Sie sind auf Seite 1von 8

Biochimica et Biophysica Acta 1793 (2009) 13351342

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta


j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / b b a m c r

Invited review

Apoptin, a tumor-selective killer


Marek Los a,b,,1, Soumya Panigrahi c,1, Iran Rashedi d, Sanat Mandal e, Joerg Stetefeld f,
Frank Essmann a, Klaus Schulze-Osthoff a
a
Interfaculty Institute for Biochemistry, University of Tbingen, D-72076 Tbingen, Germany
b
BioApplications Enterprises, Winnipeg, MB, Canada R2V 2N6
c
Department of Immunology, Lerner Research Institute/NE40, 9500 Euclid Avenue, Cleveland, OH 44195, USA
d
Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada M5G 2M9
e
Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada A1B 3V6
f
Department of Chemistry, University of Manitoba, Winnipeg, Canada

a r t i c l e i n f o a b s t r a c t

Article history: Apoptin, a small protein from chicken anemia virus, has attracted great attention, because it specically kills
Received 20 February 2009 tumor cells while leaving normal cells unharmed. The subcellular localization of apoptin appears to be crucial
Received in revised form 5 April 2009 for this tumor-selective activity. In normal cells, apoptin resides in the cytoplasm, whereas in cancerous cells
Accepted 7 April 2009
it translocates into the nucleus. The nuclear translocation of apoptin is largely controlled by its
Available online 15 April 2009
phosphorylation. In tumor cells, apoptin causes the nuclear accumulation of survival kinases including Akt
Keywords:
and is phosphorylated by CDK2. Thereby, apoptin redirects survival signals into cell death responses. Apoptin
Apoptin also binds as a multimeric complex to DNA and interacts with several nuclear targets, such as the anaphase-
Apoptosis promoting complex, resulting in a G2/M phase arrest. The proapoptotic signal of apoptin is then transduced
CDK2 from the nucleus to cytoplasm by Nur77, which triggers a p53-independent mitochondrial death pathway. In
Nur77 this review, we summarize recent discoveries of apoptin's mechanism of action that might provide intriguing
Tumor insights for the development of novel tumor-selective anticancer drugs.
2009 Elsevier B.V. All rights reserved.

1. Introduction Sequences in the 5-nontranscribed region of the CAV genome


function as promoter/enhancers [3]. The CAV genome contains three
Apoptin is a small apoptosis-inducing protein derived from partially overlapping open reading frames which produce a poly-
chicken anemia virus (CAV), which belongs to the Gyrovirus genus cistronic unspliced RNA encoding three different proteins [1,46]. All
and Circoviridae family and contains a single, minus-strand circular proteins are expressed in CAV-infected cells [5,6]. VP1 is a 51-kDa
DNA. CAV infects young chickens with the highest infectivity and capsid protein and VP2 a 24-kDa dual specicity phosphatase.
mortality on the hatch day, whereas chickens become completely Importantly, the induction of apoptosis by CAV has been attributed
resistant to CAV-related death after about two weeks of hatching [1]. to VP3, a 14-kDa non-structural protein, which is therefore also
Hematopoietic cells in the bone marrow and T-cell precursors in the referred to as apoptin [5,7,8]. Apoptin alone can induce apoptosis in a
thymus are the major targets for CAV infection. The early infection of broad range of transformed and cancer cells but not in non-
hematopoietic precursors results in the reduction of erythroid transformed or primary cells [9,10]. Recent data link apoptin's
precursor cells and megakaryocytes. This eventually leads to anemia complex mode of action to various cellular proteins, which play
and thrombocytopenia, causing intramuscular hemorrhage and death. prominent roles in both cell proliferation and cell death. It was
The infection can be propagated within the ock both vertically and reported that late in infection, three other CAV mRNAs are synthesized
horizontally. Interestingly, in chickens carrying tumors induced by due to splicing events [11]. One of them encodes a putative truncated
Rous sarcoma virus the intra-tumoral expression of apoptin causes apoptin protein consisting of the rst 58 amino acids only. The
tumor regression [2]. Thus, the coevolution of CAV and the host may signicance of these additional putative gene products for cell death
offer benets to latently infected chickens. and other CAV manifestations is still unclear.

2. Apoptin sequence and structure

Corresponding author. University of Tbingen, Interfaculty Institute for


Apoptin is composed of 121 amino acids, and does not show
Biochemistry, Hoppe-Seyler-Str. 4, D-72076 Tbingen, Germany. Tel.: +49 7071
2974159; fax: +49 7071 295565.
signicant sequence homology with known cellular proteins [1]. The
E-mail address: mjelos@gmail.com (M. Los). C-terminal end of apoptin contains a bipartite nuclear localization
1
These authors contributed equally. sequence (NLS): NLS1 spans amino acids 8288, and NLS2 residues

0167-4889/$ see front matter 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbamcr.2009.04.002
1336 M. Los et al. / Biochimica et Biophysica Acta 1793 (2009) 13351342

Fig. 1. The primary structure of apoptin. The key domains and important sequences are indicated. Lower panel shows the sequence of apoptin (UniProtKB/Swiss-Prot entry P54094).
Amino acids marked in blue or green represent the indicated domains in the upper panel. See text for details.

