262
HOX/PBX interaction as a therapeutic target in Acute Myeloid Leukaemia A thesis submitted in part requirement for the Degree of Doctor of Philosophy Raed Alharbi Microbial and cellular sciences Faculty of Health and Medical Sciences University of Surrey March 2015

HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

  • Upload
    doanh

  • View
    232

  • Download
    0

Embed Size (px)

Citation preview

Page 1: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

HOX/PBX interaction as a therapeutic target in Acute

Myeloid Leukaemia

A thesis submitted in part requirement for the

Degree of Doctor of Philosophy

Raed Alharbi

Microbial and cellular sciences

Faculty of Health and Medical Sciences

University of Surrey

March 2015

Page 2: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

I

Declaration of originality

This thesis and the work to which it refers are the results of my own efforts. Any ideas,

data, images or text resulting from the work of others (whether published or unpublished) are

fully identified as such within the work and attributed to their originator in the text, bibliography

or in footnotes. This thesis has not been submitted in whole or in part for any other academic

degree or professional qualification. I agree that the University has the right to submit my work

to the plagiarism detection service TurnitinUK for originality checks. Whether or not drafts

have been so-assessed, the University reserves the right to require an electronic version of the

final document (as submitted) for assessment as above.

The majority of this introduction is taken from my own review article (Alharbi,

Pettengell et al. 2012) with supplementary material added in sections 1.1, 1.5, 1.7.4 and 1.8.

Page 3: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

II

Summary

Acute myeloid leukaemia (AML) is a disorder characterised by the accumulation of

blast cells or progenitors of one of several non-lymphoid haematopoietic cell lineages and is

classified into two types: primary and secondary. HOX genes are over-expressed in both AML

and other cancers. This over-expression is associated with an intermediate/unfavourable

cytogenetic subset of AML. Although HOX over-expression is a common feature of AML,

conventional knockout methods have failed to fully evaluate their functions due to their

functional redundancy. We have applied an alternative approach by using a synthetic peptide

called HXR9 to antagonise the interaction between HOX proteins and their cofactor PBX,

which interacts with HOX proteins in groups 1-10.

AML cell lines derived from different AML types express different subsets of HOX

genes at different levels due to the heterogeneity of AML. It is showed for the first time that

targeting the HOX-PBX interaction using HXR9 led to cell death of the tested AML cell lines.

This cell death did not appear to be through apoptosis, as there were no signs of the caspase

activation and nuclear fragmentation. Likewise, there was also no activation of key necrotic

markers such as cypD and PARP1. Instead, cell death involved, at least in part, the expression

of c-FOS and p21 in p53-independentmanner. In addition, HXR9 caused cell death in

MEK/ERK and p38 independent pathways, but the JNK pathway exerted a resistant effect in

K562 cells. It was found that inhibiting the Ca2+ downstream mediators CaM, PKC and HO-1

significantly sensitised tested AML cell lines to HXR9. Taken together, these findings indicate

a novel cell death pathway in AML cells. In vivo modelling also showed that HXR9 could delay

tumour growth in a mouse model of AML.

Page 4: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

III

Acknowledgements

First of all, I would like to thank my first supervisor Dr. Richard Morgan for his patience

for my endless questions, great guidance throughout the last four years, and for his wonderful

help to extend my ideas and translate it to practical assays that has great impacts on both myself

and the outcome of the project. I also would like to thank Prof. Hardev Pandha for giving me

the chance to work in his group. My special thanks to Angela Boxall who trained me in my first

months in the lab. Also, I would like to thank Dr. Guy Simpson for his great help in mouse

models. I am very lucky to have such friendly colleagues and very grateful for their support and

company in the last few years.

My special thanks to my family whom I dedicate this thesis to. I am indebted to my

Mother who is an endless source of help and support throughout my life. I also would like to

thank my brothers and my little sister for their encouragement. My special thanks to my wife

Ruba and my little angels Ratille and Racille who make my life more interesting and

meaningful. I really look forward to spending all the time with you.

My sincere love is to my Dad's soul who taught me reading and writing. He was the

master teacher throughout my life. I wish he were here to see this moment that he had been

waiting for. I miss his support, encouragement and voice.

Page 5: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

IV

SUMMARY .............................................................................................................................. II

ACKNOWLEDGEMENTS .................................................................................................. III

LIST OF FIGURES ............................................................................................................ VIII

LIST OF TABLES ................................................................................................................. IX

LIST OF ABBREVIATIONS ................................................................................................. X

CHAPTER 1 INTRODUCTION ............................................................................................ 1

1.1 Overview ................................................................................................................................................... 2

1.1.1 Genetic changes in AML ....................................................................................................................... 6

1.2 HOX Genes ................................................................................................................................................ 6

1.3 HOX Cofactors ......................................................................................................................................... 8

1.4 HOX genes in haematopoiesis .................................................................................................................. 9

1.4.1 Gain of function studies ....................................................................................................................... 10

1.4.2 Loss of function studies ....................................................................................................................... 11

1.5 Upstream regulators of HOX genes ...................................................................................................... 15

1.6 HOX downstream target genes in haematopoietic cells ...................................................................... 16

1.7 The role of HOX genes in acute leukaemia ........................................................................................... 21

1.7.1 HOX fusion proteins ............................................................................................................................ 21

1.7.2 HOX over-expression in AML ............................................................................................................. 23

1.7.3 HOX gene dysregulation in acute lymphoid leukaemia (ALL) ............................................................ 25

1.7.4 HOX genes as prognostic markers ....................................................................................................... 26

1.8 Hypothesis and aims ............................................................................................................................... 28

CHAPTER 2 MATERIALS AND METHODS .................................................................. 30

2.1 Materials ................................................................................................................................................. 31

2.1.1 Reagents ............................................................................................................................................... 31

2.1.2 Instruments........................................................................................................................................... 33

2.1.3 Cell lines .............................................................................................................................................. 33

2.1.4 HXR9 and CXR9 peptide synthesis ..................................................................................................... 33

2.1.5 Mice ..................................................................................................................................................... 34

2.2 Methods ................................................................................................................................................... 34

2.2.1 General cell culture methods ................................................................................................................ 34

2.2.1.1 Routine cell culture ..................................................................................................................... 34

2.2.1.2 Cell counting and cell density calculation using a haemocytometer ........................................... 36

2.2.1.3 Cryopreservation of cell stocks ................................................................................................... 36

2.2.1.4 Revitalisation of cryopreserved cells........................................................................................... 37

Page 6: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

V

2.2.2 Gene expression analysis by real-time PCR (RT-PCR) ....................................................................... 37

2.2.2.1 mRNA extraction ........................................................................................................................ 37

2.2.2.2 Measuring mRNA concentration ................................................................................................. 38

2.2.2.3 Reverse transcription of mRNA into cDNA ............................................................................... 38

2.2.2.4 Complementary PCR primers design .......................................................................................... 39

2.2.2.5 RT-PCR ....................................................................................................................................... 42

2.2.2.6 RT-PCR data analysis ................................................................................................................. 43

2.2.3 Lactate dehydrogenase (LDH) assay ................................................................................................... 44

2.2.3.1 LDH assay data analysis ............................................................................................................. 45

2.2.4 Assessment of drug combination interaction by LDH ......................................................................... 46

2.2.5 Annexin V- PE assay ........................................................................................................................... 47

2.2.6 Caspase-3 activity assay ...................................................................................................................... 49

2.2.6.1 Caspase-3 activity data analysis .................................................................................................. 50

2.2.7 Cell cytospins and 4',6-diamidino-2-phenylindole (DAPI) staining .................................................... 50

2.2.8 Inhibition of caspases activity by z-VAD-FMK .................................................................................. 51

2.2.9 Cyclosporin A (CsA) protection assay ................................................................................................. 52

2.2.10 Necrostatin-1 (Nec-1) protection assay ........................................................................................... 53

2.2.11 Fructose protection assay ................................................................................................................. 53

2.2.12 The effect of ethilenediaminetetra-acetic acid (EDTA) on HXR9 cytotoxicity .............................. 54

2.2.13 The effect of HXR9 on mitogen activated protein kinase (MAPK) pathways ................................ 55

2.2.14 The effect of inhibition of NADPH oxidase (NOX) on HXR9 efficacy .......................................... 55

2.2.15 Role of μ-calpain in HXR9 cell killing ............................................................................................ 56

2.2.16 The effect of calmodulin (CaM) inhibition on HXR9 cytotoxicity ................................................. 56

2.2.17 The effect of protein kinase C (PKC) inhibition on HXR9 cytotoxicity ......................................... 57

2.2.18 The effect of the heme oxygenase-1 (HO-1) inhibition on HXR9 cytotoxicity ............................... 57

2.2.19 The effect of the p53 inhibition on HXR9 cytotoxicity ................................................................... 58

2.2.20 Western blotting (WB) for protein expression ................................................................................. 58

2.2.20.1 Preparation of cell lysate ............................................................................................................. 59

2.2.20.2 Measuring protein concentrations by pierce BCA assay ............................................................. 59

2.2.20.3 Sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) for protein

separation …………………………………………………………………………………………………...60

2.2.20.4 Transferring proteins to polyvinylidene fluoride (PVDF) membranes ....................................... 60

2.2.20.5 Detection of proteins by antibodies ............................................................................................. 60

2.2.21 In vivo assays ................................................................................................................................... 62

2.2.21.1 Cell preparations ......................................................................................................................... 62

2.2.21.2 Systemic injection of C1498-GFP in C57BL/6 and nude mice ................................................... 62

2.2.21.2.1 Harvesting and processing PB ............................................................................................. 62

2.2.21.2.2 Harvesting and processing BM and other organs................................................................. 62

2.2.21.3 Subcutaneous (S.C.) injection of AML cells into C57BL/6 and SCID mice .............................. 63

2.3 Statistical analysis .................................................................................................................................. 63

2.3.1 Calculation of IC50 ............................................................................................................................... 64

2.3.2 The analysis of drug combination assay data ....................................................................................... 64

2.3.3 Statistical analysis of in vivo assays ..................................................................................................... 64

CHAPTER 3 IN VITRO CYTOTOXICITY OF HXR9 ..................................................... 67

3.1 Introduction ............................................................................................................................................ 68

3.1.1 Amis of chapter 3 ................................................................................................................................. 69

3.2 Results ..................................................................................................................................................... 70

3.2.1 HOX gene expression in AML cell lines .............................................................................................. 70

Page 7: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

VI

3.2.2 HXR9 is cytotoxic on all tested AML derived cell lines ..................................................................... 73

3.2.3 DNR is cytotoxic on K562 and HL-60 ................................................................................................ 76

3.2.4 Combination effect of HXR9 and DNR ............................................................................................... 77

3.2.5 MTX induces K562 and HL-60 cell death ........................................................................................... 79

3.2.6 Combination effect of HXR9 and MTX .............................................................................................. 80

3.3 Discussion ................................................................................................................................................ 82

3.3.1 Summary of chapter ............................................................................................................................. 87

CHAPTER 4 THE MECHANISM OF HXR9 CYTOTOXICITY ................................... 88

4.1 Introduction ............................................................................................................................................ 89

4.1.1 Aims of chapter 4 ................................................................................................................................. 94

4.2 Results ..................................................................................................................................................... 96

4.2.1 HXR9 causes the up-regulation of c-FOS ............................................................................................ 96

4.2.2 HXR9 causes death of AML tested cell lines ...................................................................................... 98

4.2.3 HXR9 does not affect caspase or Bcl-2 family transcription ............................................................. 101

4.2.4 HXR9 does not activate caspase-3 in the tested AML cell lines ........................................................ 104

4.2.5 HXR9 efficacy is not affected by the general inhibition of caspases ................................................. 107

4.2.6 HXR9 does not cause nuclear fragmentation ..................................................................................... 109

4.2.7 Ca2+ chelating abrogates cell killing by HXR9 .................................................................................. 112

4.2.8 EDTA rescues cells from killing by HXR9 ....................................................................................... 114

4.2.9 Cell killing by HXR9 does not involve CypD ................................................................................... 116

4.2.10 Inhibition of RIP1 modifies the cytotoxicity of HXR9.................................................................. 118

4.2.11 HXR9 induces cell death through an ATP-independent pathway ................................................. 120

4.2.12 HXR9 induces cell death through a PARP1-independent pathway ............................................... 122

4.2.13 The effect of inhibition of MAPK pathways on HXR9 cytotoxicity ............................................. 123

4.2.13.1 HXR9 induces cell death in a MEK/ERK independent pathway .............................................. 123

4.2.13.2 HXR9-mediated cell death does not require p38 pathway signalling ....................................... 125

4.2.13.3 Inhibition of the JNK pathway sensitises K562, but not HL-60 cells to HXR9 ........................ 127

4.2.14 Blocking NOX enzymes sensitizes cells to HXR9 ........................................................................ 129

4.2.15 μ-Calpain is not required for HXR9-induced cell death ................................................................ 131

4.2.16 CaM inhibition dramatically increases the sensitivity of cells to HXR9 ....................................... 133

4.2.17 Inhibition of PKC greatly increases the sensitivity of K562 and HL-60 cells to HXR9 ............... 135

4.2.18 Simultaneous inhibition of CaM and PKC potentiates HXR9 cytotoxicity ................................... 137

4.2.19 HO-1 inhibition increases the sensitivity to HXR9 ....................................................................... 139

4.2.20 HXR9 induces p21 expression but not p53 ................................................................................... 141

4.2.21 HXR9-induces AML cell death through a p53-independent pathway ........................................... 143

4.3 Discussion .............................................................................................................................................. 144

4.3.1 Summary of chapter ........................................................................................................................... 156

CHAPTER 5 IN VIVO CYTOTOXICITY OF HXR9 ..................................................... 157

5.1 Introduction .......................................................................................................................................... 158

5.1.1 Aims of chapter 5 ............................................................................................................................... 159

5.2 Results ................................................................................................................................................... 160

5.2.1 HXR9 is cytotoxic for C1498-GFP cells ........................................................................................... 160

5.2.2 CaM blocking enhances the cytotoxicity of HXR9 ........................................................................... 161

5.2.3 Inhibition of PKC activity significantly sensitises cells to HXR9 ..................................................... 163

Page 8: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

VII

5.2.4 Concurrent inhibition of CaM and PKC significantly enhances the efficacy of HXR9 ..................... 165

5.2.5 Efficient expression of GFP in C1498 cells ....................................................................................... 167

5.2.6 Establishment a systemic C1498-GFP model in C57BL/6 mice........................................................ 168

5.2.7 Establishment a systemic C1498-GFP model in C57BL/6 and nude mice ........................................ 170

5.2.8 Development a C1498-GFP flank model in C57BL/6 mice .............................................................. 174

5.2.9 HXR9 significantly extended the survival of C1498-GFP xenograft ................................................ 175

5.2.10 Simultaneous inhibition of CaM and PKC did not sensitise C1498-GFP flank tumour to HXR9. 177

5.2.11 HXR9 did affect the tumour growth of K562 flank model ............................................................ 179

5.3 Discussion .............................................................................................................................................. 181

5.3.1 Summary of chapter ........................................................................................................................... 187

CHAPTER 6 GENERAL DISCUSSION AND FUTURE DIRECTIONS ..................... 188

6.1 Discussion .............................................................................................................................................. 189

6.2 Conclusion ............................................................................................................................................. 203

6.3 Future directions .................................................................................................................................. 204

APPENDICES ....................................................................................................................... 206

REFERENCES ..................................................................................................................... 218

PUBLICATIONS .................................................................................................................. 247

Page 9: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

VIII

List of Figures

Figure 1.1 A schematic diagram of haematopoiesis.. .............................................................................................. 3

Figure 1.2 A schematic structure of clustered HOX genes. ..................................................................................... 8

Figure 1.3 structures of AbdB-HOX, NUP98 and the predictive fusion protein NUP98-HOXA.. ....................... 22

Figure 2.1 A haemocytometer diagram.. ............................................................................................................... 36

Figure 2.2 LDH cytotoxicity assay.. ...................................................................................................................... 44

Figure 2.3 A flow cytometry plot shows different cell populations in the annexin V-PE assay.. ......................... 48

Figure 2.4 ATP depletion by fructose. .................................................................................................................. 53

Figure 3.1 HOX gene expression in KG-1, HEL92.1.7, KU812F, K562 and HL-60 cell lines.. ........................... 72

Figure 3.2 LDH assay for HXR9 and CXR9 cytotoxicity on AML derived cell lines.. ........................................ 75

Figure 3.3 LDH assay for DNR cytotoxicity on K562 and HL-60 cell lines......................................................... 76

Figure 3.4 LDH assay of HXR9 and DNR combination therapy for K562 and HL-60.. ....................................... 78

Figure 3.5 LDH assay for MTX cytotoxicity on K562 and HL-60 cell lines.. ...................................................... 79

Figure 3.6 LDH assay of HXR9/MTX combination therapy for K562 and HL-60.. ............................................. 81

Figure 4.1 A schematic diagram of cell death.. ..................................................................................................... 90

Figure 4.2 c-FOS expression in AML cell lines after treatment with HXR9.. ...................................................... 97

Figure 4.3 Detection of cell death in AML cell lines by flow cytometry.. .......................................................... 100

Figure 4.4 Analysis of transcriptional changes of several pro- and anti-apoptotic genes upon HXR9 treatment.

............................................................................................................................................................................. 103

Figure 4.5 Detection of caspase-3 activity in AML cell lines using Z-DEVD-R110.. ........................................ 106

Figure 4.6 General inhibition of caspase activity in HXR9 treated AML cell lines by z-VAD-FMK.. .............. 108

Figure 4.7 DAPI staining of AML cells upon HXR9 treatment.. ........................................................................ 111

Figure 4.8 Effect of EDTA on HXR9 cytotoxicity.. ............................................................................................ 113

Figure 4.9 LDH analysis of the effect of EDTA on HXR9 cytotoxicity.. ........................................................... 115

Figure 4.10 Effect of CsA on HXR9 cytotoxicity.. ............................................................................................. 117

Figure 4.11 Effect of RIP1 inhibition on HXR9 cytotoxicity.. ............................................................................ 119

Figure 4.12 The effect of ATP depletion on HXR9 cytotoxicity.. ...................................................................... 121

Figure 4.13 WB analysis of PARP1 activation after HXR9 treatment.. .............................................................. 122

Figure 4.14 The effect of inhibiting the MEK/ERK pathway on the cytotoxicity of HXR9. .............................. 124

Figure 4.15 The effect of inhibiting the p38 pathway on the cytotoxicity of HXR9.. ......................................... 126

Figure 4.16 The effect of inhibiting the JNK pathway on HXR9 cytotoxicity.. .................................................. 128

Figure 4.17 The effect of NOX inhibition on the cytotoxicity of HXR9.. ........................................................... 130

Figure 4.18 The effect of blocking μ-calpain on HXR9 cytotoxicity. ................................................................. 132

Figure 4.19 The effect of CaM inhibition on HXR9 sensitivity.. ........................................................................ 134

Figure 4.20 The effect of blocking of PKC on the cytotoxicity of HXR9 on K562 and HL-60 cells. ................ 136

Figure 4.21 The impact of concurrent inhibition of CaM and PKC on the cytotoxicity of HXR9.. .................... 138

Figure 4.22 The effect of HO-1 inhibition on the cytotoxicity of HXR9.. .......................................................... 140

Figure 4.23 RT-PCR analysis of p53 and p21 expression in response to HXR9 treatment.. ............................... 142

Figure 4.24 The effect of blocking p53 protein on HXR9 cytotoxicity. .............................................................. 143

Figure 5.1 LDH assay for the cytotoxicity of HXR9 on C1498-GFP cells. ........................................................ 160

Figure 5.2 The effect of CaM inhibition on the cytotoxicity of HXR9 for C1498-GFP.. ................................... 162

Figure 5.3 The impact of PKC inhibition on the sensitivity of C1498-GFP cells to HXR9. ............................... 164

Figure 5.4 The effect of CaM and PKC simultaneous inhibition on the efficacy of HXR9.. .............................. 166

Figure 5.5 Expression of GFP in C1498 cells.. ................................................................................................... 167

Figure 5.6 FACS analysis of PB from C57BL/6 mice 13 days after injection of C1498-GFP cells. .................. 168

Figure 5.7 FACS analysis of C1498-GFP cells shows engraftment in several organs of C57BL/6 and nude mice.

............................................................................................................................................................................. 172

Figure 5.8 HXR9 treatment effect on the growth of C1498-GFP xenograft in female C57BL/6 mice. .............. 176

Figure 5.9 Effect of HXR9 I.T. treatment on the growth of C1498-GFP flank xenograft in C57BL/6 mice.. .... 178

Figure 5.10 Impact of HXR9 treatment on the K562 flank-xenograft growth in SCID mice.............................. 180

Page 10: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

IX

List of Tables

Table 1.1 Classification of leukaemia. .................................................................................................................... 3

Table 1.2 AML classification according WHO classification ................................................................................. 5

Table 1.3 HOX gene studies. ................................................................................................................................. 14

Table 1.4 A summary of mammalian HOX target genes....................................................................................... 20

Table 2.1 Reagents were used in this study and their suppliers. ............................................................................ 31

Table 2.2 Cell lines used in this study. .................................................................................................................. 35

Table 2.3 cDNA synthesis mix.. ............................................................................................................................ 39

Table 2.4 HOX gene primers used for PCR amplification..................................................................................... 40

Table 2.5 Pro- and anti-apoptotic and β-actin gene primers used for PCR amplification. .................................... 42

Table 2.6 RT-PCR reaction components. This table shows the components of single RT-PCR reaction.. ........... 43

Table 2.7 Different cell populations in the annexin V-PE assay. .......................................................................... 48

Table 2.8 Antibodies used in WB and working dilutions. ..................................................................................... 61

Table 2.9 Summary of the in vitro assays performed and reagents used. .............................................................. 65

Table 3.1 IC50 values of HXR9 on AML derived cell lines with SEM. ............................................................... 75

Table 3.2 The analysis of the combination effect of HXR9 and DNR by Calcusyn software.. ............................. 78

Table 3.3 The analysis of the combination effect of HXR9 and MTX by Calcusyn software. ............................. 81

Table 3.4 The IC50 values of HXR9 with cell lines derived from different cancers. ............................................. 84

Table 4.1 The mechanism of HXR9 cytotoxicity on AML cells. ........................................................................ 156

Table 5.1 A summary of injection of C1498-GFP I.V. in C57BL/6 mice. .......................................................... 169

Table 5.2 A summary of injections of C1498-GFP I.V. in C57BL/6 and nude mice. ......................................... 173

Table 5.3 A summary of injection of C1498-GFP S.C in C57BL/6 mice. .......................................................... 174

Page 11: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

X

List of Abbreviations

7-AAD 7-Amino actinomycin D

Abd-HOXA AbdominalB-HOXA

ACPP Activatable CPP

AIF Apoptosis inducing factor

ALL Acute lymphoid leukaemia

AML Acute myeloid leukaemia

AP-1 Activator protein-1

Apaf1 Apoptosis protease-activating factor

APML Acute promyelocytic leukaemia

ATP Adenosine triphosphate

ATRA All-trans retinoic acid

BCA Bicinchonic acid

BM Bone marrow

BCA Bradford city assay

BSA Bovine serum albumin

Ca2+ Calcium ions

CaM Calmodulin

cDNA Complementary deoxyribonucleic acid

CDX Caudal-type homebox transcription factor

c-FLIP (L) Cellular FLICE (FADD-like IL-1β-converting enzyme)- inhibitory

protein (L)

CI Combination index

CML Chronic myeloid leukaemia

CPP Cell penetrating peptide

CR Complete remission

CsA Cyclosporin A

Page 12: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

XI

CXCR4 C-X-C chemokine receptor type 4

CypD Cyclophilin D

Cyt C Cytochrome C

DAPI 4',6-Diamidino-2-phenylindole

DISC Death-inducing signalling complex

DMEM Dulbecco’s Modified Eagle’s Medium

DMSO Dimethyl sulphoxide

DNR Daunorubicin

DPI Diphenyleneiodonium chloride

ED Effective dose

EDTA Ethilenediaminetetra-acetic acid

ELISA Enzyme-linked immunosorbent assay

ERK Extracellular signal-regulated kinase

FACS Fluorescence-activated cell sorting

FBS Fetal bovine serum

FLT3 Fms-like tyrosine kinase 3

GFP Green fluorescent protein

H2O2 Hydrogen Peroxide

HO-1 Heme oxygenase-1

HOX Homeobox transcription factor

HP Haemtopoietic progenitor

HSC Haematopoietic stem cell

I.T. Intratumoural

I.V. Intravenous

IC50 The half maximal inhibitory concentration

IMDM Iscove’s Modified Dulbecco’s medium

JNK Jun N-terminal kinase

Page 13: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

XII

LDH Lactate dehydrogenase

MAPK Mitogen activated protein kinase

MDS Myelodysplasia

MEIS Myeloid ectropic insertion site

MLL Mixed lineage, myeloid lymphoid, leukaemia

mPTP Mitochondrial permeability transition pore

MRD Minimal residual disease

mRNA Messenger ribonucleic acid

MTX Mitoxantrone

Nec-1 Necrostatin-1

NF-ҠB Nuclear factor-kappa B

NK Natural killer cells

NK-AML Normal karyotype AML

NOD-SCID Non-obese diabetic with severe combined immune-deficient

NOX NADPH oxidase

NPM1 3 Nucleophosmin 1

NUP98 Nucloporin 98

OS Overall survival

PARP Poly ADP ribose polymerase

PB Peripheral blood

PBS Phosphate buffered saline

PBX Pre B cell leukaemia

PCD Programmed cell death

PCR Polymerase chain reaction

PFT-α Pifithrin-α

PKC Protein kinase C

PPIX Protoporphyrin IX

Page 14: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

XIII

PS Phosphatidlyserine

PVDF Polyvinylidene fluoride

R2 Linear coefficient correlation

R9 Arginine residues

RAS Rat sarcoma

RIP1 Receptor interacting protein 1

RIP3 Receptor interacting protein 3

RNA Ribonucleic acid

Ro31-8220 Methanesulfonate salt

ROS Reactive oxygen species

RPM Revolution per minute

RPMI-1640 Roswell Park Memorial Institute

RT Room temperature

RT-PCR Real-time PCR

S.C. Subcutaneous

SCID Severe combined immune-deficient

SDS-PAGE Sodium dodecyl sulphate polyacrylamide gel electrophoresis

SEM Standard error of mean

TNF Tumour necrosis factor

TRAIL Tumour necrosis-related apoptosis-inducing ligand

VEGF Vascular endothelial growth factor

W-7 N-(6-Aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride

WB Western blotting

XIAP X linked inhibitor of apoptosis

Page 15: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

1

Chapter 1 Introduction

Page 16: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

2

1.1 Overview

Haematopoiesis is defined as the formation of new blood cells. Initially,

haematopoietic stem cells (HSCs) differentiate into multipotent haematopoietic

progenitors that undergo a gradual differentiation to give rise to array of more lineage-

restricted progenitors that ultimately produce highly specialised and differentiated

mature blood cells (Figure 1.1). Mature cells can be classified into myeloid cells

including neutrophils, eosinophils, basophils, monocytes, platelets and red cells, and

lymphoid cells including B- lymphocytes, T-lymphocytes and natural killer (NK) cells

(Orkin and Zon 2008; Doulatov, Notta et al. 2012).

Haemtological malignancies are a group of heterogeneous diseases that are

initiated by leukaemic stem cells that are able to both self-renew and differentiate like

normal HSCs, although in an abnormal fashion, thereby increasing their number and

giving rise to differentiated cells that represent the majority of cells found in the tumour.

The classification of haemtological malignancies depended on the stage of the disease

(acute or chronic leukaemia) and the immunophenotype of the cells (myeloid or

lymphoid) (Table 1.1) (Warr, Pietras et al. 2011).

Page 17: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

3

Figure 1.1 A schematic diagram of haematopoiesis. HSC differentiate to MPP that give rise to lineage-

committed progenitors which in turn are differentiated to more mature blood cells. HSC: haematopoietic

stem cell, MPP: multipotent progenitor, CLP: committed lymphoid progenitor, CMP: committed

myeloid progenitor, Pro-NK: pro-natural killer cell, MEP: megakaryocyte-erythrocyte progenitor, GMP:

granulocyte-monocyte progenitor, MP: megakaryocyte progenitor, EP: erythrocyte progenitor, GP:

granulocyte progenitor, MP: monocyte progenitor.

Table 1.1 Classification of leukaemia.

Acute Chronic

Myeloid origin Acute myeloid leukaemia Chronic myeloid leukaemia

Lymphoid origin Acute lymphoid leukaemia Chronic lymphoid leukaemia

Page 18: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

4

Acute myeloid leukaemia (AML) is a heterogeneous group of genetically and

phenotypically aggressive disorders where the differentiation of haematopoietic

progenitors (HPs) is blocked, increasing their self-renewal ability and disturbing the

normal regulation of proliferation (Frohling, Scholl et al. 2005). In the UK, AML is

the most frequent acute leukaemia in adults, accounting for 77% of cases. The median

age at presentation is 69 years and the male: female ratio is about 5: 4 (Smith, Howell

et al. 2011). The disease is commonly classified by either the French-American-British

system, or that described by the world health organization. The former is based on

morphology and maturation stage and classifies AML into eight groups (M0-M7). The

latter is also based on morphology, but also includes immunophenotyping, genetics and

clinical manifestations, and classifies AML into four main groups: AML with recurrent

genetic abnormalities, AML with myelodysplasia (MDS)-related changes, therapy-

related AML and MDS or AML that does not fit into any of these groups (Table 1.2).

Non-random chromosomal alterations, such as balanced translocations, monosomies,

trisomies, inversion and deletions have been found in the leukaemic cells of almost 55%

of AML patients, and until recently they were considered to be the most crucial

prognostic factors for complete remission (CR), likelihood of relapse, and overall

survival (OS) (Estey and Döhner 2006; Mrózek, Marcucci et al. 2007).

About 55% of AML cases have chromosomal aberrations and about 15% have

complex karyotype, three or more cytogenetic aberrations (Betz and Hess 2010).

Cytogenetic studies is used as prognostic indicator and classify AML into three

prognostic groups: favorable, intermediate and adverse. AML cases with t(15;17),

t(8;21) and t(16;16) are associated with favorable-risk group. The intermediate-risk

group includes cases with t(9;11), along with cases exhibiting loss of the Y

chromosome or gains of whole chromosome. This prognostic group also includes AML

Page 19: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

5

cases with normal karyotype that account almost 45% of AML cases. The adverse-risk

group includes complex karyotype, t(6;9), inv(3)/ t(3;3) (Roche, Zeng et al. 2004;

(Marcucci, Mrozek et al. 2005; Betz and Hess 2010; Buccisano, Maurillo et al. 2012).

Table 1.2 AML classification according WHO classification (WHO 2008).

Categories

AML with recurrent genetic abnormalities

AML with t(8;21)(q22;q22); RUNX1-RUNX1T1

AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11

APL with t(15;17)(q22;q12); PML-RARA

AML with t(9;11)(p22;q23); MLLT3-MLL

AML with t(6;9)(p23;q34); DEK-NUP214

AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2); RPN1-EVI1

AML (megakaryoblastic) with t(1;22)(p13;q13); RBM15-MKL1

Provisional entity: AML with mutated NPM1

Provisional entity: AML with mutated CEBPA

AML with myelodysplasia-related changes

Therapy-related AML

AML, not otherwise specified

AML with minimal differentiation

AML without maturation

AML with maturation

Acute myelomonocytic leukemia

Acute monoblastic/monocytic leukemia

Acute erythroid leukemia

Pure erythroid leukemia

Erythroleukemia, erythroid/myeloid

Acute megakaryoblastic leukemia

Acute basophilic leukemia

Acute panmyelosis with myelofibrosis

Down syndrome related AML

Transient abnormal myelopoiesis

Myeloid leukemia associated with Down syndrome

Page 20: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

6

Recent advances in molecular diagnosis have resulted in both gene alterations

and the dysregulation of specific genes becoming increasingly important as prognostic

elements in AML. This has helped to clarify the numerous heterogeneities of AML

subsets, particularly AML subsets showing normal karyotype AML (NK-AML)

(Dohner and Dohner 2008) and furthered understanding of the molecular mechanisms

of leukaemogenesis.

1.1.1 Genetic changes in AML

The origin of AML is associated with two distinct genetic changes, referred to

as Class I and Class II. Class I consists of mutations that enhance proliferation signal

transduction pathways and induce the proliferation of HSCs or HPs and usually affect

kinase signaling pathways, such as FLT3, KIT, NRAS/KRAS and JAK/STAT

mutations. Class I mutations take place late and cause disease progression. Class II

mutations target haematopoietic transcription factor genes leading to a block in myeloid

differentiation and conferring the self-renewal ability of HPs. These mutations take

place early and initiate the AML disease (Dohner and Dohner 2008; Renneville,

Roumier et al. 2008; Betz and Hess 2010). One of the most affected and mutated

transcription factors are homeobox (HOX) genes.

1.2 HOX Genes

The HOX genes are a family of homeodomain-containing transcription factors

(Garcia-Fernandez 2004), initially characterized in Drosophila as master regulators of

trunk and tail development during embryogenesis (Shah and Sukumar 2010).

Duplication of the original HOX gene cluster has given rise to 39 genes in mammals,

Page 21: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

7

separated into four clusters known as A, B, C, and D (Amores, Force et al. 1998;

Abramovich, Pineault et al. 2005; Shah and Sukumar 2010). These clusters are located

on four different chromosomes, HOXA (7p15), HOXB (17q21), HOXC (12q13), and

HOXD (2q31) (Rice and Licht 2007). Ancestors of the original gene in each of the

clusters are known as paralogs, and generally they show a high degree of sequence

similarity as well as functional redundancy (Figure 1.1) (He, Hua et al. 2011).

The arrangement of HOX genes into clusters allows for enhancer sharing which

enables a precise spatial and temporal coordination of expression during development.

The relative regulatory dominance also varies between HOX genes, giving rise to what

is often referred to as a 'HOX code' (Knittel, Kessel et al. 1995), and resulting in the

following distinctive criteria: (1) Temporal distribution. The expression of HOX genes

starts from the 3' end of the cluster and proceeds stepwise towards the 5' end. (2) Spatial

distribution. The 3’ most member of the cluster is expressed with a more anterior limit

than the next member and each subsequent member has a more posterior limit of

expression resulting in an overlapping series of expression domains. (3) Posterior

prevalence. In each individual cluster, the function of the posterior gene products is

dominant over the more anterior genes (He, Hua et al. 2011).

Page 22: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

8

Figure 1.2 A schematic structure of clustered HOX genes. The 39 HOX genes are located on four

different chromosomes. Homology of human HOX genes HOX-C to Drosophilia HOM-C genes is

showed by colours. Blank squares show missing genes (Previously published in Alharbi, Pettengell et al.

2012).

1.3 HOX Cofactors

DNA binding site studies suggest that HOX proteins have relatively limited

selectivity and specificity, and they need cofactor interactions in order to increase both

(Phelan, Rambaldi et al. 1995; Moens and Selleri 2006; Mann, Lelli et al. 2009). The

most important HOX-cofactors are the three amino acid loop extension proteins, which

comprise the pre B cell leukaemia (PBX) and myeloid ectropic insertion site (MEIS)

families (Moens and Selleri 2006; Mann, Lelli et al. 2009). These cofactors have

crucial roles in development and haematopoiesis (Thorsteinsdottir, Kroon et al. 2001).

For example, Pbx1 null mice die during the embryonic stage as a result of severe

haematopoietic defects (DiMartino, Selleri et al. 2001) and Meis1-deficient mice fail to

generate megakaryocytes, exhibit severe hemorrhaging and likewise die during the

embryonic stage (Hisa, Spence et al. 2004). In zebrafish, Meis1 and Pbx contribute to

Drosophilia(HOM-C)

Pb

Chromosome number

Iab Dfd Scr Antp Ubx Abd A Abd B

Antennapedia Complex (Ant-C) Bithorax Complex (BX-C)

1 32 5 6 7 10 11 12 1394HOX A

1 2 3 4 5 76 9 138

Mammalian (HOX-C)

HOX B

54 136 8 9 10 11 12

93 4 8 1110 12 13

7p15

17q21

12q13

2q31

HOX C

HOX D

3` 5`

Page 23: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

9

the production of erythropoietic cells and the inhibition of myelopoiesis (Pillay,

Forrester et al. 2010). Generally, Hox proteins 1-10 bind with Pbx1 (Shen, Chang et al.

1996), whereas Hox proteins 9-13 bind with Meis1 (Shen, Montgomery et al. 1997).

Recently, it has been reported that HOXA6 binds to MEIS1 which indicates that MEIS1

interaction is not limited to the 5' end HOXs.(Dickson, Liberante et al. 2013).

1.4 HOX genes in haematopoiesis

HOX genes are expressed in HSCs and HPs in a manner reminiscent of their

expression in early development, with lineage and differentiation stage-restricted

manners. Thus, for example, HOXB3, HOXB4, and HOXA9 are highly expressed in

uncommitted haematopoietic cells, whereas HOXB8 and HOXA10 are expressed in

myeloid committed cells. Different mammalian CD34+ cell subpopulations express at

least 22 of the 39 HOX genes (Grier, Thompson et al. 2005). Anterior '3' end' HOX

genes (HOX1-6) are highly expressed in the most primitive HSCs. Subsequently, the

anterior HOX genes are downregulated, and posterior '5' end' HOX genes are expressed

during commitment (Sauvageau, Lansdorp et al. 1994). HOX genes are highly

expressed in the most primitive HSCs and HPs, while their expression is almost absent

in CD34- cells, which are considered differentiated bone marrow (BM) cells

(Sauvageau, Lansdorp et al. 1994; Pineault, Helgason et al. 2002; Helen Wheadon

2011). The analysis of HOX gene expression in human multipotent stem cells and T-

cell progenitors showed that HOXA genes are prominently expressed during T-cell

development, in particular AbdominalB-HOXA (AbdB-HOXA) including HOXA7-

HOXA11, with only HOXB3 and HOXC3 expressed from other HOX clusters (Taghon,

Thys et al. 2003).

Page 24: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

10

1.4.1 Gain of function studies

The function of HOX genes in normal haematopoiesis has been widely studied

using gene expression analysis and knockin or knockout studies in HSCs and early HPs

(Table 1.1). Generally the over-expression of a HOX gene leads to an expansion of stem

and progenitor cell populations together with a block on differentiation. Notable

example of this include the over-expression of murine Hoxb6, which resulted in the

expansion of HSCs and myeloid progenitors, together with the inhibition of

erythropoiesis and lymphopoiesis (Fischbach, Rozenfeld et al. 2005), and over-

expression of murine Hoxb3 that resulted in several haematological abnormalities, such

as a block of B- and T-cell differentiation as well as a delay in MP proliferation

(Sauvageau, Thorsteinsdottir et al. 1997). Over-expression of human HOXC4 resulted

in expansion of early and committed myeloid and erythroid progenitors (Daga, Podesta

et al. 2000), and knockin of human HOXA5 caused an increase in the number of myeloid

progenitors and blocked erythroid differentiation (Crooks, Fuller et al. 1999; Fuller,

McAdara et al. 1999). Likewise, over-expression of HOXA10 in human cord blood or

fetal liver CD34+ HPs resulted in a significant production of blast cells and

myelopoiesis concomitant with a complete block of erythroid differentiation and a

severe reduction in B-cell development (Buske, Feuring-Buske et al. 2001). Other HOX

genes are required for the maintenance of progenitor or stem cell status and promote

their proliferation, especially HOXA9 and HOXB4. The former is the most

preferentially expressed HOX gene in human CD34+ HSCs and early HPs and is

subsequently down-regulated during differentiation. Murine Hoxa9 over-expression

enhances HSC expansion and myeloid progenitor proliferation and, with a long latency,

leads to leukaemia (Kroon, Krosl et al. 1998; Thorsteinsdottir, Mamo et al. 2002). In

Page 25: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

11

contrast to myeloid progenitors, Hoxa9 over-expression resulted in a partial inhibition

of pre-B cell differentiation, but did not affect T-cell development (Thorsteinsdottir,

Mamo et al. 2002). Hoxb4 is also highly expressed in HSCs and down-regulated during

differentiation (Sauvageau, Lansdorp et al. 1994; Pineault, Helgason et al. 2002). Its

over-expression in murine and human cell lines results in a remarkable expansion of

HSCs in vivo and in vitro without resulting in leukaemia or lineage disturbances

(Sauvageau, Thorsteinsdottir et al. 1995; Amsellem, Pflumio et al. 2003). Indeed, the

self-renewal ability of Hoxb4-transduced HSCs is 20-50 fold greater than untreated

cells, and can be increased still further by knocking down Pbx1 (Krosl, Beslu et al.

2003; Cellot, Krosl et al. 2007).

1.4.2 Loss of function studies

In addition to the knockin and over-expression approaches described above,

knockdown and deletion studies in murine models and cell lines have also been used to

evaluate the role of HOX genes in haematopoiesis. However, owing to the functional

redundancy of HOX genes, the results of knockdown assays are sometimes difficult to

interpret and do not always reflect the findings of studies where the gene has been over-

expressed. For example, it has been found that Hoxb4 null mice exhibit a significant

reduction in size and cellularity of haematopoietic organs, such as spleen and liver, and

a slight decrease in HSCs and HPs number without a significant disturbance of lineage

commitment (Brun, Björnsson et al. 2004; Bijl, Thompson et al. 2006). Likewise,

Hoxb3 null mice display a notable reduction in B-cell progenitors, and a reduction in

BM cellularity, but no significant reduction in B-cell numbers in the spleen (Ko, Kwan

Lam et al. 2007). HOXb3/b4 -/- mice have a greater reduction in HSCs and HPs, yet no

Page 26: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

12

difference in haematopoietic cell commitment (Bjornsson, Larsson et al. 2003). Using

polymerase chain reaction (PCR), it has been demonstrated that fetal liver cells (c-Kit+)

of Hoxb4 null mice expressed other Hoxb genes to a significantly higher level than

control cells (Bijl, Thompson et al. 2006). Thus, Hoxb3 and Hoxb4 can regulate the

normal function of HSCs but are not individually essential for the production of major

blood lineages because of redundancy with other members of the Hox gene family.

Bijl and colleagues (2006) found that an individual loss of Hoxb4 or even the

complete loss of the Hoxb cluster (b1-b9) did not affect the ability of murine fetal liver

HSCs to self-renew. The repopulation and differentiation potential were retained,

compared with wild-type control cells. Thus, the Hoxb cluster may not be necessary for

haematopoiesis, with members of the other Hox clusters presumably having largely

duplicate roles. Analysis of Hox gene expression in fetal liver cells (c-Kit+ Hoxb1-

Hoxb9-/-), revealed genetic interactions between members of the Hoxa, Hoxb and Hoxc

clusters, whereby these cells exhibited down-regulation of all Hoxa genes, except

Hoxa13, and up-regulation of Hoxc4, Hoxc9 and Hoxc11, also suggesting functional

redundancy and complex genetic interactions between Hox genes.

An exception to the general prevalence of functional redundancy amongst the

HOX gene family is HOXA9, the most highly expressed HOX family member in HSCs.

Hoxa9-/- mice showed significant deficiencies in myeloid and lymphoid cells

concomitant with a significant defect in repopulating ability (So, Karsunky et al. 2004;

Lawrence, Christensen et al. 2005). These deficiencies include commom myeloid

progenitors, granulocyte/monocyte progenitors, common lymphoid progenitors and

lymphoid progenitors (pro- and pre-B cells, pro-T cells) (So, Karsunky et al. 2004).

There is also a corresponding reduction in spleen cellularity and size (Magnusson, Brun

Page 27: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

13

et al. 2007). Notably, compared to the entire cluster Hoxb deficient fetal liver HSCs,

Hoxa9-/- fetal liver HSCs exhibited a more dramatic defect in repopulating ability (Bijl,

Thompson et al. 2006; Magnusson, Brun et al. 2007), and HSCs from Hoxa9/b3/b4 null

mice had the same repopulating ability as those from Hoxa9 null mice (Magnusson,

Brun et al. 2007). Gene knockdown studies have revealed that some additional HOX

genes are also essential in normal haematopoiesis. Knockdown of HOXA5 led to an

increase in erythroid progeneitors and a reduction in the number of myelomonocytic

cells (Crooks, Fuller et al. 1999; Fuller, McAdara et al. 1999), and Hoxa7 null mice

showed a reduction in megakaryocytic/erythroid progenitors as well as reticulocytosis

and thrombocytopenia (So, Karsunky et al. 2004). Knockout of Hoxb6 resulted in an

increase in early erythroid progenitors in murine BM and fetal liver cells (Kappen

2000). Likewise, Hoxc3-/- mice showed a reduction in late erythroid progenitors without

affecting the haemoglobinization size (Takeshita, Bollekens et al. 1993), and Hoxc8

deficient mice showed a significant reduction in erythroid, granulocyte and macrophage

colony formation potential (Shimamoto, Tang et al. 1999).

Page 28: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

14

Table 1.3 HOX gene studies (Previously published in Alharbi, Pettengell et al. 2012).

HOX gene Gain of function Loss of function Species

HOXA5 ↑Myeloid progenitors and

block erythroid

differentiation (Crooks,

Fuller et al. 1999; Fuller,

McAdara et al. 1999)

↑Erythroid progenitors and

↓myelomonocytic cells (Crooks,

Fuller et al. 1999; Fuller, McAdara

et al. 1999)

Human

Hoxa7 ↓ MEP, reticulocytosis and

thrombocytopenia (So, Karsunky

et al. 2004)

Mouse

Hoxa9 ↑HSCs expansion and

myeloid progenitor

proliferation.

