Flt3 receptor inhibition reduces constitutive NFjB ... - Springer Link

5 downloads 0 Views 699KB Size Report
Stéphane De Botton Æ Alain Israel Æ Pierre Fenaux Æ. Guido Kroemer. Published online: 1 August 2008. Ó Springer Science+Business Media, LLC 2008.
Apoptosis (2008) 13:1148–1161 DOI 10.1007/s10495-008-0243-4

ORIGINAL PAPER

Flt3 receptor inhibition reduces constitutive NFjB activation in high-risk myelodysplastic syndrome and acute myeloid leukemia Jennifer Grosjean-Raillard Æ Lionel Ade`s Æ Simone Boehrer Æ Maximilien Tailler Æ Claire Fabre Æ Thorsten Braun Æ Ste´phane De Botton Æ Alain Israel Æ Pierre Fenaux Æ Guido Kroemer Published online: 1 August 2008 Ó Springer Science+Business Media, LLC 2008

Abstract High-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are characterized by the activation of the anti-apoptotic transcription factor NFjB, via the IKK complex. Here, we show that constitutive activation of the receptor tyrosine kinase Flt3 is responsible for IKK activation. Chemical inhibition or knockdown of Flt3 with small interfering RNAs reduced NFjB activation in MDS and AML cell lines, as well as in

Author contributions: J.G-R. performed the experiments and analyzed the data. L.A., S.B., C.F., T.B. and S.d.B. provided bone marrow samples and essential clinical information on patients. A.I and P.F. participated in the conception of the study. G.K. conceived and directed the study. J.G-R and G.K. wrote the paper.

Electronic supplementary material The online version of this article (doi:10.1007/s10495-008-0243-4) contains supplementary material, which is available to authorized users. J. Grosjean-Raillard  L. Ade`s  S. Boehrer  M. Tailler  C. Fabre  G. Kroemer (&) INSERM, U848, Institut Gustave Roussy, PR1, 38 rue Camille Desmoulins, 94805 Villejuif, France e-mail: [email protected] J. Grosjean-Raillard  L. Ade`s  S. Boehrer  M. Tailler  C. Fabre  S. De Botton  G. Kroemer Institut Gustave Roussy, 94805 Villejuif, France J. Grosjean-Raillard  L. Ade`s  S. Boehrer  M. Tailler  C. Fabre Universite´ Paris Sud, Paris 11, 94805 Villejuif, France L. Ade`s  S. Boehrer  T. Braun  P. Fenaux Service d’He´matologie Clinique, Hoˆpital Avicenne, AP-HP, Universite´ Paris XIII, Bobigny, France A. Israel Unite´ de Signalisation Moleculaire et Activation Cellulaire, URA 2582 CNRS Institut Pasteur, 75015 Paris, France

123

primary CD34+ bone marrow cells from patients, causing apoptosis. Epistatic analysis involving the simultaneous inhibition of Flt3 and IKK suggested that both kinases act in the same anti-apoptotic pathway. An IKK2 mutant with a constitutive kinase activity and a plasma membranetethered mutant of NEMO that activates IKK1/2 prevented the cytocidal action of Flt3 inhibition. Flt3 phosphorylates IKK2 in vitro, and Flt3 inhibition reduced the phosphorylation of IKK2 in MDS or AML cell lines. IKK2 and Flt3 physically associated in MDS and AML cells, and Flt3 inhibition disrupted this interaction. Flt3 inhibition only killed CD34+ bone marrow cells from high-risk MDS and AML patients, in correlation with blast numbers and NFjB activity, yet had no lethal effect on healthy CD34+ cells or cells from low-risk MDS. These results suggest that Flt3 inhibitors might exert an anti-neoplastic effect in high-risk MDS and AML through inhibition of NFjB. Keywords IKK

Acute myeloid leukemia  Flt3  NFjB 

Abbreviations Flt3 Fms-like tyrosine kinase-3 Flt3I Flt3 inhibitor AML Acute myeloid leukemia BMMNC Bone marrow mononuclear cells NFjB Nuclear factor-jB DAPI 40 ,6-diaminidino-2-phenylindole DiOC6(3) 3,30 dihexyloxacarbocyanine iodide GAPDH Glyceraldehyde-3-phosphate dehydrogenase IjB Inhibitor of NFjB IKK IjB kinase MDS Myelodysplastic syndrome

Apoptosis (2008) 13:1148–1161

PI Z-VAD.fmk

Propidium iodide N-benzyloxycarbonyl-Val-AlaAsp-fluoromethylketone

Introduction Myelodysplastic syndrome (MDS) is characterized by ineffective hematopoiesis, refractory cytopenia and a tendency to progress into acute myeloid leukaemia (AML). A plethora of genetic aberrations have been detected in patients with MDS, and some of these genetic alterations are associated with the progression of MDS to leukemia. Activating mutations of the receptor tyrosine kinase fmslike tyrosine kinase 3 (Flt3), a member of the class III receptor tyrosine kinase family, are the most common tyrosine kinase mutations in acute myeloid leukemia (AML). Flt3 is highly expressed in most patients with AML and approximately 30% of these patients have activating mutations in Flt3. Nearly 30% of AML patients possess an internal tandem duplication (ITD) within the juxtamembrane domain and an additional 7% have point mutations such as the D835Y mutation within the catalytic domain of the kinase [1, 2]. Moreover, increased levels of Flt3 transcripts are observed in a large number of AML patients and this increased expression may also contribute to the phosphorylation and activation of Flt3 [3]. Notably, more than two-thirds of AML patients exhibit the (auto-) phosphorylation of Flt3 on tyrosine residues 589 and 591 even in the absence of activating mutations [4]. Both types of mutation result in constitutive activation of Flt3 leading to activation of downstream signaling pathways, giving cells a proliferative and survival advantage [5–7]. Flt3 phosphorylation/activation results in the activation of downstream kinase pathways, like the Ras/mitogen-activated protein kinase (MAPK), allowing abnormal cell growth and aberrant gene regulation [8–10]. Only a few studies have analyzed Flt3 alterations in patients with MDS and suggest that Flt3 mutations are rare in MDS, affecting 5–10% of high-risk MDS [1, 5, 11]. Several master regulators of proliferation and apoptosis are transactivated by the nuclear factor kappa B (NFjB) [12, 13], a transcription factor that is often activated in neoplasia. It has been reported that, in most if not all cases of AML, NFjB is constitutively activated and its target genes are upregulated [14]. Recently, it has been reported that NFjB can be activated via the Ras/phosphatidylinositol-3-kinase (PI3K)/protein kinase B pathway in AML [15]. NFjB is also constitutively active in high-risk MDS (with high or int-2 IPSS scores), yet inactive in low risk MDS (with low or int-1 IPSS) [16]. This correlates with increased spontaneous apoptosis inhibition in high-risk