111121 [12,13]. In addition, apoptin contains a putative nuclear In addition to reports that apoptin effectively induces tumor-
export sequence (NES) at residues 97105 (Fig. 1). These recognition specic apoptosis in animal models, intra-tumoral injection of
sequences drive the shuttling of apoptin in and out of the nucleus. recombinant apoptin into nude mice with xenografted human
Apoptin also harbors a short hydrophobic leucine-rich stretch (aa 33 hepatoma cells reduces tumor growth and improves tumor regression
46) that is required for self-association as well as binding of within a week of administration [22]. There are additional reports that
promyelocytic leukemia protein and other interaction partners apoptin can enhance tumor size reduction. In fact, when apoptin is
[9,14]. It is interesting to note that the biologically active form of used in combination with other chemotherapeutics, tumor regression
recombinant apoptin can form noncovalently attached globular is more dramatic compared to conditions when apoptin or che-
multimers consisting of 3040 monomers [15]. The formation of motherapeutic drugs are administered alone [7,24].
this multimeric complex appears to be facilitated through the
interaction of the proline-rich hydrophobic regions in the N-terminus 4. Mechanisms of apoptin-mediated cell death
(aa 169) of each monomer [16]. The C-terminal tip of each
monomer, on the other hand, contains a NLS with an accessible The mechanisms of cell death induced by apoptin are not clearly
phosphorylation site (Thr-108), which allows for its interaction with understood, although there is a consensus on the existence of certain
other proteins and for modication by kinases. Microinjection of molecular events. One of the key properties of apoptin is its ability to
apoptin multimers into the cytoplasm of tumor cells reveals that induce tumor-specic cell apoptosis independently of p53 [25]. Thus,
apoptin complexes translocate to the nucleus resulting in apoptotic apoptin is similarly effective in killing tumor cells that are p53-
death [15]. This conrms that apoptin multimers retain biological decient or either express wildtype or mutant p53.
activity. Unfortunately, a crystal structure of apoptin or its multimeric Apoptosis is typically mediated by intracellular cysteine proteases,
complex is not available. called caspases, that function as initiators and executioners of the
apoptotic process [26,27]. The activation of caspases is accomplished
3. Induction of apoptosis in tumor cells by apoptin by two major signaling routes: the extrinsic death receptor and the
intrinsic mitochondrial pathway. In the extrinsic pathway, ligation of
Apoptin has been reported to induce the selective death of cell death receptors by their cognate ligands triggers the recruitment of
lines derived from diverse human tumors including melanoma, the adaptor FADD and initiator caspase-8 into a death-inducing
hepatoma, lymphoma, cholangiocarcinoma, colon carcinoma, breast signaling complex. The mitochondrial pathway is initiated by the
and lung cancer and others [14,17]. Indeed, to date more than 70 release of cytochrome c into the cytosol, a process that is tightly
human tumor cell lines were shown to be susceptible to apoptin, regulated by pro- and anti-apoptotic proteins of the Bcl-2 family. Once
whereas apoptin does not induce apoptosis in a variety of normal released, cytochrome c binds to the adapter protein Apaf-1, which
cells, including human endothelial cells, hepatocytes and hemato- enables the activation of the initiator caspase-9 in a high-molecular-
poietic stem cells. Thus, the sensitivity to apoptin presumably weight complex, called the apoptosome. Both pathways then
depends on alterations related to the transformed status of the cell converge at the activation of downstream effector caspases, leading
[14]. Apoptin induces apoptosis not only in tumor cells, but also in to cell death via the cleavage of several cellular substrates [2628].
SV40-transformed broblasts or UV-irradiated cells from indivi- It is now well established that apoptin expression results in the
duals with hereditary cancer-prone syndromes [18,19]. There is activation of caspases [29]. Apoptin-mediated cell death is however
only one single report of toxicity towards some non-cancerous independent of death receptors, as cells decient in FADD or caspase-
cells [20]. Nonetheless, this study provided cell death data only for 8, the key regulators of the extrinsic apoptotic pathway [30], remain
a fetal cell type (i.e. secondary cultures of human lung broblasts) sensitive to apoptin. Moreover, blocking of death receptor CD95 with
and not for other non-transformed cell types. This raises the neutralizing antibodies does not affect apoptin-induced cell death. In
possibility that apoptin selectively affects normal cells at embryo- contrast to components of the death receptor pathway, apoptin-
nic stage, similar to apoptin's toxicity in young but not adult mediated apoptosis is strongly inuenced by regulators of the
chickens. However, the safety of apoptin is underlined by the fact mitochondrial pathway, as for instance a deciency of Apaf-1 strongly
that continuous expression of apoptin under the H2-Kb promoter protects tumor cells. Furthermore, apoptin triggers cytochrome c
in transgenic mice does not interfere with lymphocyte develop- release and the activation of caspase-9 [31].
ment and proliferation [21]. This is in contrast to the deleterious Interestingly, the role of anti-apoptotic Bcl-2 in apoptin-mediated
effects on the immune system observed in young chickens upon cell death is still a matter of debate. Some research groups reported
CAV infection. that apoptin-mediated cell death is independent of the Bcl-2 status,
M. Los et al. / Biochimica et Biophysica Acta 1793 (2009) 13351342 1337

and that co-expression of apoptin and Bcl-2 even accelerates cell that is strongly activated upon apoptin treatment is a viable candidate.
death [3234]. In contrast, Burek et al. [31] showed that both Bcl-2 and Upon transfer to cytoplasm, Nur77 may directly or indirectly cause
Bcl-xL inhibit apoptosis triggered by apoptin in several tumor cell cytochrome c release and activation of the apoptosome-dependent
lines. Notably, the proapoptotic Bcl-2 member Bax sensitizes cells to death pathway.
apoptin-induced apoptosis, whereas cells devoid of Bax are strongly
protected [31]. Thus, at least in several cell types, apoptin-induced cell 5. Apoptin-interacting molecules
death is regulated by pro- and anti-apoptotic Bcl-2 proteins and
involves the release of cytochrome c and activation of effector The cellular localization of apoptin plays crucial role for its
caspases. The apparent controversy as to the role of Bcl-2 proteins selective toxicity [12,13]. However, the targeted translocation of
may arise from the differential involvement of Nur77, a member of the apoptin into the nucleus is not sufcient to induce apoptosis in
steroid receptor family, in apoptin-induced cell death. It has been primary cells [12]. This nding suggests that for its tumor-specic
shown that Nur77 can bind to Bcl-2 and change its properties from an toxicity apoptin presumably requires additional interaction partners
anti-apoptotic to a proapoptotic molecule, leading to the activation of that activate specic signaling pathways in cancer cells. Indeed, a
mitochondrial death pathway [35]. Because Nur77 is expressed in number of molecules have been described to interact with apoptin
different cell types at very varying levels, this might explain the and appear to be important for the nuclear localization of apoptin or
opposite effects of Bcl-2 on apoptin-induced cell death. its tumor-selective cytotoxicity (Table 1).
Maddika et al. [30] indeed proposed a role of Nur77 in apoptin- A recent report [36] provides evidence that apoptin interacts with
mediated apoptosis (Fig. 2). Phosphorylated Nur77 appears to peptidyl-prolyl isomerase-like 3 (Ppil3). Ppil3 belongs to the
transmit apoptotic signal from the nucleus to mitochondria, as it cyclophilin family and shares 46% identity to cyclophilin A. Members
was shuttled from the nucleus to the cytoplasm upon transient of this family catalyze the cistrans isomerization of peptidyl-prolyl
expression of apoptin. Moreover, RNAi-mediated downregulation of imidic bonds and play a role in mitochondrial maintenance, cell cycle
Nur77 strongly protected against apoptin-triggered apoptosis [30]. It progression and apoptosis. The study showed that siRNA-mediated
is presently unclear which kinase phosphorylates Nur77, however, Akt depletion of Pipl3 leads to the migration of apoptin from cytoplasm to

Fig. 2. Proposed mechanism of action of apoptin in tumor cells. Upon heterologous expression or administration of cell-permeable constructs, apoptin associates with the regulatory
subunit of PI3K resulting in its constitutive activation, which leads to the sustained activation and nuclear translocation of Akt. The nuclear Akt activates CDK2 by both direct
phosphorylation and indirect mechanisms including the proteasome-dependent degradation of p27Kip1. The activated CDK2 phosphorylates apoptin at Thr-108 and thereby enforces
its nuclear accumulation in cancer cells. In the nucleus, apoptin can associate with different interaction partners including DEDAF, Nmi, APC/C and PML. A still unknown apoptin-
triggered event results in the phosphorylation of Nur77 and its nuclear export. In the cytoplasm Nur77 is known to modulate Bcl-2 proteins, for instance by converting Bcl-2 from an
anti-apoptotic to a proapoptotic molecule. This and additional events might induce apoptin-triggered apoptosis by the mitochondrial pathway of caspase activation.
1338 M. Los et al. / Biochimica et Biophysica Acta 1793 (2009) 13351342