Block erythroid

differentiation (Kroon,

Krosl et al. 1998;

Thorsteinsdottir, Mamo et

al. 2002).

↓↓CMP, GMP, CLP, lymphoid

progenitors, repopulating ability

and spleen cellularity and size (So,

Karsunky et al. 2004; Lawrence,

Christensen et al. 2005).

Mouse

HOXA10 ↑↑ Blast cells and

myelopoiesis, ↓ B cell

differentiation and block

erythroid differentiation

(Buske, Feuring-Buske et

al. 2001).

Human

Hoxb3 Block B- and T-cell

differentiation and a delay

in myeloid progenitor

proliferation (Sauvageau,

Thorsteinsdottir et al. 1997).

↓↓ B-cell progenitors and bone

marrow cellularity (Ko, Kwan

Lam et al. 2007).

Mouse

HOXB4/

Hoxb4

↑↑ HSCs expansion

(Sauvageau,

Thorsteinsdottir et al. 1995;

Amsellem, Pflumio et al.

2003).

↓↓ Haematopoietic organs

cellularity and size, ↓ HSCs and

HPCs and ↑ Hoxb genes (Brun,

Björnsson et al. 2004; Bijl,

Thompson et al. 2006).

Human /

mouse

Hoxb3/b4 ↓↓ HSCs and HPCs (Bjornsson,

Larsson et al. 2003).

Mouse

Hoxb6 ↑HSCs expansion and

myeloid progenitors.

↓ erythropoiesis and

lymphopoiesis (Fischbach,

Rozenfeld et al. 2005).

↑ Early erythroid progenitors

(Kappen 2000).

Mouse

Hoxc3 ↓ Erythroid progenitors (Takeshita,

Bollekens et al. 1993).

Mouse

HOXC4 ↑ Early and committed

myeloid and erythroid

progenitors (Daga, Podesta

et al. 2000).

Human

Hoxc8 ↓Erythroid, granulocyte and

macrophage colony formation

potential (Shimamoto, Tang et al.

1999).

Mouse

Page 29: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

15

1.5 Upstream regulators of HOX genes

Knockout models of HOX gene upstream regulators have helped to define their role in

normal haematopoiesis. Regulators include transcriptional activators such as mixed

lineage, myeloid lymphoid, leukaemia (MLL), and a family of caudal-type homebox

transcription factors (CDX1, CDX2, and CDX4). The existence of HOX genes in

clusters makes them particularly sensitive to changes in chromosomal organization, and

repressors of HOX transcription include genes that mediate this process, most notably

members of the polycomb group (Beuchle, Struhl et al. 2001). These regulators have

crucial roles in normal development and haematopoiesis through the regulation of HOX

genes. A number of studies demonstrated that Mll-deficient embryonic bodies and Mll-

conditional knockout mice showed a dramatic reduction in HSCs and HPs. In addition,

these embryonic bodies and the mice exhibited greatly reduced expression of a number

of Hox genes including Hoxa7, Hoxa9, Hoxa10 and other Hoxb and Hoxc genes (Ernst,

Mabon et al. 2004; Jude, Climer et al. 2007). Likewise, Cdx compound-deficient

zebrafish and murine embryonic stem cells showed dysregulation of the embryonic HPs

as well as impaired expression of Hox genes (Davidson and Zon 2006; Wang, Yabuuchi

et al. 2008). However, it has been shown that Cdxs are not essential for normal

haematopoiesis in adult mice. For example, Cdx4-deficient mice showed minimal

haematopoietic defects, though it was highly expressed in wild-type myeloid progenitor

cells (Koo, Huntly et al. 2010). In addition, human CDX2 is not expressed in normal

HSCs, or in myeloid, B-cell, or T-cell progenitors (Scholl, Bansal et al. 2007; Rawat,

Thoene et al. 2008). In addition, it has been found that HOX gene expression is also

regulated by small single-stranded RNAs (miRNAs) and the non-coding RNA

HOTAIR (Bhatlekar, Fields et al. 2014).

Page 30: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

16

miRNAs regulate HOX expression by repressing the expression of anterior genes,

thereby supporting posterior prevalence while HOTAIR was reported as a negative

regulator of HOX expression (Yekta, Tabin et al. 2008; Nakayama, Shibazaki et al.

2013).

1.6 HOX downstream target genes in haematopoietic cells

The mechanism by which HOX proteins regulate haematopoiesis is not yet fully

understood. However, genome-wide analyses after experimentally induced changes in

HOX genes expression have identified some potential downstream targets. Amongst

these are the HOX genes themselves, some of which have been shown to cross-regulate

their neighbours, or their cofactors. HOXA9, HOXA10 and HOXB4 are the most

comprehensively studied genes in this respect because of their key roles in normal

haematopoiesis and leukaemia. It is particularly noteworthy that HOXA9 positively

regulates the transcription of other HOX genes including HOXA7 and HOXA10 and its

cofactor PBX3 and MEIS1 (Faber, Krivtsov et al. 2009). A summary of HOX

downstream target genes is presented in Table 1.2.

As described above, HOXA9 is a key regulator of haematopoiesis and behaves

as an oncogene in leukaemia. It is therefore unsurprising that it activates the

transcription of genes known to regulate cell proliferation and survival. For example,

Hoxa9 directly activates the Pim1 gene, the product of which enhances proliferation by

activating c-Myb, and also exerts an anti-apoptotic effect by phosphorylating and

inactivating the BAD protein (Leverson, Koskinen et al. 1998; Hu, Passegué et al.

2007). c-Myb has also been identified as an indirect transcriptional target of Hoxa9-

Mies1 that mediates transformation in Mll-Enl leukaemia (Hess, Bittner et al. 2006).

Other HOXA9 targets include the oncogene ID2, which is up-regulated, and BIM,

Page 31: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

17

which encodes an apoptotic factor and is down-regulated (Nagel, Venturini et al. 2010).

HOXA9 also activates the CYBB gene, which encodes the Gp91phox (a phagocyte

respiratory burst oxidase protein), and is expressed in differentiated myeloid cells (Bei,

Lu et al. 2005). In mice, Hoxa9 has been shown to directly activate the transcription of

the flt3 gene, which is associated with an unfavourable prognosis of AML (Gwin, Frank

et al. 2010) and it also regulates its own cofactor, Meis1, through binding to Meis1

upstream regulator genes cerb1 and pknox1 (Hu, Fong et al. 2009). More proliferative

genes have been identified as downstream targets for Hoxa9 including Camk2d, Cdk6,

Erg, Etv6, Flt3, Foxp1, Gfi1, Kit, Lck, Lmo2, Myb and Sox4 (Huang, Sitwala et al.

2012). In the same study, it was shown that Hoxa9 down-regulates differentiation and

inflammation genes including Ifit1, Tlr4, Ccl3, Ccl4, Csf2rb, Ifngr1, Runx1, Cd28 and

Cd33. HOXA9 also regulates the anti-apoptotic gene Bcl-2 which may explain the cell

survival role of HOXA9 (Brumatti, Salmanidis et al. 2013). The fusion protein

nucloporin 98 (NUP98-HOXA9) has been found to stimulate the proliferation of HSCs

by activating the expression of other HOX genes; including HOXA9, HOXA7, MEIS1

and PBX3. It also up-regulates a number of leukaemogenic transcription factors

including EVI1 and MEF2C, and receptor tyrosine kinases including FLT3 and KIT

(Takeda, Goolsby et al. 2006).

It is also of note that there are both overlapping and opposing functions between

the closely related HOXA9 and HOXA10 transcription factors. For example, in a similar

manner to Hoxa9, Hoxa10 activates the expression of proliferative genes that result in

myeloid progenitor expansion such as Itgb3, Hif, Tgfβ2 and Fgf2 by direct binding to

their promoters (Bei, Lu et al. 2007; Magnusson, Brun et al. 2007; Shah, Wang et al.

2011; Shah, Bei et al. 2012). HOXA10 also activates the transcription of anti-apoptotic

Page 32: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

18

genes such as DUSP4 which encodes mitogen-activated protein phosphatase 2.

mitogen-activated protein phosphatase 2 in turn prevents cell death by down-regulating

JNK (Wang, Lu et al. 2007). Hoxa10 decreases erythroid differentiation and

megakaryopoiesis by activating Hoxa5 and inactivating Gata-1, respectively

(Magnusson, Brun et al. 2007), and it also induces Cdx4 expression in myeloid cells

(Bei, Huang et al. 2011).

Unlike HOXA9, HOXA10 can also exert anti-proliferative effects. For

example, in cooperation with its trimeric cofactors, HOXA10 induces p21 transcription

leading to cell cycle arrest and differentiation (Bromleigh and Freedman 2000). It also

represses CYBB transcription (Eklund, Jalava et al. 2000), thereby acting in an opposing

manner to HOXA9. In a fusion form with Nup98, Hoxa10 activates more than 400

genes including the self-renewal genes Flt3, Prnp, Hlf and Jag2 (Palmqvist, Pineault et

al. 2007).

Many HOXB4 target genes have also been identified in three studies (Lee,

Zhang et al. 2010; Oshima, Endoh et al. 2011; Fan, Bonde et al. 2012). Over-expression

of Hoxb4 resulted in transcriptional up-regulation of Meis1, Dntt, Hlf, Sox4 and Runx2,

while it down-regulated the transcription of lymphoid specific genes, such as B220, and

myeloid-specific genes, such as Hmbs (Lee, Zhang et al. 2010). Some HOXB4 targets

seem to vary in a context-dependent manner, for example, it has been found to down-

regulate the transcription of c-MYC in the HL-60 cell line leading to cell differentiation

(Pan and Simpson 2001), while it activates c-Myc transcription in murine BM cells

(Satoh, Matsumura et al. 2004). As with HOXA9, Hoxb4 activates the transcription of

its neighbouring genes, Hoxb2, Hoxb3 and Hoxb5 (Satoh, Matsumura et al. 2004).

Hoxb4 also activates activator protein-1 (AP-1) complex members Fra-1 and Jun-B,

Page 33: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

19

which leads in turn to an increase in the level of cyclin-D1 and a decrease the level of

c-Fos transcription, thereby increasing the proliferation capacity of HSCs (Krosl and

Sauvageau 2000).

Page 34: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

20

Table 1.4 A summary of mammalian HOX target genes (Alharbi, Pettengell et al. 2012).

HOX protein Targets of transcriptional activation Targets of transcriptional repression Species

HOXA9/ Hoxa9 Pim1 (Hu, Passegué et al. 2007), ID2 (Nagel,

Venturini et al. 2010), CYBB (Bei, Lu et al.

2005), HOXA7, HOXA10, PBX3, MEIS1

(Faber, Krivtsov et al. 2009). Bcl-2

(Brumatti, Salmanidis et al. 2013), Flt3

(Gwin, Frank et al. 2010), Cerb1 and Pknox1

(Bei, Lu et al. 2005), Camk2d, Cdk6, Erg,

Etv6, Foxp1, Gfi1, Kit, Lck, Lmo2, Myb and

Sox4 (Huang, Sitwala et al. 2012).

BIM (Nagel, Venturini et al. 2010)/

Itfi1, Tlr4, Ccl3, Ccl4, Csf2rb, Ifngr1,

Runx1, Cd28, Cd33 (Huang, Sitwala et

al. 2012).

Human/

mouse

Hoxa9-Meis1 c-Myb (Hess, Bittner et al. 2006). Mouse

NUP98-HOXA9 HOXA7, HOXA9, MEIS1, PBX3, EVI1,

MEF2C, FLT3 and KIT (Takeda, Goolsby et

al. 2006).

Human

HOXA10/

Hoxa10

P21 (Bromleigh and Freedman 2000),

DUSP4 (Wang, Lu et al. 2007), Itgb3 (Bei,

Lu et al. 2007), Hlf (Magnusson, Brun et al.

2007), Tgfβ2 (Shah, Wang et al. 2011), Fgf2

(Shah, Bei et al. 2012), Dusp4 (Wang, Lu et

al. 2007), Hoxa5 (Magnusson, Brun et al.

2007), Cdx4 (Bei, Huang et al. 2011).

CYBB (Eklund, Jalava et al. 2000)/

Gata1 (Magnusson, Brun et al. 2007)

Human/

mouse

Nup-Hoxa10 Flt3, Prnp, Hlf and Jag2 (Palmqvist, Pineault

et al. 2007).

Mouse

HOXB4/ Hoxb4 MEIS1, DNTT, HLF, SOX4, RUNX2 (Lee,

Zhang et al. 2010), c-MYC (Pan and Simpson

2001), Hoxb2, Hoxb3 (Satoh, Matsumura et

al. 2004), Fra-1, JunB (Krosl and Sauvageau

2000).

B220 and HMBS (Lee, Zhang et al.

2010), c-Myc (Satoh, Matsumura et al.

2004).

Human/

mouse

Page 35: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

21

1.7 The role of HOX genes in acute leukaemia

Numerous studies have now shown that HOX genes can promote the

development of AML by forming chimeric fusions with other genes, but more recent

work has also shown that their miss-expression, in particular their over-expression, is

also important in the formation of malignancy.

1.7.1 HOX fusion proteins

One of the most frequent fusion partners for HOX genes is nucloporin (NUP98),

a member of the nuclear pore family (Figure 1.2). It is localized in the nuclear

membrane and functions as a selective transporter for ribonucleic acid (RNA) and

proteins between the nucleus and cytoplasm. NUP98-HOX fusion proteins have been

reported in AML and other leukaemias. In AML, NUP98-HOXA9 is associated with a

t(7;11)(p15;p15) translocation (Borrow, Shearman et al. 1996; Nakamura, Largaespada

et al. 1996). There are eight other HOX genes that can be fused with NUP98, including

HOXA11 and HOXA13 (Fujino, Suzuki et al. 2002; Suzuki, Ito et al. 2002), HOXD11

and HOXD13 (Raza-Egilmez, Jani-Sait et al. 1998; Taketani, Taki et al. 2002), and

HOXC13 (Taketani, Taki et al. 2002). Thus only the 5’ most members of each HOX

complex have been documented to be fused with NUP98 in AML. However, Hoxb3

has been shown to be a potential leukaemogenic partner with Nup98 (Pineault,

Abramovich et al. 2004), suggesting that the ability to be a fusion NUP98 partner is not

limited to the 5’ most HOX genes. Generally, NUP98-HOX fusions induce cell

proliferation and function as transcriptional activators and NUP98 fusions with HOX

proteins are more oncogenic than fusions with other partners (Saw, Curtis et al. 2013).

Page 36: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

22

Nup98-Hox fusion proteins result in AML with a long latency, around 11-12

months (Kroon, Thorsteinsdottir et al. 2001). However, this latency can be reduced to

two months by co-over-expression of the Hox cofactor Meis1 (Pineault, Buske et al.

2003) and the receptor tyrosine kinase Flt3 (Palmqvist, Argiropoulos et al. 2006). For

example, concurrent translocation of Nup98-Hoxd13 and Flt3-ITD developed AML in

3 months (Greenblatt, Li et al. 2012). FLT3 has an essential role in the regulation of

early HPs growth, and causes increased and uncontrolled self-renewal of these cells

through a FLT3 ligand-independent pathway (Tosic, Stojiljkovic et al. 2009).

Figure 1.3 structures of AbdB-HOX, NUP98 and the predictive fusion protein NUP98-HOXA. A) A

general structure of AbdB-HOX (9-13) proteins that have been reported to fuse with NUP98. B) Structure

of normal NUP98 protein. C) Structure of predictive NUP98-HOX fusion protein. This fusion eliminates

MD and RBD from AbdB and NUP98, respectively. The arrows represent the breakpoints. MD: MEIS

domain, PM: Pbx motif, H: hexapeptide, HD: homedomain, FG: phenylalanine-glycine, GLEBS: gle2p-

binding-like motif, RBD: RNA binding domain (Previously published in Alharbi, Pettengell et al. 2012).

PMMD COOHNH2

HOX

GLEBS RBDFGNH2 COOH

NUP98

HDPMGLEBS COOHNH2

NUP98-HOX

A)

B)

C)

FG

FG FG

HDH

H

Page 37: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

23

1.7.2 HOX over-expression in AML

HOX genes may also be indirectly involved in AML through chromosomal

rearrangements that involve their upstream regulators, such as MLL. MLL fusion

proteins constitute about 10% of therapy related AML and 3% of de novo AML (Slany

2009). There are more than 64 translocation partner genes that fuse with the MLL N-

terminus (Meyer, Kowarz et al. 2009). Normally, Mll positively regulates the

transcription of Hox genes by maintaining their expression through direct binding to a

proximal promoter region (Milne, Briggs et al. 2002). MLL fusion proteins activate

HOX gene transcription more efficiently than MLL alone (Liu, Cheng et al. 2009; Slany

2009), especially the 5’ end members of the HOXA cluster, together with their co-

activator MEIS1. As a consequence, myeloid differentiation is blocked and proliferation

is enhanced (Marschalek 2011). Consistent with this proposed mechanism, it has been

reported that MLL-AF9, like NUP98-HOXA9, leads to a block in erythroid/myeloid

maturation and to erythroid hyperplasia (Abdul-Nabi, Yassin et al. 2010), and the Mll-

Enl fusion protein requires Hoxa7 and Hoxa9 for efficient immortalization of HPs

(Ayton and Cleary 2003). Conversely, a number of studies demonstrated that the

expression of HOXA genes is not crucial for MLL leukaemogenesis, yet their

expression affects disease phenotype. For instance, Hoxa7 and Hoxa9 influence AML

latency and phenotype; yet they are not essential to initiate Mll-Gas7-mediated

leukaemogenesis (So, Karsunky et al. 2004). Furthermore, suppression of HOXA9

expression in cells with a chimeric MLL-AF9 gene reduces the survival of leukaemic

cells and changes the disease phenotype, but it does not affect AML initiation (Faber,

Krivtsov et al. 2009).

Page 38: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

24

The dysregulation of another regulator of HOX genes, the CDX gene family, has

also been shown to drive the development of AML. CDX2 is expressed in the majority

of AML cases (90%), but not in normal adult haematopoiesis, and Cdx2-elevated

expression leads to AML with only a short latency period (Scholl, Bansal et al. 2007).

In contrast, the closely related CDX4 gene is expressed in 25% of AML cases, and is

expressed in normal adult haematopoiesis, and Cdx4 over-expression in murine BM

results in AML but only with a long latency period (Bansal, Scholl et al. 2006). This

latency can be accelerated in mice through cooperation of Meis1 which results in the

over-expression of a number of Hox genes including Hoxa6, Hoxa7, Hoxa9, Hoxb4,

Hoxb8 and Hoxc6 (Bansal, Scholl et al. 2006). Cdx2 expression alone is sufficient to

drive the up-regulation of a related set of HOX genes (Rawat, Thoene et al. 2008),

demonstrating the importance of the Cdx family in the dysregulation of Hox genes

during AML.

The dysregulation of HOX gene expression is also associated with the

nucleophosmin 1 (NPM1) mutation. NPM1 is a chaperone protein that shuttles between

the nucleus and cytoplasm, although its predominant localization is in the nucleus (Rau

and Brown 2009). NPM1 has a crucial role in several biological processes, such as

ribosome biogenesis, genomic stability and cell cycle progression. In adult AML,

NPM1 mutation is the most common genetic aberration, reported in about 35% of all

adult AML and approximately in 45-55% of NK-AML (Falini, Mecucci et al. 2005). In

pediatric AML, NPM1 mutations are significantly less common, occurring in 8-10% of

cases, and in about a quarter of normal karyotype cases (Brown, McIntyre et al. 2007;

Hollink, Zwaan et al. 2009). The relocation of NPM1 into the cytoplasm (NPMc+)

occurs only in AML (Falini, Bolli et al. 2009). This relocation causes up-regulation of

Page 39: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

25

a number of HOX genes, some of which are similar to those seen in AML initiated by

a MLL chimeric gene fusion, while some are distinct. Thus for example, HOXA4,

HOXA6, HOXA7, HOXA9, HOXB9 and MEIS1 are over-expressed in both contexts,

while HOXB2, HOXB3, HOXB5, HOXB6 and HOXD4 are up-regulated in NPMc+

AML only (Mullighan, Kennedy et al. 2007). It has been reported that activation of a

humanized Npm1 allele led to over-expression of Hoxa5, Hoxa7, Hoxa9 and Hoxa10,

induction of HSC self-renewal and the expansion of myelopoiesis (Vassiliou, Cooper

et al. 2011). The exact mechanism of the association between the NPM1 mutation and

the up-regulation of HOX genes is still unclear. A possible explanation is that NPM1

directly disturbs the expression of HOX genes, or alternatively, that NPM1 mutations

arrest the differentiation of early HPs in which HOX expression is up-regulated (Rau

and Brown 2009).

1.7.3 HOX gene dysregulation in acute lymphoid leukaemia (ALL)

The dysregulation of HOX genes has also been reported in ALL including both

B- and T-progenitor ALL (B-ALL and T-ALL), especially when MLL translocations

are involved. For example, human HOXA9, HOXA10 and HOXC6, and their cofactor

MEIS1 were up-regulated in both MLL-ENL T-ALL and MLL B-ALL (Ferrando,

Armstrong et al. 2003). Human HOXA9 and MEIS1, and murine Hoxa5, Hoxa9 and

Meis1 were likewise up-regulated in MLL-AF4 B-ALL (Rozovskaia, Feinstein et al.

2001; Krivtsov, Feng et al. 2008). A common signature of MLL rearrangements in

AML and ALL is the over-expression of HOXA genes including HOXA3, HOXA5,

HOXA7, HOXA9 and HOXA10 (Zangrando, Dell'Orto et al. 2009). In addition, HOX

genes can be involved in other translocations such as CALM-AF10 T-ALL, where

human HOXA5, HOXA9, HOXA10 and MEIS1 are over-expressed (Dik, Brahim et al.

Page 40: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

26

2005). HOXA proteins can also form chimeric fusion proteins with T-cell receptor in

T-ALL, which results in the global over-expression of HOXA genes (Soulier, Clappier

et al. 2005; Speleman, Cauwelier et al. 2005). Surprisingly, the aberrant expression of

CDX2 in ALL, an upstream regulator of HOX genes, is not correlated with HOX gene

dysregulation (Thoene, Rawat et al. 2009).

1.7.4 HOX genes as prognostic markers

HOX gene expression has become an important prognostic factor in AML.

Over-expression of HOX genes is associated with an intermediate/unfavorable

cytogenetic subset of AML. For example, among 6817 genes that have been

investigated in AML patients, the single gene correlated with the worst outcome and

relapse of disease as well as short survival was HOXA9 (Golub, Slonim et al. 1999).

Likewise, high expression of HOXA9 associates with low CR rate (Li, Li et al. 2013).

Correspondingly, low HOXA9 expression was found to correlate with the best outcome

and response to therapy (Andreeff, Ruvolo et al. 2008). It is also noteworthy that low

expression of HOXA4 and MEIS1 are also favorable predictors for AML patient

outcome (Zangenberg, Grubach et al. 2009). In addition, a signature of up-regulation

of HOXA6, HOXA9 and PBX3 with a low expression of MEIS1 is an independent poor

prognostic marker in NK-AML, and simultaneous knockdown of HOXA6 and HOXA9

caused cell death and AML cell lines became more sensitive to cytarabine (Dickson,

Liberante et al. 2013). Furthermore, high expression of HOXA7, HOXA9 and HOXA11

along with the cofactor PBX3 is an independent prognosis marker of adverse OS in

abnormal karyotype AML (Li, Huang et al. 2012). The global levels of HOX expression

also seem to reflect the outcome of disease, possibly reflecting the functional

redundancy exhibited by many of the HOX genes. Thus the highest levels of HOX genes

Page 41: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

27

are seen in FLT3 mutation cases, which have unfavorable outcomes, and generally the

HOX genes are expressed only at a very low level in favorable subsets of AML.

Although recent work has established the importance of HOX genes in the

development of AML, it is still not clear exactly what the functions of these genes are

beyond a general inhibition of differentiation and an increase in cell proliferation. This

lack of precise mechanistic knowledge for individual HOX genes may be owing to the

functional redundancy showed by many members of this family, making the

knockdown of single HOX genes generally difficult to interpret, and it may also help to

explain the contrast in gene knockin and knockout results. In myeloma and some solid

malignancies this has been addressed by targeting the HOX proteins as a group by

antagonizing their interactions with the PBX cofactor using the peptide inhibitor HXR9

(Morgan, Pirard et al. 2007; Shears, Plowright et al. 2008; Plowright, Harrington et al.

2009; Daniels, Neacato et al. 2010; Morgan, Plowright et al. 2010).

PBX binds to PBX motif in HOX paralogus group 1-10. HOX-PBX dimers have

more binding affinity and specificity for target DNA sequences than the HOX monomer

alone. Of note, HOX-PBX dimers are strictly involved the interaction of PBX to the

conserved hexapeptide sequence WYPWMK that is found N terminal to the

homeodomain of HOX proteins from paralogous groups 1-10 for cooperative DNA

binding by PBX and HOX partners (Chang, Shen et al. 1995; Shen, Rozenfeld et al.

1997; Medina-Martinez and Ramirez-Solis 2003). The peptide inhibitor HXR9 contains

a hexapeptide sequence mimics the HOX protein hexapeptide sequences that binds to

PBX and thereby interfering the DNA binding of HOX-PBX. HXR9 is cytotoxic to

cells, predominantly through the induction of apoptosis, which in turn depends upon a

rapid increase in expression of c-FOS (Morgan, Pirard et al. 2007; Shears, Plowright et

Page 42: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

28

al. 2008; Espinosa, Shinohara et al. 2009; Plowright, Harrington et al. 2009; Daniels,

Neacato et al. 2010). A similar approach may be useful in AML for addressing

redundant functions of HOX genes.

1.8 Hypothesis and aims

The hypothesis of this study was that AML would also be sensitive to killing by

HXR9, and indeed it may be that the HOX genes could represent a useful therapeutic

target, especially in AMLs that show high levels of HOX expression and a

correspondingly poor prognosis. In order to assess this hypothesis, three main

objectives were set up.

The first objective of this study was to investigate the cytotoxicity of HXR9 on

AML cell lines derived from different AML subtypes and if so to investigate the

combination therapy of HXR9 with AML first line chemotherapy. The strategies for

conducting this were:

To measure the expression of the whole panel of 39 HOX genes in five

AML cell lines.

To investigate the cytotoxicity of HXR9 on the five cell lines either

alone or in combination with AML first line chemotherapy.

The second objective was to investigate the mechanism of HXR9 cytotoxicity

on the AML-derived cell lines. The strategies for conducting this were:

To determine whether HXR9 induced cell death through apoptosis or

necrosis.

Page 43: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

29

To assess the involvement of some signaling pathways such as MAPK

and pro-apoptotic proteins such as p53.

The third objective was to investigate the efficacy of HXR9 against AML

disease in vivo. The strategies for conducting this were:

To establish an AML model.

To assess the efficacy of HXR9 either alone or with co-treatment with

some non-toxic inhibitors.

Page 44: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

30

Chapter 2 Materials and Methods

Page 45: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

31

2.1 Materials

2.1.1 Reagents

Table 2.1 Reagents were used in this study and their suppliers.

Reagent Supplier

SYBR® green jumpstatTM Taq ready mixTM Sigma (Dorset, UK)

Penicillin-streptomycin

μI-1640

DMEM

Trypan blue

HBSS

Histopaque®-1077 Hybri-Max

β-mercaptoethanol

Daunorubicin

Mitoxantrone

DAPI stain

z-VAD-FMK

Cyclosporin A

Necrostatin-1

Fructose

EDTA

U0126 monoethanolate

SP600125

SB 203580

DPI

Calpain inhibitor I

W-7

Ro31-8220 methanesulfonate and pifithrin-α

Page 46: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

32

Reagent Supplier

Protoporphyrin IX Sigma (Dorset, UK)

Carestream® Kodak BioMax® light film

Fixer and replenisher

Developer and replenisher

Cloned AMV first-strand synthesis kit invitrogen, Thermo scientific (Paisley, UK)

Glutamine

FBS

IMDM medium

Enz Chek® Caspase-3 Assay Kit#2

NuPAGE® LDS sample buffer

BenchmarkTM prestained protein ladder

NuPAGE novex 12% Bis Tris

DMSO

Pierce® RIPA buffer

Protease inhibitor cocktail

NuPAGE® sample reducing agent

NuPAGE® MES SDS running buffer

InvitrolonTM PVDF filter paper sandwich

Novex cryoTubeTM vials

SuperSignal® West Pico Chemiluminescent substrate

Annexin V-PE apoptosis detection kit BD pharmingen (Cambridge, UK)

Cytotoxicity detection kit Roche (Mannheim, Germany)

RNeasy mini kit and RBC lysis solution Qiagen (West Sussex, UK)

Gene Primers Eurgentec (Seraing, Belgium)

Mycoplasma detection kit lonza (Slough, UK)

Page 47: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

33

2.1.2 Instruments

Instruments used in this study were: Beckman coulter a plate reader, Miltenyibiotec a

fluorescence-activated cell sorting (FACS) machine, Lebtech a Nanodrop spectrophotometer

ND-1000, and Stratagene Mx 3005P, SHADON cytospin 4, a fluorescent microscope,

eppendorf centrifuge 5415R.

2.1.3 Cell lines

Human and mouse derived AML cell lines were used in this study. Human derived cell

lines used were primary or secondary. Primary AML cell lines derived from patients with de

novo mutations including KG-1, a cell line derived from an erythroleukaemia (Koeffler and

Golde 1978), HEL 92.1.7, an erythroleukaemia cell line (Martin and Papayannopoulou 1982)

and HL-60, a cell line obtained from acute promyelocytic leukaemia (APML) (Collins, Ruscetti

et al. 1978). Secondary AML cell lines are derived from patients that developed AML from

MDS, chronic myeloid leukaemia (CML) or following previous treatment with chemotherapy.

These include K562, a CML cell in blast crisis (Lozzio and Lozzio 1975) and KU812F, also a

CML cell in blast crisis (Kishi 1985). The mouse derived cell line G1498-GFP was a kind gift

from Dr. Justin Kline (Department of Medicine Section of Haematology/Oncology, Chicago,

USA).

2.1.4 HXR9 and CXR9 peptide synthesis

The synthesis of these peptides has been described previously (Morgan, Pirard et al.

2007). Briefly, HXR9 is a short peptide of 18 amino acids, comprising a hexapeptide sequence

and nine C-terminal arginine residues (R9) that enables cell penetration. The hexapeptide

mimics the hexapeptide sequence of HOX proteins of paralogs 1-10, this sequence functions as

a competitive inhibitor of HOX/PBX. CXR9 is a control peptide that does not have a functional

hexapeptide portion but has the R9. The stability of the peptides was increased using D-isomers

Page 48: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

34

NH2- and COOH- terminals. The half-life of both peptides is about 12 hours at the serum

(Morgan, Pirard et al. 2007). A conventional column-based technique was used to synthesise

HXR9 and CXR9 and were purified to at least 80% (Biosynthesis Inc., Lewisville, TX, USA).

The peptide sequences are as follows:

HXR9: WYPWMKKHHRRRRRRRRR (2700.06 Da)

CXR9: WYPAMKKHHRRRRRRRRR (2604.14 Da)

2.1.5 Mice

Female C57BL/6, nude and severe combined immune-deficient (SCID) mice,

approximately 6 to 8 weeks age, were purchased from Jackson laboratory, Kent, UK. These

mice were maintained in pathogen-free cages and fed with irradiated food and acidified water.

All experiments were performed under the compliance of Home Office and institutional

instructions.

2.2 Methods

2.2.1 General cell culture methods

2.2.1.1 Routine cell culture

All cell culture techniques were performed in a class II microbiological safety cabinet

and all equipment and reagent bottles were sterilised using 70% ethanol solution before use.

K562, KU812F and HEL92.1.7 cells were grown in a culture medium consisting of Roswell

Park Memorial Institute (RPMI-1640) medium complemented with 10% fetal bovine serum

(FBS), 5% L-glutamine and 5% antibiotics (1000 units of penicillin/ml and 10 mg/ml

streptomycin, P/S). KG-1 and HL-60 cells were cultured in Iscove’s Modified Dulbecco’s

medium (IMDM) supplemented with 20% FBS and 5% antibiotics. C1498-GFP cells were

maintained as a suspension culture in Dulbecco’s Modified Eagle’s Medium (DMEM)

Page 49: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

35

supplemented with 10% FBS, 5% L-glutamine and 5% antibiotics. All cell lines were cultured

in T-75 sterile cell culture flasks at 37°C in a humidified environment containing 5% CO2 and

95% air. Cells were sub-cultured routinely three times a week (Table 2.2). Mycoplasma

contamination test was carried out at monthly intervals using MycoAlert™ Mycoplasma

Detection Kit.

Table 2.2 Cell lines used in this study. This table shows sources and diseases that cell lines were derived from.

All cell lines were cultured at 37°C, 5% CO2 and split 3 times a week.

Cell line Source Disease Culure media References

KG-1 Human Erythroleukaemia IMDM, 20%FBS, P/S (Koeffler and

Golde 1978)

HEL92.1.7 Human Erythroleukaemia RPMI, 10%FBS, P/S

and L-glutmaine

(Martin and

Papayannopoulou

1982)

HL-60 Human APML IMDM, 20%FBS, P/S (Collins, Ruscetti

et al. 1978)

KU812F Human CML in blast

crisis

RPMI, 10%FBS, P/S

and L-glutmaine

(Kishi 1985)

K562 Human CML in blast

crisis

RPMI, 10%FBS, P/S

and L-glutmaine

(Lozzio and

Lozzio 1975)

C1498-GFP Murine AML DMEM, 10%FBS,

P/S and L-glutmaine

(Zhang, Gajewski

et al. 2009)

Page 50: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

36

2.2.1.2 Cell counting and cell density calculation using a haemocytometer

A 10μl aliquot of cell suspension was mixed with 90μl of trypan blue stain and 10μl of

the mixture was loaded onto the grid of a Neubauer haemocytometer. Only healthy and round

cells that were located in the four corner squares of the Neubauer haemocytometer were counted

and averaged in order to determine the cell density (Figure 2.1).

Figure 2.1 A haemocytometer diagram. It indicates the four corner squares that were used for counting cell

number; only cells that were healthy and round cells that only were counted. The number of cells in one set of 16

squares is equal to the number of cells x104/ml.

2.2.1.3 Cryopreservation of cell stocks

After calculating the cell density, a volume containing 5x106 cells was transferred into a

25ml universal tube and centrifuged at 1500rpm for 3 minutes. Then, supernatant was removed

and cell pellets were re-suspended in 1ml storage medium, which was full growth medium

supplemented with 5% (v/v) dimethyl sulphoxide (DMSO). The cell suspension was then

transferred into 1.8ml cryovials. Cells were stored at -80ºC for overnight before being

transferred to liquid nitrogen for long term storage.

Page 51: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

37

2.2.1.4 Revitalisation of cryopreserved cells

Cryopreserved cells were thawed rapidly by incubating in a 37ºC water bath for 2

minutes. Then, cells were transferred to 10ml of pre-warmed full growth medium and

centrifuged at 1500rpm for 3 minutes. The supernatant was discarded in order to remove DMSO

traces and cell pellets were re-suspended in 10ml full growth medium in a T-25 sterile cell

culture flask and cultured at 37°C in a humidified environment containing 5% CO2. After cell

growth had resumed, cells were transferred to a T-75 flask and used for experiments.

2.2.2 Gene expression analysis by real-time PCR (RT-PCR)

Gene expression analysis using SYBR greenTM was carried out in order to measure the

changes in expression of genes of interest. The RT-PCR method is characterised by its

sensitivity and specificity for the detection of the level of messenger RNA (mRNA) expression

(Marone, Mozzetti et al. 2001). This technique was performed in three stages. Firstly, mRNA

was extracted from cells of interest. Secondly, mRNA was reverse transcribed to

complementary deoxyribonucleic acid (cDNA). Thirdly, the resultant cDNA was employed as

a PCR amplification template using specific primers for the genes of the interest. During PCR

amplification, the products of the genes of the interest were measured in real-time by assessing

the fluorescence produced by SYBR green, when it binds to double-stranded DNA. The

intensity of fluorescence reflects the amount of the target gene in the sample. The relative

expression of the target genes was assessed by comparing their expression to the expression of

the housekeeping gene β-actin.

2.2.2.1 mRNA extraction

This procedure was applied in order to extract and purify mRNA that will be used in

synthesis of cDNA. mRNA was extracted using the RNeasy® Plus Mini Kit according to the

manufacturer's instructions. Cells were suspended in 5% FBS media at a concentration of 5×105

Page 52: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

38

cell/ml, seeded in 6-well plates, and treated in triplicate with the half maximal inhibitory

concentrations (IC50) of HXR9, unless otherwise specified, with the equivalent concentrations

of CXR9, or with media only. After treatment for two hours, cells were transferred into

eppendorfs. Then eppendorfs were spun at 1000rpm for five minutes and supernatants were

removed. After that, cells were lysed using RLT plus lysis buffer and β-mercaptoethanol, which

inhibits RNase. Next, cell lysates were vortexed in order to lyse more cells and yield the

maximum amount of RNA. Then, cell lysates were passed through a genomic DNA eliminator

spin column to remove the genomic DNA. Ethanol was added to cell lysates to allow RNA

binding to the RNeasy® silica gel membrane spin column. Then, cell lysates were washed using

the provided buffer RW1 to remove biomolecules that were non-specifically bound to the

membrane, such as carbohydrates, proteins, fatty acids, the small RNAs, ribosomal RNA and

transferase RNA. After that, the silica membrane was washed twice by RPE buffer to remove

ethanol. Finally, mRNA was eluted by using 35μl RNase-free water. mRNA samples were

stored at -80ºC until needed.

2.2.2.2 Measuring mRNA concentration

mRNA concentration was quantified using the Nanodrop® spectrophotometer ND-

1000, which is based on the Beer Lambert law to measure RNA concentration of 1μl of a

sample in ng/μl by measuring the ultra violet light absorbance at 260nm. RNA sample purity

was assessed using the 260/280 ratio, which should be more than two.

2.2.2.3 Reverse transcription of mRNA into cDNA

mRNA was reverse transcribed to cDNA using the Cloned AMV First-Strand cDNA

Synthesis Kit, which initiates the reverse transcription to cDNA of all mRNA molecules present

in a sample using the random primers method. mRNA samples were thawed on ice and 15μl

containing 100ng RNA was prepared in 1.5ml eppendorfs using DEPC-treated water. An

adequate volume of cDNA synthesis mix was prepared according to the manufacturer's

Page 53: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

39

instructions and 5μl was added to each RNA sample (Table 2.3). The mixture was incubated at

50°C for 30 minutes to synthesise cDNA followed by heating at 85°C for 5 minutes to terminate

the reaction. Assuming cDNA synthesis efficiency was 100%, this produced almost 100ng of

cDNA in 20μl at a concentration of 5ng/μl, which was further diluted to 2ng/μl by adding 30μl

of DEPC-treated H2O and stored at -20°C.

Table 2.3 cDNA synthesis mix. This table shows the components of the cDNA synthesis mix and volumes used

to prepare a 5μl of the solution.

cDNA synthesis mix components Volume per reaction (μl)

5× Buffer 2

10mM dNTP Mix 1

0.1M DTT 0.5

Oligo (dT)20 (50μM) 0.5

RNaseOUTTM (40U/μl) 0.5

Cloned AMV RT (15U/μl) 0.5

Total volume 5

2.2.2.4 Complementary PCR primers design

Sequences of complementary primers for PCR amplification were designed using

(http://simgene.com/Primer3). Tables 2.4 and 2.5 show the complementary primer sequences

for the whole set of HOX genes and pro- and anti-apoptotic genes, respectively, that were

assessed in this study. All used primers were stored at -20ºC and thawed on ice before use.

Page 54: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

40

Table 2.4 HOX gene primers used for PCR amplification.

Gene Forward Sequence 5’-3’ Reverse Sequence 5’-3’ Amplicon Length

(bp)

HOXA1 CTGGCCCTGGCTACGTATAA TCCAACTTTCCCTGTTTTGG 153

HOXA2 TTCAGCAAAATGCCCTCTCT TAGGCCAGCTCCACAGTTCT 176

HOXA3 ACCTGTGATAGTGGGCTTGG ATACAGCCATTCCAGCAACC 227

HOXA4 CCCTGGATGAAGAAGATCCA AATTGGAGGATCGCATCTTG 271

HOXA5 CCGGAGAATGAAGTGGAAAA ACGAGAACAGGGCTTCTTCA 193

HOXA6 AAAGCACTCCATGACGAAGG TCCTTCTCCAGCTCCAGTGT 158

HOXA7 TGGTGTAAATCTGGGGGTGT TCTGATAAAGGGGGCTGTTG 285

HOXA9 AATAACCCAGCAGCCAACTG ATTTTCATCCTGCGGTTCTG 203

HOXA10 ACACTGGAGCTGGAGAAGGA GATCCGGTTTTCTCGATTCA 159

HOXA11 CGCTGCCCCTATACCAAGTA GTCAAGGGCAAAATCTGCAT 279

HOXA13 GGATATCAGCCACGACGAAT ATTATCTGGGCAAAGCAACG 176

HOXB1 TTCAGCAGAACTCCGGCTAT CCTCCGTCTCCTTCTGATTG 157

HOXB2 CTCCCAAAATCGCTCCATTA GAAAGGAGGAGGAGGAGGAA 259

HOXB3 TATGGCCTCAACCACCTTTC AAGCCTGGGTACCACCTTCT 299

HOXB4 TCTTGGAGCTGGAGAAGGAA GTTGGGCAACTTGTGGTCTT 155

HOXB5 AAGGCCTGGTCTGGGAGTAT GCATCCACTCGCTCACTACA 189

HOXB6 ATTTCCTTCTGGCCCTCACT GGAAGGTGGAGTTCACGAAA 184

HOXB7 CAGCCTCAAGTTCGGTTTTC CGGAGAGGTTCTGCTCAAAG 249

HOXB8 GTAGGCTTCAGCTGGGACTG GGGAGCCTTTGCTTAAATCC 265

HOXB9 TAATCAAAGACCCGGCTACG CTACGGTCCCTGGTGAGGTA 198

Page 55: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

41

Gene

Forward Sequence 5’-3’

Reverse Sequence 5’-3’

Amplicon Length

(bp)

HOXB13 CTTGGATGGAGCCAAGGATA CCGCCTCCAAAGTAACCATA 234

HOXC4 CGCTCGAGGACAGCCTATAC GCTCTGGGAGTGGTCTTCAG 276

HOXC5 CAGTTACACGCGCTACCAGA AGAGAGGAAAGGCGAAAAGG 268

HOXC6 AAGAGGAAAAGCGGGAAGAG GGTCCACGTTTGACTCCCTA 190

HOXC8 CTCAGGCTACCAGCAGAACC TTGGCGGAGGATTTACAGTC 150

HOXC9 AGACGCTGGAACTGGAGAAG AGGCTGGGTAGGGTTTAGGA 190

HOXC10 CGCCTGGAGATTAGCAAGAC GGTCCCTTGGAAGGAGAGTC 289

HOXC11 CGGAACAGCTACTCCTCCTG CAGGACGCTGTTCTTGTTGA 186

HOXC12 CAAGCCCTATTCGAAGTTGC GCTTGCTCCCTCAACAGAAG 180

HOXC13 GTGGAAATCCAAGGAGGACA TTGTTGAGGGACCCACTCTC 170

HOXD1 TTCAGCACCAAGCAACTGAC TAGTGGGGGTTGTTCCAGAG 232

HOXD3 CAGCCTCCTGGTCTGAACTC ATCCAGGGGAAGATCTGCTT 176

HOXD4 TCAAATGTGCCATAGCAAGC TCCATAGGGCCCTCCTACTT 173

HOXD8 TCAAATGTTTCCGTGGATGA GCTCTTGGGCTTCCTTTTTC 290

HOXD9 TCCCCCATGTTTCTGAAAAG GGGCTCCTCTAAGCCTCACT 236

HOXD10 GCTCCTTCACCACCAACATT AAATATCCAGGGACGGGAAC 154

HOXD11 GGGGCTACGCTCCCTACTAC GCTGCCTCGTAGAACTGGTC 253

HOXD12 CGCTTCCCCCTATCTCCTAC CTTCGGGCGCATAGAACTTA 201

HOXD13 GGGGATGTGGCTCTAAATCA AACCTGGACCACATCAGGAG 265

Page 56: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

42

Table 2.5 Pro- and anti-apoptotic and β-actin gene primers used for PCR amplification.

