evading apoptosis in cancer

14

Click here to load reader

Upload: manabu

Post on 12-Dec-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Evading apoptosis in cancer

TICB-985; No. of Pages 14

Evading apoptosis in cancerKaleigh Fernald1 and Manabu Kurokawa1,2

1 Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA2 Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA

Review

Carcinogenesis is a mechanistically complex and vari-able process with a plethora of underlying geneticcauses. Cancer development comprises a multitude ofsteps that occur progressively starting with initial drivermutations leading to tumorigenesis and, ultimately,metastasis. During these transitions, cancer cells accu-mulate a series of genetic alterations that confer on thecells an unwarranted survival and proliferative advan-tage. During the course of development, however, can-cer cells also encounter a physiologically ubiquitouscellular program that aims to eliminate damaged orabnormal cells: apoptosis. Thus, it is essential that cancercells acquire instruments to circumvent programmed celldeath. Here we discuss emerging evidence indicating howcancer cells adopt various strategies to override apopto-sis, including amplifying the antiapoptotic machinery,downregulating the proapoptotic program, or both.

Regulation of apoptosis in normal and cancer cellsApoptosis is a cellular suicide program that organismshave evolved to eliminate unnecessary or unhealthy cellsfrom the body in the course of development or followingcellular stress. It involves a series of cellular events thatultimately leads to activation of a family of cysteine pro-teases called caspases. In response to various apoptoticstimuli, ‘initiator’ caspases (caspase-2, -8, -9, or -10) areactivated. Initiator caspases, in turn, cleave and activatethe zymogenic forms of ‘executioner’ caspases (e.g., cas-pase-3 or -7), resulting in the proteolytic cleavage of specificcellular substrates and, consequently, cell death (Figure 1).In this regard, the cleavage (activation) of executionercaspases is a hallmark of apoptosis. There are two routesto apoptosis: extrinsic and intrinsic. In the extrinsic path-way, initiator caspase-8 and -10 are activated through theformation of a death-inducing signal complex (DISC) inresponse to the engagement of extracellular ligands [e.g.,Fas, tumor necrosis factor (TNF)] by cell surface receptors(Figure 1). In the intrinsic pathway of apoptosis, mitochon-drial outer membrane permeabilization (MOMP) is in-volved; MOMP triggers the release of a group ofproapoptotic proteins, including cytochrome c and secondmitochondria-derived activator of caspases (SMAC), fromthe mitochondrial intermembrane space to the cytoplasm(Figure 1). In the cytoplasm, cytochrome c binds to theadaptor protein apoptotic protease-activating factor 1(APAF-1) to form a caspase-9-activating complex called

0962-8924/$ – see front matter

� 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2013.07.006

Corresponding author: Kurokawa, M. ([email protected]).Keywords: BH3; caspase; MOMP; phosphorylation; ubiquitination.

the apoptosome (Figure 1). SMAC augments cytochromec-induced caspase activation by binding and neutralizingX-linked inhibitor of apoptosis protein (XIAP), an inhibitorof caspase-3, -7, and -9 (Figure 1).

The balance between pro- and antiapoptotic B cell lym-phoma 2 (BCL-2) family proteins is a fundamental determi-nant for the initiation of MOMP. The BCL-2 family proteinsare classified based on the presence of shared blocks ofsequence homology, termed BCL-2 homology (BH). Theproapoptotic BCL-2 family members, which promoteMOMP, include BH3-only proteins [e.g., Bcl2-interactingmediator of cell death (BIM), BH3-interacting domain deathagonist (BID), and Bcl2-associated agonist of cell death(BAD), which share only a single block, the BH3 domain,of BCL-2 homology] and multi-BH domain proteins [e.g.,Bcl2-associated protein X (BAX) and Bcl2 antagonist/killer(BAK), which share BH1–BH3 domains]. Following cytotox-ic or genotoxic stress, BH3-only proteins are activated andpromote oligomerization of BAX (or BAK), resulting inMOMP, whereas prosurvival members [e.g., BCL-2, B celllymphoma extra large (BCL-XL), and induced myeloid leu-kemia cell differentiation protein (MCL-1), which contain allfour BH domains] counteract this process by sequesteringproapoptotic family members (Figure 1). Importantly, theinteractions between pro- and antiapoptotic BCL-2 familyproteins are determined with specific binding specificities[1,2]; for instance, BIM interacts with all six antiapoptoticBCL-2 family proteins as well as BAX and BAK [3], whereasphorbol-12-myristate-13-acetate-induced protein 1 (NOXA)binds to MCL-1 and BFL-1/A1 only.

Unlike normal cells, cancer cells are under constantstress, including oncogenic stress, genomic instability,and cellular hypoxia. In response to such apoptotic stimuli,the intrinsic pathway of apoptosis would normally beactivated. Yet cancer cells can often avoid this cellularresponse by disabling the apoptotic pathways. Notably,mouse genetic models have shown that genetic inactiva-tion of a BH3-only protein or a caspase can not only lead toresistance to certain proapoptotic stimuli but also acceler-ate tumor formation in mice [4–6]. Moreover, forced ex-pression of antiapoptotic BCL-2 family proteins cansignificantly augment tumor development induced by anoncogene, such as MYC, although overexpression of anantiapoptotic BCL-2 family protein alone may not resultin tumor formation similar to that seen with an oncogenealone [7–10]. Thus, it is strongly suggested that inhibitionof apoptosis plays a critical role in cancer cell survival andtumor development.

Cancer cells can modulate apoptotic pathways tran-scriptionally, translationally, and post-translationally. In

Trends in Cell Biology xx (2013) 1–14 1

Page 2: Evading apoptosis in cancer

BIMPUMABADNOXABMFHRKBIK

MOMP

SMAC

XIAP

Apoptosis

Apoptosome

PIDDosomeBCL-2BCL-XLMCL-1BCL-WBFL-1/A1BCL-B

BAX, BAK, BOK

Caspase-2

Caspase-8, -10

Cytochrome c

APAF-1

Caspase-9

Caspase-3, -7

BCL-2BCL-XLMCL-1BCL-WBFL-1/A1BCL-B

BIMPUMABADNOXABMFHRKBIK

BID → tBIDBID → tBID

DISC

An�apopto�c BCL-2 family

BH3-only proteins

BAX, BAK, BOK

Mul�-BH proteins

C

Apoptosome

pp

TRENDS in Cell Biology

Figure 1. Apoptosis is triggered in response to internal or external stimuli. The

intrinsic pathway to apoptosis is initiated by activation of BH3-only protein.

Among the BH3-only proteins, expression of which is often transcriptionally

induced following an apoptotic stimulus, BID is unique in that it is activated by

caspase cleavage (by caspase-2 or -8/10); cleaved BID is termed tBID. BH3 proteins

trigger the activation of multi-BH domain proteins by direct binding or by

inhibition of antiapoptotic BCL-2 family proteins. Once activated, multi-BH domain

proteins form oligomers that induce MOMP. MOMP prompts the proapoptotic

proteins SMAC and cytochrome c to be released to the cytoplasm, where

cytochrome c induces formation of the apoptosome. The initiator caspase-9

activated within the apoptosome initiates the activation of executioner caspase-3

and -7, resulting in apoptosis. Of note, caspase-2 is activated via formation of the

PIDDosome, a complex comprising the adaptor PIDD and RAIDD. However, despite

identification of this complex, the precise mechanism of caspase-2 activation

remains elusive. In addition, only a few cellular substrates of caspase-2, including

BID, have been identified. Colored triangles denote that the indicated gene or gene

product may be transcriptionally (blue), translationally (red), or post-translationally

(green) up- or downregulated in cancer cells (see text and Table 1).

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

some cases, cancer cells may escape apoptosis by increasingexpression of antiapoptotic genes or by decreasing expres-sion of proapoptotic genes. Alternatively, they may inhibitapoptosis by stabilizing or destabilizing anti- or proapopto-tic proteins, respectively. Moreover, cancer cells may alsoprevent apoptosis by changing the functions of anti- orproapoptotic proteins through post-translational modifica-tions such as phosphorylation. Importantly, these mecha-nisms are not mutually exclusive and cancer cells mayemploy one or multiple mechanisms to evade apoptosis.

2

In this review, we highlight the core antiapoptotic machin-ery that cancer cells may employ for survival, focusing ourdiscussion on transcriptional, translational, and post-trans-lational modifications to evade apoptosis (note that recentstudies suggest that cancer metabolism can also modulatethis machinery to protect cells from apoptosis [11,12]; how-ever, this is outside the scope of this discussion).

Transcriptional/translational regulationGenomic and epigenomic abnormalities are a characteris-tic of cancer cells. This includes gene copy number ampli-fication, gene deletion, gene silencing by DNA methylation,and activation (or inactivation) of transcription factors thathave an impact on the expression of apoptotic regulators[13]. In addition, miRNAs negatively control gene expres-sion by targeting the 30 untranslated region (30-UTR) ofmRNAs, although they could function as tumor suppres-sors or oncogenes, depending on the target messengers[14]. Thus, transcriptional and translational alterationscan be a means for cancer cells to directly raise the thresh-old for apoptotic induction at gene expression levels. In thissection, we discuss how cancer cells may increase or blockthe expression of anti- or proapoptotic gene products,respectively (see Table 1 for loss or amplification of apo-ptotic genes identified in cancer tissues).

Inducing antiapoptotic protein expression

To overcome stress signals, cancer cells frequently over-express antiapoptotic proteins, especially antiapoptoticBCL-2 family proteins. Cancer cells prevent MOMP byamplification of antiapoptotic BCL-2 family proteins. Con-sequently, inhibition of one or multiple antiapoptotic BCL-2 family proteins causes apoptosis in cancer cells but not inhealthy normal cells – it is often said that cancer cells areprimed for death or addicted to antiapoptotic BCL-2 familyproteins. Overexpression of antiapoptotic BCL-2 familyproteins is often associated with poor prognosis, recur-rence, and resistance to cancer therapeutics [15–17]. TheBCL-2 gene was originally found in B cell follicular lym-phoma, where genetic translocation resulted in constitu-tive expression of BCL-2 [18–20] (Table 1). Amplification ofthe BCL-XL gene has also been reported in various can-cers, including lung and giant-cell tumor of bone [21–23](Table 1). Likewise, frequent amplification of the MCL-1gene has been reported in breast and lung cancer samples[23,24] (Table 1).

In normal cells, MCL-1 transcription is induced uponcytokine and growth factor stimulation [25,26], whereas itssynthesis is inhibited in response to various cellular stres-ses, including DNA damage [27–29]. In particular, it wasshown that the transcription factor E2F1, known to controlproliferation and apoptosis, could repress MCL-1 geneexpression by binding directly to the MCL-1 promoter[30], resulting in increased apoptosis. Alternatively, thetranscription factors cAMP response element-binding pro-tein (CREB) and signal transducer and activator of tran-scription 3 (STAT3), which are known to activate genesresponding to prosurvival stimuli, can transactivate theMCL-1 gene [25,27]. Importantly, whereas these signalingpathways operate to maintain cellular homeostasis innormal cells, they are often deregulated in cancer cells.

Page 3: Evading apoptosis in cancer

Table 1. Cancer and apoptotic regulators

Loss or amplification

of the apoptotic geneaKinase

(phosphatase)

Siteb Outcome of

phosphorylation

Cancer connection

to phosphorylationc

Caspase-2 CK2 Ser157 Suppression of caspase activity

[122]

TRAIL resistance

Esophageal cancer cells

Colon cancer cells

Glioma cells

CaMKII

(PP1)

Ser164 Suppression of caspase activity

[123]

Metabolically regulated

CDK1/CYCLIN B1 Ser340 Suppression of caspase activity

[124]

Suppressing mitotic cell

death

Caspase-3 PKCd ND Enhancement of caspase activity

[125]

p38 MAPK

(PP2A)

Ser150 Suppression of caspase activity

[126]

Caspase-6 ARK5 Ser257 Suppression of caspase activity

[197]

FasL resistance

Colorectal cancer cells

Caspase-7 PAK2 Ser30

Thr173 Ser239

Suppression of caspase activity

[127]

Breast cancer cells

Caspase-8 ND Tyr310 SHP1 binding required for

dephosphorylation of Tyr397

and Tyr465

[128]

RSK2 Thr263 Ubiquitination and degradation

[198]

FasL resistance

p38 MAPK Ser364 Suppression of caspase activity

[126]

CDK1/CYCLIN B1 Ser387 Suppression of caspase activity

[199]

FasL resistance

Breast cancer

Colon cancer cells

SRC

LYN

(SHP1)

Tyr397

(=Tyr380)

Suppression of caspase activity

[128,129]

Colon cancer

LYN

(SHP1)

Tyr465 Suppression of caspase activity

[128]

Caspase-9 PKA Ser99

Ser183

Ser195

No effects

[130]

CDK1

DYRK1A

ERK1/2

p38 MAPK

(PP1a)

Thr125 Suppression of caspase activity

[131–136]

Suppressing mitotic cell

death

PKCz Ser144 Suppression of caspase activity

[137]

c-ABL Tyr153 Promoting caspase activation

[138]

Promoting DNA damage-

induced apoptosis

AKT Ser196

(human specific)d

Suppression of caspase activity

[139]

Prostate cancer cells

Colon cancer cells

BID CK1

CK2

Thr58

Ser61

Ser64

(mouse)

Prevents cleavage by caspase-8

[121]

ATM

ATR

Ser61

Ser64

Ser78

(mouse)

S-phase arrest in response to

DNA damage

[140]

BIMe <Loss>

Chronic myeloid leukemia

and non-small cell lung

cancer [60]

Cutaneous T cell lymphoma

[141]

ERK Ser55

Ser65

Ser73

Ser100

(mouse)

Inactivation

[4,116]

Under the control of

cytokine interleukin (IL)-3

JNK

p38 MAPK

Ser65

(mouse, rat)

Activation

[114,115,117]

ERK1/2

JNK

Ser69 Ubiquitination and degradation

by promoting phosphorylation

at Ser93/Ser94/Ser98 by RSK

[99,101,142]

Non-small cell lung

cancer cells

Chronic myeloid

leukemia cells

T cell acute lymphoblastic

leukemia cells

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

3

Page 4: Evading apoptosis in cancer

Table 1 (Continued )

Loss or amplification

of the apoptotic geneaKinase

(phosphatase)

Siteb Outcome of

phosphorylation

Cancer connection

to phosphorylationc

PKA Ser83

(mouse)

Prevents degradation

[143]

RSK1/2 Ser93

Ser94

Ser98

SCFb-TrCP-mediated

ubiquitination and degradation

following phosphorylation at

Ser69

[101]

Non-small cell lung

cancer cells

JNK Thr112

(mouse)

Activation

[4]

PUMA <Loss>

Breast, hepatocellular, non-

small cell lung,

medulloblastoma, ovarian,

renal cancers [23]

IKK Ser10 Ubiquitination and degradation

[144,145]

Under the control of

cytokine IL-3

BAD IKK Ser26

(mouse)

Inactivation

[146]

TNFa resistance

PAK1 Ser111

(mouse)

Inactivation

[147]

Malignant peripheral

nerve sheath tumor cells

Lung cancer cells

PAK1

PAK4

PAK5

PKA

PKCi

RAF1

RSK1

Ser112

(mouse)

Inactivation

[147–155]

Malignant peripheral

nerve sheath tumor cells

Lung cancer cells

Under the control of

cytokine IL-3

AKT

PKCi

p70S6K

Ser136

(mouse)

Inactivation

[112,155,156]

Lung cancer cells

Under the control of

insulin-like growth factor 1

PKA

PKCi

RSK1

Ser155

(mouse)

Inactivation

[155,157,158]

Lung cancer cells

NOXA CDK5 Ser13 Inactivation

[159]

Metabolically regulated

T cell leukemia cells

BMF JNK

ERK2

Ser74 Release BMF from dynein light

chain complexes (homologous

to BIML Thr56)

[160,161]

ERK2 Ser77 Inactivation

[161]

Melanoma cells

BIK ERK1/2 Thr124 Ubiquitination and degradation

[162]

Lung cancer cells

Colon cancer cells

BAX <Loss>

Colon cancer

[45]

PKA Ser60 Activation

[163]

GSK3 Ser163 Activation [163,164]

ERK1/2 Thr167 Inactivation via interaction with

PIN1

[165]

AKT

PKCz

(PP2A)

Ser184 Inactivation

[166–168]

Lung cancer cells

JNK

p38 MAPK

ND Activation

[169,170]

Hepatoma cells

BAK Kinase unidentified

(PTPN2)

(PTPN5)

(PTPN23)

Tyr108 Inactivation

[171]

Fibrosarcoma cells

Colon cancer cells

Kinase unidentified

(PP2A)

Ser117 Inactivation

[172]

Colon cancer cells

BCL-XL <Amplification>

Non-small cell lung cancer

Pancreas cancer [21]

Giant-cell tumor of bone

[22]

Breast cancer

Ovarian cancer

[23]

PLK3 Ser49 Cell cycle (G2) arrest

[173]

CDK1/CYCLIN B

PLK1

JNK2

Ser62 Cell cycle (G2) arrest

Inactivation

[174,175]

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

4

Page 5: Evading apoptosis in cancer

Table 1 (Continued )

Loss or amplification

of the apoptotic geneaKinase

(phosphatase)

Siteb Outcome of

phosphorylation

Cancer connection

to phosphorylationc

BCL-2 <Amplification>

B cell follicular lymphoma

[18–20]

ERK1/2

PKC

Ser70 Activation

[176,177]

Under the control of

cytokine IL-3

JNK1 Ser70 Inactivation at G2/M phase

[178]

ERK1/2

(MKP-3)

Thr 56

Thr 74

Ser 87

Protects against TNF-induced

degradation

[179,180]

CDK1/CYCLIN B ND Inactivation

[174]

MCL-1 <Amplification>

Lung cancer [23,24]

Breast cancer

Dedifferentiated

liposarcoma, esophageal

adenocarcinoma,

hepatocellular, melanoma,

ovarian, prostate, renal

[23]

CDK1

CDK2

JNK1

Ser64 Enhances binding to BIM,

NOXA, BAK

[181]

TRAIL resistance

Cholangiocarcinoma

cells

CDK1/CYCLIN B Thr92 APC/CCDC20-mediated

ubiquitination and degradation

[89]

JNK

p38 MAPK

CK2

Ser121 SCFFBW7-mediated

ubiquitination and degradation

[91]

Ovarian cancer

Non-small-cell lung

cancer

GSK3

JNK

p38 MAPK

CK2

Ser155

Ser159

Thr163

Ubiquitination and degradation

mediated by TRIM17, SCFb-TrCP,

and SCFFBW7

Inhibits USP9X binding

[81–85,90,91,97]

Breast cancer cells

Ovarian cancer

Non-small-cell lung

cancer

T cell acute

lymphoblastic leukemia

Follicular lymphomas

Diffuse large B cell

lymphomas

Multiple myeloma

Under the control of

cytokine IL-3

ERK1/2 Thr163 Prevents degradation

[86,87]

Hepatoma cells

Burkitt lymphoma cells

APAF-1 <Loss>

Melanoma

[68–70],

Colorectal cancer [71,72],

Gastric cancer [73]

Bladder cancer [74,75]

<Amplification>

Glioblastoma/

Medulloblastoma/

Astrocytoma

[182]

RSK1/2 Ser268 Inhibition

[118]

Prostate cancer cells

XIAP <Amplification>

Cervical cancer

[38]

Acute myeloid leukemia

[183]

Squamous cell cancer

[184]

Rectal cancer

[185]

AKT Ser87 Prevents ubiquitination and

degradation

[186]

Ovarian cancer cells

TBK1

IKKeSer430 Autoubiquitination and

degradation

[187]

aConfirmed at copy number, mRNA, or protein level in cancer tissues.

bHuman sites unless specified.

cDemonstrated or suggested in cancer cell lines or cancer tissues.

dNot conserved in other species.

eBIMEL.

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

In particular, because the phosphatidylinositol 3-kinase(PI3K)/AKT pathway is frequently hyperactivated in can-cer cells by mutations and other mechanisms, transcrip-tional regulation of this pathway, such as the activation ofCREB or STAT3, could promote MCL-1 expression incertain cancer cells (Figure 2). In this regard, it was also

shown that MCL-1 mRNA translation could be facilitatedby mammalian target of rapamycin complex 1 (mTORC1),a downstream target of PI3K/AKT signaling, in a mouselymphoma model (Figure 2) [31].

miRNAs also play a role in the regulation of antiapop-totic proteins. miR-15 and miR-16 inhibit BCL-2 mRNA

5

Page 6: Evading apoptosis in cancer

Oncogenic kinases(e.g.,HER2, EGFR, BCR-ABL)

MEKPI3K

ERKAKT

BIM

FOXOs

mTORC1STAT3CREB

MCL-1

Transcrip�ontransla�on

Protein stability

PUMA

Transcrip�on

Transcrip�onProtein instabilityProtein func�ons

TKI

GSK3

ERKAKT

BIM

FOXOs

CL-1

Protein stability

PUMA

Transc

Transcrip�onProtein instabi lityProtein f unc�on sSK3 T crip�on

Protein stability

TRENDS in Cell Biology

Figure 2. Oncogenic kinase signaling regulates MCL-1, BIM, and PUMA. Oncogenic kinases regulate BIM, PUMA, and MCL-1 by changing gene expression, protein stability,

and protein functions by activating two major prosurvival pathways: AKT and ERK. Prosurvival and proapoptotic pathways are denoted in blue and red, respectively.

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

translation [32]. Likewise, miR-29b, miR-101, and miR-193a-3p suppress MCL-1 mRNA translation, making cellssusceptible to apoptosis [33–35] – note that the role of miR-29b in cell death may be complex because it also targetsseveral BH3-only proteins, inhibiting apoptosis in neu-rons [36]. Expression of miR-15 and miR-16 is reduced inchronic lymphocytic leukemia [32], whereas expression ofmiR-101 is diminished in hepatocellular carcinoma [35],suggesting a possible mechanism to evade apoptosis bythese cancers. Lastly, two recent studies have shown thatexpression of miR-7 and miR-24, which inhibit XIAPmRNA, is suppressed in various cancer cells includingcervical and lung cancer cells, resulting in elevated XIAPprotein levels and blocked caspase activation [37,38]. Itwould be interesting to fully investigate whether expres-sion of these miRNAs is altered in various cancer tissuesand whether their expression patterns are associated withprognostic outcomes.

Suppressing proapoptotic protein expression

In normal cells, genotoxic and cytotoxic stress can induceexpression of proapoptotic genes, particularly some of theBH3-only proteins that trigger apoptosis. In cancer cells,this mechanism is often nullified by mutation and silencingof key apoptotic genes.

In normal cells, p53 is a critical transcription factor thatmodulates the expression of an array of genes known totrigger apoptosis following various cellular stresses. Giv-en the fact that TP53 encoding p53 is the most frequentlymutated, or inactivated, tumor suppressor gene in cancer(�50%) [39,40], apoptotic pathways induced by this gene,including BAX, p53 upregulated modulator of apoptosis(PUMA), NOXA, and APAF-1, may be widely affected incancer cells [41–44] – note that frameshift mutations inthe BAX gene have also been reported in colon cancer,which is likely to be independently of p53 [45] (Table 1).Interestingly, p53 could also activate BAX (and BAK) in a

6

transcription-independent manner in response to DNAdamage and metabolic changes [46–48]. Thus, loss ofp53 in cancer cells will elevate the threshold for MOMPby limiting BAX expression and reducing the likelihood ofBAX oligomerization.

Transcription of the BH3-only protein BIM is inducedupon growth factor withdrawal and other apoptotic stimuli[5,49]. Various transcription factors have been implicatedin BIM expression, including FOXOs [49–51], RUNX3 [52],AP-1 [53], and E2F1 [54]. Among the transcription factors,FOXOs are of particular interest because their activity canbe suppressed by two major prosurvival kinases, AKT [55]and ERK [56–59]. Thus, in cancer cells with high levels ofAKT or ERK, BIM expression is suppressed; conversely,agents that directly or indirectly inhibit AKT or ERK couldtrigger expression of BIM (Figure 2).

BIM plays an important role in cancer cells, especially,when apoptosis is induced by tyrosine kinase inhibitors(TKIs) [5,59]. Because AKT and ERK are key effectorsdownstream of oncogenic kinases [e.g., epidermal growthfactor receptor (EGFR), human epidermal growth factorreceptor 2 (HER2), and BCR-ABL], inhibition of the tyro-sine kinase could result in suppression of AKT and ERKactivity, resulting in the expression of BIM (Figure 2).Notably, a recent study showed that BIM RNA levelscan predict clinical responsiveness to targeted cancer ther-apies such as EGFR, HER2, and PI3K inhibitors; higherBIM RNA levels detected before the inhibitor treatmentsare associated with better clinical outcomes [59]. Addition-ally, a BIM deletion polymorphism has been reported incertain human populations and is significantly associatedwith innate resistance to TKI therapies in chronic myeloidleukemia and non-small cell lung cancer [60] (Table 1).Therefore, BIM induction is critical for the efficacy of TKItherapies and BIM RNA levels may be used as a biomarkerto predict cancer patients’ prognosis and susceptibility toTKI therapies.

Page 7: Evading apoptosis in cancer

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

A large somatic copy number analysis identified anotherBH3-only family member, PUMA, as one of the proapoptoticBCL-2 family genes that are most frequently deleted incancer (the study identified two genes and the other deletedgene was BOK) [23] (Table 1). PUMA was originally discov-ered as a p53 and p73 target gene [42,61]. However, FOXOscan also transactivate PUMA independently of p53 in re-sponse to growth factor withdrawal or TKI treatments [5,62](Figure 2). In this regard, a recent study has shown thatPUMA may be implicated in apoptosis triggered by FOXOsfollowing inhibition of the PI3K/AKT pathway, whereas BIMmay be primarily under the control of the ERK pathway inHER2-positive breast cancer and EGFR-mutant lung cancer(as described below, BIM is also regulated at the protein levelby ERK); for full execution of apoptosis, inhibition of bothpathways by TKIs (i.e., expression of both PUMA and BIM)may be required in these cancer cells [5]. This finding shedslight on an underappreciated role of PUMA in TKI-inducedapoptosis. Because BIM is also known to be regulated byFOXOs, how PUMA and BIM are independently regulatedremains to be fully elucidated (Figure 2).

In addition to the signaling pathways that impedeactivation of BAX and BAK, a defect in the pathwayfollowing cytochrome c release could provide cancer cellswith an additional survival advantage independent of BAXand BAK inhibition (Figure 1). Expression of some caspasegenes (caspase-2, 7, -8, and -9) is transcriptionally regulat-ed in response to E2F1 or p53 [63–65]; in cancer cells wherethese pathways are inactivated, expression of these cas-pases could be compromised, resulting in protectionagainst apoptosis. The APAF-1 gene is transactivated byp53 [43,44] and E2F1 [44,66] and its mRNA translation isinitiated through an internal ribosome entry site (IRES)[67]. Loss of APAF-1 expression has been reported inmelanoma [68–70], colorectal cancer [71,72], gastric cancer[73], and bladder cancer [74,75] and its loss is often associ-ated with poor clinical outcomes. Studies show that loss ofAPAF-1 expression in cancer cells can be secondary topromoter methylation [68,72,73,75] and loss of heteroge-neity at the APAF-1 locus [68–70,73] (Table 1).

Collectively, it is evident that the transcriptional net-work is deregulated in cancer cells, which has a significantimpact on the expression of apoptotic regulators and,consequently, contributes to tumor development and resis-tance to therapeutic agents.

Post-translational regulationVarious apoptotic regulators are modulated not only bytranscriptional/translational machinery but also by post-translational modifications, including ubiquitination andphosphorylation. Protein ubiquitination is implicated inprotein stability and is dynamically regulated through theubiquitin/proteasome pathway [76], whereas phosphoryla-tion can change protein functions [77,78]. It should benoted, however, that phosphorylation can also influenceubiquitination of the target protein by providing a dockingsite for an E3 ubiquitin ligase (phosphodegron).

The ubiquitin/proteasome pathway involves a series ofbiochemical events mediated by E1 ubiquitin-activatingenzymes, E2 ubiquitin-conjugating enzymes, and E3ubiquitin ligases [79]. In normal cells, expression levels

of an antiapoptotic protein with a short half-life (e.g., MCL-1) may readily decline by inhibition of transcription ortranslation following an apoptotic stimulus. By contrast,an apoptotic signal may prevent constant degradation of aproapoptotic protein (e.g., BIM), resulting in the accumu-lation of that protein. Importantly, cancer cells may dereg-ulate this process by enhancing the degradability of specificproapoptotic proteins or by promoting the stability ofantiapoptotic proteins. This may also be the case whencancer cells acquire resistance to a therapeutic agent. Itwas recently shown that HER2-amplified breast cancercells modulate the stabilities of pro- and antiapoptoticproteins, both upstream and downstream of MOMP, whenacquiring resistance to the HER2 inhibitor lapatinib; con-sequently, the resistant cancer cells exhibit strong protec-tion against cell death induced by HER2 inhibition [80].

Here we discuss how the stability of some apoptoticregulators (MCL-1, BIM, and BAX) is controlled in normalcells and how this stability may be altered in cancer cells,contributing to apoptotic protection. Then, we highlightsome examples where phosphorylation plays a role inaltering the functions of apoptotic regulators.

Stabilizing MCL-1

Whereas BCL-2 and BCL-XL proteins are relatively stable,MCL-1 protein is characterized by its rapid turnover[28,81]. As described above, MCL-1 transcription is stimu-lated, at least in part, by prosurvival factors such ascytokines. Hence, once its transcription is suppressed fol-lowing apoptotic stimuli, MCL-1 protein levels quicklydrop, potentially facilitating MOMP. Importantly, emerg-ing evidence strongly suggests that the rapid turnover ofMCL-1 protein may be abrogated in some cancer cells. Todate, five E3 ligases have been discovered to ubiquitinateMCL-1 for degradation, including the most recently iden-tified tripartite motif containing17 (TRIM17), which facil-itates MCL-1 degradation during neuronal apoptosis [82].It should be noted, however, that MCL-1 protein could alsobe degraded in a ubiquitination-independent manner [83],although the precise mechanism remains elusive.

Phosphorylation plays a key role in the stability of MCL-1 protein. Phosphorylation of MCL-1 at T163 followed byphosphorylation at S155/S159 targets the protein for pro-teasomal degradation, which is mediated by the E3 ligaseSkp-, Cullin-, F-box-containing complex with b-transducinrepeat-containing protein (SCFb-TrCP) [81,84]. The primingphosphorylation site T163 is a target of multiple kinases(Figure 3). An initial study identified glycogen synthasekinase 3 (GSK3) as a kinase that phosphorylates thisresidue; however, a more recent study demonstrated thatc-Jun N-terminal kinase (JNK) primarily phosphorylatesT163; in both cases, T163 phosphorylation will triggersubsequent phosphorylation of S159 (and then S155) byGSK3 [81,85]. Collectively, phosphorylation at S155/S159/T163 serves as a degron sequence for SCFb-TrCP (Table 1and Figure 3).

Paradoxically, it has also been reported that phosphor-ylation at T163 by ERK can lead to marked MCL-1 stabili-zation [86,87] (Table 1). How can we reconcile theseconclusions? Is the mechanism of MCL-1 degradationsimply cell-type specific? We speculate that it may be

7

Page 8: Evading apoptosis in cancer

P

AKT

Stabiliza�on

GSK3

SCFβ-TrCP

SCFFBW7

TRIM17

Degrada�on

Low AKT

S155S159

T163

S155S159

T163

S155S159

T163

S155S159

T163

MCL-1

PPPGSK3

JNKp38 MAPKCK2GSK3

Normal cells(A)

(B) Cancer cells

P

MCL-1

ERKP

High AKT

MCL-1

MCL-1

TRENDS in Cell Biology

Figure 3. MCL-1 stability is controlled by multiple kinases. (A) MCL-1 can be

phosphorylated at T163 by several kinases including GSK3. AKT inhibits GSK3.

Thus, at low levels of AKT, phosphorylation by GSK3 sequentially occurs at S155/

S159, targeting MCL-1 for degradation by the E3 ligases SCFb-TrCP, SCFFBW7, and

TRIM17. (B) In cancer cells with high levels of AKT and ERK, ERK is likely to be the

main kinase that phosphorylates MCL-1. Phosphorylation does not continue to

S155/S159 and MCL-1 is stabilized.

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

phosphorylation of S159 that determines the fate of MCL-1protein following T163 phosphorylation. It is known thatGSK3 activity is negatively regulated by AKT, which iscommonly upregulated in cancer cells; therefore, in cancercells with high AKT and ERK activity, MCL-1 may bephosphorylated at T163 but not at S155 and S159, whichpromotes stabilization of MCL-1 (Figure 3). Supportingthis, it is reported that the abundance of MCL-1 protein isassociated with low GSK3 activity in breast cancer tissues[88]. By contrast, in normal cells, cellular stress signals,such as growth factor deprivation, would lead to inactiva-tion of AKT (and activation of JNK), which elicits serialphosphorylation events at S155/S159/T163; this wouldresult in degradation of MCL-1 protein (Figure 3). It shouldbe noted, however, that there is at least one additional E3ligase, F-box/WD repeat-containing protein 7 (SCFFBW7),that also regulates MCL-1 stability depending on thephosphorylation status of T163 (see below). Thus, the

8

regulation of MCL-1 protein stability mediated by thisphosphorylation motif is apparently complex and themechanism remains to be fully elucidated.

A failure in the cell cycle can trigger apoptosis in normalcells. In particular, prolonged mitotic arrest can lead toactivation of the intrinsic apoptotic pathway. For thisreason, some microtubule inhibitors that induce mitoticarrest by inhibiting mitotic spindle assembly (e.g., pacli-taxel) are used clinically to treat various cancers. However,the emergence of cancer cells with intrinsic or acquiredresistance to this type of chemotherapeutic agents is anunresolved clinical problem. Three independent studiesdescribed the mechanism of MCL-1 degradation duringprolonged mitotic arrest, suggesting a possible role forMCL-1 stabilization in resistance to mitotic apoptosis.These studies have shown that, in addition to the mecha-nisms described above, two E3 ligases, anaphase-promot-ing complex/cyclosome with cell-division cycle protein 20(APC/CCDC20) and SCFFBW7, target MCL-1 for destructionduring mitosis [89–91]. APC/CCDC20 ubiquitination ofMCL-1 is mediated by phosphorylation at T92 by cyclin-dependent kinase 1 (CDK1)/CYCLIN B and the D-boxmotif during mitotic arrest [89]. Ubiquitination of MCL-1 by SCFFBW7 is mediated by two phosphodegrons on MCL-1 (S121/E125 and S159/T163), of which S159/T163 appearsto play a major role [90,91]. Interestingly, JNK, p38, andcasein kinase 2 (CK2) phosphorylate both degrons duringmitotic arrest [91], although GSK3 phosphorylation ofMCL-1 at S159/T163 may be sufficient to target MCL-1for SCFFBW7-mediated degradation [90] (Table 1).Because,unlike CDK1/CYCLIN B, these kinases are not directlycontrolled by the cell cycle, the mechanism of mitoticphosphorylation of MCL-1 remains undetermined. In ad-dition, it is unknown whether APC/CCDC20 and SCFFBW7

compete for the mitotic degradation of MCL-1 or workindependently. From the clinical viewpoint, it is interest-ing to note that mutations or loss of FBW7 (a substrate-binding component of the multisubunit E3 complexSCFFBW7) has been reported in various cancer types[92–94]. Moreover, cancer cells with low FBW7 levels haveelevated MCL-1 protein and exhibit intrinsic resistance toABT-737, a small-molecule inhibitor of BCL-2, BCL-XL,and BCL-W [90] (Box 1).

A HECT-domain containing E3 ligase, HUWE1 [alsoknown as MCL-1 ubiquitin ligase E3 (MULE)], also targetsMCL-1 for proteasomal degradation through a pathwaythat does not require MCL-1 phosphorylation by GSK3; thebinding between MCL-1 and HUWE1 is mediated throughthe BH3 domains on each protein [95]. Interestingly, it hasbeen reported that ERK activity directly or indirectlyreduces the interaction between MCL-1 and HUWE1,although the detailed molecular mechanism remains tobe elucidated [96]. It would be interesting to test whetherthe aforementioned T163 phosphorylation by ERK is im-plicated in the MCL-1–HUWE1 interaction.

Deubiquitinating enzymes (DUBs) are negative regula-tors of the ubiquitination-mediated protein degradation,because DUBs can stabilize their substrate proteins byremoving polyubiquitin chains. Ubiquitin-specific pepti-dase 9 X-linked (USP9X) reverses ubiquitination ofMCL-1 [97]. As opposed to SCFb-TrCP and SCFFBW7, the

Page 9: Evading apoptosis in cancer

Box 1. BH3 mimetics and cancer therapy

The binding between pro- and antiapoptotic BCL-2 family proteins is

mediated through a hydrophobic groove on the antiapoptotic

proteins and a BH3 domain. Several small-molecule inhibitors, so-

called BH3 mimetics (e.g., ABT-737, ATB-263, ATB-199), have been

engineered to bind specifically to the hydrophobic pocket, thereby

antagonizing the function of antiapoptotic BCL-2 family proteins.

The initial inhibitor of this class, ABT-737, and its orally bioavailable

analog ABT-263, both of which inhibit BCL-2 and BCL-XL, were a

preclinical success [188,189]. Nonetheless, the clinical success was

hampered by thrombocytopenia caused by BCL-XL inhibition. To

circumvent this problem, a BCL-2-specific inhibitor, ABT-199, was

recently developed and is now being tested in clinical trials [190].

Another challenge in BCL-2/BCL-XL inhibitors is that cancer cells can

acquire resistance to this class of inhibitors by overexpressing MCL-

1 [191]. Recently, two groups independently identified MCL-1

inhibitors through gene-expression-based or biochemical screen-

ings: Golub and colleagues identified compounds that suppress

MCL-1 transcription [192], whereas Walensky and colleagues

discovered a small molecule, MIM1, that binds to the BH3-domain

binding pocket of MCL-1 [193] – these inhibitors may overcome

resistance to BCL-2/BCL-XL inhibitors. Of note, the short protein

half-life of MCL-1 makes MCL-1-addicted cancer cells more suscep-

tible to chemotherapy than cancer cells depending on BCL-2 or BCL-

XL for survival [8]. However, given the multiple mechanisms of

MCL-1 stabilization in cancer cells (see text), it is assumed that this

approach may not work in all MCL-1-addicted cancer cells. For

instance, for cancer cells with stabilized MCL-1 protein, agents that

directly inhibit MCL-1 protein (e.g., MIM1) or promote MCL-1 protein

degradation may be an option, whereas for cancer cells with

enhanced MCL-1 mRNA levels, a transcriptional inhibitor may be

more effective.

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

binding of USP9X to MCL-1 is negatively regulated byGSK3 phosphorylation of MCL-1 at S155/S159/T163, sug-gesting that USP9X may directly counteract MCL-1 deg-radation controlled by SCFb-TrCP and SCFFBW7. USP9Xand MCL-1 protein expression levels were positively cor-related in several cancer types, including follicular lym-phomas [97]. Elevated USP9X transcription was alsoassociated with poor prognosis in multiple myelomas [97].

Destroying proapoptotic proteins

As cancer cells can attenuate MCL-1 degradation, theymay also suppress accumulation of proapoptotic proteinssuch as BIM. As with MCL-1, BIM is also characterized byits short protein half-life. BIM stability is regulated byERK phosphorylation at S69; phosphorylation of this sitepromotes BIM degradation through the ubiquitin/protea-some pathway [98,99]. A transgenic mouse study demon-strated that replacement of the ERK phosphorylation siteswith Ala prolongs BIM protein half-life and increasesapoptotic cell death in response to serum starvation [4].SCFb-TrCP, c-CBL, and TRIM2 are the E3 ligases responsi-ble for ERK-dependent BIM degradation [100–102]. Nota-bly, SCFb-TrCP-mediated BIM ubiquitination requiresphosphorylation of the degron sequence containing S93/S94/S98 by ribosomal S6 kinase (RSK), which is promotedby ERK phosphorylation at S69 [101] (Table 1). WhetherBIM ubiquitination by c-CBL and TRIM2 may also requireRSK phosphorylation remains elusive. Lastly, the ubiquitinE3 ligase complex ElonginB/C–Cullin2–CIS is also impli-cated in BIM ubiquitination and degradation [103]. BIMubiquitination mediated by ElonginB/C–Cullin2–CIS is en-hanced following paclitaxel treatment and a defect in this

ubiquitination pathway may confer resistance to this agentin cancer cells [103].

BAX is a central component of apoptosis because itsactivation is directly linked to MOMP (Figure 1). Thus,causing degradation of BAX protein may be a ‘last-minute’safeguard for cancer cells to prevent apoptosis. BAX pro-tein mainly localizes in the cytoplasm. On genotoxic orcytotoxic stress, BAX translocates to the mitochondrialouter membrane, forms oligomeric structures, and triggersMOMP [104]. A recent study has shown, however, that thesubcellular localization of BAX protein may be in a moredynamic equilibrium between the cytoplasm and the mi-tochondrial outer membrane [105]. Although the detailedmechanism of how the equilibrium may change followingapoptotic stimuli remains elusive, the BAX equilibriumhypothesis suggests there may be a mechanism that pre-vents activation of BAX on the mitochondrial outer mem-brane. Interestingly, it is known that BCL-XL can block themitochondrial translocation of BAX by redirecting BAXback to the cytoplasm [106]. Moreover, BAX degradationoccurs on the mitochondrial outer membrane followingapoptotic stimuli [107,108]. BAX is a protein with a shorthalf-life [109,110] and its rapid degradation has beenassociated with poor clinical outcomes and high tumorgrades in chronic lymphocytic leukemia and prostate can-cer [109]. The E3 ligase IBR domain-containing 2(IBRDC2), also known as p53RFP or RNF144B, was re-cently identified and shown to ubiquitinate BAX specifi-cally on the mitochondrial outer membrane [108].Overexpression of IBRDC2, however, did not block apopto-sis triggered by actinomycin D or staurosporine treatment[108], suggesting that there may be additional factors (e.g.,cofactors or other ligases) involved in this process.

Altering protein functions

In addition to modulating docking sites for E3 ligases andsubsequent ubiquitin-mediated protein degradation, phos-phorylation can also alter the function of apoptotic regu-lators. In particular, phosphorylation by prosurvivalkinases (e.g., AKT and ERK) often results in suppressionof the proapoptotic function of the target protein, whereasphosphorylation by proapoptotic kinases (e.g., GSK3 andJNK) can lead to activation of proapoptotic proteins orinhibition of antiapoptotic proteins (see Table 1 for acomprehensive list of known phosphorylation sites of apo-ptotic regulators). For instance, the proapoptotic activity ofthe BH3-only protein BAD is inhibited by several prosur-vival kinases, including PAK1 and AKT [111–113]. Like-wise, ERK phosphorylation of BIM prevents theinteraction between BIM and BAX, thereby inhibitingapoptosis, whereas phosphorylation of BIM by JNK orp38 MAP kinase promotes the proapoptotic activity ofBIM by enhancing binding to antiapoptotic BCL-2 familyproteins [4,114–117]. As described above, ERK also pro-motes suppression of BIM expression and degradation ofBIM protein [4,98,99] (Figure 2), further highlighting acritical role for ERK in BIM inhibition.

Prosurvival kinases also negatively regulate apoptoticpathways downstream of mitochondrial cytochrome c re-lease (Figure 1). Activation of APAF-1 in response tocytochrome c is inhibited via phosphorylation of APAF-1

9

Page 10: Evading apoptosis in cancer

Box 2. Outstanding questions

� Do cancer cells simply depend on the net abundance of

prosurvival BCL-2 family proteins relative to the total BH3-only

proteins? Alternatively, may the regulation of a particular BCL-2

family member selectively play a role in survival in certain

cancer types? How do the functional redundancy and

specificity of prosurvival BCL-2 family proteins affect cancer

cell survival?

� Cancer cells employ multiple mechanisms to silence the apoptotic

machinery. Letai and colleagues have developed a functional

assay, called BH3 profiling, to gauge the dependency of cancer

cells on antiapoptotic BCL-2 family proteins. Although BH3

profiling made a successful breakthrough [194], is there any other

way to predict which pathways are altered in particular cancer

cells?

� Abundance of MCL-1 is one of the mechanisms for resistance to a

BCL-2/BCL-XL inhibitor in cancer (Box 1). If a BCL-2/BCL-XL

inhibitor is used concomitantly with an MCL-1 inhibitor, can we

cure all cancers? Alternatively, will cancer cells eventually acquire

resistance to the combination of a BCL-2/BCL-XL inhibitor and an

MCL-1 inhibitor? If so, how?

� Why is the pathway downstream of mitochondrial cytochrome c

release often inhibited in cancer cells in addition to the inhibition

of MOMP [195]?

� Failure to activate caspases in response to an apoptotic stimulus

can result in caspase-independent cell death [196]. Does caspase-

independent cell death play a role in cancer cell survival?

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

by RSK [118], a kinase often amplified in prostate cancer[119] (Table 1). Leukemic tyrosine kinases, such as BCR-ABL, also inhibit APAF-1 oligomerization by modulatingthe phosphorylation status of the APAF-1 inhibitorHSP90b [120]. In addition to APAF-1, caspases are directtargets of various kinases; activation of caspases can besuppressed by direct phosphorylation (Table 1). Further-more, it is known that phosphorylation can protect somecaspase substrates from proteolytic cleavage [78]. Forexample, phosphorylation by CK1 or CK2 protects BIDfrom caspase-8, thereby blocking the activation of thisBH3-only protein and subsequent cytochrome c releasefrom mitochondria [121] (Table 1).

In summary, post-translational modifications play acritical role in the regulation of apoptosis by modulatingprotein stabilities and functions.

Concluding remarksTo survive and proliferate, cancer cells need to overridenumerous barriers that otherwise would cause apoptosis.It is conceivable that cancer cells employ multiple mecha-nisms instead of a single pathway. Given the multiplicity ofantiapoptotic mechanisms utilized by cancer cells, it is spec-ulated that agents that can globally alter cancer’s apoptoticresistance by targeting multiple pathways may provide uswith the most valuable therapeutic benefits in the future ofcancer treatment. To target cancer cells most efficiently andselectively, we must further elucidate the mechanisms ofhow they evade the apoptotic program (Box 2).

AcknowledgmentsThe authors are grateful to Leta Nutt, Joseph Opferman, MarisaBuchakjian, and Matthew Ung for critical reading and feedback on themanuscript. This work was supported by an NCI Career DevelopmentAward R00 CA140948 (to M.K.) and a cancer center core grant P30CA23108 (to the Norris Cotton Cancer Center).

10

References1 Martinou, J.C. and Youle, R.J. (2011) Mitochondria in apoptosis:

Bcl-2 family members and mitochondrial dynamics. Dev. Cell 21,92–101

2 Strasser, A. et al. (2011) Deciphering the rules of programmed celldeath to improve therapy of cancer and other diseases. EMBO J. 30,3667–3683

3 Rautureau, G.J. et al. (2012) The restricted binding repertoire of Bcl-Bleaves Bim as the universal BH3-only prosurvival Bcl-2 proteinantagonist. Cell Death Dis. 13, e443

4 Hu bner, A. et al. (2008) Multisite phosphorylation regulates Bimstability and apoptotic activity. Mol. Cell 30, 415–425

5 Bean, G.R. et al. (2013) PUMA and BIM are required for oncogeneinactivation-induced apoptosis. Sci. Signal. 6, ra20

6 Parsons, M.J. et al. (2013) Genetic deletion of caspase-2 acceleratesMMTV/c-neu-driven mammary carcinogenesis in mice. Cell DeathDiffer. http://dx.doi.org/10.1038/cdd.2013.38

7 Strasser, A. et al. (1990) Novel primitive lymphoid tumours induced intransgenic mice by cooperation between myc and bcl-2. Nature 348,331–333

8 Brunelle, J.K. et al. (2009) MCL-1-dependent leukemia cells are moresensitive to chemotherapy than BCL-2-dependent counterparts. J.Cell Biol. 187, 429–442

9 Vaux, D.L. et al. (1998) Bcl-2 gene promotes haemopoietic cell survivaland cooperates with c-myc to immortalize pre-B cells. Nature 335,440–442

10 Zhou, P. et al. (1998) Mcl-1 in transgenic mice promotes survival in aspectrum of hematopoietic cell types and immortalization in themyeloid lineage. Blood 92, 3226–3239

11 Vander-Heiden, M.G. et al. (2009) Understanding the Warburg effect:the metabolic requirements of cell proliferation. Science 324, 1029–1033

12 Buchakjian, M.R. and Kornbluth, S. (2010) The engine driving theship: metabolic steering of cell proliferation and death. Nat. Rev. Mol.Cell Biol. 11, 715–727

13 Kumar, S. and Cakouros, D. (2004) Transcriptional control of the corecell-death machinery. Trends Biochem. Sci. 29, 193–199

14 Esquela-Kerscher, A. and Slack, F.J. (2006) Oncomirs - microRNAswith a role in cancer. Nat. Rev. Cancer 6, 259–269

15 Kaufmann, S.H. et al. (1998) Elevated expression of the apoptoticregulator Mcl-1 at the time of leukemic relapse. Blood 91, 991–1000

16 Taniai, M. et al. (2004) Mcl-1 mediates tumor necrosis factor-relatedapoptosis-inducing ligand resistance in human cholangiocarcinomacells. Cancer Res. 64, 3517–3524

17 Wuilleme-Toumi, S. et al. (2005) Mcl-1 is overexpressed in multiplemyeloma and associated with relapse and shorter survival. Leukemia19, 1248–1252

18 Bakhshi, A. et al. (1985) Cloning the chromosomal breakpoint oft(14;18) human lymphomas: clustering around JH on chromosome14 and near a transcriptional unit on 18. Cell 41, 899–906

19 Cleary, M.L. and Sklar, J. (1985) Nucleotide sequence of a t(14;18)chromosomal breakpoint in follicular lymphoma and demonstrationof a breakpoint-cluster region near a transcriptionally active locus onchromosome 18. Proc. Natl. Acad. Sci. U.S.A. 82, 7439–7443

20 Tsujimoto, Y. et al. (1985) The t(14;18) chromosome translocationsinvolved in B-cell neoplasms result from mistakes in VDJ joining.Science 229, 1390–1393

21 Tonon, G. et al. (2005) High-resolution genomic profiles of human lungcancer. Proc. Natl. Acad. Sci. U.S.A. 102, 9625–9630

22 Smith, L.T. et al. (2006) 20q11.1 amplification in giant-cell tumor ofbone: Array CGH, FISH, and association with outcome. GenesChromosomes Cancer 45, 957–966

23 Beroukhim, R. et al. (2010) The landscape of somatic copy-numberalteration across human cancers. Nature 463, 899–905

24 Kendall, J. et al. (2007) Oncogenic cooperation and coamplification ofdevelopmental transcription factor genes in lung cancer. Proc. Natl.Acad. Sci. U.S.A. 104, 16663–16668

25 Wang, J.M. et al. (1999) The antiapoptotic gene mcl-1 is up-regulatedby the phosphatidylinositol 3-kinase/Akt signaling pathway througha transcription factor complex containing CREB. Mol. Cell. Biol. 9,6195–6206

26 Huang, H.M. et al. (2000) Mcl-1 is a common target of stem cell factorand interleukin-5 for apoptosis prevention activity via MEK/MAPKand PI-3K/Akt pathways. Blood 96, 1764–1771

Page 11: Evading apoptosis in cancer

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

27 Liu, H. et al. (2003) Serine phosphorylation of STAT3 is essential forMcl-1 expression and macrophage survival. Blood 102, 344–352

28 Nijhawan, D. et al. (2003) Elimination of Mcl-1 is required for theinitiation of apoptosis following ultraviolet irradiation. Genes Dev. 17,1475–1486

29 Fritsch, R.M. et al. (2007) Translational repression of MCL-1 couplesstress-induced eIF2 alpha phosphorylation to mitochondrialapoptosis initiation. J. Biol. Chem. 282, 22551–22562

30 Croxton, R. et al. (2002) Direct repression of the Mcl-1 promoter byE2F1. Oncogene 21, 1359–1369

31 Mills, J.R. et al. (2008) mTORC1 promotes survival throughtranslational control of Mcl-1. Proc. Natl. Acad. Sci. U.S.A. 105,10853–10858

32 Cimmino, A. et al. (2005) miR-15 and miR-16 induce apoptosis bytargeting BCL2. Proc. Natl. Acad. Sci. U.S.A. 102, 13944–13949

33 Mott, J.L. et al. (2007) mir-29 regulates Mcl-1 protein expression andapoptosis. Oncogene 26, 6133–6140

34 Kwon, J.E. et al. (2013) Ionizing radiation-inducible microRNA miR-193a-3p induces apoptosis by directly targeting Mcl-1. Apoptosis 18,896–909

35 Su, H. et al. (2009) MicroRNA-101, down-regulated in hepatocellularcarcinoma, promotes apoptosis and suppresses tumorigenicity.Cancer Res. 69, 1135–1142

36 Kole, A.J. et al. (2011) miR-29b is activated during neuronalmaturation and targets BH3-only genes to restrict apoptosis. GenesDev. 25, 125–130

37 Xie, Y. et al. (2012) MicroRNA-24 regulates XIAP to reduce theapoptosis threshold in cancer cells. Oncogene 32, 2442–2451

38 Liu, S. et al. (2013) MicroRNA-7 downregulates XIAP expression tosuppress cell growth and promote apoptosis in cervical cancer cells.FEBS Lett. 587, 2247–2253

39 Hollstein, D. et al. (1991) p53 mutations in human cancers. Science253, 49–53

40 Feki, I. and Irminger-Finger, I. (2004) Mutational spectrum of p53mutations in primary breast and ovarian tumors. Crit. Rev. Oncol.Hematol. 52, 103–116

41 Oda, E. et al. (2000) Noxa, a BH3-only member of the Bcl-2 family andcandidate mediator of p53-induced apoptosis. Science 288, 1053–1058

42 Nakano, K. and Vousden, K.H. (2001) PUMA, a novel proapoptoticgene is induced by p53. Mol. Cell 7, 683–694

43 Fortin, A. et al. (2001) APAF1 is a key transcriptional target for p53 inthe regulation of neuronal cell death. J. Cell Biol. 155, 207–216

44 Moroni, M.C. et al. (2001) Apaf-1 is a transcriptional target for E2Fand p53. Nat. Cell Biol. 3, 552–558

45 Rampino, N. et al. (1997) Somatic frameshift mutations in the BAXgene in colon cancers of the microsatellite mutator phenotype. Science275, 967–969

46 Mihara, M. et al. (2003) p53 has a direct apoptogenic role at themitochondria. Mol. Cell 11, 577–590

47 Chipuk, J.E. et al. (2004) PUMA couples the nuclear and cytoplasmicproapoptotic function of p53. Science 309, 1732–1735

48 Nieminen, A. et al. (2013) Myc-induced AMPK-phospho p53 pathwayactivates Bak to sensitize mitochondrial apoptosis. Proc. Natl. Acad.Sci. U.S.A. 110, E1839–E1848

49 Stahl, M. et al. (2002) The forkhead transcription factor FoxOregulates transcription of p27Kip1 and Bim in response to IL-2. J.Immunol. 168, 5024–5031

50 Dijkers, P.F. et al. (2000) Expression of the pro-apoptotic Bcl-2 familymember Bim is regulated by the forkhead transcription factor FKHR-L1. Curr. Biol. 10, 1201–1204

51 Sunters, A. et al. (2003) FoxO3a transcriptional regulation of Bimcontrols apoptosis in paclitaxel-treated breast cancer cell lines. J.Biol. Chem. 278, 49795–49805

52 Yano, T. et al. (2006) The RUNX3 tumor suppressor upregulates Bimin gastric epithelial cells undergoing transforming growth factor beta-induced apoptosis. Mol. Cell. Biol. 26, 4474–4488

53 Jin, H.O. et al. (2006) Up-regulation of Bak and Bim via JNKdownstream pathway in the response to nitric oxide in humanglioblastoma cells. J. Cell. Physiol. 206, 477–486

54 Gogada, R. et al. (2013) Bim, a proapoptotic protein, up-regulated viatranscription factor E2F1-dependent mechanism, functions as aprosurvival molecule in cancer. J. Biol. Chem. 288, 368–381

55 Gilley, J. et al. (2003) FOXO transcription factors directly activate bimgene expression and promote apoptosis in sympathetic neurons. J.Cell Biol. 162, 613–622

56 Costa, D.B. et al. (2007) BIM mediates EGFR tyrosine kinaseinhibitor-induced apoptosis in lung cancers with oncogenic EGFRmutations. PLoS Med. 4, 1669–1679

57 Cragg, M.S. et al. (2007) Gefitinib-induced killing of NSCLC cell linesexpressing mutant EGFR requires BIM and can be enhanced by BH3mimetics. PLoS Med. 4, 1681–1689

58 Gong, Y. et al. (2007) Induction of BIM is essential for apoptosistriggered by EGFR kinase inhibitors in mutant EGFR-dependentlung adenocarcinomas. PLoS Med. 4, e294

59 Faber, A.C. et al. (2011) BIM expression in treatment-naive cancerspredicts responsiveness to kinase inhibitors. Cancer Discov. 1,352–365

60 Ng, K.P. et al. (2012) A common BIM deletion polymorphism mediatesintrinsic resistance and inferior responses to tyrosine kinaseinhibitors in cancer. Nat. Med. 18, 521–528

61 Melino, G. et al. (2004) p73 Induces apoptosis via PUMAtransactivation and Bax mitochondrial translocation. J. Biol.Chem. 279, 8076–8083

62 You, H. et al. (2006) FOXO3a-dependent regulation of Puma inresponse to cytokine/growth factor withdrawal. J. Exp. Med. 203,1657–1663

63 Liedtke, C. et al. (2003) The human caspase-8 promoter sustains basalactivity through SP1 and ETS-like transcription factors and can beup-regulated by a p53-dependent mechanism. J. Biol. Chem. 278,27593–27604

64 Nahle, Z. et al. (2003) Direct coupling of the cell cycle and cell deathmachinery by E2F. Nat. Cell Biol. 4, 859–864

65 Baptiste-Okoh, N. et al. (2008) Caspase 2 is both required for p53-mediated apoptosis and downregulated by p53 in a p21-dependentmanner. Cell Cycle 7, 1133–1138

66 Furukawa, Y. et al. (2002) Apaf-1 is a mediator of E2F-1-inducedapoptosis. J. Biol. Chem. 277, 39760–39768

67 Coldwell, M.J. et al. (2000) Initiation of Apaf-1 translation by internalribosome entry. Oncogene 19, 899–905

68 Soengas, M.S. et al. (2001) Inactivation of the apoptosis effector Apaf-1in malignant melanoma. Nature 409, 207–211

69 Dai, D.L. et al. (2004) Reduced Apaf-1 expression in human cutaneousmelanomas. Br. J. Cancer 91, 1089–1095

70 Fujimoto, A. et al. (2004) Allelic imbalance of 12q22-23 associatedwith APAF-1 locus correlates with poor disease outcome in cutaneousmelanoma. Cancer Res. 64, 2245–2250

71 Umetani, N. et al. (2004) Allelic imbalance of APAF-1 locus at 12q23is related to progression of colorectal carcinoma. Oncogene 23,8292–8300

72 Zlobec, I. et al. (2007) Loss of APAF-1 expression is associated withtumour progression and adverse prognosis in colorectal cancer. Eur.J. Cancer 43, 1101–1107

73 Wang, H.L. et al. (2007) Rationales for expression and alteredexpression of apoptotic protease activating factor-1 gene in gastriccancer. World J. Gastroenterol. 13, 5060–5064

74 Christoph, F. et al. (2007) A gene expression profile of tumorsuppressor genes commonly methylated in bladder cancer. J.Cancer Res. Clin. Oncol. 133, 343–349

75 Hinz, S. et al. (2007) EZH2 polycomb transcriptional repressorexpression correlates with methylation of the APAF-1 gene insuperficial transitional cell carcinoma of the bladder. Tumour Biol.28, 151–157

76 Vucic, D. et al. (2011) Ubiquitylation in apoptosis: a post-translationalmodification at the edge of life and death. Nat. Rev. Mol. Cell Biol. 12,439–452

77 Dai, C. and Gu, W. (2010) p53 post-translational modification:deregulated in tumorigenesis. Trends Mol. Med. 16, 528–536

78 Kurokawa, M. and Kornbluth, S. (2009) Caspases and kinases in adeath grip. Cell 138, 838–854

79 Weissman, A.M. et al. (2011) The predator becomes the prey:regulating the ubiquitin system by ubiquitylation and degradation.Nat. Rev. Mol. Cell Biol. 12, 605–620

80 Kurokawa, M. et al. (2013) A network of substrates of the E3 ubiquitinligases MDM2 and HUWE1 control apoptosis independently of p53.Sci. Signal. 6, ra32

11

Page 12: Evading apoptosis in cancer

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

81 Maurer, U. et al. (2005) Glycogen synthase kinase-3 regulatesmitochondrial outer membrane permeabilization and apoptosis bydestabilization of MCL-1. Mol. Cell 21, 749–760

82 Magiera, M.M. et al. (2013) Trim17-mediated ubiquitination anddegradation of Mcl-1 initiate apoptosis in neurons. Cell DeathDiffer. 20, 281–292

83 Stewart, D.P. et al. (2010) Ubiquitin-independent degradation ofantiapoptotic MCL-1. Mol. Cell. Biol. 30, 3099–3110

84 Ding, Q. et al. (2007) Degradation of Mcl-1 by beta-TrCP mediatesglycogen synthase kinase 3-induced tumor suppression andchemosensitization. Mol. Cell. Biol. 27, 4006–4017

85 Morel, C. et al. (2009) Mcl-1 integrates the opposing actions ofsignaling pathways that mediate survival and apoptosis. Mol. Cell.Biol. 29, 3845–3852

86 Liao, M. et al. (2011) Role of bile salt in regulating Mcl-1phosphorylation and chemoresistance in hepatocellular carcinomacells. Mol. Cancer 10, 44

87 Nifoussi, S.K. et al. (2012) Thr 163 phosphorylation causes Mcl-1stabilization when degradation is independent of the adjacent GSK3-targeted phosphodegron, promoting drug resistance in cancer. PLoSONE 7, e47060

88 Ding, Q. et al. (2007) Myeloid cell leukemia-1 inversely correlateswith glycogen synthase kinase-3beta activity and associateswith poor prognosis in human breast cancer. Cancer Res. 67,4564–4571

89 Harley, M.E. et al. (2010) Phosphorylation of Mcl-1 by CDK1-cyclin B1initiates its Cdc20-dependent destruction during mitotic arrest.EMBO J. 29, 2407–2420

90 Inuzuka, H. et al. (2011) SCF(FBW7) regulates cellular apoptosisby targeting MCL1 for ubiquitylation and destruction. Nature 471,104–109

91 Wertz, I.E. et al. (2011) Sensitivity to antitubulin chemotherapeuticsis regulated by MCL1 and FBW7. Nature 471, 110–114

92 Akhoondi, S. et al. (2007) FBXW7/hCDC4 is a general tumorsuppressor in human cancer. Cancer Res. 67, 9006–9012

93 Maser, R.S. et al. (2007) Chromosomally unstable mouse tumourshave genomic alterations similar to diverse human cancers. Nature47, 966–971

94 Wood, L.D. et al. (2007) The genomic landscapes of human breast andcolorectal cancers. Science 318, 1108–1113

95 Zhong, Q. et al. (2005) Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase,catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell121, 1085–1095

96 Pervin, S. et al. (2011) Reduced association of anti-apoptotic proteinMcl-1 with E3 ligase Mule increases the stability of Mcl-1 in breastcancer cells. Br. J. Cancer 105, 428–437

97 Schwickart, M. et al. (2010) Deubiquitinase USP9X stabilizes MCL1and promotes tumour cell survival. Nature 463, 103–107

98 Ley, R. et al. (2003) Activation of the ERK1/2 signalingpathway promotes phosphorylation and proteasome-dependentdegradation of the BH3-only protein, Bim. J. Biol. Chem. 278,18811–18816

99 Luciano, F. et al. (2003) Phosphorylation of Bim-EL by Erk1/2 onserine 69 promotes its degradation via the proteasome pathway andregulates its proapoptotic function. Oncogene 22, 6785–6793

100 Akiyama, T. et al. (2003) Regulation of osteoclast apoptosis byubiquitylation of proapoptotic BH3-only Bcl-2 family member Bim.EMBO J. 22, 6653–6664

101 Dehan, E. et al. (2009) betaTrCP- and Rsk1/2-mediated degradation ofBimEL inhibits apoptosis. Mol. Cell 33, 109–116

102 Thompson, S. et al. (2011) Identification of a novel Bcl-2-interactingmediator of cell death (Bim) E3 ligase, tripartite motif-containingprotein 2 (TRIM2), and its role in rapid ischemic tolerance-inducedneuroprotection. J. Biol. Chem. 286, 19331–19339

103 Zhang, W. et al. (2008) RACK1 and CIS mediate the degradation ofBimEL in cancer cells. J. Biol. Chem. 283, 16416–16426

104 Walter, K.G. et al. (1998) Movement of Bax from the cytosol tomitochondria during apoptosis. J. Cell Biol. 139, 1281–1292

105 Schellenberg, B. et al. (2013) Bax exists in a dynamic equilibriumbetween the cytosol and mitochondria to control apoptotic priming.Mol. Cell 49, 959–971

106 Edlich, F. et al. (2011) Bcl-x(L) retrotranslocates Bax from themitochondria into the cytosol. Cell 145, 104–116

12

107 Liu, F.T. et al. (2008) Bortezomib blocks Bax degradation inmalignant B cells during treatment with TRAIL. Blood 111, 2797–2805

108 Benard, G. et al. (2010) IBRDC2, an IBR-type E3 ubiquitin ligase, is aregulatory factor for Bax and apoptosis activation. EMBO J. 29,1458–1471

109 Agrawal, S.G. et al. (2008) Increased proteasomal degradation of Baxis a common feature of poor prognosis chronic lymphocytic leukemia.Blood 111, 2790–2796

110 Fu, N.Y. et al. (2009) Baxb: a constitutively active human Bax isoformthat is under tight regulatory control by the proteasomal degradationmechanism. Mol. Cell 33, 15–29

111 Zha, J. et al. (1996) Serine phosphorylation of death agonist BAD inresponse to survival factor results in binding to 14-3-3 not BCL-X(L).Cell 87, 619–628

112 Datta, S.R. et al. (1997) Akt phosphorylation of BAD couples survivalsignals to the cell-intrinsic death machinery. Cell 91, 231–241

113 del Peso, L. et al. (1997) Interleukin-3-induced phosphorylation ofBAD through the protein kinase Akt. Science 278, 687–689

114 Putcha, G.V. et al. (2003) JNK-mediated BIM phosphorylationpotentiates BAX-dependent apoptosis. Neuron 38, 899–914

115 Becker, E.B. et al. (2004) Characterization of the c-Jun N-terminalkinase-BimEL signaling pathway in neuronal apoptosis. J. Neurosci.24, 8762–8770

116 Harada, H. et al. (2004) Survival factor-induced extracellular signal-regulated kinase phosphorylates BIM, inhibiting its association withBAX and proapoptotic activity. Proc. Natl. Acad. Sci. U.S.A. 101,15313–15317

117 Cai, B. et al. (2006) p38 MAP kinase mediates apoptosis throughphosphorylation of BimEL at Ser-65. J. Biol. Chem. 281, 25215–25222

118 Kim, J. et al. (2012) Rsk-mediated phosphorylation and 14-3-3ebinding of Apaf-1 suppresses cytochrome c-induced apoptosis.EMBO J. 31, 1279–1292

119 Clark, D.E. et al. (2005) The serine/threonine protein kinase, p90ribosomal S6 kinase, is an important regulator of prostate cancer cellproliferation. Cancer Res. 65, 3108–3116

120 Kurokawa, M. et al. (2007) Inhibition of apoptosome formation bysuppression of Hsp90b phosphorylation in tyrosine kinase-inducedleukemias. Mol. Cell. Biol. 28, 5494–5506

121 Desagher, S. et al. (2001) Phosphorylation of bid by casein kinases Iand II regulates its cleavage by caspase 8. Mol. Cell 8, 601–611

122 Shin, S. et al. (2005) Caspase-2 primes cancer cells for TRAIL-mediated apoptosis by processing procaspase-8. EMBO J. 24,3532–3542

123 Nutt, K.L. et al. (2005) Metabolic regulation of oocyte cell deaththrough the CaMKII-mediated phosphorylation of caspase-2. Cell123, 89–103

124 Andersen, J.L. et al. (2009) Restraint of apoptosis during mitosisthrough interdomain phosphorylation of caspase-2. EMBO J. 28,3216–3227

125 Voss, O.H. et al. (2005) Regulation of monocyte apoptosis by theprotein kinase Cdelta-dependent phosphorylation of caspase-3. J.Biol. Chem. 280, 17371–17379

126 Alvarado-Kristensson, M. et al. (2004) p38-MAPK signals survival byphosphorylation of caspase-8 and caspase-3 in human neutrophils. J.Exp. Med. 199, 449–458

127 Li, X. et al. (2011) Phosphorylation of caspase-7 by p21-activatedprotein kinase (PAK) 2 inhibits chemotherapeutic drug-inducedapoptosis of breast cancer cell lines. J. Biol. Chem. 286, 22291–22299

128 Jia, S.H. et al. (2008) Dynamic regulation of neutrophil survivalthrough tyrosine phosphorylation or dephosphorylation of caspase-8. J. Biol. Chem. 283, 5402–5413

129 Cursi, S. et al. (2006) Src kinase phosphorylates caspase-8 on Tyr380:a novel mechanism of apoptosis suppression. EMBO J. 25, 1895–1905

130 Martin, M.C. et al. (2005) Protein kinase A regulates caspase-9activation by Apaf-1 downstream of cytochrome c. J. Biol. Chem.280, 15449–15455

131 Allan, L.A. et al. (2003) Inhibition of caspase-9 throughphosphorylation at Thr 125 by ERK MAPK. Nat. Cell Biol. 5, 647–654

132 Dessauge, F. et al. (2006) Identification of PP1alpha as a caspase-9regulator in IL-2 deprivation-induced apoptosis. J. Immunol. 177,2441–2451

Page 13: Evading apoptosis in cancer

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

133 Allan, A.L. and Clarke, P.R. (2007) Phosphorylation of caspase-9 byCDK1/cyclin B1 protects mitotic cells against apoptosis. Mol. Cell 26,301–310

134 Laguna, A. et al. (2008) The protein kinase DYRK1A regulatescaspase-9-mediated apoptosis during retina development. Dev. Cell15, 841–853

135 Seifert, A. et al. (2008) DYRK1A phosphorylates caspase 9 at aninhibitory site and is potently inhibited in human cells byharmine. FEBS J. 275, 6268–6280

136 Seifert, A. and Clarke, P.R. (2009) p38alpha- and DYRK1A-dependentphosphorylation of caspase-9 at an inhibitory site in response tohyperosmotic stress. Cell. Signal. 21, 1626–1633

137 Brady, S.C. et al. (2005) Regulation of caspase 9 throughphosphorylation by protein kinase C zeta in response tohyperosmotic stress. Mol. Cell. Biol. 25, 10543–10555

138 Raina, D. et al. (2005) c-Abl tyrosine kinase regulates caspase-9autocleavage in the apoptotic response to DNA damage. J. Biol.Chem. 280, 11147–11151

139 Cardone, M.H. et al. (1998) Regulation of cell death protease caspase-9 by phosphorylation. Science 282, 1318–1321

140 Zinkel, S.S. et al. (2005) A role for proapoptotic BID in the DNA-damage response. Cell 122, 579–591

141 Chakraborty, A.R. et al. (2013) MAPK pathway activation leadsto Bim loss and histone deacetylase inhibitor resistance: rationaleto combine romidepsin with an MEK inhibitor. Blood 121, 4115–4125

142 Leung, K.T. et al. (2008) Activation of the JNK pathway promotesphosphorylation and degradation of BimEL–a novel mechanismof chemoresistance in T-cell acute lymphoblastic leukemia.Carcinogenesis 29, 544–551

143 Moujalled, D. et al. (2011) Cyclic-AMP-dependent protein kinase Aregulates apoptosis by stabilizing the BH3-only protein Bim. EMBORep. 12, 77–83

144 Fricker, M. et al. (2010) Phosphorylation of Puma modulates itsapoptotic function by regulating protein stability. Cell Death Dis. 1,e59

145 Sandow, J.J. et al. (2012) Cytokine receptor signaling activates anIKK-dependent phosphorylation of PUMA to prevent cell death. CellDeath Differ. 19, 633–641

146 Yan, J. et al. (2013) Inactivation of BAD by IKK inhibits TNFa-induced apoptosis independently of NF-kB activation. Cell 152,304–315

147 Ye, D.Z. et al. (2011) p21-Activated kinase 1 (Pak1) phosphorylatesBAD directly at serine 111 in vitro and indirectly through Raf-1 atserine 112. PLoS ONE 6, e27637

148 Harada, H. et al. (1999) Phosphorylation and inactivation of BAD bymitochondria-anchored protein kinase A. Mol. Cell 3, 413–422

149 Bonni, A. et al. (1999) Cell survival promoted by the Ras-MAPKsignaling pathway by transcription-dependent and -independentmechanisms. Science 286, 1358–1362

150 Fang, X. et al. (1999) Regulation of BAD phosphorylation at serine 112by the Ras-mitogen-activated protein kinase pathway. Oncogene 18,6635–6640

151 Tan, Y. et al. (1999) p90(RSK) blocks bad-mediated cell death via aprotein kinase C-dependent pathway. J. Biol. Chem. 274, 34859–34867

152 Schu rmann, A. et al. (2000) p21-activated kinase 1 phosphorylates thedeath agonist bad and protects cells from apoptosis. Mol. Cell. Biol.20, 453–461

153 Gnesutta, N. et al. (2001) The serine/threonine kinase PAK4 preventscaspase activation and protects cells from apoptosis. J. Biol. Chem.276, 14414–14419

154 Cotteret, S. et al. (2003) p21-Activated kinase 5 (Pak5) localizes tomitochondria and inhibits apoptosis by phosphorylating BAD. Mol.Cell. Biol. 23, 5526–5539

155 Jin, Z. et al. (2005) Survival function of protein kinase Ci as a novelnitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-activatedbad kinase. J. Biol. Chem. 280, 16045–16052

156 Harada, H. et al. (2001) p70S6 kinase signals cell survival as well asgrowth, inactivating the pro-apoptotic molecule BAD. Proc. Natl.Acad. Sci. U.S.A. 98, 9666–9670

157 Tan, Y. et al. (2000) BAD Ser-155 phosphorylation regulates BAD/Bcl-XL interaction and cell survival. J. Biol. Chem. 275, 25865–25869

158 Lizcano, J.M. et al. (2000) Regulation of BAD by cAMP-dependentprotein kinase is mediated via phosphorylation of a novel site, Ser155.Biochem. J. 349, 547–557

159 Lowman, X.H. et al. (2010) The proapoptotic function of Noxa inhuman leukemia cells is regulated by the kinase Cdk5 and byglucose. Mol. Cell 40, 823–833

160 Lei, K. and Davis, R.J. (2003) JNK phosphorylation of Bim-relatedmembers of the Bcl2 family induces Bax-dependent apoptosis. Proc.Natl. Acad. Sci. U.S.A. 100, 2432–2437

161 Shao, Y. and Aplin, A.E. (2012) ERK2 phosphorylation of serine 77regulates Bmf pro-apoptotic activity. Cell Death Dis. 3, e253

162 Lopez, J. et al. (2012) Src tyrosine kinase inhibits apoptosis throughthe Erk1/2- dependent degradation of the death accelerator Bik. CellDeath Dis. 19, 1459–1469

163 Arokium, H. et al. (2007) Substitutions of potentiallyphosphorylatable serine residues of Bax reveal how they mayregulate its interaction with mitochondria. J. Biol. Chem. 282,35104–35112

164 Linseman, D.A. et al. (2004) Glycogen synthase kinase-3betaphosphorylates Bax and promotes its mitochondrial localizationduring neuronal apoptosis. J. Neurosci. 24, 9993–10002

165 Shen, Z.J. et al. (2009) The peptidyl-prolyl isomerase Pin1 facilitatescytokine-induced survival of eosinophils by suppressing Baxactivation. Nat. Immunol. 10, 257–265

166 Gardai, S.J. et al. (2004) Phosphorylation of Bax Ser184 by Aktregulates its activity and apoptosis in neutrophils. J. Biol. Chem.279, 21085–21095

167 Xin, M. and Deng, X. (2006) Protein phosphatase 2A enhances theproapoptotic function of Bax through dephosphorylation. J. Biol.Chem. 281, 18859–18867

168 Xin, M. et al. (2007) Protein kinase Cz abrogates the proapoptoticfunction of Bax through phosphorylation. J. Biol. Chem. 282, 21268–21277

169 Kim, B.J. et al. (2006) JNK- and p38 kinase-mediatedphosphorylation of Bax leads to its activation and mitochondrialtranslocation and to apoptosis of human hepatoma HepG2 cells. J.Biol. Chem. 281, 21256–21265

170 Owens, T.W. et al. (2009) Apoptosis commitment and activation ofmitochondrial Bax during anoikis is regulated by p38MAPK. CellDeath Differ. 16, 1551–1562

171 Fox, J.L. et al. (2010) Tyrosine dephosphorylation is required for Bakactivation in apoptosis. EMBO J. 29, 3852–3868

172 Azad, A. et al. (2012) Blockade of the BAK hydrophobic groove byinhibitory phosphorylation regulates commitment to apoptosis. PLoSONE 7, e49601

173 Wang, J. et al. (2011) Bcl-xL phosphorylation at Ser49 by polo kinase 3during cell cycle progression and checkpoints. Cell. Signal. 23, 2030–2038

174 Terrano, D. et al. (2010) Cyclin-dependent kinase 1-mediated Bcl-xL/Bcl-2 phosphorylation acts as a functional link coupling mitotic arrestand apoptosis. Mol. Cell. Biol. 30, 640–656

175 Wang, J. et al. (2012) Phospho-Bcl-x(L)(Ser62) plays a key role at DNAdamage-induced G(2) checkpoint. Cell Cycle 11, 2159–2169

176 Ito, T. et al. (1997) Bcl-2 phosphorylation required for anti-apoptosisfunction. J. Biol. Chem. 272, 11671–11673

177 Deng, X. et al. (2000) Survival function of ERK1/2 as IL-3-activated,staurosporine-resistant Bcl2 kinases. Proc. Natl. Acad. Sci. U.S.A. 97,1578–1583

178 Yamamoto, K. et al. (1999) BCL-2 is phosphorylated and inactivatedby an ASK1/Jun N-terminal protein kinase pathway normallyactivated at G(2)/M. Mol. Cell. Biol. 19, 8469–8478

179 Dimmeler, S. et al. (1999) Dephosphorylation targets Bcl-2 forubiquitin-dependent degradation: a link between the apoptosomeand the proteasome pathway. J. Exp. Med. 189, 1815–1822

180 Breitschopf, K. et al. (2000) Posttranslational modification of Bcl-2facilitates its proteasome-dependent degradation: molecularcharacterization of the involved signaling pathway. Mol. Cell. Biol.20, 1886–1896

181 Kobayashi, S. et al. (2007) Serine 64 phosphorylation enhances theantiapoptotic function of Mcl-1. J. Biol. Chem. 282, 18407–18417

182 Johnson, C.E. et al. (2007) Differential Apaf-1 levels allow cytochromec to induce apoptosis in brain tumors but not in normal neural tissues.Proc. Natl. Acad. Sci. U.S.A. 104, 20820–20825

13

Page 14: Evading apoptosis in cancer

Review Trends in Cell Biology xxx xxxx, Vol. xxx, No. x

TICB-985; No. of Pages 14

183 Tamm, I. et al. (2004) XIAP expression correlates with monocyticdifferentiation in adult de novo AML: impact on prognosis. Hematol.J. 5, 489–495

184 Burstein, D.E. et al. (2008) Immunohistochemical detection of the X-linked inhibitor of apoptosis protein (XIAP) in cervical squamousintraepithelial neoplasia and squamous carcinoma. Ann. Diagn.Pathol. 12, 85–89

185 Moussata, D. et al. (2012) XIAP as a radioresistance factor andprognostic marker for radiotherapy in human rectaladenocarcinoma. Am. J. Pathol. 181, 1271–1278

186 Dan, H.C. et al. (2004) Akt phosphorylation and stabilization of X-linked inhibitor of apoptosis protein (XIAP). J. Biol. Chem. 279, 5405–5412

187 Nakhaei, P. et al. (2012) IkB kinase e-dependent phosphorylation anddegradation of X-linked inhibitor of apoptosis sensitizes cells to virus-induced apoptosis. J. Virol. 86, 726–737

188 Oltersdorf, T. et al. (2012) An inhibitor of Bcl-2 family proteinsinduces regression of solid tumours. Nature 435, 677–681

189 Tse, C. et al. (2008) ABT-263: a potent and orally bioavailable Bcl-2family inhibitor. Cancer Res. 68, 3421–3428

190 Souers, A.J. et al. (2013) ABT-199, a potent and selective BCL-2inhibitor, achieves antitumor activity while sparing platelets. Nat.Med. 19, 202–208

14

191 Yecies, D. et al. (2010) Acquired resistance to ABT-737 in lymphomacells that up-regulate MCL-1 and BFL-1. Blood 115, 3304–3313

192 Wei, G. et al. (2012) Chemical genomics identifies small-moleculeMCL1 repressors and BCL-xL as a predictor of MCL1 dependency.Cancer Cell 21, 547–562

193 Cohen, N.A. et al. (2012) A competitive stapled peptide screenidentifies a selective small molecule that overcomes MCL-1-dependent leukemia cell survival. Chem. Biol. 19, 1175–1186

194 Vo, T.T. et al. (2012) Relative mitochondrial priming of myeloblastsand normal HSCs determines chemotherapeutic success in AML. Cell151, 344–355

195 Parrish, A.B. et al. (2013) Cellular mechanisms controlling caspaseactivation and function. Cold Spring Harb. Perspect. Biol. 5, a008672

196 Tait, S.W. et al. (2010) Resistance to caspase-independent cell deathrequires persistence of intact mitochondria. Dev. Cell 18, 802–813

197 Suzuki, A. et al. (2004) Regulation of caspase-6 and FLIP by theAMPK family member ARK5. Oncogene 23, 7067–7075

198 Peng, C. et al. (2011) Phosphorylation of caspase-8 (Thr-263) byribosomal S6 kinase 2 (RSK2) mediates caspase-8 ubiquitinationand stability. J. Biol. Chem. 286, 6946–6954

[199] Matthess, Y. et al. (2010) Cdk1/cyclin B1 controls Fas-mediatedapoptosis by regulating caspase-8 activity. Mol. Cell. Biol. 30,5726–5740