1149

MDS myeloblasts. Inhibition of NFjB can trigger apoptosis in high-risk MDS as well as in AML [17]. NFjB activation depends on phosphorylation and degradation of inhibitor proteins collectively termed IjB, which sequester NFjB in the cytoplasm. Stimulation of the NFjB pathway leads to the rapid phosphorylation of IjBs and consequent degradation, resulting in NFjB translocation into the nucleus. NFjB molecules exist as dimers of the ‘rel’ protein family, whose members include p50/p105 (NFjB1), p52/100 (NFjB2), p65 (RelA), RelB and c-Rel. Members of the rel family are characterized by a conserved N-terminal 300 amino acid sequence, the rel homology domain. This domain enables dimerisation, DNA binding, nuclear localisation and interaction with the IjB family of proteins. The phosphorylation of IjBa on serine residues 32 and 36 is initiated by an IjB kinase (IKK) complex which, at least in the canonical NFjB activation pathway, includes a catalytic heterocomplex composed of IjB kinase (IKK)-1, IKK-2 and NEMO [12, 13, 18, 19]. According to one study, Flt3 activation by addition of the Flt3 ligand (Flt3L) can trigger NFjB activation [20]. Stimulated by these report, we investigated whether inhibition of Flt3 might inhibit NFjB activation in high-risk MDS and AML. To date, several different Flt3 kinase inhibitors have been developed and tested in vitro [21–23] and in early clinical trials with variable degrees of success [24–26]. Up to 80% of patients in these studies achieved blast reduction over a few months. Another approach for targeting Flt3 involves RNA interference (siRNA), which induced down regulation of Flt3 expression [27]. Thus, the aim of this study was to examine whether Flt3 depletion or inhibition would induce cell death through an effect on the NFjB pathway and, if so, through which molecular mechanisms.

Material and methods Patients AML, MDS patients and healthy subjects were included in our study (Table 1). Informed consent of all patients and healthy subjects was provided according to the Declaration of Helsinki. This study was approved by the Institut Gustave Roussy institutional review board. MDS patients were previously untreated except for supportive care. The diagnosis of MDS was based on peripheral blood counts, cytology of peripheral blood and bone marrow (BM) according to the WHO classification [28] and conventional cytogenetic analysis. These data evaluation of the individual International Prognostic Scoring System (IPSS) score for each patient [16]. BM aspirates were collected, after informed consent, into syringes containing media

123

1150

supplemented with EDTA. The BM mononuclear cell (BMMNC) fraction was isolated by density gradient centrifugation using Ficoll-Paque Plus (Amersham Biosciences, Sunnyvale, CA, USA) and then washed three times (10 min, 600g, 4°C) in complete medium. Cells and culture conditions The MDS cell line MOLM-13 and the AML cell lines MV4-11 and KG1 were purchased from the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, Germany. All cell lines were cultured in Roswell Park Memorial Institute 1640 medium (Gibco, Carlsbad, CA, USA) supplemented with 10% heat-inactivated fetal calf serum (FCS), 2 mM L-glutamine, 100 IU/ml penicillin and 100 lg/ml streptomycin as described [29]. BM cells were isolated and processed as follows: after density gradient (Ficoll-Paque Plus) separation of BM mononuclear cells (BMMNC), CD34+ cells were isolated by positive selection with the MiniMacs system (Miltenyi Biotec, Bergish Gladbach, Germany) and then maintained in Iscove’s modified Dulbecco’s medium (Gibco) supplemented with 10% heat-inactivated FCS. We used distinct inhibitors to suppress the enzymatic activity of Flt3, in particular the Flt3 inhibitor 2-acylaminothiophene-(3,4-di-O-methylphenyl)-3-carboxamide (Flt3I) (Calbiochem, Darmstadt, Germany). To induce Flt3 activation in KG-1 cell, Flt3 ligand (Fl3L, Calbiochem) was used overnight at a concentration of 10 lg/ml. Transfection of plasmids and knockdown of Flt3 by siRNA

Apoptosis (2008) 13:1148–1161

(Becton Dickinson, Mountain View, CA, USA) as described previously [31, 32]. Cells were stained with propidum iodine (PI, 10 lM, Sigma, St Louis, MO, USA) and 20 nM fluorochrome DiOC6(3) (Molecular Probes, Eugene, OR, USA) for 15 min at 37°C, for the determination of plasma membrane permeability and the mitochondrial transmembrane potential (DWm), respectively [33, 34]. Nuclear protein extraction and electrophoretic mobility shift assay Nuclear extracts were prepared from all cell lines after culture in the presence or absence of Flt3 inhibitor (standard treatment: 2 lM, 2 h). Cells were harvested and washed twice with ice-cold phosphate-buffered saline (PBS). Cell pellets were then lysed in hypotonic lysis buffer (10 mM N-2-hydroxyethylpiperazine-N0 -2-ethanesulfonic acid-potassium hydroxide (HEPES), 1.5 mM MgCl2, 10 mM KCl and 0.0125% NP-40, pH 7.9). After incubation on ice for 10 min, nuclei were separated from the cytosolic extracts by centrifugation (13,000g, 5 min at 4°C). The nuclear pellets were resuspended in hypertonic extraction buffer (5 mM HEPES, 1 mM MgCl2, 0.2 mM ethylenediaminetetra-acetic acid (EDTA), 0.5 M NaCl, 25% glycerol, and 0.025% NP-40, pH 7.0) for 30 min at 4°C under agitation. After centrifugation at 13,000g, 15 min at 4°C), supernatants containing nuclear proteins were removed and stored at -70°C. Nuclear extracts were examined for NFjB-binding activity by EMSA, using a non-radioactive EMSA kit (Panomics, Freemont, CA, USA). Immunofluorescence stainings

Cells were transfected with the Nucleofactor system (Amaxa, Cologne, Germany) using siRNAs specific for GFP (50 -GCAAGCTGACCCTGAAGTTCA-30 ) with no homology to human genes (EGFP siRNA; InvivoGen, San Diego, USA), a pool of siRNAs specific for Flt3 (Upstate, Lake Placid, NY, USA), two individual validated Flt3 siRNAs (Hs_Flt3_6 HP and Hs_Flt3_7 HP QIAGEN, Valencia, California, USA), siRNAs specific for p65, IKK1, IKK2 or NEMO (described in Ref. [30]) and/or plasmids VSV-IKK2 h, Flag-IKK2hSS[EE, Flag-NEMOm, Myr-NEMOm(MN)40, [31]. Cells were used 48 h after transfection. Assessment of apoptosis Cells (105) from all MDS and AML cell lines or patient cells were resuspended in 1 ml of culture medium and incubated in the presence or absence of 2 lM Flt3I and/or 50 lM Z-VAD-fmk, for 48 h. Apoptotic cells were detected by flow cytometric analysis using a FACScan