Gene Forward Sequence 5’-3’

Reverse Sequence 5’-3’

Amplicon Length

(bp)

Bad CGGAGGATGAGTGACGAGTT GATGTGGAGCGAAGGTCACT 180

Bak1 TTTTCCGCAGCTACGTTTTT GGTGGCAATCTTGGTGAAGT 248

Bax TTTGCTTCAGGGTTTCATCC CAGTTGAAGTTGCCGTCAGE 246

Bcl2 GAGGATTGTGGCCTTCTTTG ACAGTTCCACAAAGGCATCC 170

Bid CTGCAGGCCTACCCTAGAGA ACTGTCCGTTCAGTCCATCC 195

XIAP GGGGTTCAGTTTCAAGGACA CGCCTTAGCTGCTCTTCAGT 182

Apaf1 TTCTGATGCTTCGCAAACAC CTGGCAAATCTGCCTTCTTC 237

Caspase-3 TTTTTCAGAGGGGATCGTTG CGGCCTCCACTGGTATTTTA 151

Caspase-6 ATCCTCACCGGGAAACTGTG AATTGCACTTGGGTCTTTGC 161

Caspase-7 AGTGACAGGTATGGGCGTTC CGGCATTTGTATGGTCCTCT 164

Caspase-9 CTAGTTTGCCCACACCCAGT GCATTAGCGACCCTAAGCAG 172

PARP1 GCTCCTGAACAATGCAGACA CATTGTGTGTGGTTGCATGA 233

c-FOS CCAACCTGCTGAAGGAGAAG GCTGCTGATGCTCTTGACAG 232

p21 GACACCACTGGAGGGTGACT

CAGGTCCACATGGTCTTCCT 171

p53 GTGGAAGGAAATTTGCGTG CCAGTGTGATGATGGTGAGG

183

β-actin ATGTACCCTGGCATTGCCGAC GACTCGTCATACTCCTGCTTG 227

2.2.2.5 RT-PCR

Each gene of interest was tested in triplicate. The housekeeping gene β-actin was used

as an endogenous control. An adequate volume of PCR mastermix was prepared in an

eppendorf as detailed in table 2.6. This mastermix contains cDNA, SYBR® Green JumpStartTM

Taq ReadyMixTM, ROX reference dye and DEPC-treated H2O. 20μl of the PCR mastermix was

added into three wells of a Mx 3000P® 96-well plate for each gene. 5μl of 1μM of each primer

pair was added into the designated wells. Plates were then sealed with optical caps and

centrifuged at 1500rpm for 1 minute to bring all reagents to the well bottoms and remove air

Page 57: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

43

bubbles. The Plates were then run on a Stratagene Mx3005P thermal cycler. The thermal

profiles employed were: heating once for 10 minutes at 95°C, followed by 30 seconds at 95°C,

1 minute at 60°C, 30 seconds at 72°C for 40 cycles. After completing the thermal profiles, the

detected fluorescence of each reaction was converted to cycle threshold using MxPro software.

The mean of the cycle threshold values of the triplicate repeats for each gene was calculated

and used for measuring gene expression, as detailed below.

Table 2.6 RT-PCR reaction components. This table shows the components of single RT-PCR reaction. All

components were stored at -20°C and thawed on ice before use.

PCR reaction components Volume per reaction (μl)

cDNA (2ng/μl) 1

DEPC-treated H2O 6.25

SYBR® Green JumpStartTM Taq ReadyMixTM 12.5

ROX reference dye 0.25

Primers-forward and reverse mixture 5

Total volume 25

2.2.2.6 RT-PCR data analysis

A previously described relative quantification method was used to analyse RT-PCR data

(Livak and Schmittgen 2001). Two steps were performed in this analysis. Firstly, the expression

of genes of interest was normalised to the expression of the housekeeping gene β-actin.

Secondly, the normalised data of genes of interest of HXR9 or CXR9 treated samples were

compared to that of untreated control samples. Microsoft Excel was used to perform all

calculations. Experiments were repeated three times. Results were expressed as the mean of the

relative gene expression of the three experiments with error bars to show the standard error of

mean (SEM).

Page 58: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

44

2.2.3 Lactate dehydrogenase (LDH) assay

LDH is present in the cytoplasm of all cells. It is released into culture medium if the cell

membrane is permeablized. Therefore, the amount of this enzyme in the culture medium

indicates to the integrity of cell membrane and hence provides an estimation of cytotoxicity

(Haslam, Wyatt et al. 2000). LDH activity is assessed in two steps. Firstly, LDH oxidises lactate

to pyruvate by reducing NAD+ to NADH/H+. Secondly, diaphorase, a catalyst, transfers 2H

from NADH/H+ to a tetrazolium salt (INT) which results in the formation of the red salt

formazan. Hence, the amount of formazan formed directly correlates to the amount of LDH in

the culture medium and thereby to the number of damaged cells (Figure 2.2).

Figure 2.2 LDH cytotoxicity assay. LDH catalyses the reduction of NAD⁺ to NADH⁺/H⁺ through the conversion

of lactate to pyruvate. Then, diaphorase reduces the yellow tetrazolium salt INT to the red formazan salt in the

presence of NADH⁺/H⁺.

The LDH assay was used to measure the cytotoxicity of HXR9, daunorubicin (DNR)

and mitoxantrone (MTX). In HXR9 cytotoxicity determination, cells were treated with HXR9

or its negative control CXR9 for 2 hours. In DNR or MTX cytotoxicity determination, cells

were treated with either drug for 24 hours as per the following:

Cells were suspended in 5% FBS of the appropriate culture medium at a concentration of 5×105

cell/ml and seeded in 96-well flat-bottom plates at a concentration of 5×104 cell/well. Then cells

were treated with 100μl of a serial dilution of the tested drug, which have been already diluted

Page 59: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

45

in 5% FBS of the appropriate culture medium. Each plate included untreated cells (negative

control), 2% triton-treated cells (positive control), and 5% FBS medium only (background

control). Each condition was repeated six times. The plates were incubated in a humidified

incubator at 37ºC, 5% CO2 for either 2 or 24 hours according to the tested drug. After treatment,

the plates were spun at 1000rpm for 5 minutes and 100μl of supernatant was decanted from

each well into a fresh, 96-well flat-bottom plate. An equal amount of LDH reagent was prepared

according the manufacturer's instructions and added to the supernatants for two minutes, and

then LDH enzymatic activity was estimated by reading the absorbance at 492nm, using a plate

reader.

2.2.3.1 LDH assay data analysis

In order to assess the percentage of surviving cells, the cytotoxicity percentage was

calculated first. The average absorbance values of the six repeats of each condition were

calculated. Then, the average absorbance value of the background control was subtracted from

each of the other conditions average absorbance values. After that, the cytotoxicity percentage

was calculated according to the manufacturer's instructions as per the following:

Cytotoxicity% = treated value−negative control

positive control−negative control × 100

Then, the proportion of surviving cells was determined as the following:

%surviving cells = 100 - cytotoxicity

The %surviving cells was plotted on X-Y scatter diagrams using GraphPad Prism

software (California, USA) in order to create cytotoxicity curves for each cell line and drug.

Therefore, the surviving fractions of treated cells can be compared to those of untreated cells.

Additionally, the cytotoxicity results of the different cell lines can be compared.

Page 60: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

46

Each experiment was repeated at least three times (biological repeats) and each valid

experimental curve should have a linear correlation coefficient (R2) ≥ 0.95. The cytotoxicity

curves were generated using the mean %surviving cells at different drug concentrations.

Additionally, the SEM of each single dose was determined and included as an error bar on the

cytotoxicity curves.

IC50 was then calculated by CalcuSyn software (Biosoft, Cambridge, UK) as described

in statistical analysis.

2.2.4 Assessment of drug combination interaction by LDH

This assay was performed in vitro to evaluate the interaction levels (synergism, additive,

or antagonism) between HXR9 and the chemotherapeutic drugs DNR or MTX.

Cells were suspended in 5% FBS medium and seeded in 96-well flat-bottom plates at a

concentration of 5×104 cell/well. Then, cells were treated with 100μl of five increasing

concentrations (IC12.5, IC25, IC50, IC75, IC100) of each drug alone or in combination with HXR9.

For single treatments, cells were treated with HXR9 or with a chemotherapeutic drug, DNR or

MTX, for 2 or 24 hours, respectively. For combination treatment, cells were treated with either

DNR or MTX for 24 hours and HXR9 was added for the last 2 hours of treatment. Each

treatment was repeated in triplicate. Each plate included negative, positive and background

controls. After treatment, plates were spun for 5 minutes at 1000rpm, 100μl of supernatant was

decanted into new 96-well flat-bottom plates and 100μl LDH reagent was added. The

cytotoxicity was then determined as described for the LDH assay data analysis section and the

evaluation of the drug interaction between HXR9 and the chemotherapeutic drugs was

performed as described for the analysis of drug combination assay data section. The

combination assay data were plotted on column graphs using GraphPad Prism software.

Page 61: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

47

2.2.5 Annexin V- PE assay

The annexin V-PE assay was used to quantify the percentage of the apoptotic cells and

evaluate the mechanism of cell death. This assay is based on the cell membrane permeability

of the apoptotic cells. During early stages of apoptosis, the membrane bound phospholipid

phosphatidlyserine (PS) translocates from the inner surface of the cellular plasma membrane to

the external surface, where it can be detected by annexin V protein that has a high phospholipid-

binding affinity. Annexin V is conjugated to PE (a fluorochrome) and this compound is used as

a probe for cell analysis by flow cytometry (Vermes, Haanen et al. 1995). Therefore,

quantification of exposed PS using PE labelled annexin V by flow cytometry is considered a

sensitive measurement of apoptosis (Koopman, Reutelingsperger et al. 1994). In addition, 7-

amino actinomycin D (7-AAD), which is a membrane impermeable fluorescent DNA binding

stain that selectively interacts with guanine and cytosine bases in double-stranded DNA, was

used to distinguish between viable and non-viable cells (Philpott, Turner et al. 1996). Viable

cells stain negative with both reagents. Early apoptotic cells stain positive for annexin V-PE

only. Late apoptotic, necrotic, or dead cells stain positive for both stains, as shown in table 2.7.

The late apoptotic/ necrotic cells will be reflected to in this study as dead cells, because this

assay cannot definitely differentiate between these two conditions (Figure 2.3).

Cells were suspended in 5% FBS of the appropriate culture medium at a concentration

of 1×106 cell/ml and seeded in 96-well flat-bottom plates at a concentration of 1×105 cell/well.

Then cells were treated either with 100μl of a serial HXR9 dilution or with a high CXR9

concentration in triplicate. Each plate included untreated cells (negative control). The plates

were incubated in a humidified incubator at 37ºC, 5% CO2 for two hours. After two-hour

treatment, the plates were spun at 1000rpm for five minutes and supernatants were removed.

Cell pellets were washed twice with cold phosphate buffered saline (PBS) and then cells re-

suspended in 20μL of 1X binding buffer (0.1M Heps/NaOH (pH 7.4), 1.4M NaCl, 25mM

Page 62: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

48

CaCl2). Cell suspensions were transferred to culture tubes. 5μL of both annexin V-PE and 7-

AAD were added and then cell suspensions were incubated at the room temperature (RT) for

15 minutes in the dark. Then, 200μL of 1X binding buffer were added to tubes and analyzed by

a flow cytometry.

The percentage of the different cell populations, viable, early apoptotic and dead cells,

were plotted by GraphPad Prism software (California, USA) on column graphs to allow the

comparison of treated and untreated negative control cells. Each experiment was repeated at

least three times independently.

Table 2.7 Different cell populations in the annexin V-PE assay.

Annexin V 7-AAD Cell Stage

Negative Negative Viable

Positive Negative Early apoptosis

Positive Positive Late apoptosis or necrosis (dead

cells)

Figure 2.3 A flow cytometry plot shows different cell populations in the annexin V-PE assay. In the left bottom

corner are the viable cells that are negative for annexin V and 7-AAD. In the right bottom corner are the early

apoptotic cells that are stained with annexin V only. In the top part are dead cells that are stained with both annexin

V and 7-AAD.

Page 63: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

49

2.2.6 Caspase-3 activity assay

Activation of caspases is a key event in apoptosis. In particular, caspases-3/7 are

considered as executioner caspases that recognise and cleave numerous proteins containing the

DEVD sequence such as poly ADP ribose polymerase (PARP), resulting in cell apoptosis. The

caspase-3 assay is based on the ability of active caspase-3 to cleave a synthetic DEVD sequence.

DEVD is covalently linked to R110 in a compound called Z-DEVD-R110, which suppresses

the fluorescence R110. The caspase cleavage of DEVD results in R110 fluorescence, which is

measured at excitation wavelength 492nm and emission wavelength 520nm. Ac-DEVD-CHO

is a caspase-3/7 inhibitor that can be used to verify the resultant fluorescence is due to caspase-

3 activity. A reference standard can be used to quantify the R110 produced in the reaction.

Cells were suspended in the appropriate culture medium plus 5% FBS at a concentration

of 2×106 cell/ml and seeded in eppendorfs at a concentration of 1×106 cell/reaction. Cells were

treated, in triplicate, with 500μl of different HXR9 concentrations or with 500μl of an

equivalent CXR9 concentration to the highest HXR9 concentration. Each experiment included

untreated cells (negative control). The plates were then incubated in a humidified incubator at

37ºC, 5% CO2 for two hours. After the treatment for two hours, eppendorfs were spun at

1000rpm for 5 minutes and supernatants were removed. Cell pellets were then washed with

PBS and re-suspended in 100μl of 1X cell lysis buffer and incubated on ice for 30 minutes. The

lysed cells were then spun at 5000rpm for 5 minutes and 50μl of the supernatant was transferred

to a new 96-well flat bottom black plate. 50μl of the lysis buffer was added into separate wells

as non-enzyme controls in order to measure the substrate background fluorescence. As an

additional control, 1μl of 1mM Ac-DEVD-CHO inhibitor was added to selected samples that

were incubated at RT for 10 minutes in the dark. During this time, the remaining samples

(without inhibitors) were kept on ice. 50μl of 500μM Z-DEVD-R110 substrate was then added

Page 64: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

50

to all samples. The supernatants were incubated with the substrate for 30 minutes in the dark.

Serial concentrations of the reference standard were added to the plates in order to quantify

R110 fluorescence produced in the reaction. Finally the caspase-3 activity was measured by

reading the plates at 492nm by the plate reader.

2.2.6.1 Caspase-3 activity data analysis

For each individual experiment, the average fluorescent value for the different

conditions was calculated, and the lysis buffer reading (background) was subtracted from the

other average values. Then, the resultant values were plotted on column graphs using GraphPad

Prism software to compare caspase-3 activity and calculate the statistical significance between

untreated control cells and cells treated either with HXR9 or CXR9.

In order to validate each experiment, the reference standard data were plotted separately

on X-Y scatter graphs and each valid reference standard curve should have R2 ≥ 0.95.

Each experiment was performed at least in triplicate. The caspase-3 activity data were

generated by the mean of the fluorescent values of the different cell conditions. The SEM of

each single dose was determined and included on the graphs.

2.2.7 Cell cytospins and 4',6-diamidino-2-phenylindole (DAPI) staining

Cytospin is a method used to concentrate cells. After cell cytospinning, cell morphology

can be evaluated by staining. DAPI is a cell-permeable blue fluorescent stain that binds

specifically to the minor groove adenine-thymine regions of double-stranded DNA. The

binding of DAPI with DNA intensifies the fluorescence about 20 fold and this can be imaged

using a fluorescence microscope or flow cytometry. DAPI stain is used to assess apoptosis by

visualising the nuclear morphology changes such as chromosome condensation and

fragmentation (Kapuscinski 1995).

Page 65: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

51

Cells were suspended in 5% FBS culture medium at a concentration of 1×106 cell/ml

and seeded in 24-well plates at a concentration of 5×105 cell/well. Then cells were treated either

with 500μl of HXR9 the IC50 or double the IC50, in duplicate. Each plate included untreated

negative control cells. The plates were incubated in a humidified incubator at 37ºC, 5% CO2

for two hours, after which cells were transferred into eppendorfs. Harvested cells were washed

twice with PBS, and fixed in 4% formaldehyde/PBS solution.

Cytospinning was then performed. Microscope-slides, filter cards and cytofunnels were

assembled using cytospin holders and then the entire apparatus was placed in a cyto-centrifuge.

Then, 300μl of 4% formaldehyde/PBS fixed cells was added to the cytofunnels and spun at

500rpm for 5 minutes. Next, the apparatus was dis-assembled. Slides were stained with 1 drop

of 1:1000 DAPI/PBS for 5 minutes in the dark. After staining, excess DAPI/PBS was removed

and 1 drop of the mounting media 40% glycerol/PBS added. Then, slides were covered with

slip covers and sealed using nail vanish. Finally, images were captured with 20x lenses by a

fluorescent microscope.

2.2.8 Inhibition of caspases activity by z-VAD-FMK

Z-VAD-FMK is a cell permeable general caspase inhibitor. It can prevent the induction

of apoptosis through irreversible binding to caspase protease catalytic sites (Marcelli,

Cunningham et al. 1999). Pre- and co-treatment of cells with z-VAD-FMK assay was used to

investigate the role of caspases in cell death. Z-VAD-FMK was diluted in DMSO at 43mM and

stored at -20ºC.

Some wells were pre-treated with 50μM z-VAD-FMK. Cells were suspended in 5%

FBS culture media at a concentration of 1×106 cell/ml and seeded in 96-well flat-bottom plates

at a concentration of 1×105 cell/well. The plates were then pre-incubated in a humidified

incubator at 37ºC, 5% CO₂ for one hour. Meanwhile, HXR9 and CXR9 were prepared in 5%

Page 66: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

52

FBS media either with or without 50μM z-VAD-FMK. After one hour pre-incubation, cells

were treated in triplicate with 100μl of HXR9 IC50, double the IC50, or with a CXR9

concentrations equivalent to double the IC50. Additionally, each plate included untreated

negative control cells and DNR treated cells as positive control cells. The plates were then

incubated in a humidified incubator at 37ºC, 5% CO2 for two hours. After treatment for two

hours, the plates were spun at 1000rpm for five minutes and supernatants were discarded. Cell

pellets were washed twice with cold PBS and then stained with annexinV-PE as described for

the annexinV-PE assay section. Data were plotted as described for the annexinV-PE assay.

Statistical significance was assessed by comparing the data of untreated cells to those of pre-

and co-treated with 50μM z-VAD-FMK.

2.2.9 Cyclosporin A (CsA) protection assay

CsA attenuates mitochondrial permeability transition pore (mPTP) formation by

binding to cyclophilin D (cypD), which is required for mPTP formation in necrotic

mitochondrial cell death (Li, Johnson et al. 2004; Basso, Fante et al. 2005; Dube, Selwood et

al. 2012). CsA was diluted in DMSO to a stock concentration of 20mM, aliquoted and stored

at -20ºC.

In this assay cells were pre- and co-treated with or without 5μM CsA and hydrogen

peroxide (H2O2) treatment was used as a positive control. Cells were suspended in 5% FBS

culture medium at a concentration of 1×106 cell/ml and seeded in 96-well flat-bottom plates at

a concentration of 1×105 cell/well. The plates were then pre-incubated in a humidified incubator

at 37ºC, 5% CO2 for one hour. Meanwhile, HXR9 and H2O2 were prepared in 5% FBS medium

either with or without 5μM CsA. After one hour pre-incubation, cells were treated in triplicate

with 100μl of HXR9 at the IC50, double or triple the IC50 or with 20mM H2O2. Additionally,

each plate included untreated negative control cells. The plates were then incubated in a

Page 67: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

53

humidified incubator at 37ºC, 5% CO2 for two hours. After treatment for two hours, the plates

were spun at 1000rpm for 5 minutes and supernatants were discarded. Cell pellets were washed

twice with cold PBS and then stained with annexinV-PE as described for the annexinV-PE

assay. Data analysis was performed by flow cytometry.

The percentage of viable cells was plotted using GraphPad Prism software on column

graphs. Each experiment was repeated at least three times independently.

2.2.10 Necrostatin-1 (Nec-1) protection assay

Nec-1 is a selective inhibitor of receptor interacting protein 1 (RIP1) (Degterev, Hitomi

et al. 2008). RIP1 inhibition prevents a form of programmed necrosis called necroptosis

(Degterev, Huang et al. 2005). Nec-1 was diluted in DMSO to a stock concentration 40mM and

stored at -20ºC. In this experiment, some cells were pre- and co-treated with 50μM Nec-1. The

experiment and data analysis was performed as described for the CsA protection assay.

2.2.11 Fructose protection assay

Fructose is a glycolytic substrate that at high concentrations significantly sequesters

phosphate and accordingly intracellular adenosine triphosphate (ATP) is depleted (Figure 2.4).

This selective depletion of ATP protects cells from apoptosis, but leaves enough levels (15%)

of ATP to rescue cells from spontaneous necrosis (Nieminen, Saylor et al. 1994).

Figure 2.4 ATP depletion by fructose.

Page 68: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

54

Cells were pre- and co-treated with a high concentration of fructose (40mM). Fructose

was freshly dissolved in 5% FBS culture medium. Cells were suspended in 5% FBS culture

medium supplemented with or without 40mM fructose and seeded in 96-well flat-bottom plates

at a concentration of 5×104 cell/well. The plates were then incubated at 37ºC, 5% CO2 for one

hour. Serial HXR9 concentrations were prepared in 5% FBS media either with or without

40mM fructose. After an hour pre-incubation, cells were treated in triplicate with 100μl of the

different HXR9 concentrations or 2% triton as a positive control. Additionally, each plate

contained cells incubated with 5% FBS medium as a low control and background wells

contained 5% FBS medium only. After treatment for two hours, the plates were spun at

1000rpm for 5 minutes and 100μl of each supernatant was decanted into fresh, 96-well flat-

bottom plates. Finally, LDH reagents were added as described in the LDH assay section.

Similarly, data analysis was performed as for the LDH assay. The statistical significance of the

effect of fructose on the cytotoxicity of HXR9 was performed by comparing the survival

fractions of fructose treated and untreated cells.

2.2.12 The effect of ethilenediaminetetra-acetic acid (EDTA) on HXR9 cytotoxicity

EDTA is a calcium ions (Ca2+) chelator that was used to investigate the role of

extracellular Ca2+ in the cytotoxicity of HXR9. The role of Ca2+ was assessed by two different

assays, LDH and annexin V, as described for the fructose protection and CsA protection assays,

respectively. In both assays, cells were pre- and co-treated with or without 20mM EDTA

dissolved in 5% FBS medium. The statistical significance of the effect of EDTA on the

cytotoxicity of HXR9 assessed by comparing the survival fraction of EDTA treated cells to

untreated cells.

Page 69: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

55

2.2.13 The effect of HXR9 on mitogen activated protein kinase (MAPK) pathways

MAPKs are a family of protein kinases regulating many essential cellular pathways

including cell survival, proliferation and apoptosis. The most extensive studied MAPK

pathways are extracellular signal-regulated kinase (ERK), Jun N-terminal kinase (JNK) and

p38 (Dhillon, Hagan et al. 2007). The effect of HXR9 on those pathways was studied by

blocking each pathway using a specific inhibitor. The inhibitors used were U0126

monoethanolate, SP600125 and SB203580 for ERK, JNK and p38, respectively. The inhibitor-

stocks were dissolved in DMSO at concentrations 1mM, 25mM and 5mM of U0126

monoethanolate, SP600125 and SB203580, respectively. U0126 monoethanolate and

SP600125 were stored at 4ºC while SB203580 stored at -20ºC until needed.

In all of the assays that investigate the effect of HXR9 on ERK, JNK and p38 pathways,

cells were pre- and co-treated with or without the inhibitors. The working concentrations used

were 20μM, 60μM and 25μM for U0126 monoethanolate, SP600125 and SB203580,

respectively. Experiments were performed as described for the fructose protection assay;

however, data were plotted in column diagrams. The statistical significance of the effect of

blocking MAPK pathways on the cytotoxicity of HXR9 was determined by comparing the

survival fraction of the different-inhibitors-treated and untreated cells.

2.2.14 The effect of inhibition of NADPH oxidase (NOX) on HXR9 efficacy

NOX is a generator of intracellular reactive oxygen species (ROS) (Lambeth 2004). The

effects of NOX on the efficiency of HXR9 were assessed using an inhibitor called

diphenyleneiodonium chloride (DPI). DPI is widely used to block the effect of NOX in spite of

its non-specific binding (Park, Song et al. 2007). DPI stock was dissolved in DMSO at 15mM

and stored at -20ºC until used.

Page 70: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

56

Cells were pre- and co-treated with or without 60μM DPI. The assessment of DPI effect

on the cytotxicity of HXR9 was performed as described for the fructose protection assay and

data were plotted in column diagrams. The statistical significance was assessed by comparing

the survival fraction of DPI treated and untreated cells.

2.2.15 Role of μ-calpain in HXR9 cell killing

Calpains are Ca2+-dependent cysteine proteases that commonly activated in Ca2+

involved cell death mechanisms (Liu, Van Vleet et al. 2004). Among them, μ-calpain is one of

the most extensively studied in apoptosis/necrosis processes. Calpain inhibitor I (N-Acetyl-

Leu-Leu-Norleu-al) was used to inhibit the Ca2+/μ-calpain pathway. Calpain inhibitor I was

dissolved in DMSO at a concentration 20mM and stored at -20ºC until used.

Cells were pre- and co-treated with or without 60μM calpain inhibitor I. The assessment

of the role of μ-calpain in the cytotxicity of HXR9 was performed as described for the fructose

protection assay and data were plotted in column diagrams. The statistical significance was

assessed by comparing the survival fractions of calpain inhibitor I treated cells to untreated

cells.

2.2.16 The effect of calmodulin (CaM) inhibition on HXR9 cytotoxicity

CaM is one of the main intracellular regulators of Ca2+ (Yuan, Jing et al. 2011). W-7

(N-(6-Aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride) was used to inhibit

CaM and assess its role in HXR9 cytotoxicity. W-7 was dissolved in DMSO at a concentration

50mM and stored at -20ºC.

Page 71: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

57

Cells were pre- and co-treated with or without 50μM W-7. The assessment of the effect

of the inhibition of CaM on the cytotoxicity of HXR9 was performed as described for the

fructose protection assay and data were plotted in column diagrams. The statistical significance

was determined by comparing the survival fractions of W-7 treated cells to untreated cells.

2.2.17 The effect of protein kinase C (PKC) inhibition on HXR9 cytotoxicity

PKC is one of the kinases that are affected by intracellular Ca2+ concentrations. Ro31-

8220 (methanesulfonate salt) was used to inhibit and assess the role of PKC in HXR9

cytotoxicity. Ro31-8220 was dissolved in H2O at a stock concentration 20mM and stored at

4ºC. Cells were pre- and co-treated with or without 30μM Ro31-8220. The assessment of the

effect of the inhibition of PKC on the cytotxicity of HXR9 was done as performed for the

fructose protection assay and data were plotted in column diagrams. The statistical significance

was assessed by comparing the survival fractions of Ro31-8220 treated cells to untreated cells.

2.2.18 The effect of the heme oxygenase-1 (HO-1) inhibition on HXR9 cytotoxicity

HO-1 is an anti-apoptotic protein (Rushworth and MacEwan 2008). Protoporphyrin IX

(PPIX) was used to inhibit HO-1 protein and assess its role in the cytotoxicity of HXR9. PPIX

was dissolved in DMSO at a stock concentration 18mM and stored at -20ºC. Cells were pre-

and co-treated with or without 50μM of PPIX. The effect of HO-1 inhibition on HXR9

cytotoxicity was determined as described for the fructose protection assay and data were plotted

in column diagrams. The statistical significance was performed by comparing the survival

fraction of PPIX treated cells and untreated cells.

Page 72: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

58

2.2.19 The effect of the p53 inhibition on HXR9 cytotoxicity

Pifithrin-α (PFT-α) is an inhibitor of p53-dependent apoptosis and was used to block

and assess the role of p53 in the cytotoxicity of HXR9. PFT-α was dissolved in DMSO at a

stock concentration 20mM and stored at -20ºC. Cells were pre- and co-treated with or without

40μM of PFT-α. The role of p53 in HXR9 cytotoxicity was assessed as described for the

fructose protection assay and data were plotted in column diagrams. The statistical significance

was calculated by comparing the survival fraction of PFT-α treated cells and untreated cells.

2.2.20 Western blotting (WB) for protein expression

WB was conducted to assess the difference in protein expression between untreated

cells and HXR9 treated cells. This assay requires treatment of cell lines with HXR9, extraction

of cytosolic proteins, proteins separation based on their sizes and blotting on a membrane.

Finally, the proteins of interest are detected by labelled antibodies. In order to compare the

expression of protein in different samples, the same amount of protein was loaded in each well

and the same dilution of antibodies was used.

Cells were suspended in 5% FBS medium at a concentration of 1.5×106 cell/ml and

seeded in 24-well plates. Then cells were treated with 1ml of 15μM or 30μM HXR9. Each plate

included untreated cells as a negative control. Each condition was repeated three times. The

plates were incubated in a humidified incubator at 37ºC, 5% CO₂ for two hours. After treatment

for two hours, the three repeats of each condition were gathered in one 25ml universal tube and

centrifuged for 5 minutes at 1500rpm at 4ºC. Supernatants were decanted and cell pellets were

washed twice with 5ml ice cold PBS. Cell pellets were collected in eppendorfs and lysed as

described for the preparation of cell lysate.

Page 73: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

59

2.2.20.1 Preparation of cell lysate

Cell pellets containing 4.5×106 cells were suspended in 300μl RIPA solution with 10μl

of inhibitor cocktail (protease and phosphatase inhibitor) and 10μl of 1mM EDTA. Cell

suspensions were incubated on ice for 15 minutes and agitated at least twice. Cell membranes

were sheared by passing cells through 21 gauge needles attached to 1ml syringes for at least 4-

5 times. Cell suspensions were centrifuged at 13000rpm for 5 minutes at 4ºC and supernatants

were transferred to new eppendorfs. The protein concentration in cell lystaes was determined

using a bicinchonic acid (BCA) assay.

2.2.20.2 Measuring protein concentrations by pierce Bradford city assay (BCA)

This assay was used to measure protein concentrations in cell lysates. It is based on

reduction of copper Cu2+ to cuprous ion Cu+ by proteins. Cu+ then chelates with two molecules

of BCA to form a purple-coloured product that can be detected by a 96-well plate reader at

562nm. The intensity of the formed colour increases linearly with increasing protein

concentrations allowing protein concentrations to be calculated using a standard curve made of

bovine serum albumin (BSA). This assay was performed as follows:

25μl of samples were added in duplicate in 96-well flat-bottom plates. Additionally, each plate

contained a BSA series dilution added in duplicate. To each sample and standard well, 200μl

of BCA working reagent, a combination of reagents A and B in a ratio 50:1, was added. The

plates were gently agitated on a plate shaker and incubated for 30 minutes at 37ºC. After the

incubation, the plates were left at RT for 5 minutes in the dark before reading the absorbance

on a plate reader. The absorbance means for the duplicate repeats of each sample and standard

were calculated. The standard curve was then generated by the absorbance means of BSA series

dilutions and their corresponding concentrations using Microsoft Excel. Finally, the protein

concentrations of the samples were calculated using the standard curve linear regression

equation.

Page 74: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

60

2.2.20.3 Sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) for

protein separation

Cellular proteins were separated based on size using SDS-PAGE. First, 20μl samples

were prepared (13μl cell lysate, 5μl sample buffer and 2μl reducing agent) and heated at 70ºC

for 10 minutes. Second, a Novex® NuPAGE 4-12% Bis-Tris Gel, which is used to separate

proteins, was placed in an XCell SureLockTM Mini-Cell electrophoresis system that contained

800ml NuPAGE® MES SDS running buffer, which prepared by adding 20 parts of water to the

buffer. Third, 10μl of Novex® sharp pre-stained marker was loaded in the first well of every

gel. Forth, 20μl of samples were loaded in the remaining wells. Finally, protein separation was

performed by running gels at 200 volt for one hour.

2.2.20.4 Transferring proteins to polyvinylidene fluoride (PVDF) membranes

After electrophoretic separation, proteins were transferred to PVDF membranes. Firstly,

PVDF membranes were soaked in methanol for 2 minutes while filter papers were soaked in

water. Membranes were then placed in Invitrogen Electrophoresis System and covered with the

gel and then the filter papers and on top of that transfer sponges were placed. Finally, proteins

were transferred by running at 30V for 7 minutes.

2.2.20.5 Detection of proteins by antibodies

After transferring proteins, PVDF membranes were blocked in 5% blocking buffer (5%

(w/v) dried non-fat milk powder, 0.1% (v/v) Tween-20) with gentle rocking for 2 hours at RT.

Then, the membranes were incubated with primary antibodies with gentle rocking overnight at

4ºC. After overnight incubation, the membranes were washed 4 times with 0.1% Tween PBS

solution for 10 minutes at RT with gentle rocking to remove unbound antibodies. Then, the

membranes were incubated with secondary antibodies for 2 hours at RT with gentle rocking;

the antibodies used in WB are listed in table 2.8. After 2 hours incubation, membranes were

washed 3 times, as above. Then, membranes were exposed to superSignal® West Pico

Page 75: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

61

Chemiluminescent substrate according to the manufacturer's recommendation. Next,

membranes were exposed to carestream® Kodak BioMax® light film for 3 minutes to visualise

the chemiluminescence. After that, films were developed and then fixed using developer and

fixer, respectively.

Table 2.8 Antibodies used in WB and working dilutions.

Antibody Type Isotype Molecular

weight (KDa)

Working

concentration

Supplier

Primary anti-

PARP1

Monoclonal Mouse IgM 116 1/4000 Abcam

Primary anti-β-

actin

Polyclonal Rabbit IgG 42 1/10000

Secondary for

anti-PARP

Polyclonal Goat IgG 1/4000

Secondary for

anti-β-actin

Polyclonal Goat IgG 1/2000

Page 76: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

62

2.2.21 In vivo assays

2.2.21.1 Cell preparations

Cells used for in vivo experiments were C1498-GFP and K562 cell lines. Cells were

spun for 3 minutes and re-suspended and washed twice in HANKS medium. C1498-GFP and

K562 cells were then suspended in serum-free HANKS medium at density 1x107cell/ml and

5x107 cell/ml, respectively.

2.2.21.2 Systemic injection of C1498-GFP in C57BL/6 and nude mice

C57BL/6 and nude mice were warmed by a heat lamp in order to dilate the vain. C1498-

GFP cells were then injected into the tail vain (1x106 cell/mouse) using 23 gauge needles. Mice

were then culled randomly during the experiment duration using CO2 to harvest peripheral

blood (PB) and organs including BM, liver, spleen and kidney to assess the engraftment of

AML.

2.2.21.2.1 Harvesting and processing PB

PB was harvested through cardiac puncture using heparinised needles. Blood were then

suspended in 10ml PBS and washed twice. Red blood cells were lysed by harvesting cells in

RBC lysis buffer for 10 minutes. Cells were then spun and supernatant was removed. Cells

were then re-suspended in 20μL of 1X binding buffer. AML engraftment in PB was assessed

by the presence of green fluorescent protein (GFP+) cells using flow cytometry.

2.2.21.2.2 Harvesting and processing BM and other organs

BM was harvested from femurs by flushing with HANKS medium; cells were spun and

re-suspended in HANKS medium. Cells were obtained from spleen, liver and kidney by

mincing and filtration through a cell strainer (70μM). AML engraftment was then assessed as

described for PB.

Page 77: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

63

2.2.21.3 Subcutaneous (S.C.) injection of AML cells into C57BL/6 and SCID mice

C57BL/6 and SCID mice were anesthetised using isoflurane. C1498-GFP (1x106

cell/mouse) and K562 (5x106 cell/mouse) cells were then S.C. injected in the flank of C57BL/6

and SCID mice, respectively. Flank tumour sizes were measured three times a week using

calliper measurement. Tumour sizes were calculated using the formula: (length x width x

width)/2. When flank tumour became palpable, the mice were divided into two groups with one

introduced intratumourly (I.T.) with 50μl of the indicated doses of HXR9 or HXR9 + (500μM

W-7 + 300μM Ro31-8220) in the result section and the other group (control group) were

introduced with I.T. with 50μl of PBS. Mice were sacrificed when tumour size ≥ 1500mm3 or

when mice became sick.

2.3 Statistical analysis

Calcusyn software was used to determine the IC50 of the different drugs for the different

cell lines and for assessing whether the interaction between HXR9 with DNR or MTX was

synergistic, additive or antagonistic, as described below.

Student's t-test was applied for data analysis of all assays using GraphPad Prism

software. Results are expressed as the mean of three separate experiments with error bars to

show the SEM. Statistical significance was determined by comparing the different conditions

in each assay with each other and considered significant when p < 0.05 (*), highly significant

when p < 0.01 (*), or very highly significant when p < 0.001 (***) or p < 0.0001 (****).

Page 78: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

64

2.3.1 Calculation of IC50

IC50 of HXR9, DNR and MTX for different cell lines were calculated by the median-

effect equation of Chou (Chou 1974) using the Clcusyn software. The equation is:

fa/fu = (D/Dm)m

Where D is the dose of the drug; Dm is the dose need to produce a 50% effect (IC50); fa is the

fraction affected by the dose; fu is the fraction unaffected by the dose, fu = 1-fa; m is the exponent

signifying the shape (sigmoidicity) of the dose effect curve.

2.3.2 The analysis of drug combination assay data

The combination effects of HXR9 with either DNR or MTX were calculated by a

combination index (CI) equation introduced by Chou-Talalay (Chou 1991), using Calcusyn

Software. The equation is

CI = (D)1

(Dx)1+

(D)2

(Dx)2 +

(D)1(D)2

(Dx)1(Dx)2

Where CI > 1, CI = 1, CI < 1 indicate antagonism, an additive effect and synergism,

respectively. X is the inhibition percentage so for example, 50%, (Dx)1 and (Dx)2 are the

concentrations of drug 1 and drug 2 alone, respectively, that cause 50% of cell death and can

be calculated by the median-effect equation; (D)1 and (D)2 are the concentrations of the drugs

in combination that also cause 50% of cell death.

2.3.3 Statistical analysis of in vivo assays

Data were expressed as the average tumour sizes for each group at the represented day

with SEM as error bars. The statistical significance in tumour sizes was determined using two-

way anova analysis.

Page 79: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

65

Table 2.9 Summary of the in vitro assays performed and reagents used.

Assay Investigation reagent solvent Stock

concentration

Storing Working

concentration

End point

measurement

LDH HXR9, DNR

and MTX

cytotoxicity

LDH

detection

kit

-20ºC LDH activity

Combination

cytotoxicity

HXR9

interaction

with DNR or

MTX

LDH

detection

kit

-20ºC LDH activity

Annexin V Cell death

mechanism

Annexin V

and 7-

AAD

4ºC PS exposure

Caspase-3 Caspase-3

role

z-DEVD-

R110 and

Ac-

DEVD-

CHO

-20ºC LDH activity

DAPI

staining

Nuclear

fragmentation

DAPI

stain

PBS -20ºC 1:1000 Nuclear

fragmentation

General

caspase

activity

General

caspase

activity

z-VAD-

FMK

DMSO 43mM -20ºC 50μM PS exposure

CsA

protection

CypD role CsA DMSO 20mM -20ºC 50μM PS exposure

Nec-1

protection

RIP1 role Nec-1 DMSO 40mM -20ºC 50μM PS exposure

Fructose

protection

ATP role Fructose medium RT 40mM LDH activity

EDTA effect Extracellular

Ca2+

EDTA medium RT 20mM LDH activity

and PS

exposure

ERK

inhibition

EERK role U0126 DMSO 1mM 4ºC 20μM LDH activity

JNK

inhibition

JNK role SP600125 DMSO 25mM 4ºC 60μM LDH activity

p38

inhibition

p38 role SB203580 DMSO 5mM -20ºC 25μM LDH activity

Page 80: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

66

Assay Investigation reagent solvent Stock

concentration

Storing Working

conentration

End point

measurement

NOX

inhibition

NOX role DPI DMSO 15mM -20ºC 60μM LDH activity

μ-calpain μ-calpain

role

Calpain

inhibitor I

DMSO 20mM -20ºC 60μM LDH activity

CaM

inhibition

CaM role W-7 DMSO 50mM -20ºC 50μM LDH activity

PKC

inhibition

PKC role Ro31-

8220

H2O 20mM 4ºC 30μM LDH activity

Simultaneous

CaM and

PKC

inhibition

Simultaneous

CaM and

PKC

inhibition

W-7 and

Ro31-

8220

50μM W-7

and 30μM

Ro31-8220

LDH activity

p53

inhibition

p53 role PFT-α DMSO 20mM -20ºC 40μM LDH activity

HO-1

inhibition

HO-1 role PPIX DMSO 18mM -20ºC 50μM LDH activity

WB PARP1

activation

Anti-

PARP1

antibody

Blocking

buffer

-20ºC 1/4000 Protein level

Page 81: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

67

Chapter 3 In vitro cytotoxicity of HXR9

Page 82: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

68

3.1 Introduction

The dysregulation of HOX genes is a common to many types of haematological

malignancies, including AML (Ferrando, Armstrong et al. 2003), and solid

malignancies including renal (Shears, Plowright et al. 2008), ovarian (Cheng, Liu et al.

2005; Morgan, Plowright et al. 2010), lung (Abe, Hamada et al. 2006; Plowright,

Harrington et al. 2009) and other cancers. Furthermore, over-expression of HOXA9 in

AML is a poor prognostic factor (Golub, Slonim et al. 1999), whilst down-regulation

of HOX genes is a favourable prognostic factor (Andreeff, Ruvolo et al. 2008;

Zangenberg, Grubach et al. 2009). HOX genes have an oncogenic function in

leukaemias (Eklund 2007) and their over-expression in melanoma maintain cell

survival, in part through preventing c-FOS transcription (Morgan, Pirard et al. 2007).

The small cell-permeable peptide HXR9 was used to disrupt the interaction

between HOX proteins and their partner transcription factor, PBX. This resulted in cell

death after a two-hour treatment in several cancer cell lines including melanoma, renal

and ovarian cells (Morgan, Pirard et al. 2007; Shears, Plowright et al. 2008; Morgan,

Plowright et al. 2010). Therefore, it was investigated whether AML cell lines were also

sensitive to HXR9.

I also studied the effect of combining HXR9 with commonly used

chemotherapies in AML treatment, MTX and DNR (Piccaluga, Visani et al. 2002;

Minotti, Menna et al. 2004; Fernandez 2010; Larson, Campbell et al. 2012).

Page 83: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

69

3.1.1 Amis of chapter 3

To assess the expression of HOX genes in AML and the cytotoxicity of HXR9

on several AML cell lines, either alone or in combination with commonly used AML

chemotherapies.

To achieve these aims, the following experiments were performed:

1. The expression of the whole panel of 39 HOX genes was measured in five AML

cell lines (KG-1, HEL92.1.7, HL-60, KU812F and K562) by RT-PCR.

2. The cytotoxicity of HXR9 on the five AML cell lines was assessed by LDH

assay.

3. The cytotoxicity of common AML chemotherapy drugs including DNR and

MTX on two AML cell lines was assessed by LDH assay.

4. The interaction between HXR9 and DNR or MTX was conducted by LDH

assay.

Page 84: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

70

3.2 Results

3.2.1 HOX gene expression in AML cell lines

Since HXR9 disrupts the interaction of HOX (1-10)-PBX, the expression of

these HOX genes was assessed by RT-PCR. The expression of HOX genes was studied

in the five AML derived cell lines HL-60, KG-1, HEL92.1.7, KU812F and K562. This

revealed a number of differences in HOX gene expression, with HOXA and HOXB

cluster genes (Figure 3.1A and B) more highly expressed than those of HOXC and

HOXD clusters (Figure 3.1C and D). It is also noteworthy that many HOXA cluster

were highly expressed in KG-1 cell line including HOXA5, HOXA6, HOXA9 and

HOXA10. As in KG-1, HOXA9 and HOXA10 were highly expressed in the KU812F

cell line, while HOXB9 was the most highly expressed gene in K562 and HEL92.1.7.

Intriguingly, HL-60 cells show very little HOX expression.

Page 85: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

71

A)

B)

C)

Page 86: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

72

D)

Figure 3.1 HOX gene expression in KG-1, HEL92.1.7, KU812F, K562 and HL-60 cell lines. A) The most

expressed HOXA genes in the KG-1 cell line were HOXA5, HOXA6, HOXA9 and HOXA10, while HOXA9 and

HOXA10 were most highly expressed in KU812F. B) HOXB9 was the most expressed HOX gene in HEL92.1.7

and K562. C) HOXC and D) HOXD genes showed very little expression in tested AML derived cell lines. The

expression of HOX genes were represented as the logarithm of the ratio of the expression of HOX genes to the

housekeeping gene β-actin (×10 000). Graphs express the mean of three independent experiments and error bars

show the SEM.

Page 87: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

73

3.2.2 HXR9 is cytotoxic on all tested AML derived cell lines

To determine whether disrupting the interaction between HOX proteins and PBX

triggers cell death, the AML derived cell lines including KG-1, HEL 92.1.7 and HL-60, which

are derived from primary AML patients, and KU812F and K562, which are derived from

secondary AML patients, were exposed to titrations of HXR9 and its negative control CXR9

for two hours. The cytotoxic effect of HXR9 and CXR9 was then measured by LDH assay.

HXR9 had a dramatic cytotoxic effect on all tested AML cell lines, while CXR9 that lacks a

functional domain had no cytotoxicity (Figure 3.2). The IC50 values were measured by Calcusyn

software and were 4.5μM, 6.1μM, 19.9μM, 9.1μM and 10.4μM for KG-1, HEL 92.1.7, HL-60,

KU812F and K562, respectively (Table 3.1).