the nucleus. Interestingly, proline-109 in apoptin was essential for the monomers [15]. These apoptin multimers in turn can bind to DNA and
Ppil3-dependent cytoplasmic localization of apoptin [36]. These cooperatively form distinct superstructures consisting of 20 apoptin
ndings shed a new light on the regulation of apoptin's cellular multimers and approximately 3 kb DNA [16]. Whether apoptin's
localization. binding to DNA results in transcriptional events is unknown. The
Hippi, a protein interacting with huntington-interacting protein 1 nding that apoptin does neither act as a transcriptional repressor, at
(Hip 1), is also a binding partner for apoptin [37]. The HippiHip1 least in simple reporter gene assays, nor requires de novo macro-
complex can recruit caspase-8 and regulates apoptotic cell demise in molecular synthesis for apoptosis induction would, however, argue
Huntington's disease. In non-cancerous cells, apoptin colocalizes with against this possibility [12].
Hippi in the cytoplasm, whereas in tumor cells apoptin migrates to the Apoptin can also interact with a proapoptotic protein, called death
nucleus while Hippi remains in the cytoplasm [37]. The C-terminus effector domain-associated factor (DEDAF). DEDAF associates with
region of apoptin is vital for the interaction with Hippi. As mentioned death effector domain-containing proapoptotic proteins and plays a
above, however, apoptin-mediated cell death is not compromised by a role in transcriptional regulation [40]. Similar to apoptin, transient
genetic deciency of caspase-8 [30]. Nevertheless, the results suggest overexpression of DEDAF can induce cell death in cancer cells, but not
that association of apoptin with cytoplasmic Hippi or Ppil3 may play a in normal cells. The cellular localization of DEDAF is similar to that of
role in sequestering apoptin in cytoplasm and preventing apoptosis of apoptin, as both colocalize into the nucleus of tumor cells. Coexpres-
normal primary cells. sion of apoptin and DEDAF increases the apoptotic death rate as
Employing a yeast two-hybrid approach with apoptin as bait, N- compared to the effects of expression of either of them alone [40].
Myc-interacting protein (Nmi) has been found to associate with Nonetheless, whether the interaction of apoptin with DEADF is
apoptin [38]. It is an interferon-inducible protein that can interact responsible for apoptin's toxicity awaits further investigation.
with transcription factors of Myc family. Nmi-like proteins enhance Apoptin also interacts with promyelocytic leukemia protein (PML)
the activity of Myc proteins that are involved in cell proliferation, and localizes in PML nuclear bodies [41]. PML nuclear bodies are
differentiation as well as in apoptosis. The interaction of apoptin and ordered protein complexes that associate with the nuclear matrix and
Nmi has been proposed to account for the tumor specicity of play a role in apoptosis, DNA replication, repair, transcription, RNA
apoptin's cytotoxicity, because Nmi is only expressed at low levels in transport and viral replication [42]. Many proteins residing in PML
normal tissues but highly upregulated in leukemia cell lines. Apoptin nuclear bodies, including apoptin, undergo a specic posttranslational
has been hypothesized to switch the Nmi/Myc-mediated transform- modication, called sumoylation. However, the nding that a
ing activity to an apoptotic activity. However, no functional analyses sumoylation-decient mutant of apoptin can still trigger apoptosis
have been provided as yet, and the restricted expression pattern of in wildtype as well as in PML-decient cells suggest that neither
Nmi does not explain the broad antitumor activity of apoptin. sumoylation nor the interaction with PML are crucial for apoptin's
Teodoro et al. [39] have shown that apoptin associates with APC1, a proapoptotic activity. Nevertheless, although apoptin's interaction
subunit of anaphase-promoting complex/cyclosome (APC/C) and with PML appears dispensable for apoptosis, it was speculated to be
thereby inhibits the ubiquitin ligase function of APC/C. This interac- involved in CAV replication [41]. It has also been suggested that
tion is present in cancer cells but is absent in non-cancerous cells, apoptin colocalizes with FADD (Fas-associated protein with death
which suggests another possible mechanism for the tumor-selective domain) and Bcl10, a caspase recruitment domain-containing protein,
action of apoptin. Overexpression of apoptin or RNAi-mediated in cytoplasmic structures, called death effector laments [20]. Both
depletion of APC1 subunit induces apoptosis in p53-null tumor cells, FADD and Bcl10 are involved in apoptosis signaling by death receptors,
suggesting that apoptin may interfere with the APC/C pathway though the signicance of this colocalization for apoptin-induced cell
through binding to APC1. These experiments provide evidence that death is unknown.
APC/C may serve as a potential target for the development of novel Our group recently reported that, through its proline-rich
anticancer therapies. sequence (aa 8186), apoptin interacts with the SH3 domain of the
Due to its basic structure apoptin binds to naked DNA, preferen- p85 regulatory subunit of phosphoinositide 3-kinase (PI3K, Fig. 2).
tially to heterochromatin. Biophysical studies revealed that bacterially Class I PI3 kinases play a central role in various regulatory processes,
expressed apoptin forms active protein multimers consisting of 3040 such as cell growth, survival and differentiation. Surprisingly, when

Table 1
Apoptin-interacting molecules.

Molecule Biological effect References


DNA Due to its basic structure, apoptin can directly interact with DNA, preferentially with double stranded DNA and [16,59]
heterochromatin.
Akt and PI3K Apoptin interacts with the SH3-domain of the p85 regulatory subunit of PI3K and activates PI3K. This event leads to [43,44]
sustained Akt activation and presumably aids Akt in nuclear translocation.
CyclinACDK2 CyclinACDK2 complex directly phosphorylates apoptin at Thr-108 and contributes to the regulation of its subcellular [49]
localization.
Peptidyl-prolyl isomerase like 3 (Ppil3) Apoptin's interaction with Ppil3 may favor its cytoplasmic localization. [36]

Hip-interacting protein (Hippi) In non-cancerous cells, apoptin colocalizes with Hippi in the cytoplasm, whereas in tumor cells apoptin migrates to the [37]
nucleus while Hippi remains in the cytoplasm.
N-Myc interacting protein (Nmi) The functional signicance of apoptin and Nmi interaction is not well understood. [38]
APC1 (subunit of anaphase-promoting Apoptin's interaction with APC1 may lead to mitotic cell cycle arrest and apoptosis. [39]
complex/cyclosome)
Death effector domain associated factor Coexpression of apoptin and DEDAF increases apoptosis as compared to the effects of either protein alone. [40]
(DEDAF)
Promyelocytic leukemia protein (PML) SUMOylated apoptin interacts with PML in PML nuclear bodies. However, the disruption of the interaction of PML and [41]
apoptin does not affect apoptin's cytotoxicity.
Fas-associated death domain protein Upon overexpression apoptin colocalizes with FADD, a component of the death receptor pathway, in so-called death effector [20]
(FADD) laments. The signicance of this process is unknown.
Bcll0 Apoptin can colocalize with Bcl10, a regulator of apoptosis and NF-B activation. [20]
Importin-1 Importin-1 aids apoptin's nuclear translocation through the nuclear pore complex. [10,13]
M. Los et al. / Biochimica et Biophysica Acta 1793 (2009) 13351342 1339

p85 is downregulated, apoptin is shuttled out of the nucleus, thereby 7. Apoptin phosphorylation role in apoptosis induction?
imposing a negative regulation of apoptin-induced cell death [43].
Apoptin derivatives devoid of the proline-rich region do not interact In addition to nuclear targeting signals, the phosphorylation of
with p85, are unable to activate PI3K and show impaired apoptosis apoptin is crucial in transducing apoptotic signals in cancer cells.
induction. Thus, the interaction of apoptin with p85 subunit of PI3K Mapping studies provide evidence that apoptin has a phosphorylation
appears to be essential for the cytotoxic activity of apoptin [43]. In site at threonine-108 (Thr-108) adjacent to the NES [57]. Thr-108
addition to p85 regulatory subunit of PI3K, its downstream effector phosphorylation can be detected in vitro and in vivo in cell lines and
kinase Akt1 was also found as an apoptin-interacting molecule [44]. tumors. Phosphorylation at Thr-108 appears to drive tumor-specic
Although the role of PI3K/Akt pathway in cell survival is well nuclear accumulation of apoptin via inactivation of the NES. In
established, there are examples in which PI3K and Akt also promote contrast, two studies reported that Thr-108 phosphorylation only
cell death [4548]. Interestingly, as recently reported, the direct partially affects apoptin's proapoptotic activity, as an apoptin mutant
interaction of apoptin with Akt1 triggers nuclear trafcking of Akt and devoid of this phosphorylation site retains the ability to induce
initiates apoptosis instead of a survival response [43,49]. Agents such apoptosis [43,58]. Some ndings suggest that both N- and C-terminal
as etoposide, cisplatin, and interferon- induce Akt1 phosphorylation regions of apoptin have the capacity to bind DNA and induce apoptosis
that precedes the onset of caspase activation and apoptosis [50,51]. independently, and that Thr-108 phosphorylation is required only for
Importantly, transient activation of Akt1 supports cell survival, the C-terminus [58,59]. An apoptin mutant consisting of the rst 69
whereas its sustained activation leads to apoptosis [5254]. These amino acids is mainly located in the cytoplasm of human tumor cells,
observations therefore support a dual role of PI3K/Akt pathway in cell but harbors a distinct, albeit weaker, proapoptotic activity [12].
survival and apoptosis. Furthermore, there is a cluster of three threonine residues (aa 106
108) within the apoptin sequence. Thus, even though this possibility
6. Nuclear transfer of apoptin vital for its apoptotic action has not yet been tested experimentally, it is conceivable that mutation
of Thr-108 may still allow for the phosphorylation of apoptin at the
The selective toxicity of apoptin is mainly attributed to its adjacent threonine residues.
differential subcellular localization in tumor and normal cells (Fig. 3). Due to the importance of apoptin phosphorylation for its tumor-
Apoptin predominantly localizes to the nucleus of cancer cells, whereas specic action, the apoptin kinase has intensively been sought for a
its accumulation in the nucleus is severely impaired in normal cells long time. We could recently identify the mitogenic cyclin-dependent
[12]. Experimental data indicate the nucleo-cytoplasmic shuttling of kinase CDK2 as the principal kinase that directly phosphorylates
apoptin is regulated by both the bipartite NLS and the N-terminal NES apoptin at Thr-108 and is crucially required for apoptin-induced cell
[10,12,13]. The NLS of apoptin is potentially active in both normal and death [49]. As mentioned above, an early event of apoptin signaling is
cancer cells, as small amounts of apoptin can translocate to the nucleus the interference with the PI3K/Akt pathway, which does not only
in normal cells, albeit with markedly lower efciency [55,56]. Notably, a result in a very sustained activation but also the nuclear translocation
CRM1-regulated NES (aa 97105) has been identied in apoptin that of Akt. This sustained Akt activation is in contrast to the transient
appears to be active in normal cells but not in tumor cells, and thereby stimulation of kinase activity that is normally observed following Akt
retains apoptin in the cytoplasm of normal cells [10]. Apoptin also activation by growth-stimulatory signals. It is therefore likely that the
contains a leucine-rich sequence (aa 3346) that is important for its prolonged activation of Akt might result in the phosphorylation of
interaction with PML and other proteins [9,14]. Mutations that unusual substrates.
inactivate this sequence stretch not only reduce the ability of apoptin During our studies we found that apoptin-induced Akt stimulation
to associate with PML bodies, but also inhibit nuclear accumulation of results in a strong activation of CDK2, which is normally a S-phase
apoptin in tumor cells. kinase involved in cycle progression [49]. Interestingly, pharmacolo-
gical inhibitors as well as dominant-negative mutants of PI3K and Akt
not only inhibited CDK2 activation, but also protected cells from
apoptin-induced cell death. The Akt-mediated activation of CDK2 was
caused by two mechanisms, including the direct phosphorylation of
CDK2 and the phosphorylation-induced degradation of its inhibitor
p27Kip1 (Fig. 2). Furthermore, cyclin A- but not cyclin E-associated
CDK2 was responsible for apoptin phosphorylation at Thr-108 as well
as its tumor-specic localization. The crucial role of CDK2 for the
biological activity of apoptin was further substantiated by the nding
that CDK2 inhibitors as well as the genetic deciency of CDK2 strongly
protected cells against apoptin-induced apoptosis [49]. These unex-
pected ndings therefore suggest that apoptin can convert mitogenic
pathways into cell death responses, which likely account for its tumor-
specic activity.
Interestingly, a sustained activation of Akt in nucleus, unlike the
transient activation induced by growth factors at the cell membrane,
has been implicated in other cell death pathways. For instance,
prolonged nuclear Akt/CDK2 activation does not protect but
sensitizes tumor cells for apoptosis induced by anticancer agents
such as methotrexate and docetaxel [23]. Also, in primary breast
tumor cells an elevated basal CDK2 activity has been found to
Fig. 3. Differential subcellular localization of apoptin in primary and transformed cells. correlate with the sensitivity to paclitaxel [60]. In addition, elevated
(A) In normal cells, such as human primary mononuclear cells, apoptin is localized in CDK2 activity has been implicated in apoptosis in some other
the cytoplasm, as shown by the green staining of GFP-tagged apoptin and blue conditions including negative selection of thymocytes [61]. Aberrant
counterstaining using the nuclear dye DAPI. (B) In transformed cells, such as U2OS
osteosarcoma cells, apoptin resides exclusively in the cell nucleus. Costaining for
activation of CDK2 might therefore cause disturbance of cell cycle
promyelocytic leukemia protein (PML, red) reveals that a large fraction of apoptin progression and cell death. These observations not only decipher
localizes in PML nuclear bodies. mechanisms of apoptin-induced cell death, but also provide novel
1340 M. Los et al. / Biochimica et Biophysica Acta 1793 (2009) 13351342

insights for the selective killing of tumor cells. Diverse regulators of derivatives could be directly targeted to other tissues, such as brain. In
the PI3K/Akt pathway are involved in tumorigenesis and are highly this respect, apoptin conjugates that are taken up by the precursor of
active in various types of cancers. Hyperactivation of these pathways heparin-binding epidermal growth factor, also known as the
is associated with a poor clinical prognosis and contributes to drug diphtheria toxin receptor, are currently being tested [14]. This receptor
resistance. Thus, targeting the PI3K/Akt/CDK2 pathway by apoptin is constitutively present at the bloodbrain barrier and is strongly
might be responsible for its unique tumor-specic effects. We upregulated in many tumors, such as glioblastoma.
propose that apoptin hijacks cell survival pathways and redirects Several studies have used apoptin derivates coupled to protein
them towards induction of cell death, thereby establishing a novel transduction domains, including HIV-Tat, which allows for the
link between cell survival and cell death. efcient intracellular delivery of exogenously administered apoptin
[20,30,43]. However, although apoptin fused to HIV-Tat-derived
8. Clinical perspectives of apoptin peptide has been proven to efciently kill various tumor cell types
in culture, its use in preclinical and clinical settings is certainly
The ability of apoptin to induce apoptosis independently of p53 restricted. Nevertheless, HIV-Tat apoptin could be applied for the
and to switch survival pathways into proapoptotic signaling carries an treatment of squamous cell carcinoma or precancerous skin lesions.
enormous pharmacological potential. These remarkable features The efcacy of apoptin for the treatment of squamous cell carcinoma
might be directly employed for the use of apoptin in cancer therapy. has recently been demonstrated in a mouse model using its ectopic
In addition, targeting of apoptin's interaction partners might be an expression in primary oral tumor cells [67]. It might be also envisioned
approach to achieve tumor specicity and to design new antitumor to utilize the selective accumulation of apoptin in the nucleus of
therapies. Several preclinical studies have shown that apoptin cancer cells for drug development. Thus, fragments of apoptin could
improves also the efcacy of traditional therapy, while apoptin itself be coupled to radionuclides emitting low energy radioactivity in order
shows no adverse side effects. induce DNA strand breaks selectively in cancer cells.
Recent studies demonstrate that downregulation of survivin
together with the expression of apoptin signicantly increases the 9. Other tumor-selective killer proteins
percentage of apoptotic cells compared to the effects seen upon
survivin loss or apoptin expression alone [62,63]. Survivin belongs to Beside apoptin, the identication of other proapoptotic proteins
the family of inhibitors of apoptosis proteins (IAP) and is highly with cancer-selective properties raises promises for new drug
expressed in several tumors. These ndings suggest that a combined development. The molecular pathways by which these proteins
strategy using apoptin and a downregulation of survivin could be exert their tumor-selective function are intensively investigated. The
effective in controlling the growth of tumors that are otherwise cytokine TRAIL, a member of the TNF superfamily, induces apoptotic
resistant to traditional chemotherapy. Another study has shown that cell death in a wide variety of tumor cells. Preclinical studies in mice
the combined treatment of tumors with recombinant apoptin and IL- and nonhuman primates demonstrated that TRAIL induces apoptosis
18 induced a strong Th1 response against Lewis lung carcinomas that in human tumors, whereas no cytotoxicity to normal tissues was
was associated with a signicant inhibition of tumor growth [64]. found [68]. The susceptibility of tumor cells to TRAIL and the apparent
Similarly, a combined therapeutic approach with recombinant lack of activity of TRAIL in normal cells have led to the proposed use of
adenovirus expressing apoptin and etoposide showed an additive TRAIL in cancer therapy [69]. Indeed, several phase 1 and 2 studies
cytotoxicity on human osteosarcoma U2OS cells [24,33]. Also, when with TRAIL or TRAIL receptor agonists are in progress in cancer
paclitaxel treatment was combined with apoptin, the survival of p53- patients.
positive, p53-negative and p53-mutant cancer cells was compromised Interestingly, like apoptin, also the viral protein E4orf4 (adenovirus
at a signicantly lower dose of cytotoxic chemotherapy. These type 5 E4 open reading frame 4) induces p53-independent cell death
observations indicate that apoptin in combination with conventional in a wide range of tumor but not normal cells upon ectopic expression
chemotherapeutic agents offers an effective strategy for cancer [70]. In analogy to apoptin, E4orf4 is mainly localized in the nucleus
treatment [24]. and employs the survival kinase c-Src for its apoptosis signaling [71].
The tumor-selective cell death by apoptin has now been demon- Another similarity between E4orf4 and apoptin is the nding that
strated in a great number of tumor cells using various techniques both proteins obviously target the APC/C complex for their tumor-
including adenoviral transfer, transient overexpression or intro- specic induction of apoptosis. This makes the APC/C complex
duction of cell-permeable apoptin constructs. Furthermore, several certainly an attractive target for future anticancer therapeutics. In
groups have studied the effect of apoptin in different mouse modes. addition, a proapoptotic protein with structural similarity to apoptin
While all these studies conrmed a tumor-specic effect, the most has been identied in some isolates of Torque teno virus (TTV) [72,73].
challenging issue is still the development of efcient delivery forms of This protein, named TTV apoptosis-inducing protein (TAIP), triggers
apoptin at the tumor lesion. Noteborn and colleagues developed a p53-independent apoptosis in hepatocellular carcinoma and other
strategy for the use of apoptin in cancer therapy based on adenoviral tumor cells, albeit mostly with less potency than apoptin. Unlike
vectors. A single injection of the apoptin-producing adenovirus in apoptin, TAIP is localized in the cytoplasm of transformed cells.
xenografted hepatomas resulted in a considerably delay in tumor Furthermore, the high frequency of TTV in human cancer biopsies
growth [22]. Furthermore, in order to increase the transduction suggests an indirect tumor growth-stimulatory role of TTV infection
efciency some studies have investigated other viruses. A recent rather than a signicant oncosuppressive function [74].
study, for instance, showed that infection with a replication-decient Another remarkable protein with tumor-specic cytotoxic activity
fowlpox virus containing the apoptin gene resulted in a signicant has been isolated from a specic fraction of human milk and is
induction of apoptosis in hepatoma cells and, moreover, efciently therefore called HAMLET (human -lactalbumin made lethal to tumor
repressed the growth of subcutaneous tumors in mice [65]. cells). The proapoptotic activity of HAMLET was shown to reside in a
Another gene-therapeutic approach to express particular genes of molecular complex consisting of the milk protein -lactalbumin and
interest into the liver is the use of ligand conjugates of the oleic acid, which results in a change of protein conformation [75].
asialoglycoprotein receptor that is specically expressed on hepato- Similar to apoptin, HAMLET is a partly unfolded globule-like complex.
cytes. Systemic administration of the apoptin gene via the asialogly- In cancer cells, HAMLET translocates to nuclei and induces p53-
coprotein receptor into mice bearing hepatocarcinomas resulted in independent cell death of tumor cells, but not normal cells. Recently, it
signicant tumor regression, whereas normal hepatocytes were was shown that treatment with topical HAMLET has a benecial effect
clearly not affected by apoptin gene delivery [66]. In addition, apoptin on the regression of skin papillomas [76].
M. Los et al. / Biochimica et Biophysica Acta 1793 (2009) 13351342 1341

Ribonucleases have also attracted considerable attention as specic [19] Y.H. Zhang, P.J. Abrahams, A.J. van der Eb, M.H. Noteborn, The viral protein Apoptin
induces apoptosis in UV-C-irradiated cells from individuals with various
anticancer agents. Onconase, an amphibian ribonuclease from oocytes hereditary cancer-prone syndromes, Cancer Res. 59 (1999) 30103015.
of the frog Rana pipiens is of particular interest, because it is cytostatic [20] L. Guelen, H. Paterson, J. Gaken, M. Meyers, F. Farzaneh, M. Tavassoli, TAT-apoptin
and cytotoxic to various tumor cells and enhances their sensitivity to a is efciently delivered and induces apoptosis in cancer cells, Oncogene 23 (2004)
11531165.
number of cytotoxic agents [77]. Onconase has demonstrated [21] A. Pietersen, S.J. Erkeland, S.A. Rutjes, S. Philipsen, J.P. Medema, M.H. Noteborn,
signicant efcacy in phase III clinical trials of patients with malignant Continuous apoptin expression in transgenic mice does not interfere with
mesothelioma that failed prior chemotherapy. Onconase's specic lymphocyte development and proliferation, J. Med. Mol. Biol. 2 (2005)
321330.
mechanism of action is not fully understood, but it seems to target gene [22] A.M. Pietersen, M.M. van der Eb, H.J. Rademaker, D.J. van den Wollenberg, M.J.
regulation by microRNAs [77]. In conclusion, further investigations of Rabelink, P.J. Kuppen, J.H. van Dierendonck, H. van Ormondt, D. Masman, C.J. van
apoptin and related proteins with a tumor-specic proapoptotic de Velde, A.J. van der Eb, R.C. Hoeben, M.H. Noteborn, Specic tumor-cell killing
with adenovirus vectors containing the apoptin gene, Gene Ther. 6 (1999)
activity will be important not only for understanding the process of
882892.
carcinogenesis, but might eventually result in new therapeutic options [23] S. Maddika, S.R. Ande, E. Wiechec, L.L. Hansen, S. Wesselborg, M. Los, Akt-
for anticancer treatment. mediated phosphorylation of CDK2 regulates its dual role in cell cycle progression
and apoptosis, J. Cell. Sci. 121 (2008) 979988.
[24] S.J. Olijslagers, Y.H. Zhang, C. Backendorf, M.H. Noteborn, Additive cytotoxic effect
Acknowledgments of apoptin and chemotherapeutic agents paclitaxel and etoposide on human
tumour cells, Basic Clin. Pharmacol. Toxicol. 100 (2007) 127131.
We apologize to those authors whose data were not discussed due [25] S.M. Zhuang, A. Shvarts, H. van Ormondt, A.G. Jochemsen, A.J. van der Eb, M.H.
Noteborn, Apoptin, a protein derived from chicken anemia virus, induces p53-
to space restrictions. M.L., F.E. and K.S.O. acknowledge the support independent apoptosis in human osteosarcoma cells, Cancer Res. 55 (1995)
from Deutsche Forschungsgemeinschaft (SFB 773, GRK 1302) and the 486489.
Deutsche Krebshilfe. [26] M. Los, S. Wesselborg, K. Schulze-Osthoff, The role of caspases in development,
immunity, and apoptotic signal transduction: lessons from knockout mice,
Immunity 10 (1999) 629639.
References [27] C. Schwerk, K. Schulze-Osthoff, Regulation of apoptosis by alternative pre-mRNA
splicing, Mol. Cell 19 (2005) 113.
[1] M.H. Noteborn, G.F. de Boer, D.J. van Roozelaar, C. Karreman, O. Kranenburg, J.G. [28] U. Fischer, R.U. Jnicke, K. Schulze-Osthoff, Many cuts to ruin: a comprehensive
Vos, S.H. Jeurissen, R.C. Hoeben, A. Zantema, G. Koch, et al., Characterization of update of caspase substrates, Cell Death Differ. 10 (2003) 76100.
cloned chicken anemia virus DNA that contains all elements for the infectious [29] A.A. Danen-van Oorschot, A.J. van Der Eb, M.H. Noteborn, The chicken anemia
replication cycle, J. Virol. 65 (1991) 31313139. virus-derived protein apoptin requires activation of caspases for induction of
[2] S. Natesan, J.M. Kataria, K. Dhama, N. Bhardwaj, A. Sylvester, Anti-neoplastic effect apoptosis in human tumor cells, J. Virol. 74 (2000) 70727078.
of chicken anemia virus VP3 protein (apoptin) in Rous sarcoma virus-induced [30] S. Maddika, E.P. Booy, D. Johar, S.B. Gibson, S. Ghavami, M. Los, Cancer-specic
tumours in chicken, J. Gen. Virol. 87 (2006) 29332940. toxicity of apoptin is independent of death receptors but involves the loss of
[3] M.H. Noteborn, C.A. Verschueren, A. Zantema, G. Koch, A.J. van der Eb, mitochondrial membrane potential and the release of mitochondrial cell-death
Identication of the promoter region of chicken anemia virus (CAV) containing mediators by a Nur77-dependent pathway, J. Cell. Sci. 118 (2005) 44854493.
a novel enhancer-like element, Gene 150 (1994) 313318. [31] M. Burek, S. Maddika, C.J. Burek, P.T. Daniel, K. Schulze-Osthoff, M. Los, Apoptin-
[4] M.M. Miller, K.W. Jarosinski, K.A. Schat, Positive and negative regulation of chicken induced cell death is modulated by Bcl-2 family members and is Apaf-1
anemia virus transcription, J. Virol. 79 (2005) 28592868. dependent, Oncogene 25 (2005) 22132222.
[5] M.H. Noteborn, D. Todd, C.A. Verschueren, H.W. de Gauw, W.L. Curran, S. [32] A.A. Danen-Van Oorschot, A.J. van der Eb, M.H. Noteborn, BCL-2 stimulates
Veldkamp, A.J. Douglas, M.S. McNulty, E.A. van der, G. Koch, A single chicken Apoptin-induced apoptosis, Adv. Exp. Med. Biol. 457 (1999) 245249.
anemia virus protein induces apoptosis, J. Virol. 68 (1994) 346351. [33] A.A. Danen-Van Oorschot, Y. Zhang, S.J. Erkeland, D.F. Fischer, A.J. van der Eb, M.H.
[6] K.V. Phenix, B.M. Meehan, D. Todd, M.S. McNulty, Transcriptional analysis and Noteborn, The effect of Bcl-2 on Apoptin in normal vs transformed human cells,
genome expression of chicken anaemia virus, J. Gen. Virol. 75 (1994) 905909. Leukemia 13 (Suppl 1) (1999) S75S77.
[7] S. Maddika, F.J. Mendoza, K. Hauff, C.R. Zamzow, T. Paranjothy, M. Los, Cancer- [34] X. Liu, S. Elojeimy, A.M. El-Zawahry, D.H. Holman, A. Bielawska, J. Bielawski, S.
selective therapy of the future: apoptin and its mechanism of action, Cancer Biol. Rubinchik, G.W. Guo, J.Y. Dong, T. Keane, Y.A. Hannun, M. Tavassoli, J.S. Norris,
Ther. 5 (2006) 1019. Modulation of ceramide metabolism enhances viral protein apoptin's cytotoxicity
[8] M.H. Noteborn, Chicken anemia virus induced apoptosis: underlying molecular in prostate cancer, Molec. Ther. 14 (2006) 637646.
mechanisms, Vet. Microbiol. 98 (2004) 8994. [35] B. Lin, S.K. Kolluri, F. Lin, W. Liu, Y.H. Han, X. Cao, M.I. Dawson, J.C. Reed, X.K. Zhang,
[9] D.W. Heilman, J.G. Teodoro, M.R. Green, Apoptin nucleocytoplasmic shuttling is Conversion of Bcl-2 from protector to killer by interaction with nuclear orphan
required for cell type-specic localization, apoptosis, and recruitment of the receptor Nur77/TR3, Cell 116 (2004) 527540.
anaphase-promoting complex/cyclosome to PML bodies, J. Virol. 80 (2006) [36] D.H. Huo, L.N. Yi, J. Yang, Interaction with Ppil3 leads to the cytoplasmic
75357545. localization of Apoptin in tumor cells, Biochem. Biophys. Res. Commun. 372
[10] I.K.H. Poon, C. Oro, M.M. Dias, J. Zhang, D.A. Jans, Apoptin nuclear accumulation is (2008) 1418.
modulated by a CRM1-recognized nuclear export signal that is active in normal [37] C.M. Cheng, S.P. Huang, Y.F. Chang, W.Y. Chung, C.Y. Yuo, The viral death protein
but not in tumor cells, Cancer Res. 65 (2005) 70597064. Apoptin interacts with Hippi, the protein interactor of Huntingtin-interacting
[11] K. Kamada, A. Kuroishi, T. Kamahora, P. Kabat, S. Yamaguchi, S. Hino, Spliced protein 1, Biochem. Biophys. Res. Commun. 305 (2003) 359364.
mRNAs detected during the life cycle of chicken anemia virus, J. Gen. Virol. 87 [38] G.J. Sun, X. Tong, Y. Dong, Z.Z. Mei, Z.X. Sun, Identication of a protein interacting
(2006) 22272233. with apoptin from human leucocyte cDNA library by using yeast two-hybrid
[12] A.A. Danen-Van Oorschot, Y.H. Zhang, S.R. Leliveld, J.L. Rohn, M.C. Seelen, M.W. screening, ShengWu HuaXue Yu ShengWu WuLi XueBao (Shanghai) 34 (2002)
Bolk, A. Van Zon, S.J. Erkeland, J.P. Abrahams, D. Mumberg, M.H. Noteborn, 369372.
Importance of nuclear localization of apoptin for tumor-specic induction of [39] J.G. Teodoro, D.W. Heilman, A.E. Parker, M.R. Green, The viral protein Apoptin
apoptosis, J. Biol. Chem. 278 (2003) 2772927736. associates with the anaphase-promoting complex to induce G2/M arrest and
[13] H.V. Kuusisto, K.M. Wagstaff, G. Alvisi, D.A. Jans, The C-terminus of apoptin apoptosis in the absence of p53, Genes Dev. 18 (2004) 19521957.
represents a unique tumor cell-enhanced nuclear targeting module, Int. J. Cancer [40] A.A. Danen-van Oorschot, P. Voskamp, M.C. Seelen, M.H. van Miltenburg, M.W.
(2008) 29652969. Bolk, S.W. Tait, J.G. Boesen-de Cock, J.L. Rohn, J. Borst, M.H. Noteborn, Human
[14] C. Backendorf, A.E. Visser, A.G. de Boer, R. Zimmerman, M. Visser, P. Voskamp, death effector domain-associated factor interacts with the viral apoptosis agonist
Y.H. Zhang, M. Noteborn, Apoptin: therapeutic potential of an early sensor Apoptin and exerts tumor-preferential cell killing, Cell Death Differ. 11 (2004)
of carcinogenic transformation, Annu. Rev. Pharmacol. Toxicol. 48 (2008) 564573.
143169. [41] K. Janssen, T.G. Hofmann, D.A. Jans, R.T. Hay, K. Schulze-Osthoff, U. Fischer, Apoptin
[15] S.R. Leliveld, Y.H. Zhang, J.L. Rohn, M.H. Noteborn, J.P. Abrahams, Apoptin induces is modied by SUMO conjugation and targeted to promyelocytic leukemia protein
tumor-specic apoptosis as a globular multimer, J. Biol. Chem. 278 (2003) nuclear bodies, Oncogene 26 (2007) 15571566.
90429051. [42] E. Krieghoff-Henning, T.G. Hofmann, Role of nuclear bodies in apoptosis signalling,
[16] S.R. Leliveld, R.T. Dame, M.A. Mommaas, H.K. Koerten, C. Wyman, A.A. Danen-van Biochim. Biophys. Acta 1783 (2008) 21852194.
Oorschot, J.L. Rohn, M.H. Noteborn, J.P. Abrahams, Apoptin protein multimers form [43] S. Maddika, E. Wiechec, S.R. Ande, K. Poon, U. Fischer, S. Wesselborg, D.A. Jans, K.
distinct higher-order nucleoprotein complexes with DNA, Nucleic Acids Res. 31 Schulze-Osthoff, M. Los, Interaction with PI3-kinase contributes to the cytotoxic
(2003) 48054813. activity of apoptin, Oncogene 27 (2008) 30603065.
[17] M. Tavassoli, L. Guelen, B.A. Luxon, J. Gken, Apoptin: specic killer of tumor cells? [44] S. Maddika, G.H. Bay, T.J. Kroczak, S.R. Ande, S. Maddika, E. Wiechec, S.B. Gibson, M.
Apoptosis 10 (2005) 717724. Los, Akt is transferred to the nucleus of cells treated with apoptin, and it
[18] A.A. Danen-Van Oorschot, D.F. Fischer, J.M. Grimbergen, B. Klein, S. Zhuang, J.H. participates in apoptin-induced cell death, Cell Prolif. 40 (2007) 835848.
Falkenburg, C. Backendorf, P.H. Quax, A.J. Van der Eb, M.H. Noteborn, Apoptin [45] T. Aki, K. Yamaguchi, T. Fujimiya, Y. Mizukami, Phosphoinositide 3-kinase
induces apoptosis in human transformed and malignant cells but not in normal accelerates autophagic cell death during glucose deprivation in the rat
cells, Proc. Natl. Acad. Sci. U. S. A. 94 (1997) 58435847. cardiomyocyte-derived cell line H9c2, Oncogene 22 (2003) 85298535.
1342 M. Los et al. / Biochimica et Biophysica Acta 1793 (2009) 13351342

[46] B. Lu, L. Wang, C. Stehlik, D. Medan, C. Huang, S. Hu, F. Chen, X. Shi, Y. Rojanasakul, [61] G. Gil-Gomez, A. Berns, H.J. Brady, A link between cell cycle and cell death: Bax and
Phosphatidylinositol 3-kinase/Akt positively regulates Fas (CD95)-mediated Bcl-2 modulate Cdk2 activation during thymocyte apoptosis, EMBO J. 17 (1998)
apoptosis in epidermal Cl41 cells, J. Immunol. 176 (2006) 67856793. 72097218.
[47] D. Nimbalkar, M.K. Henry, F.W. Quelle, Cytokine activation of phosphoinositide 3- [62] Q. Liu, H. Fu, R. Xing, Y. Tie, J. Zhu, Z. Sun, X. Zheng, Survivin knockdown combined
kinase sensitizes hematopoietic cells to cisplatin-induced death, Cancer Res. 63 with apoptin overexpression inhibits cell growth signicantly, Cancer Biol. Ther. 7
(2003) 10341039. (2008) 10531060.
[48] S. Shack, X.T. Wang, G.C. Kokkonen, M. Gorospe, D.L. Longo, N.J. Holbrook, [63] S. Panigrahi, T. Klonisch, M. Los, The art of killing: double stroke with apoptin and
Caveolin-induced activation of the phosphatidylinositol 3-kinase/Akt pathway survivin as a novel approach in cancer therapy, Cancer Biol. Ther. 7 (2008) 10611062.
increases arsenite cytotoxicity, Mol. Cell. Biol. 23 (2003) 24072414. [64] H. Lian, N. Jin, X. Li, Z. Mi, J. Zhang, L. Sun, X. Li, H. Zheng, P. Li, Induction of an
[49] S. Maddika, S. Panigrahi, E. Wiechec, S. Wesselborg, U. Fischer, K. Schulze-Osthoff, effective anti-tumor immune response and tumor regression by combined
M. Los, Unscheduled Akt-triggered activation of CDK2 as a key effector mechanism administration of IL-18 and Apoptin, Cancer Immunol. Immunother. 56 (2007)
of apoptin's anticancer toxicity, Mol. Cell. Biol. 29 (2009) 12351248. 181192.
[50] H. Lei, P.J. Furlong, J.H. Ra, D. Mullins, R. Cantor, D.L. Fraker, F.R. Spitz, AKT [65] X. Li, N. Jin, Z. Mi, H. Lian, L. Sun, X. Li, H. Zheng, Antitumor effects of a recombinant
activation and response to interferon-beta in human cancer cells, Cancer Biol. fowlpox virus expressing Apoptin in vivo and in vitro, Int. J. Cancer 119 (2006)
Ther. 4 (2005) 709715. 29482957.
[51] D. Tang, H. Okada, J. Ruland, L. Liu, V. Stambolic, T.W. Mak, A.J. Ingram, Akt is [66] D.J. Peng, J. Sun, J.Y.Z. Wang, J. Tian, Y.H. Zhang, M.H. Noteborn, S. Qu, Inhibition of
activated in response to an apoptotic signal, J. Biol. Chem. 276 (2001) 3046130466. hepatocarcinoma by systemic delivery of Apoptin gene via the hepatic
[52] A.G. van Gorp, K.M. Pomeranz, K.U. Birkenkamp, R.C. Hui, E.W. Lam, P.J. Coffer, asialoglycoprotein receptor, Cancer Gene Ther. 14 (2007) 6673.
Chronic protein kinaseB , (PKB/c-akt) activation leads to apoptosis induced by [67] R.A. Schoop, R.J. Baatenburg de Jong, M.H. Noteborn, Apoptin induces apoptosis in
oxidative stress-mediated Foxo3a transcriptional up-regulation, Cancer Res. 66 an oral cancer mouse model, Cancer Biol. Ther. 7 (2008) 13681373.
(2006) 1076010769. [68] J. Held, K. Schulze-Osthoff, Potential and caveats of TRAIL in cancer therapy, Drug
[53] V. Nogueira, Y. Park, C.C. Chen, P.Z. Xu, M.L. Chen, I. Tonic, T. Unterman, N. Hay, Akt Resist Updat. 4 (2001) 243252.
determines replicative senescence and oxidative or oncogenic premature [69] R.W. Johnstone, A.J. Frew, M.J. Smyth, The TRAIL apoptotic pathway in cancer
senescence and sensitizes cells to oxidative apoptosis, Cancer Cell 14 (2008) onset, progression and therapy, Nat. Rev., Cancer 8 (2008) 782798.
458470. [70] R. Shtrichman, R. Sharf, H. Barr, T. Dobner, T. Kleinberger, Induction of apoptosis by
[54] M. Los, S. Maddika, B. Erb, K. Schulze-Osthoff, Switching Akt: from survival adenovirus E4orf4 protein is specic to transformed cells and requires an
signaling to deadly response, BioEssays 31 (2009) 492495. interaction with protein phosphatase 2A, Proc. Natl. Acad. Sci. U. S. A. 96 (1999)
[55] I.K. Poon, C. Oro, M.M. Dias, J.P. Zhang, D.A. Jans, A tumor cell-specic nuclear 1008010085.
targeting signal within chicken anemia virus VP3/Apoptin, J. Virol. 79 (2005) [71] C. Champagne, M.C. Landry, M.C. Gingras, J.N. Lavoie, Activation of adenovirus type
13391341. 2 early region 4 ORF4 cytoplasmic death function by direct binding to Src kinase
[56] J.L. Rohn, M.H. Noteborn, The viral death effector Apoptin reveals tumor-specic domain, J. Biol. Chem. 279 (2004) 2590525915.
processes, Apoptosis 9 (2004) 315322. [72] M.H. de Smit, M.H. Noteborn, Apoptosis-inducing proteins in chicken anemia virus
[57] J.L. Rohn, Y.-H. Zhang, R.I.J.M. Aalbers, N. Otto, J. den Hertog, N.V. Henriquez, C.J.H. and TT virus, Curr. Top. Microbiol. Immunol. 331 (2009) 131149.
van de Velde, P.J.K. Kuppen, D. Mumberg, P. Donner, M.H.M. Noteborn, A tumor- [73] S. Hino, A.A. Prasetyo, Relationship of Torque teno virus to chicken anemia virus,
specic kinase activity regulates the viral death protein apoptin, J. Biol. Chem. 277 Curr. Top. Microbiol. Immunol. 331 (2009) 117130.
(2002) 5082050827. [74] H. zur Hausen, E.M. de Villiers, TT viruses: oncogenic or tumor-suppressive
[58] Y.H. Lee, C.M. Cheng, Y.F. Chang, T.Y. Wang, C.Y. Yuo, Apoptin T108 phosphorylation properties? Curr. Top. Microbiol. Immunol. 331 (2009) 109116.
is not required for its tumor-specic nuclear localization but partially affects its [75] O. Hallgren, S. Aits, P. Brest, L. Gustafsson, A.K. Mossberg, B. Wullt, C. Svanborg,
apoptotic activity, Biochem. Biophys. Res. Commun. 354 (2007) 391395. Apoptosis and tumor cell death in response to HAMLET (human alpha-
[59] S.R. Leliveld, R.T. Dame, J.L. Rohn, M.H. Noteborn, J.P. Abrahams, Apoptin's lactalbumin made lethal to tumor cells), Adv. Exp. Med. Biol. 606 (2008) 217240.
functional N- and C-termini independently bind DNA, FEBS Lett. 557 (2004) [76] L. Gustafsson, I. Leijonhufvud, A. Aronsson, A.K. Mossberg, C. Svanborg, Treatment
155158. of skin papillomas with topical alpha-lactalbumin-oleic acid, N. Engl. J. Med. 350
[60] S. Nakayama, Y. Torikoshi, T. Takahashi, T. Yoshida, T. Sudo, T. Matsushima, Y. (2004) 26632672.
Kawasaki, A. Katayama, K. Gohda, G.N. Hortobagyi, S. Noguchi, T. Sakai, H. Ishihara, [77] H. Zhao, B. Ardelt, W. Ardelt, K. Shogen, Z. Darzynkiewicz, The cytotoxic
N.T. Ueno, Prediction of paclitaxel sensitivity by CDK1 and CDK2 activity in human ribonuclease onconase targets RNA interference (siRNA), Cell Cycle 7 (2008)
breast cancer cells, Breast Cancer Res. 11 (2009) R12. 32583261.

Das könnte Ihnen auch gefallen