123

Cell lines or patient cells (105) were allowed to adhere to polylysin-L coverslips (Sigma) and fixed in PBS containing 4% paraformaldehyde at room temperature. Cells were then permeabilized either with 0.05% Triton X-100 (Boehringer, Mannheim, Germany) or 0.1% sodium dodecyl sulfate (SDS) for 10 min, washed in PBS and stained with antibodies specific for p65 (rabbit polyclonal Ab, Santa Cruz Biotechnology, Santa Cruz, CA, USA) and revealed with a goat anti-rabbit IgG coupled with Alexa 568 (Molecular Probes). Nuclei were counterstained with DAPI (Molecular Probes) allowing for the discernment of chromatin condensation [31]. 200 cells for each slide were examined independently with a LSM 510 confocal microscope (Zeiss) at 63-fold magnification. Immunoblots Cellular lysates (50 lg protein/lane) were subjected to SDS/polyacrylamide gel electrophoresis, transferred on

78

70 76

24

25 26

85

70

23

27

55

86

14

87

87

13

22

85 58

11 12

21

69

10

56

72

9

20

56

8

61 83

77

7

18 19

78

6

84

55

5

17

57

4

69

63

3

67

59

2

15

47

1

16

Age

Number

47,XY, +8

46,XX 46,XY

46,XY, del(5q)

46,XX

45,XY, del(20q)

46,XX

46,XY

46,XY 46,XY, del(12p)

46,XX, del(11q)

46,XX

46,XY

46,XY

46,XY, del(20q)

46,XY 46,XY

Failure

46,XY

46,XY

46,XX

46,XX

46,XY

46,XY

46,XX

46,XX

46,XX

Karyotype

RAEB-2

RAEB-2 RAEB-2

RAEB-2

RAEB-2

RAEB-2

RAEB-2

RAEB-2

RAEB-1 RAEB-2

RAEB-1

RAEB-1

CMML

CMML

RCMD

RARS RCMD

RAEB-1

Pure RA

Pure RA

Pure RA

Pure RA

Control

Control

Control

Control

Control

Disease

High

INT-2 INT-2

INT-2

INT-2

INT-2

INT-2

INT-2

INT-2 High

INT-2

INT-2

INT-2

INT-1

Low

Low Low

NA

INT-1

Low

Low

Low

MDS IPSS score

Table 1 Characteristics of MDS patients with nuclear NFjB analysis

High-risk MDS

Low risk MDS

Control

16

11 16

16

11

11

12

15

13 13

11

15

11

1

2

0 1

7

0

0

0

0

0

0

0

0

0

% Bone marrow blasts

+

+

+

+

-

+

+

+ ND

+

-

+

-

-

+ +

ND

ND

-

-

-

-

-

-

-

-

Phospho-Flt3 immunostaining

66

26 65

70

24

10

17

18

76 14

56

23

19

58

9

32 84

8

5

2

3

0

0

0

0

0

0

% Nuclear NFjB staining without Flt3I

18

18 16

26

17

10

8

5

21 6

3

19

9

17

8

2 53

4

2

2

3

0

0

0

0

0

0

% Nuclear NFjB staining with Flt3I

48

41 35

37

48

49

56

65

57 59

63

72

70

56

55

62 64

68

71

76

73

75

10

8

12

17

21

% Apoptosis without Flt3I

63

64 65

63

60

69

60

71

75 70

74

80

81

65

58

65 75

76

78

82

92

84

9

15

25

22

21

% Apoptosis with Flt3I

Apoptosis (2008) 13:1148–1161 1151

123

sAML

sAML

46,complex

46,XY 36

78

sAML 46,XY, del(5q)

35

34

85

AML 47,XY, +21 33

62

AML 48,XY, +8+21 32

42

AML 46,XY 31

41

AML 46,XY, inv(16)

66

AML 46,XY 70

36 30

29

28

RA, refractory anemia; RARS, RA with ring sideroblasts; RCMD, efractory cytopenia with multilineage dysplasia; CMML, chronic myelomonocytic leukaemia; RAEB, refractory anemia with excess of blasts (5–9% BM blasts RAEB-1, 10–19% BM blasts RAEB-2); sAML, secondary aML; ND: not determined

58

56

23 25 31 54

ND

21

60 28

32

29

84

-

50

33

+

22

80

63 25 48 30

ND

31

74

45 59 50

+

11

86

44 22 82 90

+

50

82 65

11 89

17 38

+ 68

55

-

54 8 64 AML 46,XY

123

56

Disease Karyotype Age Number

Table 1 continued

MDS IPSS score

AML

70

+

% Apoptosis without Flt3I % Nuclear NFjB staining without Flt3I % Bone marrow blasts

Phospho-Flt3 immunostaining

% Nuclear NFjB staining with Flt3I

88

Apoptosis (2008) 13:1148–1161 % Apoptosis with Flt3I

1152

nitrocellulose membranes and immunochemical detection was carried out using antibodies specific for IjBa (rabbit polyclonal Ab, Santa Cruz Biotechnology, Santa Cruz, CA, USA), Phospho-IKK1-2 (Cell Signaling Technology Boston, Massachusetts, USA), IKK1 (rabbit polyclonal Ab, Santa Cruz Biotechnology, Santa Cruz, CA, USA), IKK2 (rabbit polyclonal Ab, Santa Cruz Biotechnology, Santa Cruz, CA, USA), NEMO (rabbit polyclonal Ab, Santa Cruz Biotechnology, Santa Cruz, CA, USA), phosphotyrosine (Cell Signaling Technology, Boston, Massachusetts, USA) or glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (Abcam, Cambridge, Massachusetts, USA). Immunoprecipitation Following treatment, cells were lysed in RIPA buffer (50 mM Tris HCl pH 8, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS) and 200 lg protein were immunoprecipitated with the antibody of interest for 2 h and incubated with protein G-coupled sepharose beads (Roche Applied Science, Rockford, llinois, USA) overnight at 4°C. Immunoprecipitates were obtained following several centrifugation/wash cycles in lysis buffer followed by immunoblotting. Flt3 kinase assay An IKK2-derived peptide was purchased from Merck (IKKb, HisTagÒ, Merck Chemicals Ltd, Nottingham, UK) and was biotinylated using EZ-LinkTM Sulfo-NHS-Biotin according to manufactures instruction (Pierce, Rockford, Illinois, USA). Biotinylated peptide was then evaluated for phosphorylation by Flt3 using a specific assay kit (Cell Signalling Technology for Perkin Elmer-Delfia, Boston, Massachusetts, USA). In a total reaction volume of 50 ll, 100 ng recombinant Flt3 kinase, 1.5 lM biotinylated peptide, 20 lM ATP and variable amounts of Flt3I were␣allowed to react at room temperature for 30 min. The reaction was then stopped and samples loaded on a streptavidin coated plate. The phosphorylation of biotinyated-IKK2 peptide by Flt3 kinase was detected using a phospho-tyrosine-specific mAb (P-Tyr-100), and Europium-labeled secondary antibody (Cell Signalling Technology for Perkin Elmer-Delfia, Boston, Massachusetts, USA), and the measurement of European-dependent fluorescence (excitation at 340 nm and emission at 615 nm).

Results Pharmacological inhibition of Flt3 also inhibits NFjB and induces apoptosis in MDS and AML cell lines. We chose to investigate the effects of Flt3 inhibition on two distinct

Apoptosis (2008) 13:1148–1161

1153

cell lines that both express active, phosphorylated Flt3 (on tyrosines 589/591), namely the MOLM-13 cell line (which is derived from a patient with high-risk MDS) [2, 35] and the MV4-11 cell line (which is derived from a patient with

AML) [2] (Fig. 1a). When Flt3 was immunoprecipiated from MOLM-13 cells (Fig. 1b) or MV4-11 cells (not shown), the protein was found to react with an antibody recognizing phosphotyrosine residues irrespective of their

Fig. 1 Apoptosis induction by Flt3 inhibition in MOLM-13 and MV4-11 cells. (a) Immunoblot detection of Flt3 and its activating phosphorylation in different cell lines. Extracts from the indicated cell lines were subjected to SDS PAGE and immunoblot detection of Flt3 and GAPDH as a loading control. Note the presence of a band with a reduced electrophoretic mobility in MOLM-13 and MV4-11 cells. (b) Tyrosine phosphorylation of Flt3 in MOLM-13 cells. Flt3 was immunoprecipated and the immunoprecipiated was immunoblotted for the detection of Flt3, phosphotyrosine residues and phospho-Flt3, as described in Materials and Methods. Whole cell lysates and an immunoprecipitation with non-specific rabbit IgG were included as controls. (c, d). Detection of mitochondrial signs of apoptosis after treatment with Flt3I. MOLM-13 or MV4-11 cells were cultured in the

absence or presence of Flt3I (2 lM) and/or Z-VAD-fmk (50 lM) for 18 h and then stained with the DWm-sensitive dye DiOC6(3) and the vital dye propidium iodide (PI), followed by cytofluorometric analysis. The percentage (X ± SD of triplicates) of dying (DiOC6(3)low PI-) or dead (DiOC6(3)low PI-) cells are represented for MOLM-13 cells in B and for MV4-11 cells in C. (e–g) Detection of apoptotic DNA loss after Flt3 inhibition (2 lM, 18 h). Cells were ethanol-fixed, treated with RNAse, labeled with PI and subjected to cell cycle analysis. Representative FACS histograms are shown in d (bars indicate the gate containing subdiploid cells) for MOLM-13 cells cultured as in b. Quantitative data (X ± SD of triplicates) are represented for MOLM-13 cells in e and for MV4-11 cells in f

123

1154

Apoptosis (2008) 13:1148–1161

Fig. 2 Effect of Flt3 inhibition on the NFjB system in MOLM-13 and MV4-11 cells. (a, b) Detection of activating phosphorylations of Flt3 (on tyrosines 589/591), IKK1/2 (serine 176 or 180), and IjBa degradation after addition of Flt3I (2 lM, 2 h) to the indicated cell types. (c, d) Inhibition of NFjB by Flt3, as determined by different techniques in MOLM-13 and MV4-11 cells. The abundance of NFjB

in nuclear extracts prepared from untreated or Flt3-inhibited (2 lM, 2 h) was determined by EMSA (c). Alternatively, the presence of p65 in nuclei was determined by immunofluorescence (d). Percentages refer to the frequency of cells that contain NFjB in their nuclei. All experiments were repeated at least three times, yielding similar results

surrounding peptide sequence, as well as with an antibody that specifically recognizes phosphorylated Flt3 (on tyrosines 589/591) (Fig. 1b). Addition of the Flt3 inhibitor 2acylaminothiophene-(3,4-di-O-methylphenyl)-3-carboxamide (Flt3I) [36] induced signs of apoptosis in both cell lines, namely a loss of the mitochondrial transmembrane potential (DWm, determined by staining with the DWm-sensitive dye DiOC6 (3)) that preceded the loss of viability (determined with the vital dye propidium iodide, PI) (Fig. 1c, d) and DNA degradation leading to an accumulation of cells with a subdiploid DNA content (Fig. 1e–g). Addition of the pan-caspase inhibitor Z-VAD-fmk had a minor effect on these apoptosis-associated parameters, suggesting that they occurs in a mostly caspase-independent fashion (Fig. 1e–g). MOLM-13 and MV4-11 cells exhibit constitutive activation of the NFjB pathway, and inhibition of NFjB kills these cells efficiently [30]. In both MOLM-13 and MV4-11 cells, Flt3I greatly reduced the activating phosphorylation of Flt3 (Fig. 2a), as well as the phosphorylation of the inhibitor of NFjB kinase (IKK) subunits 1/2 (IKK1/2), as observed using a phospho-neoepitope-specific antibody. Flt3I also enhanced the abundance of the IKK substrate, inhibitor of NFjB (IjBa) (Fig. 2b). Concomitant with the inhibition of IKK1/2 and IjB degradation, Flt3I reduced the nuclear presence of nuclear NFjB, as determined by by electrophoretic mobility shift assays (Fig. 2c) or immunofluorescence detection of the p65 NFjB subunit (Fig. 2d),

in both MOLM-13 and MV4-11 cells. Again, these effects were rapid, leading to strong NFjB inhibition within 1–2 h (Fig. 2c, d). Since chemical inhibitors of tyrosine kinases often (if not always) inhibit other targets than the kinase that they had originally been designed for, we attempted to exclude possible off-target effects of Flt3I by an alternative strategy of Flt3 inhibition. Knockdown of Flt3 with a pool of specific siRNAs (which was maximal 48 h after transfection) reduced the phosphorylation of IKK1/2 and increased the abundance of IjBa (Fig. 3a), as well as that of EMSAdetectable NFjB in the nucleus (Fig. 3b), while causing cell death (Fig. 3c). Note that the depletion of Flt3 was near to complete (Fig. 3) when still 30–40% of the cells were viable (Fig. 3f), indicating that the disappearance of immunodetectable Flt3 cannot be explained by massive cell death. Similar results were obtained when Flt3 was depleted by two distinct individual siRNAs, resulting in reduced IKK1/2 phosphorylation, enhanced IjBa expression as well as cell death (Fig. 3d–f). Altogether, these results indicate that Flt3 knockdown can inhibit the IKKdependent activation of NFjB and stimulate apoptosis in cell lines that express constitutively active Flt3. In a well characterized AML cell line, KG1 (which does not manifest any activating phosphorylation of Flt3, Fig. 1a), inhibition of Flt3 had no pro-apoptotic activity. Conversely, addition of the Flt3 ligand Flt3 reduced the spontaneous cell death of KG1 cells (Supplementary

123

Apoptosis (2008) 13:1148–1161

1155

Fig. 3 Knockdown of Flt3 causes NFjB inhibition and apoptosis. (a–c) Effect of a pool of Flt3-specific siRNAs. MOLM-13 and MV4-11 cells were electroporated with the indicated siRNA pool, and the expression level and phosphorylation status of Flt3, IKK1/2 and degradation of IjBa were detected by immunoblot 48 h later (a). NFjB activation was measured by EMSA (b) and the frequency of dying cells (X ± SD, n = 3) was determined by DiOC6(3)/PI staining (c). (d–e) Effect of individual Flt3-specific siRNAs. MOLM-13 and MV4-11 cells were electroporated with 2 distinct Flt3 siRNA and the expression level Flt3 was detected by immunoblot 48 h later (d), NFjB activity was measured by EMSA (f) the frequency of dying cells (X ± SD, n = 3) was determined by DiOC6(3)/PI staining (e)

Fig. 1a). In these cells, which do not activate NFjB constitutively, ligation of Flt3 with Flt3 enhanced the degradation of IjBa (Supplementary Fig.1b) and activated

NFjB (Supplementary Fig. 1c, d). Taken together, these data suggest that activation of Flt3 is intimately linked to anti-apoptotic NFjB activation.

123

1156

Apoptosis (2008) 13:1148–1161

Fig. 4 Epistatic and functional analysis of the cause-effect relationship between apoptosis induction and IKK inhibition by Flt3 depletion. (a) Knockdown of distinct subunits of the IKK complex by specific siRNAs, as determined 48 h after electroporation of MOLM-13 cells. (b) Epistatic analysis of apoptosis induction by depletion of Flt3. Cells were transfected with the indicated combination of siRNAs (using either the siRNAs used in A or a pool of Flt3-specific siRNAs used in Fig. 3a–c), and cell death (X ± SD, n = 3) was measured by DiOC6(3)/PI staining and cytofluorometry. (c) Transfection-enforced overexpression of wild type (WT) active mutants of IKK2 (CA-IKK2) and NEMO (NEMO-NM). MOLM-13 cells were transfected with vector-only or the indicated cDNAs, and

protein expression was monitored 48 h after transfection. (d) Inhibition of the cytocidal effects of Flt3 knock-down by active IKK2 and NEMO. Cells were simultaneously transfected with control siRNA or FLT-R-specific siRNA and/or the indicated IKK2 and NEMO constructs. Two days later the frequency of dead and dying cells was assessed by the DiOC6(3)/PI method. (e) Abolition of Flt3Imediated killing by activation of the IKK complex. Cells were first transfected with the indicated control constructs and normal or active versions of IKK2 or NEMO. Twenty-four h later the cells were treated for further 18 h with Flt3I, followed by cell death assessment. All results are means ± SD of triplicates of one experiment representative of three or more

Inhibition of Flt3 induces apoptosis through IKK inhibition. As shown above, pharmacological Flt3 inhibition or Flt3 knockdown causes apoptosis and inhibits NFjB. To establish a cause-effect relationship between cell death and NFjB inhibition, we employed three complementary strategies. First, we performed an epistatic analysis of the proapoptotic effects of Flt3 depletion with simultaneous inhibition of different components of the NFjB activation pathway. MOLM-13 cells were transfected with siRNAs that efficiently targeted Flt3, IKK1, IKK2 or NEMO (Fig. 4a), alone or in combination. Both the depletion of Flt3 and that of any of the component of the IKK complex (IKK1, IKK2, NEMO) induced cell death. However, combined Flt3 and IKK depletion was no more efficient

than the inhibition of Flt3 or IKK alone (Fig. 4b), suggesting that Flt3 and IKK act in the same pathway. Second, we transfected MOLM-13 cells with a constitutive active (CA) form of IKK2 (due to two point mutations in the activation loop, S177E and S181E, increasing the kinase activity of the protein) [37] or wild type (WT) IKK2 as a control. Alternatively, the cells were transfected with a version of NEMO that activates the IKK complex (due to introduction of an N-terminal myristoylation site that targets NEMO to the plasma membrane: NEMO-NM) [38] or WT NEMO (Fig. 4c). Only CA-IKK2 and NEMO-NM (but neither WT IKK2 nor WT NEMO) inhibited the cytotoxic effects of the Flt3 knockdown (Fig. 4d) and that of pharmacological Flt3 inhibition (Fig. 4e). Similar results were obtained in MOLM-13 MDS

123

Apoptosis (2008) 13:1148–1161

Fig. 5 Direct impact of Flt3 inhibition on IKK2. (a) Co-immunoprecipitation of Flt3 with IKK2. Untreated KG-1, MOLM-13 or MV4-11 cells were lysed and subjected to immunoprecipitation with an antibody specific for Flt3, followed by immunoblot detection of IKK1 and IKK2. (b) Effect of Flt3I on the interaction between Flt3 and IKK2. MOLM-13 or MV4-11 cells were left untreated or were treated with Flt3I (2 lM, 2 h), followed by immunoprecipitation of Flt3 and detection of IKK2. (c, d). Flt3I inhibits IKK2 phosphorylation on tyrosine residues by Flt3 kinase. By fluorescent kinase assay, increasing doses of Flt3I inhibited phosphorylation of IKK2 by Flt3 kinase. Immunoprecipitation revealed tyrosine of IKK2 in both MOLM-13 and MV4-11 cell lines which was inhibited by Flt3I

cells (Fig. 4) and MV4-11 AML cells (not shown), in line with the hypothesis that Flt3 depletion/inhibition kills through an inhibitory effect on the IKK complex. Moreover, these results provide unequivocal evidence that Flt3 depletion/inhibition kills the cells through a specific pathway (that involves the IKK/NF-jB pathway) rather than through non-specific effects. Third, we investigated whether Flt3 might engage in direct interactions with the IKK complex. Following immunoprecipitation of Flt3 in MOLM-13 and MV4-11

1157

cells, we detected IKK2 (but not IKK1) in the precipitate, suggesting that both kinases might interact within the same polyprotein complex (Fig. 5a). This interaction was strongly diminished upon addition of Flt3I to the cells, as seen by a reduction in the amount of IKK2 that immunoprecipitated with Flt3 (Fig. 5b). When IKK2 was immunoprecipitated, NEMO appeared to belong to the complex but no IKK1 was detected (data not shown). As a further indication for a direct regulatory interaction between Flt3 and IKK2, we found that recombinant Flt3I was able to phosphorylate an IKK2-derived peptide in vitro (Fig. 5c), demonstrating that IKK2 can be a direct substrate of Flt3. Indeed, we found that IKK2 is constitutively tyrosine-phosphorylated in MOLM-13 and MV4-11 cells. This phosphorylation was lost after treatment of the cells with Flt3I (Fig. 5d). Altogether, these results indicate that Flt3 inhibition has a direct impact on the subcellular localization and the phosphorylation status of IKK2 in MOLM-13 and MV4-11 cells, correlating with inhibition of the NFjB system. Ex vivo therapeutic effects of Flt3 inhibition on primary MDS and AML cells. The data presented here have been obtained on cell lines and hence must be interpreted with caution as to their possible clinical relevance. We therefore studied the impact of Flt3I on primary, purified CD34+ bone marrow mononuclear cells (BM-MNC) from a cohort of MDS and AML patients (Table 1). However, the vast majority of high-risk MDS and AML patients showed CD34+ BM-MNC stained positively for phospho-Flt3, correlating with constitutively active NFjB (Table 1). None of the MDS and AML patients included in this study exhibited the internal tandem mutation of Flt3 nor FLT3 Asp(835) (Table 1). CD34+ cells from normal donors or patients were cultured either alone or in the presence of Flt3I for 18 h ex vivo, followed by assessment of their spontaneous and Flt3I-induced mortality, as summarized in Table 1 and depicted in Fig. 6a, b. Flt3I had no significant cytotoxic effects on CD34+ bone marrow cells from five distinct healthy donors (Fig. 6b). As reported [17, 39–42], CD34+ BM cells from low-risk MDS patients succumbed to apoptosis spontaneously, and this enhanced baseline mortality was not accelerated by Flt3I (Fig. 6b). In contrast, CD34+ bone marrow cells from high-risk MDS patients and AML patients exhibited a significant (P \ 0.01, paired Student t-test) apoptotic response to Flt3I (Fig. 6a, b). Next, we assessed parameters of IKK and NFjB inhibition in several AML and high-risk MDS patients whose CD34+ bone marrow cells responded to Flt3I ex vivo (Fig. 6c). For both AML and high-risk MDS patients, we found that Flt3 inhibition reduced the constitutive activation of NFjB, as indicated by a loss of the nuclear p65 immunofluorescence staining (Fig. 6d, e; Table 1).

123

1158

Apoptosis (2008) 13:1148–1161

Fig. 6 Effect of Flt3 inhibition on constitutive NFjB ativation and cell death of purified CD34+ bone marrow cells from MDS and AML patients. (a) Representative DiOC6(3)/PI staining of CD34+ AML bone marrow cells cultured for 18 h in the absence or presence of Flt3I and/or Z-VAD-fmk (as in Fig. 1b). (b) Spontaneous and Flt3I induced cell death in cells from healthy normal volunteers, low-risk MDS, high-risk MDS, and AML (same patients as Table 1). (c) Effect of Flt3I on the presence of the p65 NFjB subunit of CD34+ AML bone marrow cells (same patient as in A), as determined by immunofluorescence. (d) Inhibition of NFjB activation in a cohort of patients with determined blast counts. Lines link individual patients values before and after addition of Flt3I. (e) Nuclear NF-kB in the absence or in the presence of Flt3I for the patients listed in Table 1. Statistical analyses (*\0.05; **\0.01) were performed by means of the paired Student t-test

Altogether, these data allow us to draw two major conclusions. First, constitutive NFjB activation in high-risk MDS and AML correlated with constitutive Flt3 activation (as judged from its phosphorylation status). Second, inhibition of Flt3 reduces this malignancy-associated NFjB activation, presumably through the same pathway that has been delineated for MOLM-13 and MV4-11 cell lines.

Discussion The results presented in this article indicate that Flt3 may be (one of) the upstream kinase(s) that account for constitutive NFjB activation in high-risk MDS and AML. This notion is based on the comparative analysis of Flt3 inhibition (either by means of a pharmacological inhibitor that lacks an off-target

123

effect on IKK2, Flt3I, or using distinct small-interfering RNAs) in MDS and AML cell lines or patient-derived, primary bone marrow cells. The epistatic and functional analyses, as well as the correlative data obtained on clinical samples, provide a coherent vision of the hierarchy between IKK and Flt3. Flt3 appears to be constitutively active and hence manifests an activating (auto)-phosporylation on tyrosines 589 and 591, in particular in high-risk (but less so in low-risk) MDS and AML cells. When active, Flt3 associates with IKK2 (but not or only loosely with IKK1). This step most probably occurs close to or at the plasma membrane and involves tyrosine phosphorylation of IKK2 by Flt3. It leads to activation of the IKK complex resulting in NFjB activation (through the phosphorylation and degradation of IjB). Inhibition of Flt3 strongly reduced the tyrosine phorphorylation of IKK2, dissociates the Flt3/IKK2 complex, thereby preventing NFjB activation and triggering

Apoptosis (2008) 13:1148–1161

apoptosis in those cells that are ‘‘addicted’’ to NFjB. As a caveat, it has to noted however, that the effect of Fl3I on NFjB activation and apoptosis induction do not correlate completely in clinical samples and that myeloblasts from some high-risk MDS patients did not manifest a major increment of apoptosis upon culture with Flt3I, pointing to a poorly understood heterogeneity in the patient population. Thus, a majority of AML and high-risk MDS patients follow the pattern delineated above. Nonetheless, a sizeable minority may demonstrate apoptotic responses to Flt3 inhibition without that NFjB would be involved in this process and/or activate NFjB through Flt3-independent pathways. Although these results move us one step further towards the comprehension of constitutive NFjB activation in high-risk MDS and AML, they lead us immediately to the next, yet unresolved question. If an activated Flt3 accounts for IKK activation, what are the molecular mechanisms involved in Flt3 activation? The MOLM-13 and MV4-11 cell lines have been shown to possess internal tandem duplication that activate the tyrosine kinase activity of Flt3, rendering it independent from the ligand [43]. However, activating Flt3 mutations have only been found in a minority of MDS (*5%) and AML (25%) patients [2, 11]. Nevertheless, in our cohort, most high-risk MDS and AML patients expressing phosphorylated Flt3 responded to Flt3I ex vivo, suggesting that Flt3 activation may occur independently from activating mutations in the coding region of the gene [44]. A more detailed molecular analysis of possible Flt3 gene amplifications [3], autocrine feed forward loops [4], promoter hyperactivation of Flt3 or yet-to-bediscovered upstream activators will be required to unmask the molecular etiology of Flt3 activation in these hematopoietic malignancies. Although the initial observations that AML and highrisk MDS cell survival depends on the constitutive activation of IKK suggest the clinical use of IKK inhibitors, whole-body inhibition of the classical NFjB activation pathway could have major side effects, due to the expected major toxic effects of global NFjB inhibition [45]. Indeed, agents like bortezomib that have initially been viewed as ‘‘NFjB inhibitors’’ likewise exert their anti-neoplastic effects through unrelated mechanisms, for instance by the transcriptional induction of the pro-apoptotic protein Noxa [46]. The discovery that IKK is subordinated to Flt3, at least in a fraction of AML and high-risk MDS patients, suggests the tantalizing perspective of inhibiting a kinase with a cell type-specific expression profile. Moreover, as the mouse knockout of Flt3 is viable [47], it can be expected that therapeutic Flt3 antagonists might present acceptable levels of toxicity [21, 48, 49]. The results presented here support this general idea and unravel the mechanism through which Flt3 inhibition kills malignant myeloblasts in AML and high-risk MDS.

1159 Acknowledgements We are indebted to Jalil Abdelali (Institut Gustave Roussy, Villejuif, France) for support in confocal microscopy. Guido Kroemer is supported by Agence Nationale de Recherche, Fondation de France, Cent pour Sang la Vie, Cance´ropoˆle Ile-deFrance, Institut National du Cancer, Ligue Nationale contre le Cancer, and European Community (Active p53, Apo-Sys, TransDeath, RIGHT, ChemoRes, ApopTrain). Jennifer Grosjean received a post-doctoral fellowship by Cance´ropoˆle Ile-de-France. Lionel Ades receives a scholarship from Assistance Publique-Hopitaux de Paris and Caisse Nationale d’Assurance Maladie des Professions Inde´pendantes. M.T. receives a Ph.D. fellowship from Universite´ Paris Sud, Paris 11.

References 1. Yamamoto Y, Kiyoi H, Nakano Y, Suzuki R, Kodera Y et al (2001) Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 97:2434–2439. doi:10.1182/blood.V97.8.2434 2. Yokota S, Kiyoi H, Nakao M, Iwai T, Misawa S et al (1997) Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies. A study on a large series of patients and cell lines. Leukemia 11:1605–1609. doi: 10.1038/sj.leu.2400812 3. Ozeki K, Kiyoi H, Hirose Y, Iwai M, Ninomiya M et al (2004) Biologic and clinical significance of the FLT3 transcript level in acute myeloid leukemia. Blood 103:1901–1908 Epub 2003 Nov 1906 4. Zheng R, Levis M, Piloto O, Brown P, Baldwin BR et al (2004) FLT3 ligand causes autocrine signaling in acute myeloid leukemia cells. Blood 103:267–274. Epub 2003 Sep 2011 5. Thiede C, Steudel C, Mohr B, Schaich M, Schakel U et al (2002) Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 99:4326– 4335. doi:10.1182/blood.V99.12.4326 6. Kottaridis PD, Gale RE, Frew ME, Harrison G, Langabeer SE et al (2001) The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood 98:1752–1759. doi:10.1182/blood.V98.6.1752 7. Abu-Duhier FM, Goodeve AC, Wilson GA, Gari MA, Peake IR et al (2000) FLT3 internal tandem duplication mutations in adult acute myeloid leukaemia define a high-risk group. Br J Haematol 111:190–195. doi:10.1046/j.1365-2141.2000.02317.x 8. Takahashi S, Harigae H, Kaku M, Sasaki T, Licht JD (2004) Flt3 mutation activates p21WAF1/CIP1 gene expression through the action of STAT5. Biochem Biophys Res Commun 316:85–92. doi:10.1016/j.bbrc.2004.02.018 9. Takahashi S, McConnell MJ, Harigae H, Kaku M, Sasaki T et al (2004) The Flt3 internal tandem duplication mutant inhibits the function of transcriptional repressors by blocking interactions with SMRT. Blood 103:4650–4658. Epub 2004 Feb 4624 10. Gilliland DG, Griffin JD (2002) The roles of FLT3 in hematopoiesis and leukemia. Blood 100:1532–1542. doi:10.1182/blood-2002-020492 11. Horiike S, Yokota S, Nakao M, Iwai T, Sasai Y et al (1997) Tandem duplications of the FLT3 receptor gene are associated with leukemic transformation of myelodysplasia. Leukemia 11:1442–1446. doi:10.1038/sj.leu.2400770 12. Bonizzi G, Karin M (2004) The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol 25:280–288. doi:10.1016/j.it.2004.03.008

123

1160 13. Karin M, Yamamoto Y, Wang QM (2004) The IKK NF-kappa B system: a treasure trove for drug development. Nat Rev Drug Discov 3:17–26. doi:10.1038/nrd1279 14. Guzman ML, Neering SJ, Upchurch D, Grimes B, Howard DS et al (2001) Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 98:2301–2307. doi:10.1182/blood.V98.8.2301 15. Birkenkamp KU, Geugien M, Schepers H, Westra J, Lemmink HH et al (2004) Constitutive NF-kappaB DNA-binding activity in AML is frequently mediated by a Ras/PI3-K/PKB-dependent pathway. Leukemia 18:103–112. doi:10.1038/sj.leu.2403145 16. Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P et al (1997) International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 89:2079–2088 17. Braun T, Carvalho G, Coquelle A, Vozenin MC, Lepelley P et al (2006) NF-kappaB constitutes a potential therapeutic target in high-risk myelodysplastic syndrome. Blood 107:1156–1165. doi: 10.1182/blood-2005-05-1989 18. Greten FR, Karin M (2004) The IKK/NF-kappaB activation pathway-a target for prevention and treatment of cancer. Cancer Lett 206:193–199. doi:10.1016/j.canlet.2003.08.029 19. Griessinger E, Imbert V, Lagadec P, Gonthier N, Dubreuil P et al (2007) AS602868, a dual inhibitor of IKK2 and FLT3 to target AML cells. Leukemia 21:877–885 20. Takahashi S, Harigae H, Ishii KK, Inomata M, Fujiwara T et al (2005) Over-expression of Flt3 induces NF-kappaB pathway and increases the expression of IL-6. Leuk Res 29:893–899. doi: 10.1016/j.leukres.2005.01.008 21. Levis M, Allebach J, Tse KF, Zheng R, Baldwin BR et al (2002) A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood 99:3885–3891. doi:10.1182/ blood.V99.11.3885 22. Kelly LM, Yu JC, Boulton CL, Apatira M, Li J et al (2002) CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML). Cancer Cell 1:421–432. doi:10.1016/S1535-6108(02)00070-3 23. Grundler R, Thiede C, Miething C, Steudel C, Peschel C et al (2003) Sensitivity toward tyrosine kinase inhibitors varies between different activating mutations of the FLT3 receptor. Blood 102:646–651. Epub 2003 Mar 2027 24. Smith BD, Levis M, Beran M, Giles F, Kantarjian H et al (2004) Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 103:3669–3676 Epub 2004 Jan 3615 25. Stirewalt DL, Radich JP (2003) The role of FLT3 in haematopoietic malignancies. Nat Rev Cancer 3:650–665. doi:10.1038/ nrc1169 26. Fiedler W, Mesters R, Tinnefeld H, Loges S, Staib P et al (2003) A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia. Blood 102:2763–2767 Epub 2003 Jul 2763 27. Walters DK, Stoffregen EP, Heinrich MC, Deininger MW, Druker BJ (2005) RNAi-induced down-regulation of FLT3 expression in AML cell lines increases sensitivity to MLN518. Blood 105:2952–2954. Epub 2004 Dec 2957 28. Vardiman JW, Harris NL, Brunning RD (2002) The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 100:2292–2302. doi:10.1182/blood-2002-04-1199 29. Hassan Z, Fadeel B, Zhivotovsky B, Hellstrom-Lindberg E (1999) Two pathways of apoptosis induced with all-trans retinoic and etoposide in the myeloid cell line P39. Exp Hematol 27(8): 1322–1329 30. Fabre C, Carvalho G, Tasdemir E, Braun T, Ades L et al (2007) NF-kappaB inhibition sensitizes to starvation-induced cell death in high-risk myelodysplastic syndrome and acute myeloid leukemia. Oncogene 26:4071–4083. doi:10.1038/sj.onc.1210187

123

Apoptosis (2008) 13:1148–1161 31. Castedo M, Perfettini JL, Kroemer G (2002) Mitochondrial apoptosis and the peripheral benzodiazepine receptor: a novel target for viral and pharmacological manipulation. J Exp Med 196:1121–1125. doi:10.1084/jem.20021758 32. Zamzami N, Kroemer G (2004) Methods to measure membrane potential and permeability transition in the mitochondria during apoptosis. Methods Mol Biol 282:103–115 33. Castedo M, Hirsch T, Susin SA, Zamzami N, Marchetti P et al (1996) Sequential acquisition of mitochondrial and plasma membrane alterations during early lymphocyte apoptosis. J Immunol 157:512–521 34. Metivier D, Dallaporta B, Zamzami N, Larochette N, Susin SA et al (1998) Cytofluorometric detection of mitochondrial alterations in early CD95/Fas/APO-1-triggered apoptosis of Jurkat T lymphoma cells. Comparison of seven mitochondrion-specific fluorochromes. Immunol Lett 61:157–163. doi:10.1016/S01652478(98)00013-3 35. Matsuo Y, MacLeod RA, Uphoff CC, Drexler HG, Nishizaki C et al (1997) Two acute monocytic leukemia (AML-M5a) cell lines (MOLM-13 and MOLM-14) with interclonal phenotypic heterogeneity showing MLL-AF9 fusion resulting from an occult chromosome insertion, ins(11;9)(q23;p22p23). Leukemia 11:1469–1477. doi:10.1038/sj.leu.2400768 36. Patch RJ, Baumann CA, Liu J, Gibbs AC, Ott H et al (2006) Identification of 2-acylaminothiophene-3-carboxamides as potent inhibitors of FLT3. Bioorg Med Chem Lett 16:3282–3286. doi: 10.1016/j.bmcl.2006.03.032 37. Mercurio F, Zhu H, Murray BW, Shevchenko A, Bennett BL et al (1997) IKK-1 and IKK-2: cytokine-activated IkappaB kinases essential for NF-kappaB activation. Science 278:860–866. doi: 10.1126/science.278.5339.860 38. Weil R, Schwamborn K, Alcover A, Bessia C, Di Bartolo V et al (2003) Induction of the NF-kappaB cascade by recruitment of the scaffold molecule NEMO to the T cell receptor. Immunity 18:13– 26. doi:10.1016/S1074-7613(02)00506-X 39. Boudard D, Sordet O, Vasselon C, Revol V, Bertheas MF et al (2000) Expression and activity of caspases 1 and 3 in myelodysplastic syndromes. Leukemia 14:2045–2051. doi:10.1038/ sj.leu.2401959 40. Parker JE, Mufti GJ, Rasool F, Mijovic A, Devereux S et al (2000) The role of apoptosis, proliferation, and the Bcl-2-related proteins in the myelodysplastic syndromes and acute myeloid leukemia secondary to MDS. Blood 96:3932–3938 41. Parker JE, Mufti GJ (1998) Ineffective haemopoiesis and apoptosis in myelodysplastic syndromes. Br J Haematol 101:220–230. doi:10.1046/j.1365-2141.1998.00708.x 42. Tehranchi R, Fadeel B, Forsblom AM, Christensson B, Samuelsson J et al (2003) Granulocyte colony-stimulating factor inhibits spontaneous cytochrome c release and mitochondriadependent apoptosis of myelodysplastic syndrome hematopoietic progenitors. Blood 101:1080–1086. doi:10.1182/blood-200206-1774 43. Quentmeier H, Reinhardt J, Zaborski M, Drexler HG (2003) FLT3 mutations in acute myeloid leukemia cell lines. Leukemia 17:120–124. doi:10.1038/sj.leu.2402740 44. Stam RW, den Boer ML, Schneider P, Nollau P, Horstmann M et al (2005) Targeting FLT3 in primary MLL-gene-rearranged infant acute lymphoblastic leukemia. Blood 106:2484–2490. doi: 10.1182/blood-2004-09-3667 45. Lavon I, Goldberg I, Amit S, Landsman L, Jung S et al (2000) High susceptibility to bacterial infection, but no liver dysfunction, in mice compromised for hepatocyte NF-kappaB activation. Nat Med 6:573–577. doi:10.1038/75057 46. Fernandez Y, Verhaegen M, Miller TP, Rush JL, Steiner P et al (2005) Differential regulation of noxa in normal melanocytes and melanoma cells by proteasome inhibition: therapeutic

Apoptosis (2008) 13:1148–1161 implications. Cancer Res 65:6294–6304. doi:10.1158/0008-5472. CAN-05-0686 47. McKenna HJ, Stocking KL, Miller RE, Brasel K, De Smedt T et al (2000) Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood 95:3489–3497 48. Lee BD, Sevcikova S, Kogan SC (2006) Dual treatment with FLT3 inhibitor SU11657 and doxorubicin increases survival of leukemic mice. Leuk Res 31(8):1131–1134

1161 49. O’Farrell AM, Abrams TJ, Yuen HA, Ngai TJ, Louie SG et al (2003) SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood 101:3597–3605. doi: 10.1182/blood-2002-07-2307

123