Page 88: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

74

A)

B)

C)

Page 89: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

75

D)

E)

Figure 3.2 LDH assay for HXR9 and CXR9 cytotoxicity on AML derived cell lines. The cell lines were treated

with titrations of HXR9, red curve, and CXR9, blue line, for two hours, and then the LDH enzyme activity was

measured in supernatants, which indirectly reflects cell survival. IC50s of HXR9 were calculated using Calcusyn

software. Graphs show the mean of three independent repeats that had R2 ≥ 0.95 and error bars show SEM. A)

KG-1, B) HEL 92.1.7, C) HL-60, D) KU812F and E) K562 cell lines.

Table 3.1 IC50 values of HXR9 on AML derived cell lines with SEM.

Cell line Source IC50 of HXR9 (μM) SEM

KG-1 Primary AML

patients

4.5 ± 0.353

HEL 92.1.7 6.1 ±0.720

HL-60 19.9 ±0.368

KU812F Secondary AML

patients

9.1 ±1.886

K562 10.4 ±0.281

The IC50 values represent the mean IC50 of three independent experiments that were calculated by Calcusyn

software.

Page 90: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

76

3.2.3 DNR is cytotoxic on K562 and HL-60

In attempt to investigate the combination effect of HXR9 and chemotherapeutic drugs

on AML-derived cell lines, two of the conventional chemotherapies were selected including

DNR and MTX. It was reported that CD34+ AML cells, KG-1 and HEL92.1.7, were resistant

for both drugs (Bailly, Muller et al. 1995; Bailly, Skladanowski et al. 1997). Therefore, HL-60

and K562 that are represented primary and secondary AML, respectively, were selected to

conduct the combination therapy effect.

To determine the cytotoxicity of DNR on the AML cell lines K562 and HL-60, the cell

lines were treated with a titration of DNR for 24 hours and the cytotoxicity was assessed by

LDH assay. Both cell lines were sensitive to DNR at approximately the same concentration

after 24-hour treatment with an IC50 of 17.7μM and 17.4μM for K562 and HL-60, respectively

(Figure 3.3).

Figure 3.3 LDH assay for DNR cytotoxicity on K562 and HL-60 cell lines. The cell lines were treated with a

titration of DNR (range 2.5μM - 30μM) for 24 hours. The cytotoxicity of DNR was estimated by measuring LDH

enzyme activity in cell free supernatants, which indirectly reflects cell survival. The IC50 of DNR calculated using

Calcusyn software that was 17.7μM for K562 and 17.4μM for HL-60. The Graph shows the mean of three

independent repeats that had R2 ≥ 0.95 and error bars show the SEM. The red and blue curves represent K562 and

HL-60, respectively.

Page 91: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

77

3.2.4 Combination effect of HXR9 and DNR

To determine the effect of a combination between HXR9 and DNR, the cell lines K562

and HL-60 were treated concurrently with HXR9 and DNR. The cell lines were exposed to each

drug alone or to the combination of the two drugs (Figure 3.4). In the single drug exposure, the

cell lines were exposed to HXR9 for two hours or to DNR for 24 hours. In the combination

therapy, the cell lines were exposed to DNR for 24 hours and in the last two hours HXR9 was

added. The combination effect of HXR9 and DNR was then estimated for significance using

Calcusyn software by calculating CI values, as described in the analysis of drug combination

assay data. The combination therapy had an antagonistic effect at the effective dose 50 (ED50)

on both cell lines, K562 (ED50 1.34 ± 0.75) and HL-60 (ED50 1.32 ± 0.016), while at ED90, the

combination therapy was synergistic on K562 (ED90 0.72 ± 0.12) and nearly additive on HL-

60 (ED90 0.98 ± 0.07) (Table 3.2).

Page 92: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

78

A) B)

.

Table 3.2 The analysis of the combination effect of HXR9 and DNR by Calcusyn software. HXR9 and DNR

combination effect on K562 (A) and HL-60 (B) cells.

A) HXR9 and DNR combination effect on K562.

B) HXR9 and DNR combination effect on HL-60.

ED CI Combination effect

ED50 1.34 ± 0.75 - slight antagonism

ED75 0.98 ± 0.91 ± Nearly additive

ED90 0.72 ± 0.12 ++ Moderate synergism

ED CI Combination effect

ED50 1.32 ± 0.016 -- Moderate antagonism

ED75 1.129 ± 0.045 -- Moderate antagonism

ED90 0.98 ± 0.07 ± Nearly additive

Figure 3.4 LDH assay of HXR9 and DNR combination therapy for K562 and HL-60. Drug

concentrations used were 0.25, 0.5, 1, 2, 3 times the calculated IC50 of each drug in a constant ratio,

HXR9 IC50 was 21μM and 20μM, DNR IC50 was 17.7μM and 17.4μM for K562 and HL-60,

respectively. Cells were treated with HXR9, blue bars, or DNR, red bars, for 2 hours or 24 hours,

respectively. In addition, cells were treated with DNR for 24 hours and in the last two hours HXR9 was

added, green bars, in the combination therapy. The graphs show the mean of three independent repeats

and error bars show the SEM. A) is K562 and B) is HL-60 combination therapy graphs.

Page 93: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

79

3.2.5 MTX induces K562 and HL-60 cell death

To assess the cytotoxicity of MTX on K562 and HL-60, cells were exposed to increasing

concentrations of MTX (range 2.5μM - 40μM) for 24 hours. The induction of cell death was

then determined by LDH assay. MTX induced cell death in both cell lines with an IC50 20.6μM

and 12.8μM for K562 and HL-60, respectively (Figure 3.5).

Figure 3.5 LDH assay for MTX cytotoxicity on K562 and HL-60 cell lines. The cell lines were treated with

different concentrations of MTX (range 2.5μM - 40μM) for 24 hours. Cell death was assessed by measuring LDH

enzyme activity in cell free supernatants that indirectly reflects cell survival. The IC50 of MTX was calculated

using Calcusyn software and was 20.6μM for K562 and 12.8μM for HL-60. The Graph shows the mean of three

independent repeats that had R2 ≥ 0.95 and error bars show the SEM. The red and blue curves represent K562 and

HL-60, respectively.

Page 94: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

80

3.2.6 Combination effect of HXR9 and MTX

To assess the effect of HXR9 and MTX combination therapy, K562 and HL-60 cells

were incubated with each agent alone or concurrently (Figure 3.6). Cells were treated for two

hours and 24 hours with HXR9 and MTX, respectively. In the co-administration therapy, cells

were treated for 24 hours with MTX and in the last two hours HXR9 was administrated. The

effect of the HXR9/MTX combination was then analysed by the median effect method, as

described in the analysis of drug combination assay data. Using this method, HXR9/MTX

appeared an antagonistic interaction at all drug concentrations used on both cell lines with CI

values more than 1 (Table 3.3).

Page 95: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

81

A) B)

Figure 3.6 LDH assay of HXR9/MTX combination therapy for K562 and HL-60. Drug concentrations used were

0.25, 0.5, 1, 2, 3 times the calculated IC50 of each drug in a constant ratio, the HXR9 IC50 was 21μM and 20μM,

and the MTX IC50 was 20.6μM and 12.8μM for K562 and HL-60, respectively. Cells were treated with HXR9,

blue bars, or MTX, red bars, for 2 hours or 24 hours, respectively. In addition, cells were treated with DNR for 24

hours and in the last two hours HXR9 was added, green bars, in the combination therapy. The Graphs show the

mean of three independent repeats and error bars show SEM. A) is K562 and B) is HL-60 combination therapy

graphs.

Table 3.3 The analysis of the combination effect of HXR9 and MTX by Calcusyn software.

A) HXR9 and MTX combination effect on K562.

B) HXR9 and MTX combination effect on HL-60.

ED CI Combination effect

ED50 1.47 ± 0.019 - - - Antagonism

ED75 1.62 ± 0.34 - - - Antagonism

ED90 1.53 ± 0.175 - - - Antagonism

ED CI Combination effect

ED50 2.06 ± 0.50 - - - Antagonism

ED75 1.75 ± 0.28 - - - Antagonism

ED90 1.63 ± 0.19 - - - Antagonism

Page 96: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

82

3.3 Discussion

The expression of all 39 HOX genes was evaluated in this study in five AML derived

cell lines including KG-1 and HEL92.1.7 from erythroleukaemic patients (Koeffler and Golde

1978; Martin and Papayannopoulou 1982), HL-60 from APML (Collins, Ruscetti et al. 1978),

and KU812F and K562 derived from CML patients in blast crisis (Lozzio and Lozzio 1975;

Kishi 1985). Our findings of HOX expression concur with previous studies. Whilst the myeloid

origin cell line KU812F highly expressed HOXA9 and HOXA10 genes, the erythroid cell line

HEL92.1.7 highly expressed HOXB9 gene consistent with previous findings (Giampaolo,

Sterpetti et al. 1994; Kawagoe, Humphries et al. 1999; Pineault, Helgason et al. 2002). Although

K562 originated from a CML patient in blast crisis, this cell highly expressed HOXB9 gene

which supports previous studies suggesting that K562 is in fact a human erythroleukaemia cell

line (Andersson, Jokinen et al. 1979; Andersson, Nilsson et al. 1979). Interestingly, KG-1

highly expressed HOXA genes including HOXA5, HOXA6, HOXA9 and HOXA10 even though

it is an erythroid line. It was reported that KG-1 highly expressed HOXA9 and HOXA10 genes

due to the presence of a trisomy 8 mutation (Kok, Brown et al. 2010). However, the associating

mechanism between up-regulation of HOXA genes and trisomy 8 is unclear. Intriguingly, the

APML cell line HL-60 hardly expressed HOX genes, but this is in line with previous studies in

which APML was found to be characterised by global down-regulation of HOX genes

(Thompson, Quinn et al. 2003). Notably, the most expressed subset of HOX genes were the

posterior (6-13) HOX genes. One potential explanation for their up-regulation in AML cell lines

is their ability to interrupt haematopoietic differentiation (Pineault, Abramovich et al. 2004).

Overall, the expression of HOX genes in the tested AML lines was consistent with the pre-

established notion that HOX genes are expressed in a lineage-restricted manner, whereby HOXA

and HOXB genes are expressed in myeloid and erythroid cells, respectively (Giampaolo,

Sterpetti et al. 1994; Kawagoe, Humphries et al. 1999; Pineault, Helgason et al. 2002).

Page 97: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

83

Here it is showed that the high expression of HOX genes can be used as a therapeutic

target in AML as targeting the HOX-PBX interaction with HXR9 caused cell death in the tested

AML derived cell lines. The IC50 of HXR9 for the treated AML derived cell lines ranges from

4.5μM to 20μM after a two-hour treatment. The sensitivity of the tested cell lines to HXR9

directly correlated to the level of HOX genes expression, for example, KG-1 cells that highly

expressed HOXA5, HOXA6, HOXA9 and HOXA10 were the most sensitive to HXR9 with an

IC50 4.5μM after a two-hour treatment while HL-60 cells, which show a global down-regulation

of HOX genes were the least sensitive to HXR9, with an IC50 19.9μM after a two-hour treatment.

Furthermore, there was no difference in terms of HXR9 sensitivity between the HEL92.1.7 cells

derived from a primary AML patient and KU812F and K562, derived from CML patients in

blast crisis (secondary AML) which might be because these cells express the most highly

expressed HOX gene at the same level. In general, the tested AML cell lines were more sensitive

to HXR9 than other cell lines derived from solid malignancies including melanoma, renal,

ovarian and lung cancers (Table 3.4) (Shears, Plowright et al. 2008; Plowright, Harrington et

al. 2009; Morgan, Plowright et al. 2010). The difference in the sensitivity between AML cells

and solid cancers might be because survival of AML cells is totally depending upon the HOX-

PBX interaction, or due to the abundant expression of HOX genes in AML cells compared to

the solid cancers.

Page 98: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

84

Table 3.4 The IC50 values of HXR9 with cell lines derived from different cancers.

Cell line Cell line source IC50 Treatment period Reference

KG-1, HEL92.1.7

and HL60

Primary AML 4.5μM , 6.1μM and

19.9μM

2 hours

This study

KU812F and K562 Secondary AML 9.1μM and 10.4μM

769-p and CaKi-2 Renal 40μM and 44μM (Shears, Plowright et al.

2008)

B16F10 Melanoma 20μM (Morgan, Pirard et al.

2007)

SK-OV3 Ovarian 70μM (Morgan, Plowright et

al. 2010)

A549 and H23 Lung 32.5μM and 69μM 24 hours (Plowright, Harrington

et al. 2009)

This table shows the difference in the IC50 values between AML cell lines investigated in this study and cell lines

derived from solid cancers including renal, melanoma, ovarian and lung cancers established in previous studies.

All cells were treated with HXR9 for two hours except lung cancer cells, which were treated for 24 hours.

Generally, AML cell lines were the most sensitive to HXR9.

The drug-interaction effect of HXR9 and DNR was performed using the Chou and

Talaly method of calculating the median effect dose (Chou 1991). Results showed that the

combination effect was antagonistic at ED50 in both of the tested AML cell lines K562 and HL-

60, but it was either synergistic or additive at ED90 in K562 or HL-60, respectively. DNR causes

apoptosis by blocking topoisomerase II, intercalating with DNA, and producing ROS, and leads

to nuclear factor-kappa B (NF-ҠB)-DNA binding as a cellular response to stress (Lanzi,

Gambetta et al. 1991; Boland, Foster et al. 1997; Boland, Fitzgerald et al. 2000; Laurent and

Jaffrézou 2001; Campbell, O'Shea et al. 2006; Karl, Pritschow et al. 2009). Interestingly, NF-

ҠB has an anti-apoptotic role in DNR treatment, as reducing the activity of NF-ҠB and reducing

the degradation of the NF-ҠB inhibitor IҠB using bortezomib increases the sensitivity of K562

to DNR (Wang, Wang et al. 2012). In addition, DNR causes up-regulation of PKC that reduces

DNR cytotoxicity by enhancing NF-ҠB-DNA binding and reducing ROS production (Lanzi,

Page 99: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

85

Gambetta et al. 1991; Laurent and Jaffrézou 2001; Bezombes, de Thonel et al. 2002). However,

Inhibition of PKC sensitized the AML cell line U937 to DNR (Mas, Hernandez et al. 2003). In

addition, it has been found that ROS production as a result of DNR treatment slows the kinetics

of NF-ҠB activation and DNA binding (Campbell, O'Shea et al. 2006), which might explain the

difference in effect of combining low or high doses of HXR9 and DNR. In the low dose DNR

and HXR9 combination therapy, the level of ROS production might be insufficient to delay

NF-ҠB-DNA binding long enough for HXR9 to cause the up-regulation of c-FOS that mediates

HXR9 toxicity and is also a negative regulator of NF-ҠB (Morgan, Pirard et al. 2007; Plowright,

Harrington et al. 2009; Takada, Ray et al. 2010). However, at high concentrations of both drugs,

DNR might produce more ROS that would delay the activation of NF-ҠB, allowing HXR9 to

cause an up-regulation of c-FOS that would inhibit NF-ҠB activity and ultimately lead to either

a synergistic effect in K562 or at least additive effect in HL-60. These data suggest that HXR9

is synergistic or at least additive with DNR, a drug commonly used for AML treatment (Figure

3.7).

Figure 3.7 The proposed interaction mechanism between DNR and HXR9. DNR blocks topoisomerase II that

leads to activation of the surviving pathway PKC/NF-κB, which in turn reduces necrosis by HR9 through reduction

of c-FOS activity, and the apoptotic pathway through ROS signalling that delays the activity of NF-κB.

Page 100: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

86

Unlike DNR, MTX has an antagonistic effect on HXR9. This antagonism is owing to

the conflict in the mechanism of both drugs. MTX causes apoptosis by inhibiting topoisomerase

II, which involves the production of a transient DNA double-strand break, thereby damaging

DNA that in turn activates NF-ҡB-DNA binding as a consequence of cellular stress response

(Boland, Foster et al. 1997; Boland, Fitzgerald et al. 2000; Neuhaus, Kieseier et al. 2004;

Campbell, O'Shea et al. 2006; Karl, Pritschow et al. 2009). Unlike DNR, ROS production is not

involved in MTX mediated apoptosis (Campbell, O'Shea et al. 2006). In addition, MTX leads

to activation of PKC, which is a positive upstream regulator of NF-ҡB and it is has been

reported that pre-treating cells with a PKC activator enhanced the cytotoxicity of MTX (Bhalla,

Ibrado et al. 1993; Estévez, Vieytes et al. 1996; Lin, O'Mahony et al. 2000; Mut, Amos et al.

2010; Lutzny, Kocher et al. 2013). However, PKC appeared to cause resistance to HXR9 as it

was found in this study. While c-FOS, c-JUN and EGR-1 over-expression caused relapse in

AML patients treated with MTX, c-FOS over-expression is considered as a key event in

apoptosis induced by HXR9; EGR-1 and c-JUN are also up-regulated by HXR9 (Bailly,

Skladanowski et al. 1997; Potapova, Basu et al. 2001; Staber, Linkesch et al. 2004; Morgan,

Pirard et al. 2007; Plowright, Harrington et al. 2009). Whilst disruption of NF-ҡB by short

interfering RNA caused a dramatic reduction in apoptosis upon MTX treatment, c-FOS was

reported as a negative regulator of NF-ҡB mediated stress response and c-fos-/- mice showed an

elevation in NF-ҡB activity (Ray, Kuwahara et al. 2006; Karl, Pritschow et al. 2009; Takada,

Ray et al. 2010). Likewise, MTX treatment leads to a significant reduction of c-Myc, while c-

Myc was found to be a downstream effector of c-FOS-induced apoptosis (Bhalla, Ibrado et al.

1993; Kalra and Kumar 2004). In addition, the expression of c-FOS was high in MTX-resistant

AML cell lines (Bailly, Skladanowski et al. 1997) and c-FOS positively regulates ABCG2, also

known as breast cancer resistance protein (BCRP), which mediates MTX-resistance in the

Page 101: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

87

breast cancer cell line MCF-7 (Bailey-Dell, Hassel et al. 2001; Schwabedissen, Grube et al.

2006). Taken together, these data suggest that HXR9 and MTX exerted a reciprocal inhibition.

3.3.1 Summary of chapter

In summary, this chapter studies show that:

AML cell lines derived from different AML types express different subsets of HOX

genes at different levels due to the heterogeneity of AML.

The HOX-PBX interaction is crucial for the tested AML cell lines survival and targeting

this interaction using HXR9 leads to cell death.

HXR9/DNR combination therapy is synergistic or at least additive.

HXR9/MTX combination therapy is antagonistic due to the conflict in their mechanisms

of action.

Page 102: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

88

Chapter 4 The mechanism of HXR9 cytotoxicity

Page 103: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

89

4.1 Introduction

Better understanding of the molecular aberrations in cancers resulted in the development

of several new potential therapeutic agents such as HXR9 that target HOX-PBX interaction.

However, molecular agents combined with conventional chemotherapies have given rise to

more curative therapies. In order to provide a rational basis for the selection of a synergistic

partner for HXR9, the mechanism by which HXR9 induces cell death needs to be identified.

The specificity of HXR9 for the HOX-PBX interaction was previously demonstrated in

melanoma and lung cancer cells (Morgan, Pirard et al. 2007; Plowright, Harrington et al. 2009),

as were downstream transcriptional changes. Notably, the increased expression of c-FOS was

responsible, at least in part, for the HXR9 mediated cell death (Morgan, Pirard et al. 2007;

Shears, Plowright et al. 2008; Plowright, Harrington et al. 2009; Morgan, Plowright et al. 2010;

Morgan, Boxall et al. 2012; Morgan, Boxall et al. 2014), subsequently, the expression of c-FOS

was assessed in this study. In addition, the up-regulation of other genes including ATF1, ATF3,

NRUA3, JUN, DUSP1 was also observed (Morgan, Pirard et al. 2007; Shears, Plowright et al.

2008; Morgan, Boxall et al. 2012). Previous studies showed that the general inhibition of

caspases did not completely prevent HXR9-triggered cell death, which suggests that there may

be additional mediators of cell death (Shears, Plowright et al. 2008; Morgan, Boxall et al. 2012;

Morgan, Boxall et al. 2014). However, the nature of these potential mediators of cell death by

HXR9 remains unknown. Therefore, the possible involvement of necrotic pathways in HXR9

induced cell death was investigated. In addition, the molecular mechanism of HXR9 action is

yet to be identified, subsequently, the potential role of MAPK pathways including MEK/ERK,

p38 and JNK were assessed.

Page 104: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

90

Cell death can be classified into accidental cell death and programmed cell death (PCD).

Accidental cell death results from extreme environmental disturbances including severe

hypoxia, and extreme temperature changes. Necrosis was described as an unregulated and

accidental cell death. However, it has also been suggested that necrosis might result from

regulated enzyme-catalysed biochemical reactions (Fink and Cookson 2005). Therefore,

necrosis can be categorised as PCD when it results from proteolytic reactions and called

necrotic-like PCD or regulated necrosis (Galluzzi, Kepp et al. 2014). Regulated necrosis is

classified into mitochondrial necrosis, necroptosis and parthanatos (Figure 4.1). This type of

cell death is characterised by the absence of hallmarks of apoptosis such as caspase activation,

Bcl-2 family member involvement, cyt C release, chromatin condensation, nuclear

fragmentation and PARP1 cleavage (Saraste and Pulkki 2000; Ziegler and Groscurth 2004;

Miao and Degterev 2009).

Figure 4.1 A schematic diagram of cell death. It can be classified into programmed and accidental cell death. The

former is further sub-classified into apoptosis and regulated necrosis. Apoptosis is sub-divided into extrinsic and

intrinsic pathways, both of which are caspase-dependent. Regulated necrosis, which is caspase-independent, is

subdivided into mitochondrial necrosis, necroptosis and parthanatos.

Page 105: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

91

Cells undergo apoptosis via two main pathways, extrinsic and intrinsic pathway, both

of which eventually result in caspase activation (Figure 4.1). The extrinsic pathway is initiated

by the binding of ligands to cell surface death receptors such as those of the tumour necrosis

factor (TNF) receptor family that in turn leads to activation of initiator pro-caspases such as

pro-caspase-8 and/or pro-caspase-10 (Jin and El-Deiry 2005). Active caspase-8 and-10 cleave

the executioner pro-caspases-3/7 (Huerta, Goulet et al. 2007), which in turn cleave many

substrates including PARP1 and cytoskeletal proteins, and ultimately cause cell death by

apoptosis (Vermeulen, Van Bockstaele et al. 2005). The intrinsic pathway, also called the

mitochondrial pathway, involves the release of pro-apoptotic proteins that are trapped in

mitochondria such as cyt C and apoptosis inducing factor (AIF) (Martin 2010). Mitochondrial

membrane integrity is controlled by Bcl-2 family members (Galluzzi, Kepp et al. 2014), which

include anti-apoptotic proteins such as Bcl-2 and Bcl-xL, and pro-apoptotic proteins such as

Bad, Bid, Bax and Bak1 (Jin and El-Deiry 2005). Under normal conditions, the anti-apoptotic

members of Bcl-2 and Bcl-xL heterodimerize with the pro-apoptotic members Bax and Bak1

and consequently preventing their apoptotic function (Martin 2010). During apoptosis, caspase-

8 cleaves Bid that in turn with dephosphorylated Bad promotes apoptosis by binding to the anti-

apoptotic Bcl-2 and Bcl-xL proteins (Li, Zhu et al. 1998; Ghibelli and Diederich 2010; Martin

2010). As a result, Bax and Bak1 are released and form Bax/Bak channels in the outer

mitochondrial membrane causing mitochondrial permeabilization (Martin 2010). Cyt C is then

released into the cytosol and, in the presence of ATP, binds to Apaf-1 and pro-caspase-9

forming the pro-apoptotic complex apoptosome that leads to auto-cleavage and activation of

caspase-9 and caspase-3 and -7 (Shi 2002). Then, active caspases 3, 7 and 9 proteolytically

cleave XIAP which in healthy viable cells binds and inhibits the three caspases (Vermeulen,

Van Bockstaele et al. 2005). Therefore, both extrinsic and intrinsic apoptotic pathways was

investigated to assess the involvement of apoptosis in HXR9 cell death. In addition, due its

Page 106: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

92

known role in both apoptotic pathways, p53 and its downstream effector p21 involvement in

HXR9 caused cell death were investigated.

Ca2+ is one of the main mediators of regulated necrosis and studying its role in necrosis

is common. Subsequently, assessing the involvement of extracellular Ca2+ in HXR9 induced

cell death was addressed in this study. In addition, the role of Ca2+-downstream effectors

including NOX, µ-calpain, CaM, PKC was assessed.

CypD is a member of the cyclophilin family located in the mitochondrial matrix (Elrod

and Molkentin 2013). It is considered as the most important regulator of mPTP because it is the

only protein known to be required for mitochondrial permeability transition-dependent necrosis

(Alam, Baetz et al. 2014).

Necroptosis is a type of regulated necrosis that is mediated by the interaction between

RIP1 and RIP3 to form a necrosome (Marshall and Baines 2014). RIP1 can be either a

component of a pro-survival complex inducing the expression of NF-ĸB, an apoptotic complex

enhancing caspase-dependent apoptosis, or a necrosome complex initiating necroptosis

(Vandenabeele, Grootjans et al. 2013). However, RIP1 kinase activity and necrosome formation

are only required in necroptosis (Cho, Challa et al. 2009; He, Wang et al. 2009; Zhang, Shao et

al. 2009). The molecular downstream mechanism of the necrosome that eventually results in

necroptotic cell death is yet to be identified, and it remains unclear whether mitochondria are

involved (Cho, Challa et al. 2009; Zhang, Shao et al. 2009; Tait, Oberst et al. 2013). In addition,

mixed lineage kinase domain-like protein and phosphoglycerate mutase family member 5 were

identified as RIP3 downstream mediators of necroptosis (Sun, Wang et al. 2012; Wang, Jiang

et al. 2012).

Parthanatos is a form of regulated necrosis that is PARP1-dependent and caspase-

independent. This form of necrosis is characterised by hyper-activation of PARP1 that leads to

loss of mPTP and subsequently nuclear translocation of AIF from mitochondria, loss of ATP

Page 107: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

93

and ultimately cell necrosis (Andrabi, Dawson et al. 2008). PARP1 is a DNA repair protein that

is activated in response to DNA damage by alkylating agents and Ca2+ signalling pathways

(Galluzzi, Kepp et al. 2014).

Page 108: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

94

4.1.1 Aims of chapter 4

To investigate the mechanism of HXR9 action.

To achieve this aim, the following studies were performed:

1. Assess whether HXR9 causes up-regulation of c-FOS in AML cell lines as it did in all

previously tested cancers.

2. To determine whether HXR9 induced apoptosis in AML cells the following assays were

used:

a. Annexin V/7-AAD and DAPI staining assays to assess the morphological

changes.

b. RT-PCR to measure the expression of some members of the Bcl-2 family,

initiator and executioner caspases, apoptosis protease-activating factor 1

(Apaf1), X linked inhibitor of apoptosis (XIAP) and PARP1.

c. Inhibition of either caspase-3 by Z-DEVD-R110 and general inhibition of

caspases by z-VAD-FMK to analyse the role of caspases in the cytotoxicity of

HXR9.

3. To assess whether necrosis is involved in the cytotoxicity of HXR9, the following

studies were performed:

a. Determining the effect of chelating extracellular Ca2+ by EDTA on the efficacy

of HXR9 by LDH and annexin V/7-AAD assays.

b. Assessing the role of CypD in the HXR9 cytotoxicity by inhibiting its role using

CsA.

c. Assessing the role of RIP1 in the HXR9 cytotoxicity by inhibiting its role using

Nec-1.

d. Assessing the effect of depleting ATP by fructose on the effectiveness of HXR9.

Page 109: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

95

e. Determining whether HXR9 causes activation of PARP1 protein.

4. To determine the role of MAPK pathways and their downstream targets, the following

assays were analysed:

a. Members of MAPK pathway MEK/ERK, p38 and JNK.

b. NOX proteins.

c. μ-calpain, CaM, PKC and HO-1 proteins.

5. To assess whether p53 is involved in HXR9-induced cell death, the following assays

were done:

a. Measuring the expression of p53 and its downstream target p21 mRNA.

b. Inhibiting p53 protein.

Page 110: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

96

4.2 Results

4.2.1 HXR9 causes the up-regulation of c-FOS

c-FOS up-regulation was found to be a mediator of apoptosis triggered by HXR9 in

melanoma, lung, ovarian, breast and prostate derived cell lines (Morgan, Pirard et al. 2007;

Plowright, Harrington et al. 2009; Morgan, Plowright et al. 2010; Morgan, Boxall et al. 2012;

Morgan, Boxall et al. 2014). To determine whether HXR9 also causes up-regulation of c-FOS

in AML derived cell lines, the expression of c-FOS was measured in untreated, CXR9 or HXR9

treated cells by RT-PCR. Cells were treated for two hours with the IC50s of HXR9 or equivalent

concentrations of the negative control peptide CXR9, which were 4.5μM, 6μM, 20μM, 9μM

and 10.5μM for KG-1, HEL92.1.7, HL-60, KU812F and K562, respectively. A two-hour

treatment with HXR9 resulted in a 7.5, 16.7, 3.3, 50, and 7.5 fold increase in c-FOS expression

in KG-1, HEL92.1.7, HL-60, KU812F and K562, respectively, compared to untreated cells.

However, there was no significant change in c-FOS expression between untreated cells and

CXR9 treated cells (Figure 4.1).

Page 111: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

97

Figure 4.2 c-FOS expression in AML cell lines after treatment with HXR9. AML cell lines were treated with the

following concentrations of HXR9 or CXR9: 4.5μM, 6μM, 20μM, 9μM and 10.5μM for KG-1, HEL92.1.7, HL-

60, KU812F and K562, respectively, or were untreated (control), for two hours. After a two-hour treatment, mRNA

was extracted from cells and c-FOS expression was measured by RT-PCR. Results are presented as a ratio between

c-FOS and β-actin (x10 000). A two-hour treatment with HXR9 caused an elevation of c-FOS transcript levels

comparing to untreated cells. Graphs show the mean of three independent repeats and error bars show the SEM.

*p < 0.05, **p < 0.01 and ***p < 0.001 with respect to untreated cells.

Page 112: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

98

4.2.2 HXR9 causes death of AML tested cell lines

In order to determine the mode of cell death following HXR9 treatment, an annexin V-

PE assay was used. Cell lines were treated with the IC50, double and triple the IC50 of HXR9 or

of CXR9 equivalent to the highest concentration of HXR9 for two hours and then stained with

annexin V and 7-AAD. Cells were classified either as viable, early apoptotic or dead cells

according to 7-AAD penetration and annexin V binding (Table. 2.7). After a two-hour

treatment, there was no significant change in untreated cells and CXR9 treated cells, whilst

cells treated with the IC50 of HXR9 rapidly entered apoptosis, and many were dead (Figure 4.2).

For example, in untreated KU812F cells, 95% were viable cells, 2% and 3% were in early

apoptosis or dead, respectively, while 55% were viable, 20% were early apoptotic and 30%

were dead after 9μM HXR9 treatment (Figure 4.2D). Generally, cell treatment with either

HXR9 concentration killed the majority of cells. For example, treating HL-60 cells with 20μM

HXR9 killed 55% of cells, while 27% and 18% were viable or in an early apoptotic state,

respectively (Figure 4.2C).

Page 113: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

99

A)

B)

C)

Page 114: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

100

D)

E)

Figure 4.3 Detection of cell death in AML cell lines by flow cytometry. Cells were treated with the IC50, 2xIC50,

or 3xIC50 of HXR9, and with a concentration of CXR9 equivalent to the highest concentration of HXR9 for two

hours. Cells were then stained with annexin V and 7-AAD for 15 minutes in the dark and analysed by flow

cytometry. In all the tested cell lines, the number of early apoptotic and dead cells increased with increased HXR9

concentrations. Results are expressed as the mean of three separate experiments and error bars show the SEM. *p

< 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001 with respect to untreated cells. A) KG-1, B) HEL92.1.7, C)

HL-60, D) KU812F and E) K562 cells.

Page 115: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

101

4.2.3 HXR9 does not affect caspase or Bcl-2 family transcription

RT-PCR was used to analyse the transcriptional changes of several pro- and anti-

apoptotic genes upon disruption of the HOX/PBX dimer by HXR9. This analysis revealed that

HXR9 did not cause transcriptional changes in the initiator caspase-9 and its partner Apaf1 that

together form an apoptosome complex with cytochrome C (cyt C) thereby starting the intrinsic

apoptosis pathway (Adrain and Martin 2001). In addition, executioner caspases-3, -6 and -7,

which are key mediators of apoptosis (Huerta, Goulet et al. 2007), were not up-regulated after

HXR9 treatment. Likewise, none of the pro-apoptotic members of the Bcl-2 family including

Bad, Bax, Bak1 and Bid, nor the anti-apoptotic member Bcl-2, which mediate mitochondrial

cell death (Green and Kroemer 2004), showed transcriptional changes after HXR9 treatment.

In addition, neither the transcription of PARP1 nor XIAP was affected by HXR9 treatment

(Figure 4.3).

Page 116: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

102

A)

B)

C)

Page 117: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

103

D)

E)

Figure 4.4 Analysis of transcriptional changes of several pro- and anti-apoptotic genes upon HXR9 treatment.

Cells were treated with the IC50 of HXR9, CXR9 or untreated (control) for two hours. Then, mRNA was harvested

from the AML derived cell lines KG-1, HEL92.1.7, HL-60, KU812F and K562 and gene expression was analysed

using RT-PCR. There was no significant change in any of the analysed genes. Results are presented as a ratio with

β-actin (x10 000). Statistical analysis was performed using student's t-test by comparing the relative expression of

a gene of interest in HXR9 or CXR9 treated cells to its counterpart in untreated cells. Graphs show the mean of

three independent experiments and error bars show the SEM. Results are shown for A) KG-1, B) HEL92.1.7, C)

HL-60, D) KU812F and E) K562 cells.

Page 118: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

104

4.2.4 HXR9 does not activate caspase-3 in the tested AML cell lines

In order to study the mechanism of cell death, caspase-3 activity was measured after

treating cells with increasing concentrations of HXR9 or with a CXR9 concentration that was

equivalent to the highest concentration of HXR9. A fluorescently labelled caspase-3 substrate

and Ac-DEVD-CHO inhibitor were also added. As an additional control, R110 standard curves

were generated to validate individual experiments (Appendix 1). There were no significant

changes between untreated and HXR9 treated cells with respect to caspase-3 activity in any of

the AML cell lines (Figure 4.4).

Page 119: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

105

A)

B)

C)

Page 120: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

106

D)

E)

Figure 4.5 Detection of caspase-3 activity in AML cell lines using Z-DEVD-R110. Cells were treated with IC50,

2xIC50, 3xIC50 of HXR9, with an equivalent CXR9 to the highest concentration of HXR9 or untreated (control) for

two hours. Then, cells were harvested and lysed. Next, Ac-DEVD-CHO, a caspase inhibitor, was added to some

cells as a control. The Z-DEVD-R110 substrate was then added to cells and the fluorescence was read at 492nm.

Results were then normalised to lysis buffer fluorescence reading, background levels. There was no statistical

difference in caspase-3 activity between both HXR9- or CXR9-treated cells and untreated control cells. Graphs

show the mean of three independent experiments and error bars show the SEM. A) A graph represents caspase-3

activity of KG-1, B) HEL92.1.7, C) HL-60, D) KU812F and E) K562 cells.

KU812F

Untreated 27M CXR9 9M HXR9 18M HXR9 27M HXR9100

101

102

103

104

105

106

107

HXR9

HXR9+Ac-DEVD-CHO

Treatment

Flu

ore

scence

Page 121: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

107

4.2.5 HXR9 efficacy is not affected by the general inhibition of caspases

In order to further study the role of caspases in HXR9-induced cell death, K562 and

HL-60 cells were pre-treated with or without the general inhibitor of caspase activity z-VAD-

FMK. DNR that has been shown to activate caspases was used as a positive control (Liu, Kelsey

et al. 2002; Kim, Park et al. 2003). Results revealed that 50μM z-VAD-FMK pre-treatment

dramatically increased the percentage of viable cells in DNR treated cells, p < 0.0001. However,

there was no statistical difference in terms of the percentage of viable cells between z-VAD-

FMK non- or pre-treated cells. For example, the percentage of viable cells was 62% and 58%

for z-VAD-FMK non- or pre-treated K562 cells, respectively (Figure 4.5). Likewise, pre-

treatment with z-VAD-FMK did not cause significant changes in the early apoptotic and dead

cells comparing with z-VAD-FMK non-treated cells (Appendix 2). This suggests that HXR9

kills cells in a caspase-independent pathway.

Page 122: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

108

A)

B)

Figure 4.6 General inhibition of caspase activity in HXR9 treated AML cell lines by z-VAD-FMK. Cells were

pre-treated either with or without 50μM z-VAD-FMK for one hour, and then treated with the IC50, or 2xIC50 of

HXR9 for two hours or with 17.5μM DNR for 24 hours, which was used as a positive control. Next, cells were

harvested and stained with annexin V and 7-AAD for 15 minutes in the dark and analysed by flow cytometry. Data

showed that there was no statistical difference in terms of sensitivity to HXR9 between pre-treated cells with or

without 50μM z-VAD-FMK. Graphs show the mean of three independent experiments and error bars show the

SEM. ****p < 0.0001 with respect to z-VAD-FMK untreated cells. A) K562 and B) HL-60 cells.

Page 123: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

109

4.2.6 HXR9 does not cause nuclear fragmentation

In order to assess changes in nuclear morphology upon HXR9 treatment, AML derived

cell lines were stained with the fluorescent stain DAPI and analysed by a fluorescence

microscope. Changes in the nuclear morphology such as chromatin condensation and nuclear

fragmentation are markers of late stage apoptosis (Collins, Schandl et al. 1997; Ziegler and

Groscurth 2004; Huerta, Goulet et al. 2007). It was found that treatment of cells with the IC50s

of HXR9 did not result in changes to the nuclear morphology when comparing to untreated

cells in all investigated cell lines (Figure 4.6). This suggests that HXR9-induces cell death in a

nuclear fragmentation-independent pathway.

Page 124: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

110

Untreated IC50 HXR9

KG-1

HEL92.1.7

HL-60

Page 125: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

111

Untreated IC50 HXR9

Figure 4.7 DAPI staining of AML cells upon HXR9 treatment. Cells were treated with the IC50s of HXR9 for 2

hours. Cells were then harvested, fixed in 4% formaldehyde, and cytospan. Fixed cells were stained with DAPI

for 5 minutes at the dark. Finally, stained cells were analysed by a fluorescent microscope at 20x lenses.

KU812F

K562

Page 126: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

112

4.2.7 Ca2+ chelating abrogates cell killing by HXR9

As HXR9 seems to cause cell death in a caspase-independent manner, a number of

alterative pathways were investigated. Ca2+ is known to mediate caspase-independent cell death

(Dong, Li et al. 2005; Festjens, Vanden Berghe et al. 2006). Therefore, to analyse the role of

Ca2+ in HXR9-induced cell death, the extracellular Ca2+ chelator EDTA was used. Interestingly,

chelating extracellular Ca2+ rapidly attenuated the cytotoxicity of HXR9 and rescued cells from

cell death. For example, the percentage of viable cells increased from 50% to 80%, ****p <

0.0001, upon co-treatment with 20mM EDTA in 20μM HXR9 treated K562 cells (Figure 4.7A).

Likewise, in 20μM HXR9 treated HL-60 cells, the percentage of viable cells increased threefold

in 20mM EDTA co-treated cells compared to untreated cells (Figure 4.7B). In addition, co-

treatment with EDTA dramatically decreased the percentage of early apoptotic and dead cells

in both cell lines (Appendix 3).

Page 127: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

113

A)

B)

Figure 4.8 Effect of EDTA on HXR9 cytotoxicity. Cells were pre-treated with or without 20mM EDTA for one

hour, and then treated with 10μM, 20μM and 40μM HXR9, prepared in media either with or without 20mM EDTA,

for two hours. Next, cells were harvested and stained with annexin V and 7-AAD for 15 minutes in the dark and

analysed by flow cytometry. Results showed that Pre- and co-treatment of cells with 20mM EDTA significantly

increased the % viable cells and thereby inhibited the cytotoxicity of HXR9. Graphs show the mean of three

independent experiments of viable cells, non-stained cells with either stain, and error bars show the SEM. ***p <

0.001 and ****p < 0.0001 with respect to cells not exposed to 20mM EDTA. A) K562 and B) HL-60 cells.

Page 128: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

114

4.2.8 EDTA rescues cells from killing by HXR9

In order to confirm the previous results that showed chelating extracellular Ca2+ reduced

the cytotoxicity of HXR9, an LDH assay was performed to analyse the effect of EDTA pre-

and co-treatment. The results of the LDH assay were consistent with the results obtained using

the annexin V assay. Chelating extracellular Ca2+ by EDTA markedly reduced the levels of

LDH enzyme detected in K562 and HL-60 cells. In addition, HXR9 did not have an IC50 up to

the highest concentration used, 50μM (Figure 4.8). These results showed that EDTA prevented

cell killing by HXR9.

Page 129: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

115

A)

B)

Figure 4.9 LDH analysis of the effect of EDTA on HXR9 cytotoxicity. Cells were pre-treated with or without

20mM EDTA for one hour, and then with varying concentrations of HXR9 for two hours prepared in media either

with or without 20mM EDTA, red and blue curves, respectively. LDH activity was then measured in the

supernatants, which indirectly reflects the fraction of surviving cells. Pre- and co-treatment of cells with 20mM

EDTA significantly attenuated the cytotoxicity of HXR9. Graphs show the mean of three independent experiments

and error bars show the SEM. *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001 with respect to cells not

treated with 20mM EDTA. A) K562 and B) HL-60 cells.

Page 130: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

116

4.2.9 Cell killing by HXR9 does not involve CypD

To assess whether HXR9 causes mitochondrial necrotic cell death through activation of

CypD that in turn leads to formation of mPTP, cells were treated with CsA that binds to CypD

and inhibits mitochondrial cell death (Li, Johnson et al. 2004; Basso, Fante et al. 2005; Dube,

Selwood et al. 2012). H2O2, which triggers mPTP formation and mitochondrial necrotic cell

death, was used as a positive control for this assay (Baines, Kaiser et al. 2005; Nakagawa,

Shimizu et al. 2005; Schinzel, Takeuchi et al. 2005; Jobe, Wilson et al. 2008). Cell death was

measured using the annexin V assay. Treatment with 5μM CsA significantly increased the

percentage of viable cells exposed to H2O2 from 44.9% ± 1.7 to 69.2% ± 1.34, ****p < 0.0001,

and from 0.92% ± 0.3 to 34.2% ± 2.4, ****p < 0.0001, of H2O2-treated K562 and HL-60,

respectively. However, CsA at 5μM could not rescue K562 cells from death caused by HXR9

treatment, and interestingly, concurrent CsA treatment with HXR9 significantly decreased the

survival of HXR9 treated HL-60 cells (Figure 4.9, appendix 4). Taken together, it can be

concluded that CypD activation is not required for HXR9-induced K562 and HL-60 cell death.

Page 131: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

117

A)

B)

Figure 4.10 Effect of CsA on HXR9 cytotoxicity. Cells were pre-treated with or without 5μM CsA for one hour,

and then with the IC50, 2xIC50, 3xIC50 of HXR9, 20mM H2O2 (positive control), or were left untreated (negative

control) for two hours with or without 5μM CsA. The cells were then harvested and stained with annexin V and

7-AAD for 15 minutes in the dark and analysed by flow cytometry. Results showed no statistical difference in the

proportion of viable cells between K562 cells (A) incubated with or without 5μM CsA, while incubation with 5μM

CsA led to a significant decrease in proportion of viable HL-60 cells (B). Graphs show the mean of three

independent experiments and error bars show the SEM. *p < 0.05, ***p < 0.001 and ****p < 0.0001 with respect

to 5μM CsA untreated cells.

Page 132: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

118

4.2.10 Inhibition of RIP1 modifies the cytotoxicity of HXR9

Activation of RIP1 is a key event in necroptosis (Holler, Zaru et al. 2000). To assess

whether HXR9 causes necroptosis through activation of RIP1, the effect of RIP1 inhibition by

its specific inhibitor Nec-1 on HXR9 cytotoxicity was evaluated using the annexin V assay.

Nec-1 has been reported to inhibit H2O2-induced cell death; therefore H2O2 was used as a

positive control for this assay (Hanus, Zhang et al. 2013). Treatment with 50mM Nec-1 rescued

cells from 20mM H2O2 mediated cell killing. Nec-1 pre- and co-incubation with 20mM H2O2-

treated K562 cells significantly increased the percentage of viable cells from 34.5% ± 3.4 to

67.1% ± 2.3 and decreased the percentage of early apoptotic and dead cells from 41.3% ± 2.5

to 27.2% ± 1.4 and from 24.6% ± 3.6 to 7.3% ± 0.25, respectively, compared to Nec-1untreated

cells (Figure 4.10A). In HXR9-treated K562 cells, there was no significant impact of inhibition

of RIP1 on the cytotoxicity of increasing concentrations of HXR9 (Figure 4.10A). However,

Nec-1 at 50μM significantly changed the cytotoxicity of all concentrations of HXR9 on HL-60.

For example, at 40µM HXR9 treatment, Nec-1 significantly increased the percentage of viable

cells from 3.7% ± 1.21 to 9.2% ± 1.73 and dramatically delayed cell death by increasing the

percentage of early apoptotic cells from 16.3% ± 1.2 to 46.4% ± 2.4 and decreasing the

percentage of dead cells from 80% ± 1.1 to 44.3% ± 2.4, compared to Nec-1 untreated cells

(Figure 4.10B). Therefore, it can be concluded that the inhibition RIP1 has an impact on the

cytotoxicity of HXR9 and this effect depends on the cell type.

Page 133: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

119

A)

B)

Figure 4.11 Effect of RIP1 inhibition on HXR9 cytotoxicity. Cells were pre-treated with or without 50μM Nec-1

for one hour, and then with increasing concentrations of HXR9, with an equivalent CXR9 concentration to the

highest concentration of HXR9, 20mM H2O2 (positive control), or were untreated (negative control), in media

supplemented either with or without 50μM Nec-1 for two hours. Cells were then harvested and stained with

annexin V and 7-AAD for 15 minutes in the dark and analysed by flow cytometry. Treatment with 50μM Nec-1

did not affect the sensitivity of K562 cells (A) to HXR9, while for HL60 (B) it significantly increased the

percentage of viable and early apoptotic cells and decreased the percentage of dead cells. Graphs show the mean

of three independent experiments and error bars show the SEM. *p < 0.05, **p < 0.01, ***p < 0.001 and ****p <

0.0001 with respect to 50μM Nec-1 untreated cells.

Page 134: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

120

4.2.11 HXR9 induces cell death through an ATP-independent pathway

High levels of fructose have been found to cause ATP depletion (Nieminen, Saylor et

al. 1994; Latta, Künstle et al. 2000). ATP is necessary for PARP1-dependent necrosis and

caspase-dependent apoptosis (Los, Mozoluk et al. 2002; Latta, Künstle et al. 2007). To analyse

whether ATP has a role in HXR9-induced AML cell death, the impact of ATP depletion by a

high concentration of fructose on the cytotoxicity of HXR9 was measured by the LDH assay

using an enzyme-linked immunosorbent assay (ELISA). ATP depletion did not reduce the

cytotoxicity of HXR9 on K562 and HL-60 (Figure 4.11). These results were consistent with a

caspase-independent and PARP1-independent pathway.

Page 135: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

121

A)

B)

Figure 4.12 The effect of ATP depletion on HXR9 cytotoxicity. Cells were pre-incubated with or without

40mM fructose for one hour, and then with a range of HXR9 concentrations (2.5μM – 50μM) for two hours

prepared in media either with or without 40mM fructose, red and blue curves, respectively. HXR9 cytotoxicity

was then measured by assessing LDH enzyme activity in cell-free supernatants. ATP depletion did not inhibit

HXR9-induced death in either (A) K562 or (B) HL-60 cells. Graphs show the mean of three independent

experiments and error bars show the SEM.

Page 136: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

122

4.2.12 HXR9 induces cell death through a PARP1-independent pathway

The activation of PARP-1 results in ATP depletion, which is a key event in necrotic cell

death (Bouchard, Rouleau et al. 2003; Kim, Zhang et al. 2005). To assess the involvement of

PARP1 in HXR9 induction of AML cell death, the expression of PARP1 protein was estimated

by WB assay. By comparing the band density of treated cells to their untreated counterparts, it

can be concluded that HXR9 did not cause PARP1 activation (Figure 4.12). These findings

concur with those of the ATP depletion assay which suggests that HXR9 treatment results in

cell death independently of ATP depletion.

Figure 4.13 WB analysis of PARP1 activation after HXR9 treatment. HL-60 and K562 cells were treated with

increasing concentrations of HXR9 for two hours and then PARP1 protein expression was examined by WB. β-

actin protein was used as a loading control. There was no difference in band densities between treated and treated

cells after a two hour-treatment with HXR9.

Page 137: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

123

4.2.13 The effect of inhibition of MAPK pathways on HXR9 cytotoxicity

The results presented earlier in this chapter showed that chelating extracellular Ca2+ by

EDTA prevented HXR9 cytotoxicity. Potential downstream effectors of Ca2+-mediating

necrosis are MAPK pathway members and therefore the effect of HXR9 treatment on three key

components of this pathway, MEK/ERK, p38 and JNK was evaluated (Dhillon, Hagan et al.

2007).

4.2.13.1 HXR9 induces cell death in a MEK/ERK independent pathway

Ca2+ enhances the activation of the MEK/ERK pathway (Egea, Espinet et al. 1999;

Huang, Maher et al. 1999; Lallemend, Lefebvre et al. 2003). To test whether the MEK/ERK

pathway has a role in HXR9 cytotoxicity, this pathway was blocked by a specific inhibitor,

U0126, and the cytotoxicity of HXR9 was examined using the LDH assay. H2O2 that causes

cell death through activation of the MEK/ERK pathway was used as a positive control (Chung,

Jeong et al. 2002; Ray, Huang et al. 2012). Treatment with 20μM U0126 significantly inhibited

the cytotoxicity of H2O2 (Appendix 5). However, U0126 could not attenuate the cytotoxicity of

HXR9 on K562 or HL-60 cells (Figure 4.13). These findings suggest that MEK/ERK pathway

is not required for HXR9-mediated cell death of these cells.

Page 138: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

124

A)

B)

Figure 4.14 The effect of inhibiting the MEK/ERK pathway on the cytotoxicity of HXR9. Cells were pre-treated

with or without 20μM of the MEK/ERK inhibitor U0126 for one hour, and then with increasing concentrations of

HXR9 prepared in media with or without 20μM U0126, red and blue curves, respectively. The proportion of

surviving cells was then measured using the LDH assay. Pre- and co-treatment of cells with 20μM U0126 did not

prevent the cytotoxicity of HXR9 in (A) K562 or (B) HL60 cells. Graphs show the mean of three independent

experiments and error bars show the SEM.

Page 139: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

125

4.2.13.2 HXR9-mediated cell death does not require p38 pathway signalling

Ca2+ positively regulates the p38 pathway (Huang, Maher et al. 1999; Hsu, Huang et al.

2007). To evaluate the role of the p38 pathway in the HXR9-mediated cell killing, its specific

inhibitor SB302580 was used and the cytotoxicity of HXR9 measured by LDH assay. The p38

pathway is involved in H2O2 cytotoxicity (Pinkus, Weiner et al. 1996; Tobiume, Matsuzawa et

al. 2001; Yoon, Kim et al. 2001; Carvalho, Evelson et al. 2004), and therefore H2O2 was used

as a positive control for this assay. Blocking p38 remarkably inhibited the cytotoxicity of H2O2

(Appendix 6), but not HXR9 on the AML cell lines (Figure 4.14).

Page 140: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

126

A)

B)

Figure 4.15 The effect of inhibiting the p38 pathway on the cytotoxicity of HXR9. Cells were pre-treated with or

without 25μM of the p38 inhibitor SB302580 for one hour, and then with a titration of HXR9 prepared in media

with or without 25μM SB302580, red and blue curves, respectively. The cytotoxicity of HXR9 was analysed by

measuring LDH enzyme activity in cell-free supernatants. Pre- and co-treatment of cells with 25μM SB302580

did not affect the cytotoxicity of HXR9 in either (A) K562 or (B) HL-60 cells. Statistical significance was tested

using the student's t-test. Graphs show the mean of three independent experiments and error bars show the SEM.

Page 141: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

127

4.2.13.3 Inhibition of the JNK pathway sensitises K562, but not HL-60 cells to HXR9

Like the MEK/ERK and p38 pathways, the JNK pathway is activated by Ca2+ signalling

(Huang, Maher et al. 1999; Son, Lee et al. 2010; Jan, Su et al. 2013). To test the involvement

of JNK pathway for HXR9-induced cell death, it was inhibited using the specific inhibitor

SP600125. Due its ability to induce cell death through activation of the JNK pathway, H2O2

was used as a positive control for this assay (Pinkus, Weiner et al. 1996; Tobiume, Matsuzawa

et al. 2001; Yoon, Kim et al. 2001; Carvalho, Evelson et al. 2004; Kadowaki, Nishitoh et al.

2005). SP600125 at 60μM efficiently abrogated the cytotoxicity of H2O2 (Appendix 7).

Interestingly, inhibiting the JNK pathway resulted in inconsistent results in terms of HXR9

cytotoxicity, significantly sensitizing K562 cells, but not HL-60 cells to killing by HXR9.

Blocking the JNK pathway sharply decreased the percentage of surviving K562 cells from 56%

± 1.7 to 35.4 ± 2.5, ***p < 0.001, after 20μM HXR9 treatment (Figure 4.15).

Page 142: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

128

A)

B)

Figure 4.16 The effect of inhibiting the JNK pathway on HXR9 cytotoxicity. Cells were pre-incubated in media

supplemented with or without 60μM of the JNK inhibitor SP600125 for one hour, and then treated with increasing

concentrations of HXR9 prepared in media supplemented with or without 60μM SP600125, red and blue curves,

respectively. The percentage of surviving cells was measured using the LDH assay. Graphs show the mean of three

independent experiments and error bars show the SEM. *p < 0.05, **p < 0.01 and ***p < 0.001 with respect to

60μM SP600125 untreated cells.

Page 143: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

129

4.2.14 Blocking NOX enzymes sensitizes cells to HXR9

Previous studies have reported that members of NOX enzyme family such as NOX1

and NOX5 are regulated by Ca2+ (Bánfi, Molnár et al. 2001; Bánfi, Tirone et al. 2004; Brueckl,

Kaestle et al. 2006; Valencia and Kochevar 2007). To test whether NOXs have a role in HXR9

mediated cell killing, they were inhibited using DPI. Inhibition of NOX activity by DPI

remarkably sensitised both K562 and HL-60 cells to HXR9. For example, after 20μM HXR9

treatment, the proportion of surviving K562 and HL-60 cells decreased from 57.9% ± 3.54 to

32.6% ± 0.21, *p < 0.05, and from 55.2% ± 1.9 to 26.6 % ± 2.7, *p < 0.05, respectively,

compared to DPI untreated cells (Figure 4.16). These results show that NOX enzymes exert an

anti-necrotic effect in HXR9 treatment.

Page 144: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

130

A)

B)

Figure 4.17 The effect of NOX inhibition on the cytotoxicity of HXR9. Cells were pre-treated with or without

60μM of the NOX inhibitor, DPI, for one hour, and then with a range of HXR9 concentrations (2.5μM – 50μM)

prepared in media with or without 60μM DPI, red and blue curves, respectively. Next, the cytotoxicity of HXR9

was assessed using the LDH assay. NOX inhibition significantly sensitised both (A) K562 and (B) HL-60 cells to

HXR9. Graphs show the mean of three independent experiments and error bars show the SEM. *p < 0.05 and **p

< 0.01 with respect to 60μM DPI untreated cells.

Page 145: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

131

4.2.15 μ-Calpain is not required for HXR9-induced cell death

μ-Calpain or calpain type I is a downstream effector of Ca2+ (Mathiasen, Sergeev et al.

2002). To analyse whether HXR9 caused AML cell death through the activation of μ-Calpain,

its activity was blocked by the specific inhibitor calpain inhibitor I and assessed using the LDH

assay. Due to its ability to activate μ-Calpain, H2O2 was used as a positive control for this assay

(McClung, Judge et al. 2009; Pallepati and Averill-Bates 2011). Calpain inhibitor I significantly

inhibited the cytotoxicity of H2O2 (Appendix 8). However, calpain inhibitor I at 60μM could

not abrogate the cytotoxicity of HXR9 on tested AML cell lines, K562 and HL-60 (Figure 4.17).

Page 146: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

132

A)

B)

Figure 4.18 The effect of blocking μ-calpain on HXR9 cytotoxicity. Cells were pre-treated with or without 60μM

calpain inhibitor I for one hour, and then for two hours with increasing concentrations of HXR9 prepared in media

with or without 60μM calpain inhibitor I, red and blue curves, respectively. Next, the cytotoxicity of HXR9 was

assessed in cell-free supernatants using the LDH. μ-calpain inhibition did not affect HXR9 cytotoxicity in either

(A) K562 or (B) HL-60 cells. Graphs show the mean of three independent experiments and error bars show the

SEM.

Page 147: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

133

4.2.16 CaM inhibition dramatically increases the sensitivity of cells to HXR9

CaM is one of the main Ca2+ concentration sensor (Balshaw, Xu et al. 2001; Hoeflich

and Ikura 2002; Yuan, Jing et al. 2011). To assess the involvement of CaM in the cytotoxicity

of HXR9, its role was blocked by the specific inhibitor W-7. Interestingly, treating cells with

50μM W-7 alone did not affect cells, yet co-treatment of cells with HXR9 significantly reduced

cell survival. Co-treatment of K562 and HL-60 cells with 20μM HXR9 and 50μM W-7 sharply

decreased the percentage of surviving cells from 57% ± 1.6 to 15% ± 1.8, ****p < 0.0001, and

from 61% ± 2.4 to 26% ± 2.1, ***p < 0.001, respectively (Figure 4.18). It also reduced the IC50

of HXR9 on K562 cells by threefold, from 21μM to 7μM, and on HL-60 cells by twofold, from

24μM to 13μM (Figure 4.18C).

Page 148: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

134

A)

B)

C)

IC50

Cell line/condition HXR9 HXR9 + 50μM W-7

K562 21.4μM ± 3.1 7μM ± 2.9

HL-60 24μM ± 1.7 13μM ± 1.2

Figure 4.19 The effect of CaM inhibition on HXR9 sensitivity. Cells were pre-treated with or without 50μM

W-7 for one hour, and then for two hours with increasing concentrations of HXR9 prepared in media with

or without 50μM W-7, red and blue curves, respectively. The LDH assay was used to measure cytotoxicity.

W-7 dramatically increased the sensitivity to HXR9 of both (A) K562 cells and (B) HL-60 cells. Graphs

show the mean of three independent experiments and error bars show the SEM. C) A table shows the

difference in HXR9 IC50 between 50μM W-7-treated and untreated cells. *p < 0.05, **p < 0.01, ***p < 0.001

and ****p < 0.0001 with respect to 50μM W-7 untreated cells.

Page 149: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

135

4.2.17 Inhibition of PKC greatly increases the sensitivity of K562 and HL-60 cells to

HXR9

PKC is a family of kinases that are regulated by intracellular Ca2+ (Zhu, Fang et al. 1999;

Reyland 2007). To analyse the impact of blocking PKC on HXR9-induced AML cell death, the

specific inhibitor Ro31-8220 was used and the cytotoxicity of HXR9 was assessed with the

LDH assay. 30μM Ro31-8220 was not cytotoxic for AML cells, yet it significantly increased

HXR9-mediated cell killing. In K562 cells, Ro31-8220 remarkably sensitized cells to a very

low concentration of HXR9, 1.25μM, *p < 0.05 (Figure 4.19A). In addition, it caused a huge

reduction in the IC50 of HXR9, by 4.3 fold, from 21.4μM ± 1.5 in K562-HXR9 only treated

cells to 5μM ± 2.9 (Figure 4.19C). Similarly, inhibition of PKC in HL-60 increased cell killing

by HXR9 and decreased the IC50 by 2.7 fold from 29.3μM ± 1.9 to 10.7μM ± 0.8 (Figure 4.19B

and C). These results showed that PKC activity protects against HXR9 cytotoxicity.

Page 150: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

136

A)

B)

C)

Figure 4.20 The effect of blocking of PKC on the cytotoxicity of HXR9 on K562 and HL-60 cells. Cells were pre-

treated with or without 30μM Ro31-8220 for one hour, and then for two hours with different concentrations of

HXR9 in media supplemented with or without 30μM Ro31-8220, red and blue curves, respectively. The LDH

assay was then used to measure the cytotoxicity of HXR9 in cell-free media. Ro31-8220 co-treatment significantly

reduced cell survival. Graphs show the mean of three independent experiments for (A) K562 cells and (B) HL-60

cells and error bars show the SEM. *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001 with respect to 30μM

Ro31-8220 untreated cells. C) Differences in the IC50 values between HXR9-only treated cells and HXR9 + 30μM

Ro31-8220 treated cells.

IC50

Cell line/condition HXR9 HXR9 + 30μM Ro31-8220

K562 21.4μM ± 1.5 5μM ± 2.9

HL-60 29.3μM ± 1.9 10.7μM ± 0.8

Page 151: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

137

4.2.18 Simultaneous inhibition of CaM and PKC potentiates HXR9 cytotoxicity

To test whether simultaneous inhibition of CaM and PKC produced a further

sensitisation to HXR9, K562 and HL-60 were exposed to the inhibitors of both proteins and

treated with a titration of HXR9 and the cytotoxicity was measured using the LDH assay. The

concurrent regimen significantly increased the sensitivity to HXR9. For example, at 10μM

HXR9 treatment, the concurrent inhibition of both proteins dramatically decreased the

percentage of surviving cells by 55%, ****p < 0.0001, and 57%, ****p < 0.0001, for K562 and

HL-60, respectively, comparing with HXR9-only treated cells (Figure 4.20A and B). In

addition, the IC50 of HXR9 was reduced by 4.3 and 3.1 fold for K562 and HL-60, respectively

(Figure 4.20C).

Page 152: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

138

A)

B)

C)

IC50

Cell line/condition HXR9 HXR9 + (50μM W-7 30μM Ro31-8220)

K562 21.1μM ± 4.5 4.9μM ± 3.5

HL-60 14.8μM ± 3.9 4.8μM ± 2.7

Figure 4.21 The impact of concurrent inhibition of CaM and PKC on the cytotoxicity of HXR9. Cells were pre-

incubated with or without 50μM W-7 and 30μM Ro31-8220 for one hour, and then for two hours with a range of

HXR9 concentrations (0.625μM – 50μM) prepared in media with or without the inhibitors, red and blue curves,

respectively. The cytotoxicity of HXR9 was assessed using the LDH assay. The concurrent inhibition of CaM and

PKC significantly increased the HXR9-mediated cell death. Graphs show the mean of three independent

experiments with (A) K562 cells and (B) HL-60 cells, and error bars show the SEM. **p < 0.01, ***p < 0.001 and

****p < 0.0001 with respect to W-7 and Ro31-8220 untreated cells. C) The difference in the IC50s between HXR9-

only treated cells and HXR9 + (50μM W-7 + 30μM Ro31-8220) treated cells.

Page 153: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

139

4.2.19 HO-1 inhibition increases the sensitivity to HXR9

HO-1 acts as an anti-apoptotic protein in AML (Rushworth and MacEwan 2008). In

addition, HO-1 is a downstream effector of NOXs and PKC (Tsoyi, Jang et al. 2011; Schroder,

Zhang et al. 2012; Lee, Yang et al. 2012; Nguyen, Kim et al. 2013). To assess whether the

inhibition of HO-1 could increase the sensitivity to HXR9, HO-1 activity was blocked by its

specific inhibitor PPIX and the sensitivity of AML cells to HXR9 assessed. HO-1 inhibition

resulted in a significant reduction in the survival of both cell lines (Figure 4.21). PPIX decreased

the IC50 of HXR9 by 2.2 fold and 1.4 fold for K562 and HL-60, respectively (Figure 4.21C).

Therefore, HO-1 activity inhibits HXR9-mediated cell killing.

Page 154: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

140

A)

B)

C)

IC50

Cell line/condition HXR9 HXR9 + 50μM PPIX

K562 23.7μM ± 3.4 10.9μM ± 4.2

HL-60 22.5μM ± 5.1 16μM ± 1.4

Figure 4.22 The effect of HO-1 inhibition on the cytotoxicity of HXR9. Cells were pre-treated with or without

50μM PPIX for one hour and then for two hours with HXR9 in media with or without 50μM PPIX, red and blue

curves, respectively. Cell death was assessed using the LDH assay. Graphs show the mean of three independent

experiments for (A) K562 and (B) HL-60 cells, and error bars show the SEM. *p < 0.05, **p < 0.01, ***p < 0.001

and ****p < 0.0001 with respect to 50μM PPIX untreated cells. C) Differences in the IC50s between HXR9-only

treated cells and HXR9 + 50μM PPIX treated cells.

Page 155: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

141

4.2.20 HXR9 induces p21 expression but not p53

To analyse whether HXR9 causes up-regulation of p53 and its downstream effector p21,

the expression of both genes was analysed by RT-PCR. Results showed that HXR9 did not

affect the expression of p53 in K562 or HL-60 cells, but it did lead to a significant increase in

the expression of p21. For example, the expression of p21 in K562 cells increased by 2.5, **p

< 0.01, and 4.5, ****p < 0.0001, fold after treatment with 15μM and 30μM of HXR9,

respectively (Figure 4.22B).

Page 156: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

142

A)

B)

Figure 4.23 RT-PCR analysis of p53 and p21 expression in response to HXR9 treatment. RNA extracted from

cells after a two-hour treatment with 15μM and 30μM HXR9, or 30μM CXR9. The expression of p53 and p21

genes are given as ratio to the expression of the housekeeping gene β-actin (×10 000). Graphs show the mean of

three independent experiments and error bars show the SEM. **p < 0.01, ***p < 0.001 and ****p < 0.0001 with

respect to untreated cells.

Page 157: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

143

4.2.21 HXR9-induces AML cell death through a p53-independent pathway

To further investigate the role of p53 in HXR9-mediated cell killing, the activity of p53

protein was blocked by PFT-α, and the cytotoxicity of HXR9 was measured by LDH assay.

Blocking p53 protein did not affect the sensitivity of AML cells to HXR9 (Figure 4.23).

A)

B)

Figure 4.24 The effect of blocking p53 protein on HXR9 cytotoxicity. Cells were pre-treated with or without

40μM PFT-α for one hour, and then for two hours with a titration of HXR9 prepared in media supplemented with

or without 40μM PFT-α, red and blue curves, respectively. Graphs show the mean of three independent

experiments for (A) K562 and (B) HL-60 cells, and error bars show the SEM.

Page 158: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

144

4.3 Discussion

HXR9 has previously been shown to induce cell death in a range of solid malignancies,

and here it is demonstrated that HXR9 also induces AML cell death. The efficacy of HXR9

could be improved by better understanding its mechanism of action, allowing synergistic

therapies to be identified.

Antagonism of the HOX/PBX interaction resulted in a remarkable up-regulation in the

expression of c-FOS. The induction of c-FOS expression by HXR9 was previously documented

to be responsible, at least partially, for triggering cell death in human melanoma, renal, lung,

ovarian, breast and prostate cancer cell lines (Morgan, Pirard et al. 2007; Shears, Plowright et

al. 2008; Plowright, Harrington et al. 2009; Morgan, Plowright et al. 2010; Morgan, Boxall et

al. 2012; Morgan, Boxall et al. 2014), and knocking down c-FOS expression partially rescued

melanoma and prostate cancer cell lines from HXR9-induced apoptosis (Morgan, Pirard et al.

2007; Morgan, Boxall et al. 2014). c-FOS is a component of the AP-1 transcription activating

complex, and upon dimerisation with its binding partner JUN, regulates cell proliferation and

cell cycle progression (Durchdewald, Angel et al. 2009). It has been shown to be a proto-

oncogene in a variety of human tumours including those of the bone, brain and skin (Gamberi,

Benassi et al. 1998; Silvestre, Gil et al. 2010; Guinea-Viniegra, Zenz et al. 2012). However, a

reduction in c-FOS expression in epithelial ovarian carcinoma was found to be an independent

marker of shorter progression-free and OS (Mahner, Baasch et al. 2008). In addition, c-FOS

over-expression has a pro-apoptotic function in vitro, and caused a delay in tumour growth and

metastasis of the ovarian cancer OvCa cells in vivo by dysregulating adhesion proteins

(Oliveira-Ferrer, Rossler et al. 2014). It was also found to be involved in the induction of the

human ovarian carcinoma cell line A2780 in response to fenrtinide treatment (Appierto, Villani

et al. 2003). Moreover, c-FOS exerted a pro-apoptotic function by repressing the anti-apoptotic

gene cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein (L) (c-FLIP (L))

Page 159: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

145

which resulted in sensitising prostate cells to tumour necrosis-related apoptosis-inducing ligand

(TRAIL) (Zhang, Zhang et al. 2007). It also induced human hepatoma Huh7 apoptosis by

mediating c-MYC expression (Kalra and Kumar 2004). c-Fos over-expression resulted in cell

cycle arrest by up-regulating the expression of the nuclear protein CHOP and the cell cycle

inhibitors p16INK4A and p57KIP2; suppressing rat sarcoma (Ras)-mediated malignancy that

ultimately led to apoptosis of murine epithelial hepatocytes (Mikula, Gotzmann et al. 2003). Its

prolonged expression also suppressed cell cycle entry of murine BM cells (Okada, Fukuda et

al. 1999). Furthermore, c-Fos enhanced hepatocyte apoptosis by directly up-regulating the

major isoform of bim gene BimEL (Kitamura, Ogawa et al. 2003).

Externalisation of PS was seen in all tested AML cell lines after HXR9 treatment. This

finding was consistent with previous studies in different cancer cell lines upon HXR9 treatment

(Morgan, Pirard et al. 2007; Shears, Plowright et al. 2008; Plowright, Harrington et al. 2009;

Daniels, Neacato et al. 2010; Morgan, Plowright et al. 2010; Morgan, Boxall et al. 2012;

Morgan, Boxall et al. 2014). In viable cells, PS is localised on the inner leaflet of the cell surface

and its transfer to the outer leaflet is thought to be an engulfment signal by macrophages (Fink

and Cookson 2005). It is well-known that the exposure of PS to the outer leaflet of the cell

surface is a distinctive feature for apoptosis and results from the activation of caspase-3

(Mandal, Moitra et al. 2002; Bonomini, Dottori et al. 2004). Therefore, the binding-ability of

annexin V to PS was used specifically to detect and quantify apoptotic cells. However, the outer

leaflet exposure of PS before the damage of the plasma membrane was reported in thymocytes

at the prelethal stage of necrosis, oncosis (Waring, Lambert et al. 1999). In addition, oncotic

L1210 cells, a mouse leukaemia cell line, showed annexin V+ /PI- phenotype after serum amine

oxidase exposure (Bonneau and Poulin 2000), and exposure of Jurkat cells to different oncotic

stimuli led to externalisation of PS, annexin V+ /7-ADD-, to the outer leaflet before the loss of

cell membrane integrity (Lecoeur, Prevost et al. 2001; Lecoeur, de Oliveira-Pinto et al. 2002).

Page 160: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

146

Likewise, caspase-8 deficient and Bcl-2 over-expressing Jurkat cells exhibited PS exposure

during the oncotic stage (Krysko, de Ridder et al. 2004). H2O2 also was reported to cause PS

externalisation in a cypD-dependent pathway, i.e. mitochondrial necrosis (Jobe, Wilson et al.

2008). Therefore, externalisation of PS does not actually differentiate between apoptosis and

necrosis (Fink and Cookson 2005; Verheij 2008; Blankenberg and Strauss 2012).

The hallmark of apoptosis is caspase activation, and this was not found to be necessary

in HXR9-induced cell death. These data are consistent with the likely mechanism of HXR9-

induced cell death in the renal cancer cell lines Caki-2 and 769-P, in malignant B cell lines and

some prostate cell lines (Shears, Plowright et al. 2008; Daniels, Neacato et al. 2010; Morgan,

Boxall et al. 2014). However, the general inhibition of caspases decreased the cytotoxicity of

HXR9 on the tested renal, ovarian, breast and prostate cancer cell lines, although this decrease

did not completely abrogate the effect of HXR9 (Shears, Plowright et al. 2008; Morgan,

Plowright et al. 2010; Morgan, Boxall et al. 2014). This may be due to the presence of other

mediators of HXR9-induced cell death. The results of this study support this. For example,

HXR9 treatment of AML cells did not change the expression of PARP1 at the RNA or protein

level. Nuclear fragmentation of apoptotic cells results from the cleaving of the cytoskeletal

protein lamin by caspase-3 or -6 (Kagawa, Go et al. 2001; Ruchaud, Korfali et al. 2002), which

might explain the absence of nuclear fragmentation in HXR9 treated AML cell lines. In

addition, the expression of the anti-apoptotic genes Bcl-2 and XIAP that were known as targets

of active caspases (Jin and El-Deiry 2005; Vermeulen, Van Bockstaele et al. 2005) were not

affected by HXR9 treatment.

The findings of this study suggest that mitochondrial apoptosis is not required for

HXR9-mediated cell death. For example, an unchanged ratio between the anti-apoptotic Bcl-2

and the pro-apoptotic Bax and Bak1 may reflect the intactness of mitochondrial membranes,

since the presence of excess pro-apoptotic members of the Bcl-2 family enhances mitochondrial

Page 161: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

147

permeabilization (Martin 2010). Furthermore, the mitochondrial release of cyt C would

ultimately lead to caspase activation. In addition, the cleavage and activation of Bid that

mediates mitochondrial apoptosis depends on the activation of caspase-8, which was not

required for HXR9 cytotoxicity. Likewise, HXR9 seemed to induce cell death in a Bax-

independent process, since Bax-dependent apoptosis involves either caspase-9 or p53 activity,

neither of which was needed for HXR9 mediated-cell killing (Janssen, Pohlmann et al. 2007;

Gogada, Prabhu et al. 2011; Meng, Zhang et al. 2012).

The absence of apoptotic-hallmarks activation, including caspase activation, Bcl-2

family involvement, chromatin condensation and nuclear fragmentation suggest that HXR9

induces AML cell death through a necrotic pathway.

EDTA is a general cation-chelating agent, despite being widely used as an extracellular

Ca2+ chelator (Dong, Li et al. 2005; Allen, Gemel et al. 2011; de Araújo Leite and Marques-

Santos 2012). For example, EDTA can form complexes with cations such as Na+, Mg+, Fe2+,

Cu2+, Zn2+ that can be easily removed from the extracellular environment (Flora and Pachauri

2010). Interestingly, the cell penetrating peptide (CPP) of HXR9 is R9 that is a highly cationic

sequence (Mitchell, Steinman et al. 2000; Sarko, Beijer et al. 2010; Regberg, Srimanee et al.

2012; Bechara and Sagan 2013). In addition, the binding of annexin V to PS is a Ca2+ dependent

reaction (Vermes, Haanen et al. 1995). Therefore, the abrogation of HXR9 effect by EDTA

might be due to the chelating of HXR9 and the absence of Ca2+ in annexin V/7-AAD assay.

CypD knockout studies have demonstrated its crucial role in mitochondrial necrosis by

Ca2+ overload and oxidative stress, but is not required for apoptosis (Li, Johnson et al. 2004;

Baines, Kaiser et al. 2005; Nakagawa, Shimizu et al. 2005; Schinzel, Takeuchi et al. 2005). In

addition, CypD is involved in Ca2+-independent, RIP3- and p53-dependent mPTP necrosis

(Lerch, Halangk et al. 2013; Tian, Xu et al. 2013; Pei, Shang et al. 2014). Accordingly, HXR9

Page 162: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

148

might mediate necrosis of AML in an mPTP-independent pathway, or at least, in a CypD-

independent mechanism.

In this study it was shown that HXR9 led to cell death through a necroptotic pathway,

at least in part, through RIP1 activity in HL-60 cells, whilst K562 cell death was RIP1-

independent. It is possible though that the effect of blocking RIP1 HXR9-induced HL-60 cell

death could reflect a sub-effective dose of the RIP1 inhibitor, as Nec-1 showed some toxic

effects at higher doses. The latter explanation is consistent with the fact that Nec-1 partially

protected HL-60 cells from HXR9. Alternatively, other mediators in addition to RIP1 may be

required in HXR9-induced cell death. It is noteworthy that a RIP1-independent cell death

pathway does not rule out a potential role for necroptosis since it was reported that viral

infection resulted in RIP3-dependent but RIP1-independent necroptosis through TNF mediated

necroptosis (Zhang, Shao et al. 2009; Upton, Kaiser et al. 2010). Therefore, the role of RIP3 in

HXR9-induced K562 cell death requires further investigation.

The presence of ATP is crucial for caspase-dependent apoptosis and PARP1 hyper-

activation. In caspase-dependent apoptosis, ATP is involved in the formation of the apoptosome

complex that mediates apoptotic signalling by cleaving and activating executioner caspases

(Jochen, Richter et al. 2002). It was reported that ATP depletion by fructose in primary rat and

mouse hepatocytes abrogated cyt C release and activation of executioner caspases, and DNA

fragmentation by TNF-mediated apoptosis, and that these events were restored after ATP

repletion (Latta, Kunstle et al. 2000; Jochen, Richter et al. 2002). In addition, the presence of

ATP is essential for chromatin condensation and nuclear fragmentation (Tsujimoto 1997). ATP

depletion by fructose prevented caspase activation and subsequently shifted cell death toward

necrosis in the leukaemic Jurkat and K562 cells (Leist, Single et al. 1999; Verrax, Dejeans et

al. 2011). Likewise, PARP1 involves ATP in the transfer of poly (ADP-ribose) from NAD+

(Devalaraja-Narashimha and Padanilam 2009; Ethier, Tardif et al. 2012). Taking together, the

Page 163: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

149

depletion of ATP elicits the role of caspases and PARP1 hyper-activation in the mechanism of

HXR9-induced cell death of AML cells. It is well-known that fructose at high concentrations

traps intracellular ATP and maintains a sufficient cytoplasmic concentration to prevent cell

necrosis (Latta, Künstle et al. 2000; Latta, Künstle et al. 2007; Speicher, Köhler et al. 2012).

The results of this study revealed that ATP depletion by a high concentration of fructose did

not prevent HXR9-induced necrotic death of the tested AML cells, which also supports a

caspase- and PARP1- independent mechanism for HXR9-induced cell death.

Despite its well-known crucial role in TNF-induced necrosis, PARP1 was dispensable

in TNF-mediating necroptosis (Xu, Huang et al. 2006). Accordingly, it can be concluded that

TNF activates necroptosis and necrosis in distinct pathways, although PARP1 was also shown

to be a downstream effector of the necrosome in TRAIL-induced necroptosis (Jouan-Lanhouet,

Arshad et al. 2012). In this study, it was reported that HXR9-induced necrosis in a PARP1-

independent mechanism. While the JNK pathway is involved in PARP1-induced necrosis,

HXR9-induces necrosis of AML cells in JNK-independent pathway (Degterev, Huang et al.

2005; Xu, Huang et al. 2006; Jog, Dinnall et al. 2009; Ethier, Tardif et al. 2012; Sosna, Voigt

et al. 2014). Since PARP1 is a crucial player in mPTP, the results of PARP1 activation in this

study support a mitochondria-independent pathway for HXR9-induced necrosis (Abramov and

Duchen 2008).

The MAPK family of protein kinases consists of four members; ERK, p38, JNK and the

big mitogen activated protein kinase. MAPK proteins regulate different cellular pathways

including cell survival, proliferation and cell death (Chen and Sommer 2009). In this study

inhibition of MEK/ERK and p38 did not affect HXR9-induced cell necrosis. JNK inhibition

also did not affect the efficacy of HXR9 in HL-60, although it increased the cytotoxicity of

HXR9 for K562. This effect of JNK inhibition might be due to the expression of the BCR-ABL

oncoprotein in K562 cells that is reported to induce leukaemogenesis through activation of JNK

Page 164: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

150

signalling pathway (Hess, Pihan et al. 2002; Hantschel, Warsch et al. 2012; Chen, Gallipoli et

al. 2013). In addition, the JNK pathway is reported to be an inducer of clonal evolution of

Fanconi anaemia to AML (Li, Sejas et al. 2007) and sustained activation of the JNK pathway

with AKT/FOXO signalling maintains AML cells in an undifferentiated state (Sykes, Lane et

al. 2011). In FL15.12 cells, a haemopoietic pro-B cell line, IL-3 promoted cell survival by

phosphorylating BAD through JNK signalling (Yu, Minemoto et al. 2004). More recently, it

was reported that co-inhibition of the TNF-JNK pathway increased the sensitivity of primary

human AML cells in vitro to NF-ĸB inhibitors (Volk, Li et al. 2014), and that the JNK pathway

promoted proliferation and metastasis in several cancers including those of the liver and skin

(Chen, Nomura et al. 2001; Han, Moon et al. 2009; Bettermann, Vucur et al. 2010; Ke, Harris

et al. 2010; Ebelt, Cantrell et al. 2013).

NOXs are a family of proteins that are considered to be a primary resource of ROS in

living cells and regulated by intracellular Ca2+ signalling (Yu, Kim et al. 2006; Morgan, Kim et

al. 2008; El Jamali, Valente et al. 2010; Hole, Darley et al. 2011; Jiang, Zhang et al. 2011;

Pandey, Gratton et al. 2011) and mediate necrotic cell death (Ouyang, Shi et al. 2012). DPI is

a widely used NOX inhibitor, though it is also a broad inhibitor of other flavoenzymes including

nitric oxide synthase and xanthine oxidase (Li, Ragheb et al. 2003; Park, Song et al. 2007). In

this study DPI increased the efficacy of HXR9 on the tested AML cells, suggesting that NOXs

has survival roles upon HXR9 treatment. In line with these findings, it was found that DPI

mediated the apoptosis of the pancreatic cancer cell line PANC-1 by blocking the survival

pathway NOX4/ROS/AKT (Mochizuki, Furuta et al. 2006). DPI was also shown to mediate

p53-dependent apoptosis (Kim, Song et al. 2007; Park, Song et al. 2007). In addition, NOXs

were found to be regulated by PKC and CaM kinases that were identified in this study as

mediators of cell resistance to HXR9 (Inoguchi, Sonta et al. 2003; Cai, Torreggiani et al. 2010;

Pandey, Gratton et al. 2011).

Page 165: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

151

Calpains are a family of 14 cysteine proteases that are regulated by Ca2+. The most

extensively studied members of this family are μ-calpain or calpain I that is present in the

cytosol and is stimulated by micro-molar Ca2+ concentrations, and m-calpain or calpain II that

is present in the cell membrane and is activated by mille-molar concentration of Ca2+ (Kar,

Chakraborti et al. 2009; Szydlowska and Tymianski 2010). In this study, the potential role of

μ-calpain was studied because it was reported a mediator of Ca2+-dependent necrosis

(Mathiasen, Sergeev et al. 2002; Zhivotovsky and Orrenius 2011; Ouyang, Shi et al. 2012).

However, HXR9-induced necrosis of AML cells in a μ-calpain-independent manner. This

might be explained by the caspase-independent pathway of HXR9-mediated cell death, as μ-

calpain was shown to cleave and activate pro-caspases-3/9 in human lung adenocarcinoma cells

(Liu, Xing et al. 2009). This finding is also consistent with the suggestion that HXR9-induced

cell death does not require mitochondria, since μ-calpain was found to cause cleavage and

activation of the pro-apoptotic members of Bcl-2 family Bax and Bid (Chen, He et al. 2001;

Mandic, Viktorsson et al. 2002; Toyota, Yanase et al. 2003; Sanchez-Gomez, Alberdi et al.

2011). It has also been shown that μ-calpain is a downstream mediator of PARP1 in the release

of AIF that ultimately leads to mitochondria-dependent necrosis (Cao, Xing et al. 2007; Liu,

Xing et al. 2009; Vosler, Sun et al. 2009; Storr, Carragher et al. 2011).

CaM is a small 17KDa intracellular protein that is considered to be the main regulator

and Ca2+-binding protein in all eukaryotic cells (Wang, Li et al. 2010; Berchtold and Villalobo

2014). In this study, the CaM antagonist W-7 enhanced HXR9-induced necrosis. CaM exerts

surviving effects by positively regulating pro-survival and proliferative proteins, and inhibition

of CaM alone or in combination therapy promotes cell death in many cancer models. CaM

inhibition sensitised human lung cancer H1299 cells to TRAIL-induced apoptosis by enhancing

death-inducing signalling complex (DISC)-triggered apoptosis and/or preventing the

phosphorylation of the survival protein AKT, leading to a reduction in expression of the anti-

Page 166: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

152

apoptotic protein c-FLIP (Hwang, Min et al. 2009). CaM also regulates the PI3K survival

pathway (Lasbury, Durant et al. 2009), and inhibition of CaM attenuated the ERK1/2-dependent

survival pathway and up-regulated the Bax- and caspase-3-dependent pathway (Bartolomé, de

las Cuevas et al. 2007). CaM can also maintain NF-ĸB activity by phosphorylating and

degrading its binding inhibitor IĸB (Shumway, Berchtold et al. 2002; Tano and Vazquez 2011).

In vitro and in vivo studies of cholangiocarcinoma showed that the inhibition of CaM prevented

the phosphorylation of AKT and c-FLIP recruitment to DISC, thereby increasing the

recruitment and activation of caspase-2, caspase-8 and caspase-9. This in turn leads to cleavage

and activation of caspase-3, releasing cyt C and triggering apoptosis (Ahn, Pan et al. 2003;

Pawar, Ma et al. 2009; Jing, Yuan et al. 2011; Jing, Yuan et al. 2012).

PKCs are a family of protein kinases that regulate several cellular functions including

cell cycle progression, cellular proliferation, differentiation, survival and apoptosis. The PKC

family is composed of 12 members and is classified into four classes according to structural

and regulatory characteristics including the conventional/classical PKCs, the novel PCKs, the

atypical PKCs and PKC-related kinases. Of these, only the conventional PKCs are Ca2+-

dependent (Bosco, Melloni et al. 2011; Shen, Kim et al. 2012; Parker, Justilien et al. 2014). Of

the 12 PKCs, 7 are anti-apoptotic proteins including PKCα, PKCβ, PKCι, PKCγ, PKCζ, PKCε

(Reyland 2007). In this study, the general inhibitor of PKC Ro31-8220 increased the sensitivity

of the tested AML cell lines to HXR9. This supports the findings that NOX inhibition also

sensitized cells to HXR9, as PKCα and PKCβ are positive regulators for NOXs (Shiraki, Oyama

et al. 2012; Paneni, Beckman et al. 2013). Up-regulation of PKCα and PKCι were documented

in breast cancer stem cells, human glioma (Desai, Pillai et al. 2011; Tam, Lu et al. 2013). In

addition, PKC was found to increase the stability of the anti-apoptotic protein c-FLIP (Kaunisto,

Kochin et al. 2009). PKCι was documented to cause cell cycle progression by phosphorylating

CdK7 and CdK2 (Win and Acevedo-Duncan 2009; Pillai, Desai et al. 2011), and in the cancer

Page 167: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

153

prostate cell line LNCaP, PKC promoted cell survival through the

PI3K/PKD1/PKC/ERK1/2/NF-ҡB signalling pathway (Chen, Giridhary et al. 2011). It was also

reported that PKC enhanced cell survival and invasion by inducing the expression of the growth

factor bone morphogenic protein-4, components of the Ras signalling pathway, and matrix

metalloproteinase 2 and 9 (Hwang, Yun et al. 2011; Shimizu, Kayamori et al. 2012; Lonic,

Powell et al. 2013). During TRAIL-mediated apoptosis, PKC activation was reported to prevent

DISC formation and consequently inhibited the apoptosis of HeLa cells (Harper, Hughes et al.

2003). In ALL, PKC inhibition sensitised cells to Ca2+-mobilizers (Zhu, Fang et al. 1999).

Furthermore, inhibition of PKC resulted in a dephosphorylation and activation of Bad

concurrently with a down-regulation of Bcl-2 and ultimately to mitochondrial dysfunction and

caspase-dependent apoptosis (Win and Acevedo-Duncan 2009; Desai, Pillai et al. 2011;

Whang, Jo et al. 2013).

The concurrent inhibition of CaM and PKCs alongside with HXR9 treatment resulted

in AML cell death at very low IC50 in compared to HXR9-alone. This might result from the

inhibition of both cell survival pathways such as PI3K/AKT, and anti-apoptotic proteins such

as c-FLIP and anti-apoptotic members of Bcl-2 family.

HO-1 protein is an important cytoprotective enzyme that has anti-apoptotic properties

and promotes cancer cell survival, angiogenesis and drug-resistance (Jozkowicz, Was et al.

2007; Lee, Chun et al. 2011; Park, Jung et al. 2011; Lee, Yang et al. 2012). It is constitutively

expressed in primary human AML samples, cell lines and multiple myeloma cell lines (Barrera,

Rushworth et al. 2012; Herrmann, Kneidinger et al. 2012). Inhibiting HO-1 increased the

sensitivity of AML cells to HXR9. This is consistent with a number of other findings. For

example, c-FOS is suppressed by HO-1 (Sun 2008), and PKC promotes cell survival through

two different pathways that both involve HO-1, for example, through PKC/p38/Nrf2/HO-1 or

PKC/ROS/PI3K/AKT/Nrf2/HO-1 (Tsoyi, Jang et al. 2011; Lee, Yang et al. 2012; Nguyen, Kim

Page 168: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

154

et al. 2013). In addition, NOX4 was reported to induce HO-1 expression through the ROS/Nrf2

pathway (Schroder, Zhang et al. 2012). HO-1 was also found to reduce the expression of p21

that was found to play a pro-necrotic role following HXR9 treatment in this study (Was,

Sokolowska et al. 2011). In addition, HO-1 prevents TNF-induced apoptosis in both c-FLIP

and NF-ҡB knockdown AML cells (Rushworth and MacEwan 2008; Rushworth, Zaitseva et

al. 2010). HO-1 also protected AML cells from the front-line chemotherapeutic drugs DNR and

cytarabine and its inhibition enhanced the efficacy of both drugs (Heasman, Zaitseva et al.

2011).

p53 is one of the most extensively studied cell death proteins, and stimulates c-FOS

transcription in cells undergoing p53-dependent apoptosis (Elkeles, Juven-Gershon et al. 1999).

The results of this study showed that HXR9 did not change the expression of p53 at the RNA

or at the protein level. These results were consistent with the mechanism of HXR9 killing of

breast cancer cells that is p53-independent (Morgan, Boxall et al. 2012). In addition, p53 protein

directly interacts with the anti-apoptotic members of the Bcl-2 family, Bcl-2 and Bcl-XL,

resulting in the release of the pro-apoptotic members Bak and Bax, leading to caspase-

dependent apoptosis (Mihara, Erster et al. 2003; Vaseva and Moll 2009; Chakraborty,

Mazumdar et al. 2014; Chi 2014). p53 also up-regulates the transcription of, and physically

interacts with Bak, Bax and Bad resulting in mitochondrial apoptosis (Chipuk, Maurer et al.

2003; Chipuk, Kuwana et al. 2004; Leu, Dumont et al. 2004; Jiang, Du et al. 2006; Pietsch, Leu

et al. 2007; Wenzel, Wunderlich et al. 2012; Chi 2014; Lee, Lee et al. 2014; Saha, Bhattacharjee

et al. 2014). However, none of the Bcl-2 family members or Apaf1, a mediator of p53-dependent

apoptosis, changed in expression during HXR9-mediated AML cell necrosis. Furthermore,

calpain, an inducer of p53 apoptosis, was not required for HXR9-induced AML cell death (Del

Bello, Moretti et al. 2007; Bukowska, Lendeckel et al. 2012; Woo, Xue et al. 2012). More

recently, the role of p53 in cypD- and PARP1-dependent programmed necrosis was identified

Page 169: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

155

(Vaseva, Marchenko et al. 2012; Montero, Dutta et al. 2013; Pei, Shang et al. 2014). However,

neither cypD nor PARP1 were involved in HXR9-induced AML necrosis. Taken together, these

findings suggest that HXR9-indcues AML necrosis in a p53-independent manner.

p21 is a member of the family of cyclin kinase inhibitors that include p27 and p57

(Kwon, Jovanovic et al. 2003; Masgras, Carrera et al. 2012). It was originally recognised as a

main regulator of the progression of the cell cycle (Harper, Adami et al. 1993). However, p21

also regulates a number of other cellular functions, including apoptosis, cell proliferation and

autophagy (Asada, Yamada et al. 1999; Dong, Li et al. 2005; Luo, Zou et al. 2011). Although

it was thought that the regulation of p21 was totally dependent on p53, p21 can in fact be

regulated by several transcription factors in a p53-independent manner (Liu and Huang 2006;

Lafarga, Cuadrado et al. 2009; Shin, Kim et al. 2011; Han, Kim et al. 2012; Masgras, Carrera

et al. 2012). For example, the early growth response-1 protein can induce the transcription of

p21 independently of p53 (Choi, Kim et al. 2008). The early growth response-1 gene is induced

in lung cancer cells after HXR9 treatment (Plowright, Harrington et al. 2009). Interestingly,

p21 has been shown to be involved in p53-independent necrotic cell death (Ussat, Werner et al.

2002; Kwon, Jovanovic et al. 2003). The results of this study show that the induction of p21 in

response to HXR9 was p53-independent.

Page 170: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

156

4.3.1 Summary of chapter

In the summary, HXR9-mediated cell death in AML cells was characterised by the

absence of the well-known features of apoptosis including caspase activation, the up-regulation

of Bcl-2 family-members, chromatin condensation and nuclear fragmentation. The mechanism

was also p53-independent. However, the mechanism of cell death involved, at least in part, c-

FOS and p21 over-expression. In addition, RIP1 was partially involved in the cell death of HL-

60, but not K562 cells, and the JNK pathway exerted pro-survival signalling in the HXR9-

mediated killing of K562 cells. Likewise, CaM, PKC and HO-1 exerted resistance to HXR9-

mediated cell death (Table 4.1).

Table 4.1 The mechanism of HXR9 cytotoxicity on AML cells.

Causes up-regulation of Involves Independent of Induced by inhibition of

c-FOS

p21

RIP1 (in HL-60) Caspases

Bcl-2-family members

Nuclear fragmentation

CypD

ATP

PARP-1 activation

MEK/ERK pathway

p38 pathway

µ-calpain

p53

JNK pathway (in K562)

NOX

CaM

PKC

HO-1

Page 171: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

157

Chapter 5 In vivo cytotoxicity of HXR9

Page 172: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

158

5.1 Introduction

In vitro cytotoxicity of HXR9 on the tested AML cell lines was proven. In addition, this

study data revealed that the concurrent inhibition of CaM and PKC dramatically increased the

sensitivity of the tested AML cell lines to HXR9. Likewise, in vivo efficacy of HXR9 to

eradicate solid tumours of different origins including melanoma, lung, ovarian, breast and

prostate was demonstrated (Morgan, Pirard et al. 2007; Plowright, Harrington et al. 2009;

Morgan, Plowright et al. 2010; Morgan, Boxall et al. 2012; Morgan, Boxall et al. 2014). In the

previous studies, HXR9 was administrated via different routes including intravenously (I.V.),

I.T. or intraperitoneally. In addition, HXR9 did not show adverse side effects on PB and BM

after 10 days of daily I.V. administration at 15mg/kg/day (Morgan, Pirard et al. 2007).

However, the in vivo efficacy of HXR9 on AML cell lines and the side effect of I.V.

administration of high doses of HXR9 have not been studied, yet.

To determine the cytotoxicity of high doses of HXR9 on normal PB, a mouse strain with

the whole blood cells was used, C57BL/6. Due to the intact immunity of C57BL/6 mice, the

mouse AML cell line C1498 that is originated from C57BL/6 mouse strain, a syngeneic tumour

model, labelled with GFP (C1498-GFP) was used to evade the immune system and engraft in

mice. In addition, SCID mice were used to engraft the human AML cell line K562.

Page 173: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

159

5.1.1 Aims of chapter 5

To assess the efficacy of HXR9 against AML disease in vivo either alone or in

combination with the simultaneous inhibition of CaM and PKC, and to assess the adverse effect

of high doses of HXR9 on normal PB.

The main objectives were:

1. To determine the cytotoxicity of HXR9 either alone or with the simultaneous

inhibition of CaM and PKC on the C1498-GFP cell line in vitro.

2. To establish an AML mouse model by engrafting C1498-GFP or K562 cells in either

C57BL/6 or SCID mice, respectively.

3. To treat these models with high doses of HXR9.

Page 174: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

160

5.2 Results

5.2.1 HXR9 is cytotoxic for C1498-GFP cells

To test whether HXR9 induces the cell death of the murine AML cell line C1498-GFP,

C1498-GFP cells were treated with a series of HXR9 concentrations for two hours, and

cytotoxicity was determined by LDH assay using ELISA. HXR9 induced C1498-GFP cell death

(Figure 5.1) with an IC50 15.8μM ± 2.6 that determined using Calcusyn software.

Figure 5.1 LDH assay for the cytotoxicity of HXR9 on C1498-GFP cells. Cells were exposed to a titration of

HXR9 for two hours and the cytotoxicity of HXR9 was assessed by measuring LDH enzyme activity in cell-free

supernatants. The graph shows the mean of three independent repeats that had R2 ≥ 0.95 and error bars show the

SEM.

Page 175: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

161

5.2.2 CaM blocking enhances the cytotoxicity of HXR9

To assess whether the inhibition of CaM produces the same effect on the cytotoxicity

of HXR9 as its inhibition in human AML cell lines, CaM was blocked in the murine AML cell

line C1498-GFP cells using the W-7 inhibitor. As with human AML cells, blocking CaM

increased the sensitivity of C1498-GFP cells to HXR9. For example, co-treatment of C1498-

GFP cells with 10μM HXR9 and 50μM W-7 significantly decreased the percentage of surviving

cells from 72% ± 6.2 to 37% ± 3.3, **p < 0.01, compared to HXR9-only treated cells (Figure

5.2). In addition, co-treatment caused a dramatic decrease in the IC50 of HXR9 by two folds,

from 14.4μM ± 1.2 to 7.5μM ± 2.3 (Figure 5.2).

Page 176: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

162

IC50

Cell line/ condition HXR9 HXR9+50μM W-7

C1498-GFP 14.4μM ± 1.2 7.5μM ± 2.3

Figure 5.2 The effect of CaM inhibition on the cytotoxicity of HXR9 for C1498-GFP. Cells were pre-incubated

with or without 50μM W-7 for one hour and then with HXR9 for two hours, with or without 50μM W-7, red and

blue curves, respectively. The cytotoxicity of HXR9 was then measured by LDH assay in cell-free media. Co-

treatment with 50μM W-7 significantly increased the cytotoxicity of HXR9 on C1498-GFP cells. The graph shows

the mean of three independent experiments and error bars show the SEM. *p < 0.05, **p < 0.01 and ***p < 0.001

with respect to 50μM W-7 untreated cells. The table shows the difference in the IC50 between HXR9-only treated

cells and HXR9 + 50μM W-7 treated cells.

Page 177: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

163

5.2.3 Inhibition of PKC activity significantly sensitises cells to HXR9

To assess the effect of inhibition of PKC activity on the sensitivity of C1498-GFP cells

to HXR9, PKC activity was blocked using Ro31-8220 and the sensitivity to HXR9 was

measured by LDH. Ro31-8220 at 30μM dramatically enhanced cell killing by HXR9. For

example, co-treatment of cells with 30μM Ro31-8220 resulted in a huge reduction, p < 0.0001,

in the percentage of surviving cells from 89% ± 3.5 to 58% ± 3.3, compared to cells treated

with 5μM HXR9 alone (Figure 5.3). Likewise, co-treatment with Ro31-8220 decreased the IC50

of HXR9 by twofold from 14.2μM ± 1.9 to 6.6μM ± 2.1 compared to HXR9-only treated cells

(Figure 5.3).

Page 178: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

164

IC50

Cell line/ condition HXR9 HXR9+30μM Ro31-8220

C1498-GFP 14.2μM ± 1.9 6.6μM ± 2.1

Figure 5.3 The impact of PKC inhibition on the sensitivity of C1498-GFP cells to HXR9. Cells were pre-treated

with or without 30μM Ro31-8220 for one hour and then with HXR9 for two hours, with or without 30μM Ro31-

8220, red and blue curves, respectively. The sensitivity of C1498-GFP cells to HXR9 was then assessed by LDH

assay using ELISA. Results show that blocking PKC dramatically increases the sensitivity of C1498-GFP cells to

HXR9. Graphs show the mean of three independent experiments and error bars show the SEM. *p < 0.05 and

****p < 0.0001 with respect to 30μM Ro31-8220 untreated cells. The table shows the difference in the IC50

between HXR9-only treated cells and HXR9 + 30μM Ro31-8220 treated cells.

Page 179: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

165

5.2.4 Concurrent inhibition of CaM and PKC significantly enhances the efficacy of

HXR9

To assess the impact of the concurrent inhibition of CaM and PKC on the sensitivity of

C1498-GFP cells to HXR9, both kinases were concurrently blocked and cells were treated with

increasing concentrations of HXR9 and the cytotoxicity of HXR9 was analysed using the LDH

assay. The simultaneous inhibition of both proteins remarkably sensitised C1498-GFP cells to

HXR9. For example, at 5μM HXR9 treatment, the inhibition of CaM and PKC hugely reduced

the percentage of surviving cells from 83% ± 3.9 to 30.4% ± 4.4 (p < 0.0001), compared to

HXR9-only treated cells (Figure 5.4). Likewise, the concurrent blocking of CaM and PKC

remarkably decreased the IC50 of HXR9 by approximately tenfold, from 14.2μM ± 1.3 to 1.5μM

± 1.4 (Figure 5.4).

Page 180: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

166

IC50

Cell line/ condition HXR9 HXR9+(30μM Ro31-8220+50μM

W-7)

C1498-GFP 14.2μM ± 1.3 1.5μM ± 1.4

Figure 5.4 The effect of CaM and PKC simultaneous inhibition on the efficacy of HXR9. Cells were pre-treated

with or without the combination of the inhibitors, 50μM W-7 and 30μM Ro31-8220, for one hour and then with

HXR9 for two hours, with or without both inhibitors (red and blue curves, respectively). The sensitivity of C1498-

GFP cells to HXR9 was then estimated by measuring LDH enzyme activity in cell-free media using ELISA. The

simultaneous blocking of CaM and PKC remarkably enhanced the efficacy of HXR9. The graph shows the mean

of three independent experiments and error bars show the SEM. *p < 0.05, **p < 0.01 and ****p < 0.0001 with

respect to W-7 and Ro31-8220 untreated cells. The table shows the difference in the IC50 between HXR9-only

treated cells and HXR9 + (50μM W-7 + 30μM Ro31-8220) treated cells.

Page 181: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

167

5.2.5 Efficient expression of GFP in C1498 cells

The murine AML cell line C1498 was previously transduced to express GFP in order to

monitor tumourgenicity (Zhang, Gajewski et al. 2009). The expression of GFP was confirmed

by fluorescence microscopy, and flow cytometry, which revealed that approximately 95% of

C1498 cells express GFP (Figure 5.5).

A) B)

Figure 5.5 Expression of GFP in C1498 cells. A) Detection of GFP expression, green fluorescence, by fluorescent

microscope (x10). B) Analysis of GFP expression in C1498 by flow cytometry. The percentage of C1498 was

calculated as the percentage of cells that express GFP.

Page 182: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

168

5.2.6 Establishment a systemic C1498-GFP model in C57BL/6 mice

To establish a murine AML model to assess the cytotoxicity of HXR9 in vivo, C57BL/6

mice (n=10) were inoculated I.V. with 1x106 C1498-GFP cells on day 0 (Table 5.1). The

leukaemia burden was monitored in PB by harvesting blood from random mice on days 3, 7,

13, 17 and 20 (Appendix 9), and estimated by calculating the percentage of GFP+ cells within

the peripheral white blood cells (Figure 5.6). GFP+ cells were found to have engrafted in 2 of

10. This engraftment rate was too low to study the effect of HXR9 in vivo.

A) B)

Figure 5.6 FACS analysis of PB from C57BL/6 mice 13 days after injection of C1498-GFP cells. C1498-GFP

cells were injected in C57BL/6 mice on day 0 and PB was collected from two mice. Peripheral blood mononuclear

cells were analysed by FACS. A) A plot shows no engraftment of C1498-GFP cells. B) A plot shows engraftment

of C1498-GFP cells.

Page 183: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

169

Table 5.1 A summary of injection of C1498-GFP I.V. in C57BL/6 mice.

Mouse strain C57BL/6

Number of mice 10

Cell line C1498-GFP

Cell inoculation number 1x106 cell/mouse

Inoculation site I.V.

Harvested organ PB

Experiment length (days) 20

Engraftment rate 2 out of 10

(1 in day 13 and 1 in day 17)

Page 184: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

170

5.2.7 Establishment a systemic C1498-GFP model in C57BL/6 and nude mice

As the first xenograft model did not work, two further mouse strains, C57BL/6 (n=10)

and nude mice (n=5), were inoculated with 1x106 C1498-GFP cells I.V. on day 0 (Table 5.2).

In this experiment, the leukaemia burden was monitored by calculating the percentage of GFP+

cells within the whole population of white blood cells in many sites including PB, BM, liver,

kidney and spleen on days 8, 15, 22, 25, 28 and 35 (Figure 5.7 and appendix 10). Results showed

that the first engraftment site was the liver in both strains. However, cells were engrafted in 3

and 2 mice in C57BL/6 and nude strains, respectively, and two C57BL/6 mice died in the day

22 after injection. The low engraftment rate, the late stage engraftment of cells in PB and the

death of two mice suggested that this xenograft model was not appropriate for analysing the

efficacy of HXR9 in vivo.

Page 185: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

171

A) C57BL/6 mice

Page 186: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

172

B) Nude mice

Figure 5.7 FACS analysis of C1498-GFP cells shows engraftment in several organs of C57BL/6 and nude mice.

C1498-GFP cells were inoculated I.V. in both strains on day 0. Several sites including PB, BM, liver, kidney and

spleen were harvested and white blood cells were purified as described in methods. Then, the engraftment of

C1498-GFP cells was analysed by FACS. A) A plot shows C1498-GFP cells engraftment in organs from a

C57BL/6 mouse, B) a nude mouse.

Page 187: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

173

Table 5.2 A summary of injections of C1498-GFP I.V. in C57BL/6 and nude mice.

Mouse strain C57BL/6 Nude

Number of mice 10 5

Cell line C1498-GFP

Cell inoculation number 1x106 cell/mouse

Inoculation site I.V.

Harvested organs PB, BM, liver, kidney and spleen

1st engraftment site Liver

Experiment length (days) 28 35

Engraftment rate 3 out of 10

(2 and 1 in days 22 and 28,

respectively)

2 out of 5

(1 in day 22 and 1 in day 28)

Page 188: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

174

5.2.8 Development a C1498-GFP flank model in C57BL/6 mice

As the I.V. inoculation of C1498-GFP cells was not appropriate to establish it as an

AML model to study the efficacy of HXR9 in vivo, the flank xenograft attempt was done to

assess its efficacy to be as a model. C57BL/6 mice (n=5) were injected S.C. with 1x106 C1498-

GFP cells on day 0 (Table 5.3). The leukaemic tumours started to be measured on day 7 and all

mice had tumours by day 20. Therefore, the flank xenograft was used to conduct in vivo

experiments.

Table 5.3 A summary of injection of C1498-GFP S.C in C57BL/6 mice.

Mouse strain C57BL/6

Number of mice 5

Cell line C1498-GFP

Cell inoculation number 1x106 cell/mouse

Inoculation site S.C.

Experiment length (days) 20

Engraftment rate 100%

Page 189: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

175

5.2.9 HXR9 significantly extended the survival of C1498-GFP xenograft

Due to its successful engraftment, the flank xenograft of C1498-GFP cells in C57BL/6

mice was established in order to assess the effect of HXR9 in vivo. C1498-GFP cells were

implanted S.C. in the flank, 1x106 cell/mouse. When tumours became palpable, the mice were

divided into two cohorts of 9 mice each with one cohort treated with 50mg/kg HXR9 three

times/week I.T. and the other with PBS as a control. Comparison of tumour growth delay

between the different cohorts at each time point showed that there was no statistical difference

(p = 0.23) in tumour sizes although the mean tumour volume on day 12 was less than half that

of the control group (Figure 5.8A). OS data showed that HXR9-treated mice survived

significant longer than their counterpart (p = 0.036), 22% compared to 0% for HXR9 and PBS

treated mice, respectively (Figure 5.8B).

Page 190: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

176

A)

B)

Figure 5.8 HXR9 treatment effect on the growth of C1498-GFP xenograft in female C57BL/6 mice. Mice were

injected with 1x106 C1498-GFP cells S.C. in the flank. When xenografts became palpable, day 0, mice were treated

I.T. with 50mg/kg HXR9 (n=9) or PBS (n=9) three times/week. Mice were culled when tumour sizes reached ≥

1500 mm3. A) Mean changes in xenograft volumes following HXR9 or PBS treatment and error bars show SEM.

B) The corresponding survival curve for mice treated with HXR9 or PBS shown in A.

Page 191: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

177

5.2.10 Simultaneous inhibition of CaM and PKC did not sensitise C1498-GFP flank

tumour to HXR9

To assess whether the concurrent inhibition of CaM and PKC in vivo could enhance the

efficacy of HXR9 as it did in vitro, the same drug combination was used in the C1498-GFP

flank tumour. Flank tumours were initiated in C57BL/6 mice by S.C. administration of C1498-

GFP cells, 1x106cell/mouse. When tumours became measureable, mice were divided into four

groups. Two control groups were treated with either PBS or CaM and PKC inhibitors, 500μM

W-7 and 300μM Ro31-8220, respectively; the other groups were either treated with 50mg/kg

HXR9 alone or 50mg/kg HXR9 plus the inhibitors simultaneously for three times/week.

Tumour growth data demonstrated that there were no statistical variances among the different

groups at any time point. Although the mean tumour volume in HXR9 and the inhibitor treated

mice on days 21, 23 and 25 showed an entire absence of tumour, this data represented the

tumour volume of one mouse out of 10 mice (Figure 5.9A). Consequently, the OS of the treated

groups with HXR9 alone or HXR9 and the inhibitors were not statistically significant different

compared to the OS of the control groups (Figure 5.9B).

Page 192: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

178

A)

B)

Figure 5.9 Effect of HXR9 I.T. treatment on the growth of C1498-GFP flank xenograft in C57BL/6 mice. C1498-

GFP cells (1x106) were inoculated S.C. in the mice flanks. When tumours became palpable, mice were split into

four groups, two control groups were administrated PBS (n=9) and CaM and PKC inhibitors (n=10), 500μM W-7

and 300μM Ro31-8220, respectively; and the other two groups were administrated either 50mg/kg HXR9 alone

(n=10) or 50mg/kg HXR9 + (500μM W-7 and 300μM Ro31-8220) (n=10) I.T. for three times/week. When tumours

reached ≥ 1500 mm3, mice were excised. A) Changes in tumour volumes resulting from treatment with PBS,

500μM W-7 and 300μM Ro31-8220, 50mg/kg HXR9 or 50mg/kg HXR9 + (500μM W-7 and 300μM Ro31-8220)

and error bars show SEM. B) Corresponding survival curve for mouse groups shown in A.

Page 193: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

179

5.2.11 HXR9 did affect the tumour growth of K562 flank model

As an additional in vivo model, human AML cell line K562 was used in the SCID mice.

K562 cells were inoculated S.C. in the flank of SCID mice at 1x106 cell/mouse. When tumours

became measurable, the mice were split into two groups of 6 mice each. One group was injected

I.T. with 100mg/kg HXR9 three times/week, except on the 10 and 12 mice were treated with

300mg/kg and 200mg/kg HXR9, respectively. The control group was injected with PBS.

Tumour growth delay and median survival data revealed that there was no statistical difference

between the two groups (Figure 5.10). Of note, 300mg/kg and 200mg/kg HXR9 led to the death

of two mice each.

Page 194: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

180

A)

B)

Figure 5.10 Impact of HXR9 treatment on the K562 flank-xenograft growth in SCID mice. The xenograft was

initiated by S.C. injection of 1x106 K562 cells in the flank. When tumours became measureable, day 0, mice were

treated I.T. with HXR9 (n=6) or PBS (n=6) three times/week. HXR9-treated mice were treated with 100mg/kg

overall the experiment except on day 7 and 10 where they were treated with 300mg/kg and 200mg/kg respectively.

Mice were euthanized when tumour volumes reached ≥ 1500 mm3. A) Mean values of tumour volumes of HXR9-

treated or PBS-treated mice and error bars show SEM. B) OS of HXR9- and PBS-treated mice.

Page 195: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

181

5.3 Discussion

Leukaemic mouse models were established four decades ago after the development of

SCID mice. This model is characterised by the lack of active T and B lymphocytes (Bosma and

Carroll 1991). Subsequently, the non-obese diabetic with severe combined immunodeficiency

(NOD-SCID) model was developed; the mice lack functional T and B lymphocytes and have

fewer NKs (Shultz, Schweitzer et al. 1995). Another model is nude mice, which are

characterised by the absence of functional T lymphocytes and a partial absence of B

lymphocytes. There are also other leukaemic mouse models available including NOD-SCID-

β2 null and NOD-SCID-IL-2Rγ null. The leukaemic xenotransplantations in mice can be

established by different administrations including I.V., intrafemoral and S.C. inoculations. The

intrafemoral injection of leukaemic cells into the BM is the most common and effective

xenotransplantation administration. These preclinical models are used to assess the anti-cancer

efficacy of drugs.

One of the aims of this chapter was to measure the cytotoxicity of HXR9 and its

cytotoxic efficacy on AML cells in vivo. Therefore, C57BL/6 mouse strain that has a full

functional immune system and the murine AML cell line C1498 labelled with GFP were used

for this purpose. The in vitro cytotoxicity assay showed the sensitivity of C1498-GFP to HXR9

and this sensitivity was further induced by either inhibiting CaM or PKC activity alone or

concurrently, which indicates that C1498-GFP cells respond to HXR9 as human AML cells.

The systemic injection of C1498-GFP in C57BL/6 mice resulted in only a low engraftment rate.

This low engraftment in PB might be the result of C1498-GFP cell lysis by NKs and CD8+

cytotoxic lymphocytes (Boyer, Orchard et al. 1995). In addition, the level of engraftment

measured in this study is typically seen in AML xenotransplantation experiments (Sanchez,

Perry et al. 2009).

Page 196: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

182

As a result, the systemic xenotransplantation was redone in two mouse strains including

C57BL/6 and nude mice, monitoring engraftment in the PB, BM, liver, kidney and spleen. This

experiment showed engraftment of C1498-GFP cells in all harvested organs with low

engraftment in PB and BM. Extramedullary infiltration, the presence and proliferation of

leukaemic cells in sites beyond the medullary spaces of BM including liver, kidney and spleen,

is a common symptom in AML and associated with poor prognosis and poor OS (Ravindranath,

Steuber et al. 1991; Creutzig, Harbott et al. 1995; Rubnitz, Raimondi et al. 2002; Chang,

Brandwein et al. 2004). Extramedullary infiltration was also reported in mouse

xenotransplantations (Krivtsov, Twomey et al. 2006; Sanchez, Perry et al. 2009). The causes of

extramedullary infiltration are not fully understood. However, the interaction between the

chemokine stromal cell-derived factor-1 alpha produced from human and murine liver, kidney

and spleen, and its receptor C-X-C chemokine receptor type 4 (CXCR4), is highly expressed

by AML and other leukaemic cells is considered as a crucial step in AML cell proliferation and

trafficking, AML disease progression and extramedullary infiltration (Mohle, Schittenhelm et

al. 2000; Kollet, Spiegel et al. 2001; Kollet, Shivtiel et al. 2003; Kato, Niwa et al. 2011). In

addition, targeting CXCR4 prevented the engraftment of AML cells in the BM and spleen of

transplanted mice (Tavor, Petit et al. 2004; Zhang, Patel et al. 2012). Obviously, the overall

engraftment rate was relatively low in both strains which may due to some immune activity in

both strains or due to technical issues such as failure to inject cells in the tail vain.

The I.V. injection of C1498-GFP cells in C57BL/6 and nude mice was clinically

relevant to AML disease. However, this systemic injection has many limitations. For example,

the low engraftment rate which could be overcome by pre-treating mice with a combination

regimen of total body irradiation and a pharmacological immunosuppressive agents. It is well-

documented that total body irradiation of recipient mice sustained engraftment by decreasing

the homing ability to the marrow microenvironment, decreasing the repopulating ability and

Page 197: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

183

causing cell death of the resident stem and progenitor cells and accordingly allowing injected

cells to engraft (Kahl, Mielcarek et al. 2004). In addition, the combination regimen inhibited

graft rejection by host T-cells which increased the engraftment rate (Mielcarek, Torok-Storb et

al. 2011; Shan and Ma 2013). Furthermore, the high engraftment rate of C1498-GFP cells in

extramedullary organs and the low engraftment rate in PB was a limitation due to the lack of

non-invasive monitoring system of the progression of AML which could be overcome by using

bioluminescence imaging. This technique can rapidly and noninvasively detect disease

progression from early stages by measuring light emission from cells labelled with the

bioluminescent reporter genes.

Due to the low systemic engraftment rate; the lack of radiation and bioluminescence

imaging facilities, the S.C. engraftment model was used in this study. This model is not a

realistic representation of AML, but it is an acceptable model for the preclinical evaluation of

drugs (Vinante, Rigo et al. 1999; He, Liu et al. 2003; Siegler, Kalberer et al. 2005; Motiwala,

Majumder et al. 2009; Zhang, Zhang et al. 2012; Chen, Yang et al. 2013; Park, Mishra et al.

2013; Sukhai, Prabha et al. 2013; Li, Li et al. 2014; Yang, Yu et al. 2014). The flank model in

C57BL/6 mice showed 100% engraftment rate that indicated to the usefulness of this model to

assess the efficacy of HXR9. At the first experiment, the concentration of HXR9 used was

50mg/kg, which was the half of the maximum concentration that previously used (Morgan,

Plowright et al. 2010; Morgan, Boxall et al. 2012; Morgan, Boxall et al. 2014). The I.T injection

of HXR9 showed that HXR9 was effective in delaying the growth of C1498-GFP cell tumour.

At the second experiment, the concurrent inhibition of CaM and PKC was introduced with the

same concentration of HXR9, 50mg/kg. Similar to the first experiment, using concurrent

inhibition of CaM and PKC did not sensitize tumours to 50mg/kg HXR9. This result was

consistent with in vitro results that showed the concurrent inhibition of CaM and PKC was not

cytotoxic itself, but increased the sensitivity to HXR9.

Page 198: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

184

The human AML cell line K562 and SCID mice were used as a second in vivo model.

At 100mg/kg, HXR9 was not effective in reducing tumour sizes of treated mice. In addition, at

300mg/kg and 200mg/kg, HXR9 caused the death of two mice at each concentration. This effect

might be owing to the systemic leakage of HXR9 through tumour microvessels which might

result in HXR9 mediated toxicity. The acute toxicity of HXR9 might result from the cationic

R9 which is well-known for its nonspecific interactions and ability to penetrate most organs

(Vives, Schmidt et al. 2008; Sarko, Beijer et al. 2010).

The ethics regulations might underestimate the in vivo cytotoxicity of HXR9 on AML

cells. According to the ethics mice were culled when tumours reached 1500 mm3, this action

might underestimate the difference in tumour sizes between different mouse groups, treated and

untreated mice. This is one of the limitations of using the flank model for AML

xenotransplantation and could be overcome by using a systemic in vivo model.

The main challenge of treating cancer is drug resistance. To understand the mechanisms

of chemoresistance, the structure of solid tumours should be known. Solid tumours are

heterogenous environment compose of cancer and stromal cells. The stromal cells includes

fibroblasts, immune, endothelial and mesothelial cells, adipocytes and extracellular matrix

(Castells, Thibault et al. 2012). The resistance to therapy is complicated and caused by several

factors. Some of resistance factors are cell intrinsic ''biochemical'' including genetic mutations

that affect drug uptake, over-expression of antiapoptotic and/or survival genes (Wilson,

Longley et al. 2006; Bleau, Hambardzumyan et al. 2009). Other factors are cell extrinsic

''physiological'' that resulted from the stromal microenvironment (Williams, den Besten et al.

2007; Eckstein, Servan et al. 2009). For example, tumours characterised by their extracellular

acidity and their alkaline or neutral intracellular environments (Tredan, Galmarini et al. 2007).

The extracellular pH influences the cellular uptake and thereby the efficacy of chemotherapies.

For example, increasing the extracellular acidity by I.V. glucose administration, through an

Page 199: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

185

unknown mechanism, promoted the uptake and the efficacy of weak acid drugs such as

chlorambucil, while it decreased the tumour growth delay of the small-cell lung carcinoma cell

line 54A by the weak base doxorubicin (Gerweck, Vijayappa et al. 2006). In contrast, in vitro

and systemic in vivo alkalinisation of extracellular environment by sodium biocarbonate

increased the uptake and the efficiency of weak base drugs including anthracyclines, taxanes

and alkylating agents on the breast cancer cell line MCF-7, whereas it reduced the cytotoxicity

of weak acid drugs including cyclophosphamide, 5-fluorouracil and chlorambucil (Mahoney,

Raghunand et al. 2003; Raghunand, Mahoney et al. 2003). It is noteworthy that HXR9 is

composed of the functional peptide and R9 which is one of the cell-penetrating peptides that

characterised by its cationic feature (Jin, Zhang et al. 2013). Therefore, the tumour extracellular

acidity might affect the uptake and the in vivo efficacy of HXR9 on the tested cell lines

including C1498-GFP and K562 cells. Tumour microenvironments are also characterised by

the presence of some hypoxia regions (Tredan, Galmarini et al. 2007; De Bock, Mazzone et al.

2011). Hypoxia was documented to cause in vivo drug resistance by activating the expression

angiogenic, anti-apoptotic and survival genes through triggering the over-expression of hypoxia

inducible factor-1 (Pouyssegur, Dayan et al. 2006). hypoxia inducible factor-1 in turn induces

the expression of NF-ĸB that activates the survival pathway by enhancing PI3K/Akt (Eltzschig

and Carmeliet 2011). NF-ĸB also mediates the expression of the inflammatory molecules such

INF-γ and TNF-α that induces the expression of the angiogenic factor vascular endothelial

growth factor (VEGF) from endothelial cells and cancer cells (Eltzschig and Carmeliet 2011).

VEGF is a mediator of the over-expression of the anti-apoptotic proteins including Bcl-2 and

XIAP (Nör, Christensen et al. 1999; Lee, Shanafelt et al. 2005). However, the inhibition of

VEGF expression in a S.C. model of K562 led to a decrease in the tumour microvessels and

caused cell apoptosis (He, Liu et al. 2003). Hypoxia was also reported to cause up-regulation

of P-glycoprotein, which is responsible of drug efflux (Comerford, Wallace et al. 2002).

Page 200: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

186

Tumours cause infiltration and accumulation of immune cells such as macrophages in tumour

microenvironments (De Palma and Lewis 2011). This subset of macrophages called tumour

associated macrophages and functions as a tumour growth inducer by increasing tumour

vascularisation and angiogenesis through over-expressing VEGF (Stockmann, Doedens et al.

2008). In addition, those macrophages facilitate tumour growth by providing

immunosuppressive microenvironment through expressing anti-inflammatory molecules such

as interleukin 10, transforming growth factor β (Hao, Lu et al. 2012). The concurrent treatment

regimen of tumour associated macrophages and chemotherapy caused a significant reduction

in tumour sizes (DeNardo, Brennan et al. 2011). Adhesion of stromal cells to cancer cells also

promoted the survival of cancer cells (Correia and Bissell 2012). Resistance to new therapies

is multi-factorial and tumour microenvironment has an integral part in it.

The discrepancy between in vitro and in vivo response of both tested mouse and human

AML cell lines C1498-GFP and K562, respectively, indicated that tumour microenvironment

might played a crucial role in the in vivo chemoresistance to HXR9. However, this potential

explanation does not exclude the acquired chemoresistance that resulted from cell intrinsic

mutations. To understand and improve the in vivo efficacy of HXR9, more molecular assays

and the physiology of tumour microenvironment are needed to be investigated. Of note, the

S.C. injection does not realistically reflect the nature of AML disease. Therefore, there is a need

to develop a reliable preclinical in vivo model for the assessment of the efficacy of HXR9.

Page 201: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

187

5.3.1 Summary of chapter

In summary, in vitro results showed that HXR9 has a significant anti-tumour activity on

C1498-GFP cells that was sharply promoted by either inhibition of CaM/PKC or both of which.

However, S.C. injection of C1498-GFP mouse model results demonstrated that this cell line

was chemo-resistant to either HXR9-alone treatment or combination treatment of HXR9 and

concurrent inhibition of CaM and PKC. The chemoresistance to HXR9 was also seen in K562

S.C. implemented into SCID mice. In addition, HXR9 resulted in death of some mice at high

concentrations. Finally, the attempt to develop a systemic mouse model for C1498-GFP cells

in C57BL/6 and nude mice resulted in low engraftment rate.

Page 202: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

188

Chapter 6 General Discussion and Future Directions

Page 203: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

189

6.1 Discussion

AML is considered to be the most frequent acute leukaemia to affect adult patients.

Conventionally, AML is classified cytogenetically according to the cell karyotypes into three

prognostic groups: favourable prognostic group including t(8;21), inv(16) and t(15;17), an

adverse prognostic group including complex karyotypes, trisomy 8 and CML-blast crisis, and

an intermediate prognostic group including most other abnormalities and NK-AML (Roche,

Zeng et al. 2004). Of note, about 45% of AML cases exhibit normal karyotypes (Marcucci,

Mrozek et al. 2005). The treatment regimen for all prognostic groups has not been changed

substantially over the last 4 decades, with the exception of APML. The current standard

induction therapy is a combination of DNR for 3 days with cytarabine for 7 days in a so-called

''3+7 regimen''. This regimen usually achieves 70%-75% CR in young (< 60 years), and 50%

CR in old patients. A post-induction, consolidation regimen consisting of high cytarabine doses

or alternatively allogeneic transplantation improves the outcome of AML patients. However,

the outcome of this regimen is heterogonous and the majority of patients will relapse and die

even after achieving clinical CR (Burnett 2012; Patel and Levine 2012).

The significant heterogeneity in the outcome of AML patients might be due to several

reasons. First, the current CR criteria are broad and totally dependent on microscopic counting

of the number of blast cells in BM to determine if the make-up ≤ 5% of the cell population, and

has not changed substantially since 1956 (Hourigan and Karp 2013). This limitation could be

overcome by including the detection of AML-specific molecular stable targets using PCR or

flow cytometry in CR criteria. The detection of AML cells after chemotherapy at levels under

the sensitivity level of the microscopic detection is known as minimal residual disease (MRD)

(Ossenkoppele and Schuurhuis 2013). Second, the low specificity of the current treatment

regimen also plays a role in the heterogeneity of response. To overcome this limitation, AML

patients should be treated with more cell-specific drugs. Third, there is heterogeneity of AML

Page 204: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

190

cells with respect to treatment sensitivity; for example, favourable prognostic groups respond

better than poor prognostic groups to the currently used chemotherapy.

Currently, the evaluation of relapse risk of AML (except for APML) patients in initial

clinical CR is based on the average clinical results of large historical populations who had the

same pre-treatment chromosomal and molecular abnormalities at the presentation time

(Mrozek, Marcucci et al. 2012; O'Donnell, Abboud et al. 2012). This assessment method lacks

the evaluation of the induction treatment efficiency by patient-individualised-evaluation and

can be improved by including MRD detection to guide decisions to the most-suitable post-

induction treatment. MRD is currently applicable in CML to detect the BCR-ABL fusion that

is present in almost all CML cases (Cross, White et al. 2012; O'Hare, Zabriskie et al. 2012). In

AML, MRD is now only used in APML to detect the PML-RARA fusion (Grimwade,

Jovanovic et al. 2009; Chendamarai, Balasubramanian et al. 2012). However, MRD is not

involved in the rest of AML cases; therefore, finding genes that are over-expressed in AML

cells is still an open area for more studies. For example, the most common mutated genes in

AML, including NPM1 and FLT3, might be potential detectable by MRD (Kristensen, Møller

et al. 2011; Buccisano, Maurillo et al. 2012). The detection of molecular targets offers a higher

sensitive than the current microscopic detection and presents a more-personalised approach to

diagnosis. It was reported that detection of the NPM1mut MRD preceded haematological relapse

by a median time of 8 weeks (Schnittger, Kern et al. 2009). Analogously, therapeutic

intervention after the detection of molecular relapse by MRD improved clinical outcomes

(Rubnitz, Inaba et al. 2010; Inaba, Coustan-Smith et al. 2012). It is noteworthy that HOX gene

over-expression is a common event in many AML subtypes and is strongly associated with a

poor prognosis, as previously mentioned in the introduction, and thus they are potential targets

in MRD.

Page 205: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

191

The heterogeneity of AML is due to the presence of various cytogenetic abnormalities

in 55% of AML cases including numerical aberrations, structural aberrations and balanced

translocations (Buccisano, Maurillo et al. 2012). In addition, molecular studies revealed a

diversity of molecular markers that further sub-classified the majority of NK-AML patients

(Marcucci, Mrozek et al. 2005). The discovery of molecular markers in NK-AML leads to the

hypothesis that leukaemogenesis results from the interaction of many mutations. These lead to

either an increase in cell proliferation (class I mutations) or a block of cell differentiation (class

II mutations), ultimately leading to AML (Dohner and Dohner 2008; Renneville, Roumier et

al. 2008; Betz and Hess 2010).

The burgeoning molecular understanding of AML could be valuable in many ways,

including the further sub-classification of the disease and the development of more personal

prognostic predictions that could form the cornerstone for future therapeutic regimens.

Currently, all-trans retinoic acid (ATRA) is the only widely used molecularly targeted therapy

in AML, for the treatment of APML characterised by the presence of t(15;17)/PML-RARA.

Understanding the molecular mechanism, signalling pathways and cooperating mutations will

pave the way for new potential molecular drugs that hopefully will result in more personalised

treatment.

The diversity of cytogenetic and molecular aberrations is one of the main obstacles to

the development of targeted therapies for AML, and the definition of the common genetic

abnormalities in patients with different kinds of mutations may lead the way to subtype specific

therapies. MLL fusion proteins were reported in 10% of therapy-related AML and 3% of de

novo AML (Slany 2009). Core binding factor translocations including CBFB/MYH11 and

RUNX1/RUNX1T1 fusions constitute 6% and 10% of all AML cases, respectively (Haferlach

2008). NUP98-HOX fusion proteins were also reported in AML (Fujino, Suzuki et al. 2002;

Suzuki, Ito et al. 2002; Taketani, Taki et al. 2002). Advances in molecular techniques also

Page 206: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

192

defined genetic aberrant expressions in AML. For example, the dysregulation of CDX2 and

CDX4 occur in 90% and 25% of AML cases, respectively (Bansal, Scholl et al. 2006; Scholl,

Bansal et al. 2007). Wilms' tumour gene 1 over-expression was seen in 80%-90% of AML cases

(Ostergaard, Olesen et al. 2004; Weisser, Kern et al. 2005), and the NPM1 mutation is present

in 35% of all AML cases and approximately 55% of NK-AML (Falini, Mecucci et al. 2005;

Thiede, Koch et al. 2006; Schlenk, Döhner et al. 2008). FLT3 mutations are present in 25% of

all AMLs and in 70% of NK-AMLs (Thiede, Steudel et al. 2002; Yanada, Matsuo et al. 2005).

The NRAS mutation was reported in 11% of AMLs (Bacher, Haferlach et al. 2006). These

aberrations can be found alone or in combination. For example, FLT3 is often coincident with

NPM1 or MLL mutations (Döhner, Schlenk et al. 2005; Olesen, Nyvold et al. 2005).

The above-mentioned combinations of FLT3 with NPM1 or MLL indicates that genetic

and cytogenetic aberrations cannot be assessed independently, and may in fact reflect the early

mechanism of leukaemogenesis and narrow down the potential genetic and signalling pathways

that are suitable for therapeutic approaches. An in depth analysis of AML subtypes suggest that

HOX genes can be one of the common denominators of different cytogenetic and molecular

aberrations. HOX gene over-expression was reported in AML with chromosomal rearrangement

including MLL fusion proteins and MYST3-CREBBP translocation (Ayton and Cleary 2003;

Camos, Esteve et al. 2006; Slany 2009). Mll fusion was found to function in AML by

collaborating with Ras to drive the over-expression of Hoxa9 and activate Raf (Ono, Kumagai

et al. 2009). Hox proteins can also form fusion proteins with Nup98 that mediate AML

formation in combination with flt3 (Borrow, Shearman et al. 1996; Nakamura, Largaespada et

al. 1996; Raza-Egilmez, Jani-Sait et al. 1998; Kroon, Thorsteinsdottir et al. 2001; Fujino,

Suzuki et al. 2002; Suzuki, Ito et al. 2002; Taketani, Taki et al. 2002; Pineault, Abramovich et

al. 2004; Palmqvist, Argiropoulos et al. 2006). In addition, NUP98-HOXA9 protein was

significantly associated with KRAS and Wilms' tumour gene 1 mutations (Chou, Chen et al.

Page 207: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

193

2009). Hox genes are regulated by the Cdx family in AML (Bansal, Scholl et al. 2006; Rawat,

Thoene et al. 2008),and, likewise, gene profiling studies showed a strong association between

FLT3 mutations and HOXA7, HOXA9 and HOXA10 expression (Roche, Zeng et al. 2004;

Eklund 2011). Over-expression of several HOX genes was also seen in the presence of NPM1

mutations (Mullighan, Kennedy et al. 2007; Vassiliou, Cooper et al. 2011). These studies

suggested that HOX genes are novel targets for molecular targeted therapies in various AML

subtypes.

Targeting a group of HOX proteins by disrupting their interactions with their cofactor

PBX was previously achieved using the small cell-permeable peptide HXR9. This approach

illustrated the importance of HOX genes in the tumorigenesis and survival of the cancer cells

derived from several solid cancers (Morgan, Pirard et al. 2007; Shears, Plowright et al. 2008;

Plowright, Harrington et al. 2009; Morgan, Plowright et al. 2010; Morgan, Boxall et al. 2012;

Morgan, Boxall et al. 2014). Therefore, this study was carried out based on the hypothesis that

AML cells would be sensitive to killing by HXR9 and indeed HOX genes might be a useful

therapeutic target in a subset of AML patients that over-express HOX genes.

In order to test this hypothesis I measured HOX gene expression in AML cell lines

derived from different AML subtypes and assessed their susceptibility in vitro to HXR9

treatment either alone or in combination with first line AML chemotherapies (chapter 3). I also

performed a comprehensive study of the mechanism of cell killing by HXR9, (chapter 4) and

assessed the in vivo efficacy of HXR9 to kill AML cells either alone or in combination with

non-toxic reagents (chapter 5).

The AML cell lines used in this study represented various AML sybtypes and prognoses.

These included AML patients with de novo mutations (primary AML), including KG-1,

HEL92.1.7 and HL-60. In addition, cell lines were derived from CML patients in blast crises

(secondary AML), including K562 and KU812F. KG-1 is an erythroleukaemia cell line

Page 208: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

194

characterised by the expression of CD34 and the presence of a complex karyotype, and is

consequently associated with a poor prognosis (Koeffler and Golde 1978; Ahmed, Laverick et

al. 1999; Mrózek, Tanner et al. 2003; Mrozek 2008). HEL92.1.7 is also an erythroleukaemia

cell line carrying the Janus kinase 2 mutation that is associated with a poor prognosis: 80% of

patients relapse (Martin and Papayannopoulou 1982; Illmer, Schaich et al. 2007; Fiskus,

Verstovsek et al. 2011). HL-60 is an APML cell line associated with a favourable prognosis

and characterised by the presence of t(15;17)/(PML-RARA) (Collins, Ruscetti et al. 1978;

Dores, Devesa et al. 2012). Both K562 and KU812F are characterised by the presence of the

Philadelphia (Ph) chromosome t(9;22)/(BCR-ABL) and derived from CML patients in blast

crisis, which is indicative of a poor prognosis (Lozzio and Lozzio 1975; Kishi 1985; Roche,

Zeng et al. 2004; Rucker, Sander et al. 2006; Weisberg, Wright et al. 2008).

The expression of HOX genes in the tested AML lines was consistent with the

established idea that HOX genes are expressed in a lineage-restricted manner, whereby HOXA

and HOXB genes are expressed in myeloid (KU812F) and erythroid (HEL92.1.7 and K562)

cells, respectively. The exception to this was the erythroid cell line KG-1, which expressed

HOXA genes that were linked to the presence of trisomy 8. In addition, the APML cell line HL-

60 was characterised by a global down-regulation of HOX genes. It is noteworthy that the

CD34+ cell line KG-1, which is characterised by a poor response to conventional

chemotherapies, strongly expresses a set of HOX genes compared with the CD34- cell line HL-

60 that represents AML with a favourable prognosis (Ahmed, Laverick et al. 1999; Marone,

Scambia et al. 2002; Dores, Devesa et al. 2012). This difference in the prognosis is associated,

at least in part, to the expression of HOX genes since the high and low expression of HOX genes

corresponds to a poor and favourable prognosis, respectively (Golub, Slonim et al. 1999;

Andreeff, Ruvolo et al. 2008; Zangenberg, Grubach et al. 2009). In addition, the comparison

between KG-1 and HL-60 in terms of HOX expression is consistent with the previous

Page 209: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

195

observation that HOX genes are highly expressed in CD34+ cells but show virtually no

expression in CD34- cells (Sauvageau, Lansdorp et al. 1994; Pineault, Helgason et al. 2002).

The limitation of assessing the expression of HOX genes was the lack of normal CD34+ cells,

which could be addressed in future studies.

The functional redundancy of HOX genes obscures our understanding of their

importance in the survival and proliferation of cancer cells, even though their over-expression

has been reported in different kinds of cancers. In this study I showed for the first time that the

high expression of HOX genes could form the basis of individualised medicine in AML as

antagonising the interaction between a set of HOX proteins and their cofactor PBX by the small

cell-permeable peptide HXR9 resulted in cell death. I also found that the susceptibility of the

tested AML cell lines to HXR9 was correlated with the expression level of HOX genes. The

most susceptible cell line was KG-1 that highly expresses a set of HOX genes, while HL-60

showed very low HOX gene expression had the lowest susceptibility to HXR9. Interestingly,

the expression of CD34 is considered to be a marker of a poor prognosis, an adverse clinical

course and a low CR rate of AML (Repp, Schaekel et al. 2003). In addition, CD34+ AML cells

were reported to be resistant to the current chemotherapies, yet CD34+ might predict high

sensitivity to HXR9 since HOX genes are highly expressed in CD34+ cells (Bailly, Muller et al.

1995; Bailly, Skladanowski et al. 1997). Likewise, gene expression profiling of CD34+ cells in

CML patients in chronic phase reveals a higher expression of HOXA9, which may suggest that

HXR9 could be used in combination with imatinib, the first line treatment for CML, which

targets the BCR-ABL fusion protein (Diaz-Blanco, Bruns et al. 2007; Palandri, Castagnetti et

al. 2008; Hehlmann 2012). Consistent with this suggestion, the cell lines that express BCR-

ABL fusion, KU812F and K562, were sensitive to HXR9. In addition, it was reported that Bcr-

Abl cooperated with Nup98-Hox proteins to progress to the ultimate phase of CML that is blast

crisis (Mayotte, Roy et al. 2002; Ito, Kwon et al. 2010; Di Giacomo, Pierini et al. 2014).

Page 210: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

196

Analogously, the sensitivity of Ph+ AML cells to HXR9 might suggest that HXR9 could be

used in a combination with imatinib to kill ALL cells that express both BCR-ABL fusion and

HOX genes (Redaelli, Piazza et al. 2009). Since HL-60 is sensitive to HXR9, a rational

approach may be to combine HXR9 with the current first line treatment for APML, ATRA

(Tallman and Altman 2009).

Combination therapy is a common approach to treating drug-resistant cancer, and

depends on a synergistic interaction between drugs that presumably have complementary

modes of action. Multiple DNA-damaging agents were reported to activate NF-ҡB, such as

anthracyclines, alkalyting agents and topoisomerase inhibitors (Boland, Fitzgerald et al. 2000;

Laurent and Jaffrézou 2001; Campbell, O'Shea et al. 2006). However, NF-ҡB is negatively

regulated by c-FOS, which is the main pro-apoptotic downstream target of HXR9 (Ray,

Kuwahara et al. 2006; Takada, Ray et al. 2010). In addition, DNA-damage is known to stimulate

PKC activity that was found to have an antagonistic, anti-apoptotic effect on HXR9 in this study

(Johnson, Lu et al. 2002; Bluwstein, Kumar et al. 2013). The findings of this study therefore

suggest that DNA-damaging agents that activate NF-ҡB and PKC might not be suitable

combination partners to HXR9. The molecular mechanism of HXR9 is yet to be fully

characterised. The specificity of HXR9 to target the HOX-PBX interaction and modify the

transcription of some genes were previously assessed, and the mode of cell death of the tested

solid cancers showed that HXR9 led to cell death by either apoptosis or necrosis (Morgan,

Pirard et al. 2007; Shears, Plowright et al. 2008; Plowright, Harrington et al. 2009; Morgan,

Plowright et al. 2010; Morgan, Boxall et al. 2012; Morgan, Boxall et al. 2014). However, the

findings of this study indicate that there is difference, possibly unique mechanism of cell death

by HXR9 in AML.

Page 211: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

197

The study of cell death was carried out by assessing the changes in various cellular

compartments after HXR9 treatment. For example, nuclear morphology was assessed by DAPI

staining. The limitation of this assay was the lack of a positive control for nuclear

fragmentation. The cytosolic changes were studied by analysing the changes in different

members including the pro-apoptotic member of the Bcl-2 family, XIAP, Apaf1 and caspase

activation. In addition, the mitochondrial role was investigated by measuring the expression of

the pro-apoptotic and anti-apoptotic genes of the Bcl-2 family including Bad, Bax, Bak1 and

Bcl-2. All the above-mentioned studies revealed that HXR9-induced cell death through an

apoptosis-independent pathway, involving programmed necrosis.

Ca2+ and ROS are two main regulators of necrosis (Festjens, Vanden Berghe et al.

2006). The role of extracellular Ca2+ was assessed using EDTA. However, EDTA is a general-

cation inhibitor that might chelate HXR9 since R9 is a cation sequence. EGTA is another cation

chelator used in a preliminary experiment, data not shown. EGTA increased the sensitivity to

HXR9, although this might be the effect of its alkaline solvent NaOH that might increase the

pH of the extracellular environment and thereby strengthen the interaction between the

polycation HXR9 and the acidic cell surface (Raghunand, Mahoney et al. 2003). To the best of

my knowledge, there is no specific cell impermeable Ca2+ chelator. In addition, intracellular

Ca2+ concentration was not measured in this study, although Ca2+ downstream mediators of

necrosis were.

Programmed necrosis is classified into various types such as mitochondrial necrosis,

parthanatos and necroptosis (Andrabi, Dawson et al. 2008; Elrod and Molkentin 2013; Marshall

and Baines 2014). The assessment of CypD and PARP1 activation suggested that HXR9-

induced necrosis in mitochondria- and parthanatos-independent pathways, respectively. Of

note, the limitation of PARP1 activation assay was the lack of a positive control. However,

RIP1 is involved, at least in part, in HL-60 necroptosis and the absence of any RIP1 inhibition

Page 212: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

198

effect on K562 does not exclude a necroptotic pathway since necroptosis can be RIP1-

independent, but RIP3-dependent. Therefore, a RIP1 inhibition assay might suggest the need to

block the role of RIP1 and RIP3 by short interfering RNA or a pharmacological inhibitor to

analyse whether HXR9-induced necrosis in the only necroptosis-dependent pathway or whether

there is another pathway involved.

ATP depletion was achieved by incubating cells with a high concentration of fructose,

which is known to deplete intracellular ATP. The limitation of this assay was the lack of a

positive control. In addition, it might be better to measure intracellular ATP using more specific

kits in the future.

Mitochondria do not seem necessary for HXR9-induced death in the tested AML cells.

Mitochondria participate in apoptosis and some forms of necrosis. In apoptosis, it is involved

in caspase- and Bax-dependent pathways, yet neither pathway was essential in HXR9-induced

AML cell death (Martin 2010; Gogada, Prabhu et al. 2011). In necrosis, mitochondria are

indispensable for CypD- and PARP1-dependent necrosis, but neither CypD nor PARP1

activation was involved in HXR9-induced AML cell necrosis (Andrabi, Dawson et al. 2008;

Elrod and Molkentin 2013) .

HXR9 caused cell death in c-FOS-dependent pathway, at least in part, in previously

tested cell lines that derived from different kinds of solid cancers (Morgan, Pirard et al. 2007;

Shears, Plowright et al. 2008; Plowright, Harrington et al. 2009; Morgan, Plowright et al. 2010;

Morgan, Boxall et al. 2012; Morgan, Boxall et al. 2014). Likewise, c-FOS up-regulation was

seen in the tested AML cell lines. In an attempt to understand more about the molecular

mechanism that is involved in HXR9 cell killing, MAPK pathways were studied using

pharmacological inhibitors to block their potential role in HXR9 treatment. It was found that

HXR9 induced necrosis of the tested AML cells in a MAPK-independent pathway. As Ca2+ is

Page 213: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

199

a main inducer of necrosis, the role of some of the Ca2+ downstream effectors that might

regulate HXR9-induced necrosis was also analysed. For example, μ-Calpain was not involved

in HXR9-induced necrosis. Interestingly, the other analysed Ca2+ downstream mediators

including NOXs, CaM, PKC and HO-1 had an anti-necrotic effect, as their inhibition

accelerated the necrosis of the tested AML cells. In addition, it was found that c-FOS induced

p21 expression in p53-independent pathway.

The anti-necrotic effect of NOXs, CaM, PKC and HO-1 after HXR9 treatment for AML

cells might direct the selection of combination drugs that would be used in the future with

HXR9. For example, cisplatin was reported to result in cell apoptosis through pathways that

involve the activation of PKC, NOXs and JNK (Persaud, Hoang et al. 2005; Kim, Lee et al.

2010; Itoh, Terazawa et al. 2011; Pabla, Dong et al. 2011; Pereira, Igea et al. 2013). In addition,

c-FOS over-expression was highly associated with cisplatin resistance (Muscella, Urso et al.

2009). Likewise, the alkylating agents treosulfan and etoposide were reported to induce

apoptosis in a PKC-dependent manner (Schmidmaier, Oellerich et al. 2004; Day, Wu et al.

2009; Zhao, Duan et al. 2009). Therefore, cisplatin, treosulfan and etoposide might have

antagonistic effects on HXR9 treatment. However, drugs that inhibit NOXs, CaM, PKC and

HO-1 might produce synergistic effect upon combination with HXR9.

ROS seems to exert an anti-necrotic effect in HXR9 treated AML cells. The primary

sources of ROS are mitochondria, NOXs, and some enzymes and growth factors (Festjens,

Vanden Berghe et al. 2006; Morgan, Kim et al. 2008; Jiang, Zhang et al. 2011). The results of

this study suggested that mitochondria are not required for HXR9 cytotoxicity. However,

inhibition of NOXs enhanced the efficacy of HXR9. In addition, the results of this study showed

that inhibition of PKC, which is an upstream regulator for HO-1 and JNK pathway and

downstream effector for ROS also augmented the effect of HXR9 (Liu, Nishitoh et al. 2000;

Kadowaki, Nishitoh et al. 2005; Tsoyi, Jang et al. 2011). Therefore, HXR9 might increase the

Page 214: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

200

expression of anti-oxidant transcription factors and it will be informative to investigate their

expression and measure ROS levels after HXR9 treatment.

The in vitro results were promising and were followed up by in vivo models to assess

the efficacy of HXR9 in inducing the necrosis of AML cells. In addition, one of the aims of this

study was to assess the cytotoxicity of HXR9 on normal PB; subsequently, the mouse AML

cell line C1498-GFP and the C57BL/6 mouse strain that has a full immune system were used.

However, using both this strain and a nude mouse strain for I.V. injection of cell lines and

HXR9 had several limitations. There was a low engraftment rate of AML cells, which could be

a result of multiple factors. These include the presence of either a full or partial immune system

in C57BL/6 and nude mice, respectively. This limitation could be overcome by eradicating the

immune system by pre-treating mice with immunosuppressive agents and total body irradiation.

Alternatively, the most common mouse strains for in vivo AML models that are characterised

by the lack of the immune system are NOD/SCID or NOD/SCID gamma mice that have a high

rate of engraftment could be used (Shan and Ma 2013). The low engraftment rate could also be

explained by the low number of injected cells or a failure to inject cells in tail vein. Another

limitation is the presence of extramedullary infiltration that could be monitored using a

bioluminescence imaging system.

The S.C. model of AML cells is an acceptable model for preclinical evaluations of new

agents, but it does not recapitulate the BM microenvironment. Recent studies demonstrated

multiple differences among different stromal cells originating from different organs. The

differences include phenotypes, proliferation, the expression levels of SDF-1, VEGF and

TGFβ1 (Dmitrieva, Minullina et al. 2012; De Luca, Verardi et al. 2013). As stromal cells play

an integral part in chemoresistance, this difference in tumour microenvironments should be

considered in terms of assessing the in vivo efficacy of HXR9 to induce AML necrosis.

Page 215: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

201

Despite the heterogeneity of the S.C. and BM tumour microenvironments, the in vivo

chemoresistance to HXR9 could be a result of cell intrinsic or extrinsic factors. Therefore, the

potential causes of chemoresistance should be studied in order to improve the in vivo efficacy

of HXR9. Chemoresistance studies could be performed using a short-duration, ex vivo/co-

culture model, and could allow the role of stromal cells in HXR9 resistance to be determined.

Possible mechanisms of resistance could include an interaction with AML cells, and or the

expression of specific cytokines, growth factors and matrix proteins (Garrido, Appelbaum et al.

2001; Konopleva, Konoplev et al. 2002; van Gosliga, Schepers et al. 2007; Schuringa and

Schepers 2009; Civini, Jin et al. 2013; Lopez, Garcia et al. 2014).

The specificity of HXR9 to target HOX-PBX interaction was previously studied

(Morgan, Pirard et al. 2007; Plowright, Harrington et al. 2009). However, one of the main

limitations of CPPs, which is the R9 sequence in HXR9 peptide, its non-specific uptake. In

other words, CPPs are able to penetrate all tissues and organs, regardless of the administration

route. This drawback can be overcome by binding targeting peptides with an R9 sequence that

will produce a selective delivery system. For example, CXCR4 that is over-expressed in many

cancers including AML can be targeted by a ligand that is bound to CPPs in order to inhibit the

non-specific interaction of CPPs. This approach was done by attaching CXCR4 ligand to TAT,

a CPP, and the anticancer peptide p53-activating peptide that led to increase the specificity of

the delivery system and the efficacy of the anticancer peptide comparing to unguided CPP

(Snyder, Saenz et al. 2005). Analogously, the specificity can be established by creating

activatable CPP (ACPP) that constitute of a polycationic CPP linked to a neutralising polyanion

motif via a cleavable linker. This structure blocks the electrostatic interaction of the ACPP with

the negative charge cell surface by decreasing the charge of ACPP to approximately zero and

ultimately blocks uptake into cells. Proteolysis of the cleavable linker by a specific protease

releases the polycationic ACPP domain and enables it to enter cells. Many cancer associated

Page 216: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

202

proteases are known, which are highly expressed in cancer tissues, but are absent or expressed

at low concentrations in normal tissues such as metalloproteases (Vives, Schmidt et al. 2008;

Regberg, Srimanee et al. 2012). For example, binding doxorubicin to ACPP that was sensitive

to metalloproteases 2/9 effectively decreased the toxicity of doxorubicin and inhibited the

proliferation of HT-1080 cells (Shi, Gao et al. 2012).

Page 217: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

203

6.2 Conclusion

In vitro results showed that HOX-PBX dimer might be a potential therapeutic target in

a subset of AML that over-express HOX genes. This may potentially help development

of personalised medicine for AML disease. The comprehensive analysis of the

mechanism of HXR9 cytotoxicity may guide in the selection of the potential combination

drugs. For example, agents that cause NOX, CaM, PKC activation should be avoided and

vise versa. A proper in vivo systemic model should be used to assess the in vivo efficacy

of HXR9 cytotoxicity, along with conducting more in vitro assays to investigate the

mechanism of HXR9 in order to choose potential synergistic agents.

Page 218: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

204

6.3 Future directions

Combination therapy is becoming increasingly important in cancer treatment. The

findings of this study reveal that HXR9 is able to trigger the necrosis of K562 and KU812F

cells that express the BCR-ABL fusion protein; and HL-60 that expresses PML-RARA. It may

therefore be valuable to assess combination therapy with HXR9 and first line treatment drugs

such as imatinib and ATRA that target BCR-ABL and PML-RARA fusion proteins,

respectively.

Resistance to cell killing by HXR9 may involve anti-necrotic proteins, and thus agents

that target such proteins are potential synergistic agents with HXR9. For example, tamoxifen

and trifluoperazine, CaM antagonists, might be used in a combination with HXR9 (Ahn, Pan et

al. 2003; Wang, Li et al. 2010). Likewise, enzastaurin that inhibits PKC activity and increased

p21 levels in multiple myeloma cells could have potential synergistic effect with HXR9 (Raab,

Breitkreutz et al. 2009). The HO-1 inhibitors, pegylated zinc-protoporphyrin and styrene-

maleic-acid-copolymer-micelle-encapsulated particles, which showed apoptotic effects in

AML cell lines are also potential synergistic drugs with HXR9 (Herrmann, Kneidinger et al.

2012). Arsenic trioxide exerts its cytotoxicity by inhibiting the JNK pathway thereby increasing

the activity of c-FOS and p21 proteins; it will also be valuable to assess its combinatory effect

with HXR9 (Huang, Liu et al. 2006; Liu and Huang 2008).

Silencing c-FLIP in combination with HXR9 treatment seems an interesting

combination to perform since c-FLIP is negatively regulated by c-FOS (Zhang, Zhang et al.

2007). In addition, c-FLIP is a downstream effector for CaM and PKC (Hwang, Min et al. 2009;

Kaunisto, Kochin et al. 2009). It is also able to inhibit apoptosis and necrosis by suppressing

procaspase-8 and RIP1, respectively (Day, Huang et al. 2008; Wang, Du et al. 2008; He and

He 2013).

Page 219: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

205

The mechanism of cell killing by HXR9 requires further investigation. To understand

how RIP1 activity can cause c-FOS over-expression after HXR9 treatment, at least in HL-60

cells, the upstream regulators of c-FOS and downstream effectors of RIP1 such as apoptosis

signal-regulating kinase 1 might be involved in this mechanism (Liu, Nishitoh et al. 2000;

Vanlangenakker, Vanden Berghe et al. 2012). Such investigations may also include some c-

FOS upstream regulators, including FOS related kinase and ribosomal S6 kinase 2 (Zhang,

Zhang et al. 2007). In addition, p21 downstream effectors might be assessed. It is noteworthy

to assess the role of RIP3 in HXR9 cytotoxicity since necroptosis was involved, at least in HL-

60 cells.

A co-culture model to assess the efficacy of HXR9 in inducing AML necrosis needs to

be conducted. Such a model can mimic the BM microenvironment in in vivo models. AML cells

could be co-cultured either in direct or indirect contact with stromal cells such as human MS-5

cells to evaluate the influence of stromal cells and the secretome in the efficacy of HXR9.

A systemic in vivo model is needed to be established to assess the efficacy of HXR9 on

AML disease. In this study, the nude mouse strain was used, but the growth rate was low which

might due to the immune activity. In addition, C57BL/6 and SCID flank models do not

represent AML disease. Therefore, it is noteworthy to establish a systemic in vivo model in

other mouse mouse strains including NOG or NOD, the most common AML models, which are

characterised by the lack of T and B lymphocytes, impaired NK cells and antigen-presenting

cells (Shan and Ma 2013).

Page 220: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

206

Appendices

Page 221: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

207

A)

B)

C)

Page 222: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

208

D)

E)

Appendix 1. R110 standard curves for the caspase-3 assay. R110 standard curves were used as additional controls

for caspases-3 activity to show that free R110 reagent was fluorescent and its fluorescence correlated with its

concentration. A range of concentrations (5μM -25μM) of R110 were prepared by diluting 5mM R110 into 1×

reaction buffer. Then, R110 fluorescence was measured at 492nm. To validate individual experiments, R2 should

be ≥ 0.95. Graphs show the mean of three independent experiments and error bars show the SEM.

Page 223: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

209

A)

B)

Appendix 2. General inhibition of caspase activity in HXR9 treated AML cell lines by z-VAD-FMK. Cells were

pre-treated either with or without 50 μM z-VAD-FMK for one hour. Then, cells were treated with the IC50 and 2×

IC50 of HXR9 for two hours, or with 17.5 μM of DNR for 24 hours, which was used as a positive control. Next,

cells were harvested and stained with annexin V and 7-AAD for 15 minutes in the dark and analysed by flow

cytometry. There was no statistical difference in terms of sensitivity to HXR9 between pre-treated cells with or

without 50μM z-VAD-FMK. Graphs show the mean of three independent experiments and error bars show the

SEM. *p < 0.05, **** p < 0.0001 with respect to z-VAD-FMK untreated cells. A) Inhibition of caspase activity

by z-VAD-FMK in K562 and B) HL-60 cells.

Page 224: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

210

A)

B)

Appendix 3. Effect of EDTA on HXR9 cytotoxicity. Cells were pre-treated with or without 20mM EDTA for one

hour. Then, cells were treated with 10μM, 20μM, 40μM HXR9 that prepared in media either with or without

20mM EDTA or with an equivalent CXR9 concentration to the highest concentration of HXR9 for two hours. The

cells were then harvested and stained with annexin V and 7-AAD for 15 minutes in the dark and analysed by flow

cytometry. Pre- and co-treatment of cells with 20mM EDTA significantly inhibited the cytotoxicity of HXR9

compared to EDTA untreated counterparts. Graphs show the mean of three independent experiments and error

bars show the SEM. *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001 with respect to 20mM EDTA treated

cells. A) K562 cells and B) HL-60 cells.

Page 225: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

211

A)

B)

Appendix 4. The Effect of CsA on HXR9 cytotoxicity. Cells were pre-treated with or without 5μM CsA for one

hour. Then cells were treated with 1×, 2×, or 3× IC50 of HXR9, with an equivalent CXR9 concentration to the

highest concentration of HXR9, with 20mM H2O2 as a positive control, or were untreated (negative control). Next,

cells were harvested and stained with annexin V and 7-AAD for 15 minutes in the dark and analysed by flow

cytometry. Results show 5μM CsA leads to a significant decrease in % viable cells and an increase in % early

apoptotic and dead cells of HXR9 treated cells comparing to their counterparts. Graphs show the mean of three

independent experiments and error bars show the SEM. *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001

with respect to 5μM CsA untreated cells. A) K562 and B) HL-60 cells.

Page 226: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

212

A)

B)

Appendix 5. The effect of blocking the MEK/ERK pathway in H2O2 cytotoxicity. Cells were pre-treated with or

without 20μM of the MEK/ERK inhibitor U0126 for one hour. Next, cells were treated with 20mM H2O2 for two

hours and the cytotoxicity was measured by LDH assay. U0126 significantly inhibited the cytotoxicity of H2O2.

The graphs show the mean of three independent experiments and the error bars show the SEM. **p < 0.01 and

****p < 0.0001 with respect to 20μM U0126 untreated cells. A) K562 and B) HL-60 cells.

Page 227: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

213

A)

B)

Appendix 6. The effect of inhibiting the p38 pathway on H2O2 cytotoxicity. Cells were pre-treated with or without

25 μM SB302580 for one hour, and were then treated with 20mM H2O2 for two hours. Cytotoxicity was measured

using the LDH assay. Blocking the p38 pathway significantly reduced the cytotoxicity of H2O2. The graph shows

the mean of three independent experiments and the error bars show the SEM. **p < 0.01 and ***p < 0.001 with

respect to 25μM SB302580 untreated cells. A) K562 and B) HL-60 cells.

Page 228: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

214

Appendix 7. The impact of JNK pathway inhibition in the cytotoxicity of H2O2 in HL-60 cells. Cells were pre-

incubated either with or without 60μM SP600125 for one hour, and then incubated with 20mM H2O2 in media

supplemented with or no supplemented with 60μM SP600125 (red and blue bars, respectively). Next, the

cytotoxicity was assessed using the LDH assay. 60μM SP600125 dramatically decreased the cytotoxicity of H2O2.

The graph shows the mean of three independent experiments and the error bars show the SEM. ***p < 0.001 with

respect to 60μM SP600125 untreated cells.

0

25

50

75

100HL-60

20mM

% S

urv

ival cells

H2O2

H2O2+60M SP600125

***

Page 229: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

215

A)

B)

Appendix 8. The effect of blocking μ-calpain on the cytotoxicity of H2O2. Cells were pre-incubated with or

without 60μM calpain inhibitor I for one hour, and then incubated with 20mM H2O2 prepared in media with or

without calpain inhibitor I (red and blue bars, respectively). The cytotoxicity of H2O2 was then measured using the

LDH assay. Blocking μ-calpain significantly inhibited the cytotoxicity of H2O2. Graphs show the mean of three

independent experiments and error bars show the SEM. **p < 0.01 with respect to calpain inhibitor I untreated

cells. A) K562 and B) HL-60 cells.

Page 230: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

216

Appendix 9. Establishment of a systemic C1498-GFP model in C57BL/6 mice, only PB was harvested.

Harvesting day Number of mice harvested Engraftment rate

3 2 0

7 2 0

13 2 1

17 2 1

20 2 0

Page 231: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

217

Appendix 10.

A) Establishment a systemic C1498-GFP model in C57BL/6, PB, BM, kidney, liver and spleen were harvested.

B) Establishment a systemic C1498-GFP model in C57BL/6, PB, BM, kidney, liver and spleen were harvested.

Harvesting day Number of mice harvested Engraftment rate

8 2 0

15 2 1

22 2 1

25 2 0

28 2 1

Harvesting day Number of mice harvested Engraftment rate

8 1 0

15 1 0

22 1 1

28 1 0

35 1 1

Page 232: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

218

References

Page 233: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

219

Abdul-Nabi, A. M., E. R. Yassin, et al. (2010). "In vitro transformation of primary human

CD34+ cells by AML fusion oncogenes: early gene expression profiling reveals possible drug

target in AML." PLoS ONE 5(8): e12464.

Abe, M., J. I. Hamada, et al. (2006). "Disordered expression of HOX genes in human non-small

cell lung cancer." Oncology Reports 15(4): 797-802.

Abramov, A. Y. and M. R. Duchen (2008). "Mechanisms underlying the loss of mitochondrial

membrane potential in glutamate excitotoxicity." Biochimica et Biophysica Acta

(BBA) - Bioenergetics 1777(7–8): 953-964.

Abramovich, C., N. Pineault, et al. (2005). "Hox Genes: From Leukemia to Hematopoietic

Stem Cell Expansion." Annals of the New York Academy of Sciences 1044(1): 109-

116.

Adrain, C. and S. J. Martin (2001). "The mitochondrial apoptosome: a killer unleashed by the

cytochrome seas." Trends in Biochemical Sciences 26(6): 390-397.

Ahmed, N., L. Laverick, et al. (1999). "Effect of all-trans retinoic acid on chemotherapy

induced apoptosis and down-regulation of Bcl-2 in human myeloid leukaemia CD34

positive cells." Leukemia Research 23(8): 741-749.

Ahn, E. Y., G. Pan, et al. (2003). "The combination of calmodulin antagonists and interferon-

gamma induces apoptosis through caspase-dependent and -independent pathways in

cholangiocarcinoma cells." American Journal of Pathology 163(5): 2053-2063.

Alam, M. R., D. Baetz, et al. (2014). "Cyclophilin D and myocardial ischemia–reperfusion

injury: A fresh perspective." Journal of Molecular and Cellular Cardiology(0).

Alharbi, R. A., R. Pettengell, et al. (2012). "The role of HOX genes in normal hematopoiesis

and acute leukemia." Leukemia

Allen, M. J., J. Gemel, et al. (2011). "Atomic Force Microscopy of Connexin40 Gap Junction

Hemichannels Reveals Calcium-dependent Three-dimensional Molecular Topography

and Open-Closed Conformations of Both the Extracellular and Cytoplasmic Faces."

Journal of Biological Chemistry 286(25): 22139-22146.

Amores, A., A. Force, et al. (1998). "Zebrafish hox clusters and vertebrate genome evolution."

Science 282(5394): 1711-1714.

Amsellem, S., F. Pflumio, et al. (2003). "Ex vivo expansion of human hematopoietic stem cells

by direct delivery of the HOXB4 homeoprotein." Nat Med 9(11): 1423-1427.

Andersson, L. C., M. Jokinen, et al. (1979). "INDUCTION OF ERYTHROID-

DIFFERENTIATION IN THE HUMAN LEUKEMIA CELL-LINE K562." Nature

278(5702): 364-365.

Andersson, L. C., K. Nilsson, et al. (1979). "K562 - HUMAN ERYTHROLEUKEMIC CELL-

LINE." International Journal of Cancer 23(2): 143-147.

Andrabi, S. A., T. M. Dawson, et al. (2008). "Mitochondrial and Nuclear Cross Talk in Cell

Death Parthanatos." Mitochondria and Oxidative Stress in Neurodegenerative

Disorders 1147: 233-241.

Andreeff, M., V. Ruvolo, et al. (2008). "HOX expression patterns identify a common signature

for favorable AML." Leukemia 22(11): 2041-2047.

Appierto, V., M. G. Villani, et al. (2003). "Involvement of c-Fos in fenretinide-induced

apoptosis in human ovarian carcinoma cells." Cell Death Differ 11(3): 270-279.

Asada, M., T. Yamada, et al. (1999). "Apoptosis inhibitory activity of cytoplasmic

p21(Cip1/WAF1) in monocytic differentiation." Embo Journal 18(5): 1223-1234.

Ayton, P. M. and M. L. Cleary (2003). "Transformation of myeloid progenitors by MLL

oncoproteins is dependent on Hoxa7 and Hoxa9." Genes & Development 17(18): 2298-

2307.

Bacher, U., T. Haferlach, et al. (2006). Implications of NRAS mutations in AML: a study of

2502 patients.

Bailey-Dell, K. J., B. Hassel, et al. (2001). "Promoter characterization and genomic

organization of the human breast cancer resistance protein (ATP-binding cassette

transporter G2) gene." Biochimica et Biophysica Acta (BBA) - Gene Structure and

Expression 1520(3): 234-241.

Page 234: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

220

Bailly, J. D., C. Muller, et al. (1995). "Lack of Correlation between Expression and Function

of P-Glycoprotein in Acute Myeloid-Leukemia Cell-Lines." Leukemia 9(5): 799-807.

Bailly, J. D., A. Skladanowski, et al. (1997). "Natural resistance of acute myeloid leukemia cell

lines to mitoxantrone is associated with lack of apoptosis." Leukemia 11(9): 1523-

1532.

Baines, C. P., R. A. Kaiser, et al. (2005). "Loss of cyclophilin D reveals a critical role for

mitochondrial permeability transition in cell death." Nature 434(7033): 658-662.

Balshaw, D. M., L. Xu, et al. (2001). "Calmodulin Binding and Inhibition of Cardiac Muscle

Calcium Release Channel (Ryanodine Receptor)." Journal of Biological Chemistry

276(23): 20144-20153.

Bánfi, B., G. Molnár, et al. (2001). "A Ca2+-activated NADPH Oxidase in Testis, Spleen, and

Lymph Nodes." Journal of Biological Chemistry 276(40): 37594-37601.

Bánfi, B., F. Tirone, et al. (2004). "Mechanism of Ca2+ Activation of the NADPH Oxidase 5

(NOX5)." Journal of Biological Chemistry 279(18): 18583-18591.

Bansal, D., C. Scholl, et al. (2006). "Cdx4 dysregulates Hox gene expression and generates

acute myeloid leukemia alone and in cooperation with Meis1a in a murine model."

Proceedings of the National Academy of Sciences of the United States of America

103(45): 16924-16929.

Barrera, L. N., S. A. Rushworth, et al. (2012). "Bortezomib induces heme oxygenase-1

expression in multiple myeloma." Cell Cycle 11(12): 2248-2252.

Bartolomé, F., N. de las Cuevas, et al. (2007). "Impaired apoptosis in lymphoblasts from

Alzheimer’s disease patients: Cross-talk of Ca2+/calmodulin and ERK1/2 signaling

pathways." Cellular and Molecular Life Sciences 64(11): 1437-1448.

Basso, E., L. Fante, et al. (2005). "Properties of the Permeability Transition Pore in

Mitochondria Devoid of Cyclophilin D." Journal of Biological Chemistry 280(19):

18558-18561.

Bechara, C. and S. Sagan (2013). "Cell-penetrating peptides: 20 years later, where do we

stand?" FEBS letters 587(12): 1693-1702.

Bei, L., W. Huang, et al. (2011). "HoxA10 Activates CDX4 Transcription and Cdx4 Activates

HOXA10 Transcription in Myeloid Cells." Journal of Biological Chemistry 286(21):

19047-19064.

Bei, L., Y. Lu, et al. (2007). "Identification of a HoxA10 Activation Domain Necessary for

Transcription of the Gene Encoding β3 Integrin during Myeloid Differentiation."

Journal of Biological Chemistry 282(23): 16846-16859.

Bei, L., Y. Lu, et al. (2005). "HOXA9 Activates Transcription of the Gene Encoding gp91Phox

during Myeloid Differentiation." Journal of Biological Chemistry 280(13): 12359-

12370.

Berchtold, M. W. and A. Villalobo (2014). "The many faces of calmodulin in cell proliferation,

programmed cell death, autophagy, and cancer." Biochimica Et Biophysica Acta-

Molecular Cell Research 1843(2): 398-435.

Bettermann, K., M. Vucur, et al. (2010). "TAK1 Suppresses a NEMO-Dependent but NF-κB-

Independent Pathway to Liver Cancer." Cancer Cell 17(5): 481-496.

Betz, B. L. and J. L. Hess (2010). "Acute Myeloid Leukemia Diagnosis in the 21st Century."

Archives of Pathology & Laboratory Medicine 134(10): 1427-1433.

Beuchle, D., G. Struhl, et al. (2001). "Polycomb group proteins and heritable silencing of

Drosophila Hox genes." Development 128(6): 993-1004.

Bezombes, C., A. de Thonel, et al. (2002). "Overexpression of Protein Kinase Cζ Confers

Protection Against Antileukemic Drugs by Inhibiting the Redox-Dependent

Sphingomyelinase Activation." Molecular Pharmacology 62(6): 1446-1455.

Bhalla, K., A. Ibrado, et al. (1993). "High-dose mitoxantrone induces programmed cell death

or apoptosis in human myeloid leukemia cells." Blood 82(10): 3133-3140.

Bhatlekar, S., J. Fields, et al. (2014). "HOX genes and their role in the development of human

cancers." Journal of Molecular Medicine 92(8): 811-823.

Page 235: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

221

Bijl, J., A. Thompson, et al. (2006). "Analysis of HSC activity and compensatory Hox gene

expression profile in Hoxb cluster mutant fetal liver cells." Blood 108(1): 116-122.

Bjornsson, J. M., N. Larsson, et al. (2003). "Reduced Proliferative Capacity of Hematopoietic

Stem Cells Deficient in Hoxb3 and Hoxb4." Mol. Cell. Biol. 23(11): 3872-3883.

Blankenberg, F. G. and H. W. Strauss (2012). "Recent Advances in the Molecular Imaging of

Programmed Cell Death: Part I-Pathophysiology and Radiotracers." Journal of Nuclear

Medicine 53(11): 1659-1662.

Bleau, A.-M., D. Hambardzumyan, et al. (2009). "PTEN/PI3K/Akt Pathway Regulates the Side

Population Phenotype and ABCG2 Activity in Glioma Tumor Stem-like Cells." Cell

stem cell 4(3): 226-235.

Bluwstein, A., N. Kumar, et al. (2013). "PKC signaling prevents irradiation-induced apoptosis

of primary human fibroblasts." Cell Death & Disease 4.

Boland, M. P., K. A. Fitzgerald, et al. (2000). "Topoisomerase II Is Required for Mitoxantrone

to Signal Nuclear Factor κB Activation in HL60 Cells." Journal of Biological

Chemistry 275(33): 25231-25238.

Boland, M. P., S. J. Foster, et al. (1997). "Daunorubicin activates NF kappa B and induces

kappa B-dependent gene expression in HL-60 promyelocytic and Jurkat T lymphoma

cells." Journal of Biological Chemistry 272(20): 12952-12960.

Bonneau, M. J. and R. Poulin (2000). "Spermine oxidation leads to necrosis with plasma

membrane phosphatidylserine redistribution in mouse leukemia cells." Experimental

Cell Research 259(1): 23-34.

Bonomini, M., S. Dottori, et al. (2004). "Increased platelet phosphatidylserine exposure and

caspase activation in chronic uremia." Journal of Thrombosis and Haemostasis 2(8):

1275-1281.

Borrow, J., A. M. Shearman, et al. (1996). "The t(7;11)(p15;p15) translocation in acute myeloid

leukaemia fuses the genes for nucleoporin NUP96 and class I homeoprotein HOXA9."

Nat Genet 12(2): 159-167.

Bosco, R., E. Melloni, et al. (2011). "Fine Tuning of Protein Kinase C (PKC) Isoforms in

Cancer: Shortening the Distance from the Laboratory to the Bedside." Mini-Reviews

in Medicinal Chemistry 11(3): 185-199.

Bosma, M. J. and A. M. Carroll (1991). "THE SCID MOUSE MUTANT - DEFINITION,

CHARACTERIZATION, AND POTENTIAL USES." Annual Review of Immunology

9: 323-350.

Bouchard, V. J., M. Rouleau, et al. (2003). "PARP-1, a determinant of cell survival in response

to DNA damage." Experimental Hematology 31(6): 446-454.

Boyer, M. W., P. J. Orchard, et al. (1995). "DEPENDENCY ON INTERCELLULAR-

ADHESION MOLECULE RECOGNITION AND LOCAL INTERLEUKIN-2

PROVISION IN GENERATION OF AN IN-VIVO CD8(+) T-CELL IMMUNE-

RESPONSE TO MURINE MYELOID-LEUKEMIA." Blood 85(9): 2498-2506.

Bromleigh, V. C. and L. P. Freedman (2000). "p21 is a transcriptional target of HOXA10 in

differentiating myelomonocytic cells." Genes & Development 14(20): 2581-2586.

Brown, P., E. McIntyre, et al. (2007). "The incidence and clinical significance of

nucleophosmin mutations in childhood AML." Blood 110(3): 979-985.

Brueckl, C., S. Kaestle, et al. (2006). "Hyperoxia-Induced Reactive Oxygen Species Formation

in Pulmonary Capillary Endothelial Cells In Situ." American Journal of Respiratory

Cell and Molecular Biology 34(4): 453-463.

Brumatti, G., M. Salmanidis, et al. (2013). "HoxA9 regulated Bcl-2 expression mediates

survival of myeloid progenitors and the severity of HoxA9-dependent leukemia."

Oncotarget 4(11): 1933-1947.

Brun, A. C. M., J. M. Björnsson, et al. (2004). "Hoxb4-deficient mice undergo normal

hematopoietic development but exhibit a mild proliferation defect in hematopoietic

stem cells." Blood 103(11): 4126-4133.

Buccisano, F., L. Maurillo, et al. (2012). "Prognostic and therapeutic implications of minimal

residual disease detection in acute myeloid leukemia." Blood 119(2): 332-341.

Page 236: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

222

Bukowska, A., U. Lendeckel, et al. (2012). "Physiologic and Pathophysiologic Role of Calpain:

Implications for the Occurrence of Atrial Fibrillation." Cardiovascular Therapeutics

30(3): e115-e127.

Burnett, A. K. (2012). "Treatment of acute myeloid leukemia: are we making progress?"

Hematology-American Society Hematology Education Program: 1-6.

Buske, C., M. Feuring-Buske, et al. (2001). "Overexpression of HOXA10 perturbs human

lymphomyelopoiesis in vitro and in vivo." Blood 97(8): 2286-2292.

Cai, W. J., M. Torreggiani, et al. (2010). "AGER1 regulates endothelial cell NADPH oxidase-

dependent oxidant stress via PKC-delta: implications for vascular disease." American

Journal of Physiology-Cell Physiology 298(3): C624-C634.

Camos, M., J. Esteve, et al. (2006). "Gene expression profiling of acute myeloid leukemia with

translocation t(8;16)(p11;p13) and MYST3-CREBBP rearrangement reveals a

distinctive signature with a specific pattern of HOX gene expression." Cancer Research

66(14): 6947-6954.

Campbell, K., J. O'Shea, et al. (2006). "Differential regulation of NF-kappaB activation and

function by topoisomerase II inhibitors." BMC Cancer 6(1): 101.

Cao, G. D., J. Xing, et al. (2007). "Critical role of calpain I in mitochondrial release of

apoptosis-inducing factor in ischemic neuronal injury." Journal of Neuroscience

27(35): 9278-9293.

Carvalho, H., P. Evelson, et al. (2004). "Mitogen-activated protein kinases modulate H2O2-

induced apoptosis in primary rat alveolar epithelial cells." Journal of Cellular

Biochemistry 92(3): 502-513.

Castells, M., B. Thibault, et al. (2012). "Implication of Tumor Microenvironment in

Chemoresistance: Tumor-Associated Stromal Cells Protect Tumor Cells from Cell

Death." International Journal of Molecular Sciences 13(8): 9545-9571.

Cellot, S., J. Krosl, et al. (2007). "Sustained in vitro trigger of self-renewal divisions in

Hoxb4(hi)Pbx1(lo) hematopoietic stem cells." Experimental Hematology 35(5): 802-

816.

Chakraborty, S., M. Mazumdar, et al. (2014). "Restoration of p53/miR-34a regulatory axis

decreases survival advantage and ensures Bax-dependent apoptosis of non-small cell

lung carcinoma cells." FEBS letters 588(4): 549-559.

Chang, C. P., W. F. Shen, et al. (1995). "Pbx Proteins Display Hexapeptide-Dependent

Cooperative DNA-Binding with a Subset of Hox Proteins." Genes & Development

9(6): 663-674.

Chang, H., J. Brandwein, et al. (2004). "Extramedullary infiltrates of AML are associated with

CD56 expression, 11q23 abnormalities and inferior clinical outcome." Leukemia

Research 28(10): 1007-1011.

Chen, J., K. V. Giridhary, et al. (2011). "A protein kinase C/protein kinase D pathway protects

LNCaP prostate cancer cells from phorbol ester-induced apoptosis by promoting

ERK1/2 and NF-kappa B activities." Carcinogenesis 32(8): 1198-1206.

Chen, K., X. Y. Yang, et al. (2013). "Pinellia pedatisecta Agglutinin Targets Drug Resistant

K562/ADR Leukemia Cells through Binding with Sarcolemmal Membrane Associated

Protein and Enhancing Macrophage Phagocytosis." PLoS ONE 8(9).

Chen, M., P. Gallipoli, et al. (2013). "Targeting Primitive Chronic Myeloid Leukemia Cells by

Effective Inhibition of a New AHI-1BCR-ABL-JAK2 Complex." Jnci-Journal of the

National Cancer Institute 105(6): 405-423.

Chen, M., H. P. He, et al. (2001). "Bid is cleaved by calpain to an active fragment in vitro and

during myocardial ischemia/reperfusion." Journal of Biological Chemistry 276(33):

30724-30728.

Chen, N. Y., M. Nomura, et al. (2001). "Suppression of skin tumorigenesis in c-Jun NH2-

terminal kinase-2-deficient mice." Cancer Research 61(10): 3908-3912.

Chen, Y. and C. Sommer (2009). "The Role of Mitogen-Activated Protein Kinase (MAPK) in

Morphine Tolerance and Dependence." Molecular Neurobiology 40(2): 101-107.

Page 237: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

223

Chendamarai, E., P. Balasubramanian, et al. (2012). Role of minimal residual disease

monitoring in acute promyelocytic leukemia treated with arsenic trioxide in frontline

therapy.

Cheng, W., J. Liu, et al. (2005). "Lineage infidelity of epithelial ovarian cancers is controlled

by HOX genes that specify regional identity in the reproductive tract." Nat Med 11(5):

531-537.

Chi, S.-W. (2014). "Structural insights into the transcription-independent apoptotic pathway of

p53." Bmb Reports 47(3): 167-172.

Chipuk, J. E., T. Kuwana, et al. (2004). "Direct activation of Bax by p53 mediates

mitochondrial membrane permeabilization and apoptosis." Science 303(5660): 1010-

1014.

Chipuk, J. E., U. Maurer, et al. (2003). "Pharmacologic activation of p53 elicits Bax-dependent

apoptosis in the absence of transcription." Cancer Cell 4(5): 371-381.

Cho, Y., S. Challa, et al. (2009). "Phosphorylation-Driven Assembly of the RIP1-RIP3

Complex Regulates Programmed Necrosis and Virus-Induced Inflammation." Cell

137(6): 1112-1123.

Choi, B. H., C. G. Kim, et al. (2008). "P21(Waf1/Cip1) expression by curcumin in U-87MG

human glioma cells: Role of early growth response-1 expression." Cancer Research

68(5): 1369-1377.

Chou, T.-C. (1974). "Relationships between inhibition Constants and Fractional Inhibition in

Enzyme-Catalyzed Reactions with Different Numbers of Reactants, Different Reaction

Mechanisms, and Different Types and Mechanisms of Inhibition." Molecular

Pharmacology 10(2): 235-247.

Chou, T. C. (1991). THE MEDIAN-EFFECT PRINCIPLE AND THE COMBINATION

INDEX FOR QUANTITATION OF SYNERGISM AND ANTAGONISM.

Chou, W. C., C. Y. Chen, et al. (2009). "Acute myeloid leukemia bearing t(7;11)(p15;p15) is a

distinct cytogenetic entity with poor outcome and a distinct mutation profile:

comparative analysis of 493 adult patients." Leukemia 23(7): 1303-1310.

Chung, Y. W., D. W. Jeong, et al. (2002). "H2O2-induced AP-1 activation and its effect on

p21(WAF1/CIP1)-mediated G2/M arrest in a p53-deficient human lung cancer cell."

Biochemical and Biophysical Research Communications 293(4): 1248-1253.

Civini, S., P. Jin, et al. (2013). "Leukemia cells induce changes in human bone marrow stromal

cells." Journal of Translational Medicine 11.

Collins, J. A., C. A. Schandl, et al. (1997). "Major DNA Fragmentation Is a Late Event in

Apoptosis." Journal of Histochemistry & Cytochemistry 45(7): 923-934.

Collins, S. J., F. W. Ruscetti, et al. (1978). "TERMINAL DIFFERENTIATION OF HUMAN

PROMYELOCYTIC LEUKEMIA-CELLS INDUCED BY DIMETHYL-

SULFOXIDE AND OTHER POLAR COMPOUNDS." Proceedings of the National

Academy of Sciences of the United States of America 75(5): 2458-2462.

Comerford, K. M., T. J. Wallace, et al. (2002). "Hypoxia-inducible Factor-1-dependent

Regulation of the Multidrug Resistance (MDR1) Gene." Cancer Research 62(12):

3387-3394.

Correia, A. L. and M. J. Bissell (2012). "The tumor microenvironment is a dominant force in

multidrug resistance." Drug Resistance Updates 15(1-2): 39-49.

Creutzig, U., J. Harbott, et al. (1995). "CLINICAL-SIGNIFICANCE OF SURFACE-

ANTIGEN EXPRESSION IN CHILDREN WITH ACUTE MYELOID-LEUKEMIA

- RESULTS OF STUDY AML-BFM-87." Blood 86(8): 3097-3108.

Crooks, G. M., J. Fuller, et al. (1999). "Constitutive HOXA5 Expression Inhibits Erythropoiesis

and Increases Myelopoiesis From Human Hematopoietic Progenitors." Blood 94(2):

519-528.

Cross, N. C. P., H. E. White, et al. (2012). "Standardized definitions of molecular response in

chronic myeloid leukemia." Leukemia 26(10): 2172-2175.

Daga, A., M. Podesta, et al. (2000). "The retroviral transduction of HOXC4 into human CD34+

cells induces an in vitro expansion of clonogenic and early progenitors." Experimental

Hematology 28(5): 569-574.

Page 238: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

224

Daniels, T. R., I. I. Neacato, et al. (2010). "Disruption of HOX activity leads to cell death that

can be enhanced by the interference of iron uptake in malignant B cells." Leukemia

24(9): 1555-1565.

Davidson, A. J. and L. I. Zon (2006). "The caudal-related homeobox genes cdx1a and cdx4 act

redundantly to regulate hox gene expression and the formation of putative

hematopoietic stem cells during zebrafish embryogenesis." Developmental Biology

292(2): 506-518.

Day, T. W., S. Huang, et al. (2008). "c-FLIP knockdown induces ligand-independent DR5-,

FADD-, caspase-8-, and caspase-9-dependent apoptosis in breast cancer cells."

Biochemical Pharmacology 76(12): 1694-1704.

Day, T. W., C. H. Wu, et al. (2009). "Etoposide Induces Protein Kinase C delta- and Caspase-

3-Dependent Apoptosis in Neuroblastoma Cancer Cells." Molecular Pharmacology

76(3): 632-640.

de Araújo Leite, J. C. and L. F. Marques-Santos (2012). "Extracellular Ca2+ influx is crucial

for the early embryonic development of the sea urchin Echinometra lucunter." Journal

of Experimental Zoology Part B: Molecular and Developmental Evolution 318(2): 123-

133.

De Bock, K., M. Mazzone, et al. (2011). "Antiangiogenic therapy, hypoxia, and metastasis:

risky liaisons, or not?" Nature Reviews Clinical Oncology 8(7): 393-404.

De Luca, A., R. Verardi, et al. (2013). "Comparative Analysis of Mesenchymal Stromal Cells

Biological Properties." ISRN Stem Cells 2013: 9.

De Palma, M. and C. E. Lewis (2011). "CANCER Macrophages limit chemotherapy." Nature

472(7343): 303-304.

Degterev, A., J. Hitomi, et al. (2008). "Identification of RIP1 kinase as a specific cellular target

of necrostatins." Nature Chemical Biology 4(5): 313-321.

Degterev, A., Z. Huang, et al. (2005). "Chemical inhibitor of nonapoptotic cell death with

therapeutic potential for ischemic brain injury." Nat Chem Biol 1(2): 112-119.

Del Bello, B., D. Moretti, et al. (2007). "Cross-talk between calpain and caspase-3/-7 in

cisplatin-induced apoptosis of melanoma cells: a major role of calpain inhibition in cell

death protection and p53 status." Oncogene 26(19): 2717-2726.

DeNardo, D. G., D. J. Brennan, et al. (2011). "Leukocyte Complexity Predicts Breast Cancer

Survival and Functionally Regulates Response to Chemotherapy." Cancer Discovery

1(1): 54-67.

Desai, S., P. Pillai, et al. (2011). "PKC-ι promotes glioblastoma cell survival by

phosphorylating and inhibiting BAD through a phosphatidylinositol 3-kinase

pathway." Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 1813(6):

1190-1197.

Devalaraja-Narashimha, K. and B. J. Padanilam (2009). "PARP-1 Inhibits Glycolysis in

Ischemic Kidneys." Journal of the American Society of Nephrology 20(1): 95-103.

Dhillon, A. S., S. Hagan, et al. (2007). "MAP kinase signalling pathways in cancer." Oncogene

26(22): 3279-3290.

Di Giacomo, D., V. Pierini, et al. (2014). "Blast crisis Ph plus chronic myeloid leukemia with

NUP98/HOXA13 up-regulating MSI2." Molecular Cytogenetics 7.

Diaz-Blanco, E., I. Bruns, et al. (2007). "Molecular signature of CD34+ hematopoietic stem

and progenitor cells of patients with CML in chronic phase." Leukemia 21(3): 494-

504.

Dickson, G. J., F. G. Liberante, et al. (2013). "HOXA/PBX3 knockdown impairs growth and

sensitizes cytogenetically normal acute myeloid leukemia cells to chemotherapy."

Haematologica 98(8): 1216-1225.

Dik, W. A., W. Brahim, et al. (2005). "CALM-AF10+ T-ALL expression profiles are

characterized by overexpression of HOXA and BMI1 oncogenes." Leukemia 19(11):

1948-1957.

DiMartino, J. F., L. Selleri, et al. (2001). "The Hox cofactor and proto-oncogene Pbx1 is

required for maintenance of definitive hematopoiesis in the fetal liver." Blood 98(3):

618-626.

Page 239: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

225

Dmitrieva, R. I., I. R. Minullina, et al. (2012). "Bone marrow- and subcutaneous adipose tissue-

derived mesenchymal stem cells Differences and similarities." Cell Cycle 11(2): 377-

383.

Dohner, K. and H. Dohner (2008). "Molecular characterization of acute myeloid leukemia."

Haematologica 93(7): 976-982.

Döhner, K., R. F. Schlenk, et al. (2005). Mutant nucleophosmin (NPM1) predicts favorable

prognosis in younger adults with acute myeloid leukemia and normal cytogenetics:

interaction with other gene mutations.

Dong, C., Q. Li, et al. (2005). "A novel apoptosis pathway activated by the carboxyl terminus

of p21." Blood 105(3): 1187-1194.

Dores, G. M., S. S. Devesa, et al. (2012). Acute leukemia incidence and patient survival among

children and adults in the United States, 2001-2007.

Doulatov, S., F. Notta, et al. (2012). "Hematopoiesis: A Human Perspective." Cell stem cell

10(2): 120-136.

Dube, H., D. Selwood, et al. (2012). "A mitochondrial-targeted cyclosporin A with high binding

affinity for cyclophilin D yields improved cytoprotection of cardiomyocytes."

Biochemical Journal 441(3): 901-907.

Durchdewald, M., P. Angel, et al. (2009). "The transcription factor Fos: a Janus-type regulator

in health and disease." Histology and Histopathology 24(11): 14451-11461.

Ebelt, N. D., M. A. Cantrell, et al. (2013). "c-Jun N-Terminal Kinases Mediate a Wide Range

of Targets in the Metastatic Cascade." Genes & cancer 4(9-10): 378-387.

Eckstein, N., K. Servan, et al. (2009). "Hyperactivation of the Insulin-like Growth Factor

Receptor I Signaling Pathway Is an Essential Event for Cisplatin Resistance of Ovarian

Cancer Cells." Cancer Research 69(7): 2996-3003.

Egea, J., C. Espinet, et al. (1999). "Calcium Influx Activates Extracellular-regulated

Kinase/Mitogen-activated Protein Kinase Pathway through a Calmodulin-sensitive

Mechanism in PC12 Cells." Journal of Biological Chemistry 274(1): 75-85.

Eklund, E. A. (2007). "The role of HOX genes in malignant myeloid disease." Current Opinion

in Hematology 14(2): 85-89.

Eklund, E. A. (2011). "The Role of Hox Proteins in Leukemogenesis: Insights Into Key

Regulatory Events in Hematopoiesis." Critical Reviews in Oncogenesis 16(1-2, Sp. Iss.

SI): 65-76.

Eklund, E. A., A. Jalava, et al. (2000). "Tyrosine Phosphorylation of HoxA10 Decreases DNA

Binding and Transcriptional Repression during Interferon γ-induced Differentiation of

Myeloid Leukemia Cell Lines." Journal of Biological Chemistry 275(26): 20117-

20126.

El Jamali, A., A. J. Valente, et al. (2010). "Regulation of phagocyte NADPH oxidase by

hydrogen peroxide through a Ca2+/c-Abl signaling pathway." Free Radical Biology

and Medicine 48(6): 798-810.

Elkeles, A., T. Juven-Gershon, et al. (1999). "The c-fos Proto-Oncogene Is a Target for

Transactivation by the p53 Tumor Suppressor." Molecular and Cellular Biology 19(4):

2594-2600.

Elrod, J. W. and J. D. Molkentin (2013). "Physiologic Functions of Cyclophilin D and the

Mitochondrial Permeability Transition Pore." Circulation Journal 77(5): 1111-1122.

Eltzschig, H. K. and P. Carmeliet (2011). "Hypoxia and Inflammation." New England Journal

of Medicine 364(7): 656-665.

Ernst, P., M. Mabon, et al. (2004). "An Mll-Dependent Hox Program Drives Hematopoietic

Progenitor Expansion." Current Biology 14(22): 2063-2069.

Espinosa, A. V., M. Shinohara, et al. (2009). "Regulator of calcineurin 1 modulates cancer cell

migration in vitro." Clinical & Experimental Metastasis 26(6): 517-526.

Estévez, M. D., M. R. Vieytes, et al. (1996). "Mitoxantrone induces nonimmunological

histamine release from rat mast cells." Inflammation Research 45(3): 113-117.

Estey, E. and H. Döhner (2006). "Acute myeloid leukaemia." The Lancet 368(9550): 1894-

1907.

Page 240: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

226

Ethier, C., M. Tardif, et al. (2012). "PARP-1 Modulation of mTOR Signaling in Response to a

DNA Alkylating Agent." PLoS ONE 7(10).

Faber, J., A. V. Krivtsov, et al. (2009). "HOXA9 is required for survival in human MLL-

rearranged acute leukemias." Blood 113(11): 2375-2385.

Falini, B., N. Bolli, et al. (2009). "Altered nucleophosmin transport in acute myeloid leukaemia

with mutated NPM1: molecular basis and clinical implications." Leukemia 23(10):

1731-1743.

Falini, B., C. Mecucci, et al. (2005). "Cytoplasmic Nucleophosmin in Acute Myelogenous

Leukemia with a Normal Karyotype." New England Journal of Medicine 352(3): 254-

266.

Fan, R., S. Bonde, et al. (2012). "Dynamic HoxB4 regulatory network during embryonic stem

cell differentiation to hematopoietic cells." Blood.

Fernandez, H. F. (2010). "New Trends in the Standard of Care for Initial Therapy of Acute

Myeloid Leukemia." Hematology-American Society Hematology Education Program:

56-61.

Ferrando, A. A., S. A. Armstrong, et al. (2003). "Gene expression signatures in MLL-

rearranged T-lineage and B-precursor acute leukemias: dominance of HOX

dysregulation." Blood 102(1): 262-268.

Festjens, N., T. Vanden Berghe, et al. (2006). "Necrosis, a well-orchestrated form of cell

demise: Signalling cascades, important mediators and concomitant immune response."

Biochimica Et Biophysica Acta-Bioenergetics 1757(9-10): 1371-1387.

Fink, S. L. and B. T. Cookson (2005). "Apoptosis, pyroptosis, and necrosis: Mechanistic

description of dead and dying eukaryotic cells." Infection and Immunity 73(4): 1907-

1916.

Fischbach, N. A., S. Rozenfeld, et al. (2005). "HOXB6 overexpression in murine bone marrow

immortalizes a myelomonocytic precursor in vitro and causes hematopoietic stem cell

expansion and acute myeloid leukemia in vivo." Blood 105(4): 1456-1466.

Fiskus, W., S. Verstovsek, et al. (2011). "Heat Shock Protein 90 Inhibitor Is Synergistic with

JAK2 Inhibitor and Overcomes Resistance to JAK2-TKI in Human Myeloproliferative

Neoplasm Cells." Clinical Cancer Research 17(23): 7347-7358.

Flora, S. J. S. and V. Pachauri (2010). "Chelation in Metal Intoxication." International Journal

of Environmental Research and Public Health 7(7): 2745-2788.

Frohling, S., C. Scholl, et al. (2005). "Genetics of myeloid malignancies: pathogenetic and

clinical implications." J Clin Oncol 23: 6285-6295.

Fujino, T., A. Suzuki, et al. (2002). "Single-translocation and double-chimeric transcripts:

detection of NUP98-HOXA9 in myeloid leukemias withHOXA11 or HOXA13 breaks

of the chromosomal translocation t(7;11)(p15;p15)." Blood 99(4): 1428-1433.

Fuller, J. F., J. McAdara, et al. (1999). "Characterization of HOX Gene Expression During

Myelopoiesis: Role of HOX A5 in Lineage Commitment and Maturation." Blood

93(10): 3391-3400.

Galluzzi, L., O. Kepp, et al. (2014). "Molecular mechanisms of regulated necrosis." Seminars

in Cell & Developmental Biology 35(0): 24-32.

Gamberi, G., M. S. Benassi, et al. (1998). "C-myc and c-fos in human osteosarcoma: Prognostic

value of mRNA and protein expression." Oncology 55(6): 556-563.

Garcia-Fernandez, J. (2004). "Hox, ParaHox, ProtoHox: facts and guesses." Heredity 94(2):

145-152.

Garrido, S. M., F. R. Appelbaum, et al. (2001). "Acute myeloid leukemia cells are protected

from spontaneous and drug-induced apoptosis by direct contact with a human bone

marrow stromal cell line (HS-5)." Experimental Hematology 29(4): 448-457.

Gerweck, L. E., S. Vijayappa, et al. (2006). "Tumor pH controls the in vivo efficacy of weak

acid and base chemotherapeutics." Molecular Cancer Therapeutics 5(5): 1275-1279.

Ghibelli, L. and M. Diederich (2010). "Multistep and multitask Bax activation." Mitochondrion

10(6): 604-613.

Page 241: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

227

Giampaolo, A., P. Sterpetti, et al. (1994). "Key functional role and lineage-specific expression

of selected HOXB genes in purified hematopoietic progenitor differentiation." Blood

84(11): 3637-3647.

Gogada, R., V. Prabhu, et al. (2011). "Resveratrol Induces p53-independent, X-linked Inhibitor

of Apoptosis Protein (XIAP)-mediated Bax Protein Oligomerization on Mitochondria

to Initiate Cytochrome c Release and Caspase Activation." Journal of Biological

Chemistry 286(33): 28749-28760.

Golub, T. R., D. K. Slonim, et al. (1999). "Molecular Classification of Cancer: Class Discovery

and Class Prediction by Gene Expression Monitoring." Science 286(5439): 531-537.

Green, D. R. and G. Kroemer (2004). "The Pathophysiology of Mitochondrial Cell Death."

Science 305(5684): 626-629.

Greenblatt, S., L. Li, et al. (2012). "Knock-in of a FLT3/ITD mutation cooperates with a

NUP98-HOXD13 fusion to generate acute myeloid leukemia in a mouse model." Blood

119(12): 2883-2894.

Grier, D. G., A. Thompson, et al. (2005). "The pathophysiology of HOX genes and their role

in cancer." Journal of Pathology 205(2): 154-171.

Grimwade, D., J. V. Jovanovic, et al. (2009). "Prospective Minimal Residual Disease

Monitoring to Predict Relapse of Acute Promyelocytic Leukemia and to Direct Pre-

Emptive Arsenic Trioxide Therapy." Journal of Clinical Oncology 27(22): 3650-3658.

Guinea-Viniegra, J., R. Zenz, et al. (2012). "Differentiation-induced skin cancer suppression

by FOS, p53, and TACE/ADAM17." Journal of Clinical Investigation 122(8): 2898-

2910.

Gwin, K., E. Frank, et al. (2010). "Hoxa9 Regulates Flt3 in Lymphohematopoietic Progenitors."

The Journal of Immunology 185(11): 6572-6583.

Haferlach, T. (2008). "Molecular Genetic Pathways as Therapeutic Targets in Acute Myeloid

Leukemia." ASH Education Program Book 2008(1): 400-411.

Han, J., S. Kim, et al. (2012). "TPA-induced p21 expression augments G2/M arrest through a

p53-independent mechanism in human breast cancer cells." Oncology Reports 27(2):

517-522.

Han, Y. H., H. J. Moon, et al. (2009). "JNK and p38 inhibitors increase and decrease apoptosis,

respectively, in pyrogallol-treated calf pulmonary arterial endothelial cells."

International Journal of Molecular Medicine 24(5): 717-722.

Hantschel, O., W. Warsch, et al. (2012). "BCR-ABL uncouples canonical JAK2-STAT5

signaling in chronic myeloid leukemia." Nature Chemical Biology 8(3): 285-293.

Hanus, J., H. Zhang, et al. (2013). "Induction of necrotic cell death by oxidative stress in retinal

pigment epithelial cells." Cell Death Dis 4: e965.

Hao, N. B., M. H. Lu, et al. (2012). "Macrophages in Tumor Microenvironments and the

Progression of Tumors." Clinical & Developmental Immunology.

Harper, J. W., G. R. Adami, et al. (1993). "THE P21 CDK-INTERACTING PROTEIN CIP1

IS A POTENT INHIBITOR OF G1 CYCLIN-DEPENDENT KINASES." Cell 75(4):

805-816.

Harper, N., M. A. Hughes, et al. (2003). "Protein kinase C modulates tumor necrosis factor-

related apoptosis-inducing ligand-induced apoptosis by targeting the apical events of

death receptor signaling." Journal of Biological Chemistry 278(45): 44338-44347.

Haslam, G., D. Wyatt, et al. (2000). "Estimating the number of viable animal cells in multi-

well cultures based on their lactate dehydrogenase activities." Cytotechnology 32(1):

63-75.

He, H., X. Hua, et al. (2011). "Epigenetic regulations in hematopoietic Hox code." Oncogene

30(4): 379-388.

He, M. X. and Y. W. He (2013). "A role for c-FLIPL in the regulation of apoptosis, autophagy,

and necroptosis in T lymphocytes." Cell Death and Differentiation 20(2): 188-197.

He, R., B. Liu, et al. (2003). "Inhibition of K562 leukemia angiogenesis and growth by

expression of antisense vascular endothelial growth factor (VEGF) sequence." Cancer

Gene Therapy 10(12): 879-886.

Page 242: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

228

He, S., L. Wang, et al. (2009). "Receptor Interacting Protein Kinase-3 Determines Cellular

Necrotic Response to TNF-α." Cell 137(6): 1100-1111.

Heasman, S. A., L. Zaitseva, et al. (2011). "Protection of acute myeloid leukaemia cells from

apoptosis induced by front-line chemotherapeutics is mediated by haem oxygenase-1."

Oncotarget 2(9): 658-668.

Hehlmann, R. (2012). "How I treat CML blast crisis." Blood 120(4): 737-747.

Helen Wheadon, J. M. R., Edwina Dobbin, Glenda J. Dickson, Pamela M. Corrigan, Robin W.

Freeburn, and Alexander Thompson (2011). "Differential Hox Expression in Murine

Embryonic Stem Cell Models of Normal and Malignant Hematopoiesis." STEM

CELLS AND DEVELOPMENT.

Herrmann, H., M. Kneidinger, et al. (2012). "The Hsp32 Inhibitors SMA-ZnPP and PEG-ZnPP

Exert Major Growth-Inhibitory Effects on CD34(+)/CD38(+) and CD34(+)/CD38(-)

AML Progenitor Cells." Current Cancer Drug Targets 12(1): 51-63.

Hess, J. L., C. B. Bittner, et al. (2006). "c-Myb is an essential downstream target for homeobox-

mediated transformation of hematopoietic cells." Blood 108(1): 297-304.

Hess, P., G. Pihan, et al. (2002). "Survival signaling mediated by c-Jun NH2-terminal kinase in

transformed B lymphoblasts." Nat Genet 32(1): 201-205.

Hisa, T., S. E. Spence, et al. (2004). "Hematopoietic, angiogenic and eye defects in Meis1

mutant animals." EMBO J 23(2): 450-459.

Hoeflich, K. P. and M. Ikura (2002). "Calmodulin in Action: Diversity in Target Recognition

and Activation Mechanisms." Cell 108(6): 739-742.

Hole, P. S., R. L. Darley, et al. (2011). Do reactive oxygen species play a role in myeloid

leukemias?

Holler, N., R. Zaru, et al. (2000). "Fas triggers an alternative, caspase-8-independent cell death

pathway using the kinase RIP as effector molecule." Nature Immunology 1(6): 489-

495.

Hollink, I. H. I. M., C. M. Zwaan, et al. (2009). "Favorable prognostic impact of NPM1 gene

mutations in childhood acute myeloid leukemia, with emphasis on cytogenetically

normal AML." Leukemia 23(2): 262-270.

Hourigan, C. S. and J. E. Karp (2013). "Minimal residual disease in acute myeloid leukaemia."

Nature Reviews Clinical Oncology 10(8): 460-471.

Hsu, S.-S., C.-J. Huang, et al. (2007). "Anandamide-induced Ca2+ elevation leading to p38

MAPK phosphorylation and subsequent cell death via apoptosis in human

osteosarcoma cells." Toxicology 231(1): 21-29.

Hu, Y.-L., S. Fong, et al. (2009). "HOXA9 Modulates Its Oncogenic Partner Meis1 To

Influence Normal Hematopoiesis." Molecular and Cellular Biology 29(18): 5181-5192.

Hu, Y.-L., E. Passegué, et al. (2007). "Evidence that the Pim1 kinase gene is a direct target of

HOXA9." Blood 109(11): 4732-4738.

Huang, H. S., Z. M. Liu, et al. (2006). "Opposite effect of ERK1/2 and JNK on p53-independent

p21(WAF1/CIP1)supercript stop activation involved in the arsenic trioxide-induced

human epidermoid carcinoma A431 cellular cytotoxicity." Journal of Biomedical

Science 13(1): 113-125.

Huang, S., V. M. Maher, et al. (1999). "Involvement of Intermediary Metabolites in the

Pathway of Extracellular Ca2+-Induced Mitogen-Activated Protein Kinase Activation

in Human Fibroblasts." Cellular Signalling 11(4): 263-274.

Huang, Y., K. Sitwala, et al. (2012). "Identification and characterization of Hoxa9 binding sites

in hematopoietic cells." Blood 119(2): 388-398.

Huerta, S., E. J. Goulet, et al. (2007). "Screening and Detection of Apoptosis." Journal of

Surgical Research 139(1): 143-156.

Hwang, M. K., Y. K. Min, et al. (2009). "Calmodulin inhibition contributes to sensitize TRAIL-

induced apoptosis in human lung cancer H1299 cells." Biochemistry and Cell Biology-

Biochimie Et Biologie Cellulaire 87(6): 919-926.

Hwang, Y. P., H. J. Yun, et al. (2011). "Suppression of phorbol-12-myristate-13-acetate-

induced tumor cell invasion by piperine via the inhibition of PKC alpha/ERK1/2-

dependent matrix metalloproteinase-9 expression." Toxicology Letters 203(1): 9-19.

Page 243: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

229

Illmer, T., M. Schaich, et al. (2007). "Tyrosine kinase mutations of JAK2 are rare events in

AML but influence prognosis of patients with CBF-leukemias." Haematologica-the

Hematology Journal 92(1): 137-138.

Inaba, H., E. Coustan-Smith, et al. (2012). "Comparative Analysis of Different Approaches to

Measure Treatment Response in Acute Myeloid Leukemia." Journal of Clinical

Oncology 30(29): 3625-3632.

Inoguchi, T., T. Sonta, et al. (2003). "Protein Kinase C–Dependent Increase in Reactive Oxygen

Species (ROS) Production in Vascular Tissues of Diabetes: Role of Vascular NAD(P)H

Oxidase." Journal of the American Society of Nephrology 14(suppl 3): S227-S232.

Ito, T., H. Y. Kwon, et al. (2010). "Regulation of myeloid leukaemia by the cell-fate

determinant Musashi." Nature 466(7307): 765-U713.

Itoh, T., R. Terazawa, et al. (2011). "Cisplatin induces production of reactive oxygen species

via NADPH oxidase activation in human prostate cancer cells." Free Radical Research

45(9): 1033-1039.

Jan, C.-R., J.-A. Su, et al. (2013). "Mechanism of maprotiline-induced apoptosis: Role of

[Ca2+]i, ERK, JNK and caspase-3 signaling pathways." Toxicology 304(0): 1-12.

Janssen, K., S. Pohlmann, et al. (2007). "Apaf-1 and caspase-9 deficiency prevents apoptosis

in a Bax-controlled pathway and promotes clonogenic survival during paclitaxel

treatment." Blood 110(10): 3662-3672.

Jiang, F., Y. Zhang, et al. (2011). "NADPH Oxidase-Mediated Redox Signaling: Roles in

Cellular Stress Response, Stress Tolerance, and Tissue Repair." Pharmacological

Reviews 63(1): 218-242.

Jiang, P., W. Du, et al. (2006). "The bad guy cooperates with good cop p53: Bad is

transcriptionally up-regulated by p53 and forms a bad/p53 complex at the mitochondria

to induce apoptosis." Molecular and Cellular Biology 26(23): 9071-9082.

Jin, E. L., B. Zhang, et al. (2013). "Acid-Active Cell-Penetrating Peptides for in Vivo Tumor-

Targeted Drug Delivery." Journal of the American Chemical Society 135(2): 933-940.

Jin, Z. Y. and W. S. El-Deiry (2005). "Overview of cell death signaling pathways." Cancer

Biology & Therapy 4(2): 139-163.

Jing, G., K. Y. Yuan, et al. (2012). "Reduced CaM/FLIP binding by a single point mutation in

c-FLIPL modulates Fas-mediated apoptosis and decreases tumorigenesis." Laboratory

Investigation 92(1): 82-90.

Jing, G., K. Y. Yuan, et al. (2011). "Tamoxifen enhances therapeutic effects of gemcitabine on

cholangiocarcinoma tumorigenesis." Laboratory Investigation 91(6): 896-904.

Jobe, S. M., K. M. Wilson, et al. (2008). "Critical role for the mitochondrial permeability

transition pore and cyclophilin D in platelet activation and thrombosis." Blood 111(3):

1257-1265.

Jochen, F. A., J. Richter, et al. (2002). "Fructose inhibits apoptosis induced by reoxygenation

in rat hepatocytes by decreasing reactive oxygen species via stabilization of the

glutathione pool." Biochimica Et Biophysica Acta-Molecular Cell Research 1542(1-

3): 82-94.

Jog, N. R., J.-A. Dinnall, et al. (2009). "Poly(ADP-Ribose) Polymerase-1 Regulates the

Progression of Autoimmune Nephritis in Males by Inducing Necrotic Cell Death and

Modulating Inflammation." The Journal of Immunology 182(11): 7297-7306.

Johnson, C. L., D. M. Lu, et al. (2002). "Regulation of p53 stabilization by DNA damage and

protein kinase C." Molecular Cancer Therapeutics 1(10): 861-867.

Jouan-Lanhouet, S., M. I. Arshad, et al. (2012). "TRAIL induces necroptosis involving

RIPK1/RIPK3-dependent PARP-1 activation." Cell Death and Differentiation 19(12):

2003-2014.

Jozkowicz, A., H. Was, et al. (2007). "Heme oxygenase-1 in tumors: Is it a false friend?"

Antioxidants & Redox Signaling 9(12): 2099-2117.

Jude, C. D., L. Climer, et al. (2007). "Unique and Independent Roles for MLL in Adult

Hematopoietic Stem Cells and Progenitors." Cell stem cell 1(3): 324-337.

Kadowaki, H., H. Nishitoh, et al. (2005). "Amyloid beta induces neuronal cell death through

ROS-mediated ASK1 activation." Cell Death and Differentiation 12(1): 19-24.

Page 244: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

230

Kagawa, S., J. Go, et al. (2001). "Deficiency of caspase-3 in MCF7 cells blocks bax-mediated

nuclear fragmentation but not cell death." Clinical Cancer Research 7(5): 1474-1480.

Kahl, C., M. Mielcarek, et al. (2004). "Radiation dose determines the degree of myeloid

engraftment after nonmyeloablative stem cell transplantation." Biology of Blood and

Marrow Transplantation 10(12): 826-833.

Kalra, N. and V. Kumar (2004). "c-Fos Is a Mediator of the c-Myc-induced Apoptotic Signaling

in Serum-deprived Hepatoma Cells via the p38 Mitogen-activated Protein Kinase

Pathway." Journal of Biological Chemistry 279(24): 25313-25319.

Kappen, C. (2000). "Disruption of the homeobox gene Hoxb-6 in mice results in increased

numbers of early erythrocyte progenitors." American Journal of Hematology 65(2):

111-118.

Kapuscinski, J. (1995). "DAPI - A DNA-SPECIFIC FLUORESCENT-PROBE." Biotechnic &

Histochemistry 70(5): 220-233.

Kar, P., T. Chakraborti, et al. (2009). "μ-Calpain mediated cleavage of the Na+/Ca2+ exchanger

in isolated mitochondria under A23187 induced Ca2+ stimulation." Archives of

Biochemistry and Biophysics 482(1–2): 66-76.

Karl, S., Y. Pritschow, et al. (2009). "Identification of a novel pro-apopotic function of NF-

kappa B in the DNA damage response." Journal of Cellular and Molecular Medicine

13(10): 4239-4256.

Kato, I., A. Niwa, et al. (2011). "Identification of Hepatic Niche Harboring Human Acute

Lymphoblastic Leukemic Cells via the SDF-1/CXCR4 Axis." PLoS ONE 6(11).

Kaunisto, A., V. Kochin, et al. (2009). "PKC-mediated phosphorylation regulates c-FLIP

ubiquitylation and stability." Cell Death and Differentiation 16(9): 1215-1226.

Kawagoe, H., R. K. Humphries, et al. (1999). "Expression of HOX genes, HOX cofactors, and

MLL in phenotypically and functionally defined subpopulations of leukemic and

normal human hematopoietic cells." Leukemia 13(5): 687-698.

Ke, H. N., R. Harris, et al. (2010). "The c-Jun NH2-Terminal Kinase 2 Plays a Dominant Role

in Human Epidermal Neoplasia." Cancer Research 70(8): 3080-3088.

Kim, H. J., J. H. Lee, et al. (2010). "Roles of NADPH Oxidases in Cisplatin-Induced Reactive

Oxygen Species Generation and Ototoxicity." Journal of Neuroscience 30(11): 3933-

3946.

Kim, K., J. Song, et al. (2007). "NADPH oxidase inhibitor diphenyleneiodonium induces ROS-

independent p53 expression and apoptosis in human RPE cells." Febs Journal 274: 154-

154.

Kim, M. Y., T. Zhang, et al. (2005). "Poly(ADP-ribosyl)ation by PARP-1: 'PAR-laying'

NAD(+) into a nuclear signal." Genes & Development 19(17): 1951-1967.

Kim, Y.-H., J.-W. Park, et al. (2003). "Bcl-2 overexpression prevents daunorubicin-induced

apoptosis through inhibition of XIAP and Akt degradation." Biochemical

Pharmacology 66(9): 1779-1786.

Kishi, K. (1985). "A NEW LEUKEMIA-CELL LINE WITH PHILADELPHIA-

CHROMOSOME CHARACTERIZED AS BASOPHIL PRECURSORS." Leukemia

Research 9(3): 381-&.

Kitamura, C., Y. Ogawa, et al. (2003). "Transient co-localization of c-Jun N-terminal kinase

and c-Jun with heat shock protein 70 in pulp cells during apoptosis." Journal of Dental

Research 82(2): 91-95.

Knittel, T., M. Kessel, et al. (1995). "A conserved enhancer of the human and murine Hoxa-7

gene specifies the anterior boundary of expression during embryonal development."

Development 121(4): 1077-1088.

Ko, K.-H., Q. L. Kwan Lam, et al. (2007). "Hoxb3 deficiency impairs B lymphopoiesis in

mouse bone marrow." Experimental Hematology 35(3): 465-475.

Koeffler, H. P. and D. W. Golde (1978). "ACUTE MYELOGENOUS LEUKEMIA - HUMAN

CELL LINE RESPONSIVE TO COLONY-STIMULATING ACTIVITY." Science

200(4346): 1153-1154.

Kok, C. H., A. L. Brown, et al. (2010). "Gene expression analysis reveals HOX gene

upregulation in trisomy 8 AML." Leukemia 24(6): 1239-1243.

Page 245: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

231

Kollet, O., S. Shivtiel, et al. (2003). "HGF, SDF-1, and MMP-9 are involved in stress-induced

human CD34(+) stem cell recruitment to the liver." Journal of Clinical Investigation

112(2): 160-169.

Kollet, O., A. Spiegel, et al. (2001). "Rapid and efficient homing of human CD34(+)CD38(-

/low)CXCR4(+) stem and progenitor cells to the bone marrow and spleen of

NOD/SCID and NOD/SCID/B2m(null) mice." Blood 97(10): 3283-3291.

Konopleva, M., S. Konoplev, et al. (2002). "Stromal cells prevent apoptosis of AML cells by

up-regulation of anti-apoptotic proteins." Leukemia 16(9): 1713-1724.

Koo, S., B. J. Huntly, et al. (2010). "Cdx4 is dispensable for murine adult hematopoietic stem

cells but promotes MLL-AF9-mediated leukemogenesis." Haematologica 95(10):

1642-1650.

Koopman, G., C. Reutelingsperger, et al. (1994). "Annexin V for flow cytometric detection of

phosphatidylserine expression on B cells undergoing apoptosis." Blood 84(5): 1415-

1420.

Kristensen, T., M. B. Møller, et al. (2011). "NPM1 mutation is a stable marker for minimal

residual disease monitoring in acute myeloid leukaemia patients with increased

sensitivity compared to WT1 expression*." European Journal of Haematology 87(5):

400-408.

Krivtsov, A. V., Z. Feng, et al. (2008). "H3K79 Methylation Profiles Define Murine and Human

MLL-AF4 Leukemias." Cancer Cell 14(5): 355-368.

Krivtsov, A. V., D. Twomey, et al. (2006). "Transformation from committed progenitor to

leukaemia stem cell initiated by MLL-AF9." Nature 442(7104): 818-822.

Kroon, E., J. Krosl, et al. (1998). "Hoxa9 transforms primary bone marrow cells through

specific collaboration with Meis1a but not Pbx1b." EMBO J 17(13): 3714-3725.

Kroon, E., U. Thorsteinsdottir, et al. (2001). "NUP98-HOXA9 expression in hemopoietic stem

cells induces chronic and acute myeloid leukemias in mice." Embo Journal 20(3): 350-

361.

Krosl, J., N. Beslu, et al. (2003). "The Competitive Nature of HOXB4-Transduced HSC Is

Limited by PBX1: The Generation of Ultra-Competitive Stem Cells Retaining Full

Differentiation Potential." Immunity 18(4): 561-571.

Krosl, J. and G. Sauvageau (2000). "AP-1 complex is effector of Hox-induced cellular

proliferation and transformation." Oncogene 19(45): 5134-5141.

Krysko, O., L. de Ridder, et al. (2004). "Phosphatidylserine exposure during early primary

necrosis (oncosis) in JB6 cells as evidenced by immunogold labeling technique."

Apoptosis 9(4): 495-500.

Kwon, Y. H., A. Jovanovic, et al. (2003). "The Cdk inhibitor p21 is required for necrosis, but

it inhibits apoptosis following toxin-induced liver injury." Journal of Biological

Chemistry 278(32): 30348-30355.

Lafarga, V., A. Cuadrado, et al. (2009). "p38 Mitogen-Activated Protein Kinase- and HuR-

Dependent Stabilization of p21(Cip1) mRNA Mediates the G(1)/S Checkpoint."

Molecular and Cellular Biology 29(16): 4341-4351.

Lallemend, F., P. P. Lefebvre, et al. (2003). "Substance P protects spiral ganglion neurons from

apoptosis via PKC-Ca2+-MAPK/ERK pathways." Journal of Neurochemistry 87(2):

508-521.

Lambeth, J. D. (2004). "NOX enzymes and the biology of reactive oxygen." Nat Rev Immunol

4(3): 181-189.

Lanzi, C., R. A. Gambetta, et al. (1991). "PROTEIN-KINASE-C ACTIVATION BY

ANTHRACYCLINES IN SWISS 3T3 CELLS." International Journal of Cancer 47(1):

136-142.

Larson, S. M., N. P. Campbell, et al. (2012). "High dose cytarabine and mitoxantrone: an

effective induction regimen for high-risk Acute Myeloid Leukemia (AML)." Leukemia

& Lymphoma 53(3): 445-450.

Lasbury, M. E., P. J. Durant, et al. (2009). "Effects of Decreased Calmodulin Protein on the

Survival Mechanisms of Alveolar Macrophages during Pneumocystis Pneumonia."

Infection and Immunity 77(8): 3344-3354.

Page 246: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

232

Latta, M., G. Künstle, et al. (2000). "Metabolic Depletion of Atp by Fructose Inversely Controls

Cd95- and Tumor Necrosis Factor Receptor 1–Mediated Hepatic Apoptosis." The

Journal of Experimental Medicine 191(11): 1975-1986.

Latta, M., G. Künstle, et al. (2007). "ATP-Depleting Carbohydrates Prevent Tumor Necrosis

Factor Receptor 1-Dependent Apoptotic and Necrotic Liver Injury in Mice." Journal of

Pharmacology and Experimental Therapeutics 321(3): 875-883.

Laurent, G. and J.-P. Jaffrézou (2001). "Signaling pathways activated by daunorubicin." Blood

98(4): 913-924.

Lawrence, H. J., J. Christensen, et al. (2005). "Loss of expression of the Hoxa-9 homeobox

gene impairs the proliferation and repopulating ability of hematopoietic stem cells."

Blood 106(12): 3988-3994.

Lecoeur, H., L. M. de Oliveira-Pinto, et al. (2002). "Multiparametric flow cytometric analysis

of biochemical and functional events associated with apoptosis and oncosis using the

7-aminoactinomycin D assay." Journal of Immunological Methods 265(1-2): 81-96.

Lecoeur, H., M. C. Prevost, et al. (2001). "Oncosis is associated with exposure of

phosphatidylserine residues on the outside layer of the plasma membrane: A

reconsideration of the specificity of the annexin V/Propidium iodide assay." Cytometry

44(1): 65-72.

Lee, B. W., S. W. Chun, et al. (2011). "Lithospermic acid B protects beta-cells from cytokine-

induced apoptosis by alleviating apoptotic pathways and activating anti-apoptotic

pathways of Nrf2-HO-1 and Sirt1." Toxicology and Applied Pharmacology 252(1): 47-

54.

Lee, H., H. Lee, et al. (2014). "ERBB3 knockdown induces cell cycle arrest and activation of

Bak and Bax-dependent apoptosis in colon cancer cells." Oncotarget 5(13): 5138-5152.

Lee, H. M., H. Zhang, et al. (2010). "Downstream targets of HOXB4 in a cell line model of

primitive hematopoietic progenitor cells." Blood 116(5): 720-730.

Lee, S. E., H. Yang, et al. (2012). "Induction of Heme Oxygenase-1 Inhibits Cell Death in

Crotonaldehyde-Stimulated HepG2 Cells via the PKC-delta-p38-Nrf2 Pathway." PLoS

ONE 7(7).

Lee, Y. K., T. D. Shanafelt, et al. (2005). "VEGF receptors on chronic lymphocytic leukemia

(CLL) B cells interact with STAT 1 and 3: implication for apoptosis resistance."

Leukemia 19(4): 513-523.

Leist, M., B. Single, et al. (1999). "Inhibition of mitochondrial ATP generation by nitric oxide

switches apoptosis to necrosis." Experimental Cell Research 249(2): 396-403.

Lerch, M. M., W. Halangk, et al. (2013). "Preventing Pancreatitis by Protecting the

Mitochondrial Permeability Transition Pore." Gastroenterology 144(2): 265-269.

Leu, J. I. J., P. Dumont, et al. (2004). "Mitochondrial p53 activates Bak and causes disruption

of a Bak-Mcl1 complex." Nature Cell Biology 6(5): 443-450.

Leverson, J. D., P. J. Koskinen, et al. (1998). "Pim-1 Kinase and p100 Cooperate to Enhance

c-Myb Activity." Molecular Cell 2(4): 417-425.

Li, D.-P., Z.-Y. Li, et al. (2013). "HOXA9 Gene Expression in Acute Myeloid Leukemia." Cell

Biochemistry and Biophysics 67(3): 935-938.

Li, G., X. Li, et al. (2014). "CD123 targeting oncolytic adenoviruses suppress acute myeloid

leukemia cell proliferation in vitro and in vivo." Blood Cancer Journal 4.

Li, H., H. Zhu, et al. (1998). "Cleavage of BID by Caspase 8 Mediates the Mitochondrial

Damage in the Fas Pathway of Apoptosis." Cell 94(4): 491-501.

Li, J., D. P. Sejas, et al. (2007). "TNF-alpha induces leukemic clonal evolution ex vivo in

Fanconi anemia group C murine stem cells." Journal of Clinical Investigation 117(11):

3283-3295.

Li, N. Y., K. Ragheb, et al. (2003). "DPI induces mitochondrial superoxide-mediated

apoptosis." Free Radical Biology and Medicine 34(4): 465-477.

Li, Y., N. Johnson, et al. (2004). "Cyclophilin-D promotes the mitochondrial permeability

transition but has opposite effects on apoptosis and necrosis." Biochem. J. 383(1): 101-

109.

Page 247: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

233

Li, Z., H. Huang, et al. (2012). "Up-regulation of a HOXA-PBX3 homeobox-gene signature

following down-regulation of miR-181 is associated with adverse prognosis in patients

with cytogenetically abnormal AML." Blood 119(10): 2314-2324.

Lin, X., A. O'Mahony, et al. (2000). "Protein Kinase C-θ Participates in NF-κB Activation

Induced by CD3-CD28 Costimulation through Selective Activation of IκB Kinase β."

Molecular and Cellular Biology 20(8): 2933-2940.

Liu, F. T., S. M. