39]. In contrast to AICD, ACAD requires cleavage to tBid in order to migrate to the mito-
does not require TCR restimulation and is independent of chondria and induce apoptosis. Therefore Bid might serve as
death receptor engagement. Several in vivo studies show an amplifier of a low caspase or proteolytic activity within
that the T cell contraction phase at the end of the immune the cell. Another example of a BH3-only protein controling
response, when ACAD takes place, may be largely indepen- the health status of the cell is Bad. Bad is phosphorylated
dent of CD95 signaling [5,115]. by the kinase PKB/AKT [116,117]. PKB/AKT exhibits anti-
ACAD is also referred to as death by neglect, death by apoptotic functions and phosphorylation of Bad keeps the
cytokine withdrawal or passive cell death. These terms reflect molecule in an inactive conformation. Diminished PKB/AKT
the common finding that deprivation of survival factors ini- activity is followed by a dephosphorylation and migration of
tiates T cell death. Nevertheless, ACAD is an active and Bad to the mitochondria where it cooperates with Bax and
cell autonomous program [5]. Several studies suggest that Bak to induce apoptosis [17,116,117]. The BH3-only protein
ACAD is regulated by the intrinsic cell death pathway and Bim also has to be modified, most likely phosphorylated, to
involves members of the Bcl-2 family (Fig. 5). In general, execute its apoptotic function [17]. In addition to posttransla-
the Bcl-2 family members are known to have pro- and anti- tional modification, regulation of Bim transcription by TCR
apoptotic functions within a cell [17]. On the structural level stimulation has been described [118].
the Bcl-2 family is divided into three subgroups: The first In general BH3-only proteins serve as stress sensors and
group exhibits anti-apoptotic activities and shares the BH1-4 control the viability of a cell. In the absence or the pres-
homology domains. Bcl-2 and Bcl-XL are members of this ence of certain stimuli they get activated and fulfill their
group. The second group with its members Bax and Bak is pro-apoptotic function. From studies with Bax/Bak double
clearly pro-apoptotic and characterized by three BH domains knock out mice we know that BH3-only proteins need Bax
(BH1-3). The BH3-only proteins are the third group of the and Bak to execute apoptosis thereby following a defined
Bcl-2 family proteins and share the BH3-domain, a nine hierarchy [119]. There is an ongoing debate about how BH3-
amino acid sequence. In contrast to the other groups, BH3- only proteins induce apoptosis (details reviewed elsewhere)

Fig. 5. ACAD is mediated by Bim and declining Bcl-2 levels. Inactive Bim is sequestered by the dynein motor complex and is bound to microtubules. Bcl-2
levels are sufficient to inactivate the pro-apoptotic function of Bax and Bak (left). Induction of cell death, e.g. by growth factor deprivation, leads to the release
of Bim from microtubules and allows its migration to the mitochondrion. At the peak of an immune response Bcl-2 levels decline and activated Bim can now
cooperate with Bax and Bak to override the anti-apoptotic function of Bcl-2 and to induce the release of cytochrome c (cyt c) from the mitochondria. cyt c,
APAF1 and procaspase-9 form the apoptosome and activate caspase-9. As a consequence, caspase-9 activates caspase-3/7 which then induces cell death.
60 D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64

[17]. The ratio between pro- and anti-apoptotic Bcl-2 family when foreign antigen is present only at very low concen-
members determines whether the cell dies or survives. trations. AICD, in contrast, is the prevailing mechanism if
It is important to note that Bcl-2 proteins influence the antigens are present at higher concentrations for example
strength and the prolongation of the immune response. during chronic infections or in autoimmune diseases. Thus,
Expression of a Bcl-2 transgene increases the T cell response AICD might serve to eliminate autoreactive T cells and main-
in vivo [120]. T cells from mice challenged with different tain peripheral tolerance towards self-antigens in the steady
antigens show a decrease in Bcl-2 and Bcl-XL -levels at the state. Such self-antigens are systemic and mostly present
peak of the immune response compared to non-activated T at higher concentrations. AICD controls T cell homeosta-
cells [5,120,121]. Interestingly, memory cells which are not sis by balancing autoimmunity and the ability to mount a
deleted in the T cell contraction phase show elevated Bcl-2 proper immune response. The different thresholds of reactiv-
levels that might protect them from ACAD [122]. In a T cell ity towards antigen concentrations are probably the central
activation model in which the superantigen staphylococcus discrimination between ACAD and AICD. In summary, two
enterotoxin B (SEB) is used to activate T cells, transgenic different regulatory pathways may control T cell homeostasis
expression of Bcl-2 inhibits ACAD. This is clearly not the under different physiological conditions: ACAD in response
case in lpr-mice which are bearing a CD95-mutation [5]. to foreign antigens and AICD in response to self-antigens.
This further supports the finding that Bcl-2 family members
and the CD95-system regulate different apoptosis pathways
within T cells. Several groups have reported a CD95/CD95L- Reviewers
independent apoptosis pathway after application of SEB in
vivo [5,13,123,124]. Some studies, however, show exactly the Christian Widmann, Ph.D., Associate Professor, Lausanne
opposite effect [125,126]. These controversial findings might University, Department of Physiology, Bugnon 7/9, CH-1005
arise from different doses of SEB or different experimen- Lausanne, Switzerland.
tal models used in the studies. Nevertheless, these findings Ingo Schmitz, Ph.D., University of Duesseldorf, Depart-
led to the hypothesis that T cell deletion in the presence of ment of Molecular Medicine, Universitätsstrasse 1, D-40225
low amounts of antigen might be preferentially mediated via Duesseldorf, Germany.
ACAD whereas high antigen load results in AICD depending Klaus-Michael Debatin, M.D., University Children’s Hos-
on CD95/CD95L engagement. pital, Erythstrasse 24, D-89070 Ulm, Germany.
The complexity is further increased by the finding that
T cells which are deficient for the BH3-only protein Bim
persist after SEB-injection in vivo [5]. Interestingly, these Acknowledgements
Bim-deficient T cells show the same downregulation of Bcl-2
as their wild-type counterparts, which indicates that down- We thank Inna Lavrik, Lucie Dörner and Karsten Gülow
regulation of Bcl-2 alone – by a ROS-dependent mechanism for critically reading the manuscript and are grateful to Heidi
[69] – is insufficient to induce ACAD. This suggests a two- Sauter for expert secretary assistance. This work was sup-
step mechanism for the induction of ACAD involving Bcl-2 ported by grants from the Deutsche Forschungsgemeinschaft,
and Bim (Fig. 5). As Bim levels change only marginally upon the Wilhelm Sander-Stiftung, the Deutsche Krebshilfe and
T cell activation it is tempting to speculate that Bim is some- the European Community.
how activated, released from the dynein motor complex at
the microtubules and fulfills its pro-apoptotic functions when
Bcl-2 levels are decreasing. The molecular details of Bim References
activation are currently not fully understood. In summary
ACAD is limiting T cell responses in vivo and is indepen- [1] Krammer PH, Arnold R, Lavrik IN. Life and death in peripheral T
dent of death receptor signaling. ACAD is executed via the cells. Nat Rev Immunol 2007;7:532–42.
[2] Krammer PH. CD95’s deadly mission in the immune system. Nature
intrinsic, mitochondrial pathway where decreasing Bcl-2 lev- 2000;407:789–95.
els cannot compensate the pro-apoptotic activity of activated [3] Strasser A, Pellegrini M. T-lymphocyte death during shutdown of an
Bim. immune response. Trends Immunol 2004;25:610–5.
[4] Stranges PB, Watson J, Cooper CJ, et al. Elimination of antigen-
presenting cells and autoreactive T cells by Fas contributes to
prevention of autoimmunity. Immunity 2007;26:629–41.
11. Conclusions [5] Hildeman DA, Zhu YN, Mitchell TC, et al. Activated T cell death in
vivo mediated by proapoptotic Bcl-2 family member Bim. Immunity
Two concepts of activated T cell death exist: AICD which 2002;16:759–67.
is dependent on TCR-restimulation and ACAD which is an [6] Brunner T, Mogil RJ, LaFace D, et al. Cell-autonomous Fas
autonomous, intrinsic death program. Both death programs (CD95)/Fas-ligand interaction mediates activation-induced apoptosis
in T-cell hybridomas. Nature 1995;373:441–4.
control T cell homeostasis and it largely depends on the [7] Dhein J, Walczak H, Baumler C, Debatin KM, Krammer PH.
antigen which program is activated. ACAD might be the Autocrine T-cell suicide mediated by APO-1/(Fas/CD95). Nature
dominating mechanism at the end of the immune response 1995;373:438–41.
D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64 61

[8] Krueger A, Fas SC, Baumann S, Krammer PH. The role of CD95 [29] Zhang J, Bardos T, Shao Q, et al. IL-4 potentiates activated
in the regulation of peripheral T-cell apoptosis. Immunol Rev T cell apoptosis via an IL-2-dependent mechanism. J Immunol
2003;193:58–69. 2003;170:3495–503.
[9] Klas C, Debatin KM, Jonker RR, Krammer PH. Activation inter- [30] Refaeli Y, Van Parijs L, Alexander SI, Abbas AK. Interferon [Gamma]
feres with the APO-1 pathway in mature human T cells. Int Immunol is required for activation-induced death of T lymphocytes. J Exp Med
1993;5:625–30. 2002;196:999–1005.
[10] Owen-Schaub LB, Yonehara S, Crump III WL, Grimm EA. DNA [31] Karin M, Lin A. NF-␬ B at the crossroads of life and death. Nat
fragmentation and cell death is selectively triggered in activated Immunol 2002;3:221–7.
human lymphocytes by Fas antigen engagement. Cell Immunol [32] De SE, Zazzeroni F, Papa S, et al. Induction of gadd45beta by
1992;140:197–205. NF-kappaB downregulates pro-apoptotic JNK signalling. Nature
[11] Peter ME, Kischkel FC, Scheuerpflug CG, Medema JP, Debatin 2001;414:308–13.
KM, Krammer PH. Resistance of cultured peripheral T cells towards [33] Mittal A, Papa S, Franzoso G, Sen R. NF-␬ B-dependent regulation
activation-induced cell death involves a lack of recruitment of FLICE of the timing of activation-induced cell death of T lymphocytes. J
(MACH/caspase 8) to the CD95 death-inducing signaling complex. Immunol 2006;176:2183–9.
Eur J Immunol 1997;27:1207–12. [34] Gulow K, Kaminski M, Darvas K, Suss D, Li-Weber M, Krammer PH.
[12] Radvanyi LG, Shi YF, Vaziri H, et al. CD28 costimulation inhibits HIV-1 trans-activator of transcription substitutes for oxidative signal-
TCR-induced apoptosis during a primary T cell response. J Immunol ing in activation-induced T cell death. J Immunol 2005;174:5249–
1996;156:1788–98. 60.
[13] Van Parijs L, Refaeli Y, Lord JD, Nelson BH, Abbas AK, Balti- [35] Cui H, Matsui K, Omura S, et al. Proteasome regulation of activation-
more D. Uncoupling IL-2 signals that regulate T cell proliferation, induced T cell death. PNAS 1997;94:7515–20.
survival, and Fas-mediated activation-induced cell death. Immunity [36] Arnold R, Brenner D, Becker M, Frey C, Krammer PH. How
1999;11:281–8. T lymphocytes switch between life and death. Eur J Immunol
[14] Baumann S, Dostert A, Novac N, et al. Glucocorticoids inhibit 2006;36:1654–8.
activation-induced cell death (AICD) via direct DNA-dependent [37] Brenner D, Golks A, Kiefer F, Krammer PH, Arnold R. Activation or
repression of the CD95 ligand gene by a glucocorticoid receptor suppression of NFkB by HPK1 determines sensitivity to activation-
dimmer. Blood 2005;106:617–25. induced cell death. EMBO J 2005;24:4279–90.
[15] Schuh K, Twardzik T, Kneitz B, Heyer J, Schimpl A, Serfling E. [38] Wan YSY, DeGregori J. The survival of antigen-stimulated T cells
The interleukin 2 receptor alpha chain/CD25 promoter is a target requires NF␬ B-mediated inhibition of p73 expression. Immunity
for nuclear factor of activated T cells. J Exp Med 1998;188:1369– 2003;18:331–42.
73. [39] Hildeman DA, Zhu YN, Mitchell TC, Kappler J, Marrack P. Molecular
[16] Benczik M, Gaffen SL. The interleukin (IL)-2 family cytokines: sur- mechanisms of activated T cell death in vivo. Curr Opin Immunol
vival and proliferation signaling pathways in T lymphocytes. Immunol 2002;14:354–9.
Invest 2004;33:109–42. [40] Ruland J, Mak TW. From antigen to activation: specific signal trans-
[17] Strasser A. The role of BH3-only proteins in the immune system. Nat duction pathways linking antigen receptors to NF-␬ B. Sem Immunol
Rev Immunol 2005;5:189–200. 2003;15:177–83.
[18] Lenardo MJ. lnterleukin-2 programs mouse ␣␤ T lymphocytes for [41] Kane LP, Lin J, Weiss A. It’s all relative: NF-␬ B and CD28
apoptosis. Nature 1991;353:858–61. costimulation of T-cell activation. Trends Immunol 2002;23:413–
[19] Refaeli Y, Van Parijs L, London CA, Tschopp J, Abbas AK. Biochem- 20.
ical mechanisms of IL-2-regulated Fas-mediated T cell apoptosis. [42] Schmitz ML, Dienz O, Bacher S. NF-␬ B activation pathways induced
Immunity 1998;8:615–23. by T cell costimulation. FASEB J 2003;17:2187–93.
[20] Liao XC, Littman DR. Altered T cell receptor signaling and dis- [43] Ghosh S, Karin M. Missing pieces in the NF-␬ B puzzle. Cell
rupted T cell development in mice lacking Itk. Immunity 1995;3:757– 2002;109:S81–96.
69. [44] Hayden MS, Ghosh S. Signaling to NF-␬ B. Genes Dev 2004;
[21] Sommers CL, Rabin RL, Grinberg A, Tsay HC, Farber J, Love PE. 18:2195–224.
A role for the Tec family tyrosine kinase Txk in T cell activation and [45] Thome M. CARMA1, BCL-10 and MALT1 in lymphocyte develop-
thymocyte selection. J Exp Med 1999;190:1427–38. ment and activation. Nat Rev Immunol 2004;4:348–59.
[22] Miller AT, Berg LJ. Defective fas ligand expression and activation- [46] Matsumoto R, Wang D, Blonska M, et al. Phosphorylation of
induced cell death in the absence of IL-2-Inducible T cell kinase 5. J CARMA1 plays a critical role in T cell receptor-mediated NF-␬ B
Immunol 2002;168:2163–72. activation. Immunity 2005;23:575–85.
[23] Li-Weber M, Laur O, Krammer PH. Novel Egr/NF-AT composite [47] Rueda D, Thome M. Phosphorylation of CARMA1: the link(er) to
sites mediate activation of the CD95 (APO-1/Fas) ligand promoter in NF-␬ B activation. Immunity 2005;23:551–3.
response to T cell stimulation. Eur J Immunol 1999;29:3017–27. [48] Sommer K, Guo B, Pomerantz JL, et al. Phosphorylation of
[24] Xiao S, Matsui K, Fine A, et al. FasL promoter activation by IL- the CARMA1 linker controls NF-␬ B activation. Immunity
2 through SP1 and NFAT but not Egr-2 and Egr-3. Eur J Immunol 2005;23:561–74.
1999;29:3456–65. [49] Rawlings DJ, Sommer K, Moreno-Garcia ME. The CARMA1 sig-
[25] Radvanyi LG, Shi YF, Mills GB, Miller RG. Cell cycle progression nalosome links the signalling machinery of adaptive and innate
out of G1 sensitizes primary-cultured nontransformed T cells to TCR- immunity in lymphocytes. Nat Rev Immunol 2006;6:799–812.
mediated apoptosis. Cell Immunol 1996;170:260–73. [50] Sun LJ, Deng L, Ea CK, Xia ZP, Chen ZJJ. The TRAF6 ubiquitin lig-
[26] Marks-Konczalik J, Dubois S, Losi JM, et al. IL-2-induced activation- ase and TAK1 kinase mediate IKK activation by BCL10 and MALT1
induced cell death is inhibited in IL-15 transgenic mice. PNAS in T lymphocytes. Mol Cell 2004;14:289–301.
2000;97:11445–50. [51] Kamata H, Honda S, Maeda S, Chang LF, Hirata H, Karin M.
[27] Ku CC, Murakami M, Sakamoto A, Kappler J, Marrack P. Control of Reactive oxygen species promote TNF␣-induced death and sus-
homeostasis of CD8+ memory T cells by opposing cytokines. Science tained JNK activation by inhibiting MAP kinase phosphatases. Cell
2000;288:675–8. 2005;120:649–61.
[28] Zhang X, Sun S, Hwang I, Tough DF, Sprent J. Potent and selective [52] Pham CG, Bubici C, Zazzeroni F, et al. Ferritin heavy chain upreg-
stimulation of memory-phenotype CD8+ T cells in vivo by IL-15. ulation by NF-␬ B inhibits TNF␣-induced apoptosis by suppressing
Immunity 1998;8:591–9. reactive oxygen species. Cell 2004;119:529–42.
62 D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64

[53] Schulze-Luehrmann J, Santner-Nanan B, Jha MK, Schimpl A, Avots [72] Debatin KM, Fahrig-Faissner A, Enenkel-Stoodt S, Kreuz
A, Serfling E. Hematopoietic progenitor kinase 1 supports apoptosis W, Benner A, Krammer PH. High expression of APO-1
of T lymphocytes. Blood 2002;100:954–60. (CD95) on T lymphocytes from human immunodefi-
[54] Kiefer F, Tibbles LA, Anafi M, et al. HPK1, a hematopoi- ciency virus-1-infected children. Blood 1994;83:3101–
etic protein kinase activating the SAPK/JNK pathway. EMBO J 3.
1996;15:7013–25. [73] Bidere N, Su HC, Lenardo MJ. Genetic disorders of programmed cell
[55] Arnold R, Liou J, Drexler HCA, Weiss A, Kiefer F. Caspase-mediated death in the immune system. Annu Rev Immunol 2006;24:321–52.
cleavage of hematopoietic progenitor kinase 1 (HPK1) converts [74] Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA,
an activator of NF␬ B into an inhibitor of NF␬ B. J Biol Chem Nagata S. Lymphoproliferation disorder in mice explained by defects
2001;276:14675–84. in Fas antigen that mediates apoptosis. Nature 1992;356:314–7.
[56] Liou J, Kiefer F, Dang A, et al. HPK1 is activated by lympho- [75] Nagata S. Mutations in the Fas antigen gene in lpr mice. Sem Immunol
cyte antigen receptors and negatively regulates AP-1. Immunity 1994;6:3–8.
2000;12:399–408. [76] Takahashi T, Tanaka M, Brannan CI, et al. Generalized lymphoprolif-
[57] Liu SK, Smith CA, Arnold R, Kiefer F, McGlade CJ. The adaptor erative disease in mice, caused by a point mutation in the Fas ligand.
protein Gads (Grb2-related adaptor downstream of Shc) is implicated Cell 1994;76:969–76.
in coupling hemopoietic progenitor kinase-1 to the activated TCR. J [77] Radvanyi LG, Raju K, Spaner D, Mills GB, Miller RG. Interleukin-
Immunol 2000;165:1417–26. 2 reverses the defect in activation-induced apoptosis in T cells from
[58] Arnold R, Patzak IM, Neuhaus B, et al. Activation of hematopoi- autoimmune lpr mice. Cell Immunol 1998;183:1–12.
etic progenitor kinase 1 involves relocation, autophosphorylation, [78] Gorak-Stolinska P, Truman JP, Kemeny DM, Noble A. Activation-
and transphosphorylation by protein kinase D1. Mol Cell Biol induced cell death of human T-cell subsets is mediated by Fas and
2005;25:2364–83. granzyme B but is independent of TNF-␣ 21. J Leukocyte Biol
[59] Le Bras S, Foucault I, Foussat A, Brignone C, Acuto O, Deckert M. 2001;70:756–66.
Recruitment of the actin-binding protein HIP-55 to the immunological [79] Sytwu HK, Liblau RS, McDevitt HO. The roles of Fas/APO-1 (CD95)
synapse regulates T cell receptor signaling and endocytosis. J Biol and TNF in antigen-induced programmed cell death in T cell receptor
Chem 2004;279:15550–60. transgenic mice. Immunity 1996;5:17–30.
[60] Chen YR, Meyer CF, Ahmed B, Yao ZB, Tan TH. Caspase-mediated [80] Green DR, Droin N, Pinkoski M. Activation-induced cell death in T
cleavage and functional changes of hematopoietic progenitor kinase cells. Immunol Rev 2003;193:70–81.
1 (HPK1). Oncogene 1999;18:7370–7. [81] Janssen EM, Droin NM, Lemmens EE, et al. CD4(+) T-cell help con-
[61] Arnold R, Frey CR, Muller W, Brenner D, Krammer PH, Kiefer F. trols CD8(+) T-cell memory via TRAIL-mediated activation-induced
Sustained JNK signaling by proteolytically processed HPK1 medi- cell death. Nature 2005;434:88–93.
ates IL-3 independent survival during monocytic differentiation. Cell [82] Muppidi JR, Siegel RM. Ligand-independent redistribution of Fas
Death Differ 2007;14:568–75. (CD95) into lipid rafts mediates clonotypic T cell death. Nat Immunol
[62] Brenner D, Golks A, Becker M, et al. Caspase-cleaved HPK1 induces 2004;5:182–9.
CD95L-independent activation-induced cell death in T and B lym- [83] Devadas S, Das J, Liu C, et al. Granzyme B is critical for T
phocytes. Blood 2007;110:3968–77. cell receptor-induced cell death of type 2 helper T cells. Immunity
[63] Campbell KJ, Rocha S, Perkins ND. Active repression of anti- 2006;25:237–47.
apoptotic gene expression by ReIA(p65) NF-␬ B. Mol Cell [84] Peter ME, Krammer PH. The CD95 (APO-1/Fas) DISC and beyond.
2004;13:853–65. Cell Death Differ 2003;10:26–35.
[64] Shishodia S, Aggarwal BB. Guggulsterone inhibits NF-␬ B and [85] Bentele M, Lavrik I, Ulrich M, et al. Mathematical modeling
I kappa B alpha kinase activation, suppresses expression of anti- reveals threshold mechanism in CD95-induced apoptosis. J Cell Biol
apoptotic gene products, and enhances apoptosis. J Biol Chem 2004;166:839–51.
2004;279:47148–58. [86] Lavrik IN, Golks A, Baumann S, Krammer PH. Caspase-2 is activated
[65] Lissy NA, Davis PK, Irwin M, Kaelin WG, Dowdy SF. A com- at the CD95 death-inducing signaling complex in the course of CD95-
mon E2F-1 and p73 pathway mediates cell death induced by TCR induced apoptosis. Blood 2006;108:559–65.
activation. Nature 2000;407:642–5. [87] Lavrik I, Golks A, Krammer PH. Death receptor signalling. J Cell Sci
[66] Vallabhapurapu S, Ryseck RP, Malewicz M, Weih DS, Weih F. Inhi- 2005;118:265–7.
bition of NF-kappa B in T cells blocks lymphoproliferation and [88] Thome M, Tschopp J. Regulation of lymphocyte proliferation and
partially rescues autoimmune disease in gld/gld mice. Eur J Immunol death by flip. Nat Rev Immunol 2001;1:50–8.
2001;31:2612–22. [89] Krueger A, Schmitz I, Baumann S, Krammer PH, Kirchhoff S. Cellu-
[67] Kaminski M, Kiessling M, Suess D, Krammer PH, Gulow K. Novel lar FLICE-inhibitory protein splice variants inhibit different steps of
role for mitochondria: protein kinase C theta dependent oxidative caspase-8 activation at the CD95 death-inducing signaling complex.
signalling organelles in activation induced T cell death. Mol Cell Biol J Biol Chem 2001;276:20633–40.
2007;27:3625–39. [90] Scaffidi C, Schmitz I, Krammer PH, Peter ME. The role of c-
[68] Jackson SH, Devadas S, Kwon J, Pinto LA, Williams MS. T cells FLIP in modulation of CD95-induced apoptosis. J Biol Chem
express a phagocyte-type NADPH oxidase that is activated after T 1999;274:1541–8.
cell receptor stimulation. Nat Immunol 2004;5:818–27. [91] Djerbi M, Darreh-Shori T, Zhivotovsky B, Grandien A. Characteri-
[69] Hildeman DA, Mitchell T, Aronow B, Wojciechowski S, Kappler J, zation of the human FLICE-inhibitory protein locus and comparison
Marrack P. Control of Bcl-2 expression by reactive oxygen species. of the anti-apoptotic activity of four different FLIP isoforms. Scand
PNAS 2003;100:15035–40. J Immunol 2001;54:180–9.
[70] Schieven GL, Mittler RS, Nadler SG, et al. ZAP-70 tyrosine [92] Golks A, Brenner D, Fritsch C, Krammer PH, Lavrik IN. c-FLIPR,
kinase, CD45, and T cell receptor involvement in UV- and H2 O2 - a new regulator of death receptor-induced apoptosis. J Biol Chem
induced T cell signal transduction. J Biol Chem 1994;269:20718– 2005;280:14507–13.
26. [93] Schmitz I, Weyd H, Krueger A, et al. Resistance of short term activated
[71] Baumler CB, Bohler T, Herr I, Benner A, Krammer PH, Debatin T cells to CD95-mediated apoptosis correlates with de novo protein
KM. Activation of the CD95 (APO-1/Fas) system in T cells from synthesis of c-FLIPshort. J Immunol 2004;172:2194–200.
human immunodeficiency virus type-1-infected children. Blood [94] Kirchhoff S, Muller WW, Krueger A, Schmitz I, Krammer PH.
1996;88:1741–6. TCR-mediated up-regulation of c-FLIPshort correlates with resis-
D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64 63

tance toward CD95-mediated apoptosis by blocking death-inducing [115] van Parijs L, Peterson DA, Abbas AK. The Fas/Fas ligand pathway
signaling complex activity. J Immunol 2000;165:6293–300. and Bcl-2 regulate T cell responses to model self and foreign antigens.
[95] Schmitz I, Krueger A, Baumann S, Schulze-Bergkamen H, Kram- Immunity 1998;8:265–74.
mer PH, Kirchhoff S. An IL-2-dependent switch between CD95 [116] Datta SR, Dudek H, Tao X, et al. Akt phosphorylation of BAD
signaling pathways sensitizes primary human T cells toward CD95- couples survival signals to the cell-intrinsic death machinery. Cell
mediated activation-induced cell death. J Immunol 2003;171:2930– 1997;91:231–41.
6. [117] Zha JP, Harada H, Yang E, Jockel J, Korsmeyer SJ. Serine phospho-
[96] Fas SC, Baumann S, Krueger A, et al. In vitro generated rylation of death agonist BAD in response to survival factor results in
human memory-like T cells are CD95 type II cells and resistant binding to 14-3-3 not BGL-X(L). Cell 1996;87:619–28.
towards CD95-mediated apoptosis. Eur J Immunol 2006;36:2894– [118] Bouillet P, Purton JF, Godfrey DI, et al. BH3-only Bcl-2 family mem-
903. ber Bim is required for apoptosis of autoreactive thymocytes. Nature
[97] Budd RC, Yeh WC, Tschopp J. cFLIP regulation of lymphocyte acti- 2002;415:922–6.
vation and development. Nat Rev Immunol 2006;6:196–204. [119] Zong WX, Lindsten T, Ross AJ, MacGregor GR, Thompson CB.
[98] Scaffidi C, Schmitz I, Zha J, Korsmeyer SJ, Krammer PH, Peter ME. BH3-only proteins that bind pro-survival Bcl-2 family members fail
Differential modulation of apoptosis sensitivity in CD95 type I and to induce apoptosis in the absence of Bax and Bak. Genes Dev
type II cells. J Biol Chem 1999;274:22532–8. 2001;15:1481–6.
[99] Villalba M, Kasibhatla S, Genestier L, Mahboubi A, Green DR, Alt- [120] Strasser A, Harris AW, Cory S. bcl-2 transgene inhibits T cell death
man A. Protein kinase C␪ cooperates with calcineurin to induce Fas and perturbs thymic self-censorship. Cell 1991;67:889–99.
ligand expression during activation-induced T cell death. J Immunol [121] Mitchell T, Kappler J, Marrack P. Bystander virus infection prolongs
1999;163:5813–9. activated T cell survival. J Immunol 1999;162:4527–35.
[100] Manicassamy S, Sun Z. The critical role of protein kinase C-␪ [122] Grayson JM, Zajac AJ, Altman JD, Ahmed R. Cutting edge: increased
in Fas/Fas ligand-mediated apoptosis. J Immunol 2007;178:312– expression of Bcl-2 in antigen-specific memory CD8(+) T cells. J
9. Immunol 2000;164:3950–4.
[101] Holtz-Heppelmann CJ, Algeciras A, Badley AD, Paya CV. Transcrip- [123] Gonzalo JA, Tarazona R, Schuurman HJ, et al. A single injection
tional regulation of the human FasL promoter-enhancer region. J Biol of Staphylococcus aureus enterotoxin B reduces autoimmunity in
Chem 1998;273:4416–23. MRL/lpr mice. Clin Immunol Immunopathol 1994;71:176–82.
[102] Kasibhatla S, Genestier L, Green DR. Regulation of Fas-ligand [124] Hildeman DA, Mitchell T, Teague TK, et al. Reactive oxygen species
expression during activation-induced cell death in T lymphocytes via regulate activation-induced T cell apoptosis. Immunity 1999;10:
nuclear factor kappa B. J Biol Chem 1999;274:987–92. 735–44.
[103] Latinis KM, Norian LA, Eliason SL, Koretzky GA. Two NFAT tran- [125] Ettinger R, Panka DJ, Wang JK, Stanger BZ, Ju ST, Marshak-
scription factor binding sites participate in the regulation of CD95 Rothstein A. Fas ligand-mediated cytotoxicity is directly responsible
(Fas) ligand expression in activated human T cells. J Biol Chem for apoptosis of normal CD4+ T cells responding to a bacterial super-
1997;272:31427–34. antigen. J Immunol 1995;154:4302–8.
[104] Li-Weber M, Laur O, Hekele A, Coy J, Walczak H, Krammer PH. [126] Renno T, Hahne M, Tschopp J, MacDonald HR. Peripheral T cells
A regulatory element in the CD95 (APO-1/Fas) ligand promoter undergoing superantigen-induced apoptosis in vivo express B220 and
is essential for responsiveness to TCR-mediated activation. Eur J upregulate Fas and Fas ligand. J Exp Med 1996;183:431–7.
Immunol 1998;28:2373–83.
[105] Manicassamy S, Gupta S, Huang Z, Sun Z. Protein kinase C-␪-
mediated signals enhance CD4+ T cell survival by up-regulating
Biographies
Bcl-xL. J Immunol 2006;176:6709–16.
[106] Saibil SD, Jones RG, Deenick EK, et al. CD4+ and CD8+ T cell Dirk Brenner is a junior post-doc at the DKFZ, Heidel-
survival is regulated differentially by protein kinase C␪, c-Rel, and
berg, Germany, in the division of Peter H. Krammer in the
protein kinase B. J Immunol 2007;178:2932–9.
[107] Villalba M, Bushway P, Altman A. Protein kinase C-theta medi- subgroup of Rüdiger Arnold. He studied biochemistry at the
ates a selective T cell survival signal via phosphorylation of BAD. universities of Bonn and Witten/Herdecke (Germany) and
J Immunol 2001;166:5955–63. Stanford and Harvard (USA). He joined the division of Peter
[108] Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. H. Krammer for his Ph.D. thesis and is working since then
Th17: an effector CD4 T cell lineage with regulatory T cell ties.
on signal transduction pathways influencing sensitivity and
Immunity 2006;24:677–88.
[109] Varadhachary AS, Perdow SN, Hu CG, Ramanarayanan M, Salgame resistance to activation-induced cell death in lymphocytes.
P. Differential ability of T cell subsets to undergo activation-induced
Peter H. Krammer is head of the Division of Immuno-
cell death. Proc Natl Acad Sci USA 1997;94:5778–83.
[110] Roberts AI, Devadas S, Zhang XR, et al. The role of activation- genetics and speaker of the Tumor Immunology Program
induced cell death in the differentiation of T-helper-cell subsets. at the DKFZ, Heidelberg, Germany. He studied medicine at
Immunol Res 2003;28:285–93. the universities of Freiburg, Germany, St. Louis, USA, and
[111] Zhang XR, Zhang LY, Devadas S, Li L, Keegan AD, Shi YF. Recip- Lausanne, Switzerland. After his M.D. he joined the Basel
rocal expression of TRAIL and CD95L in Th1 and Th2 cells: role
Institute of Immunology, Switzerland, where he focused on T
of apoptosis in T helper subset differentiation. Cell Death Differ
2003;10:203–10. cell biology. In 1976, he moved to the DKFZ to continue his
[112] Zhang XH, Brunner T, Carter L, et al. Unequal death in T helper cell work on T cell function and characterized the death recep-
(Th)1 and Th2 effectors: Th1, but not Th2, effectors undergo rapid tor CD95 (APO-1/Fas). Peter H. Krammer is interested in
Fas/FasL-mediated apoptosis. J Exp Med 1997;185:1837–49. the general mechanisms influencing life and death decisions
[113] Lord SJ, Rajotte RV, Korbutt GS, Bleackley RC. Granzyme B: a
in lymphocytes. The research in his division is focused on
natural born killer. Immunol Rev 2003;193:31–8.
[114] Sharma V, Delgado M, Ganea D. VIP protects Th2 cells by downreg- molecular mechanisms involving CD95, regulatory T cells,
ulating granzyme B expression. Vip Pacap Rel Peptides: Gene Ther cytokine gene expression (e.g. IL-4) and T cell receptor sig-
2006;1070:540–4. naling in health and disease.
64 D. Brenner et al. / Critical Reviews in Oncology/Hematology 66 (2008) 52–64

Rüdiger Arnold is group leader at the DKFZ, Heidel- and Clinical Research, Bad Nauheim, Germany, where he
berg, Germany, in the division of Peter H. Krammer. He studied signal transduction mechanisms in hematopoietic
studied biology at the universities of Darmstadt, Germany, cells in the group of Friedemann Kiefer. He moved to Hei-
and Giessen, Germany, and joined the department of Rainer delberg in 2002 to continue his work on signal transduction,
Renkawitz, Giessen, Germany to undertake his Ph.D. on where his group studies mechanisms of activation, prolifera-
mechanisms of gene regulation. He performed his postdoc- tion and differentiation of leukocytes.
toral studies at the Max-Planck-Institute for Physiological

You might also like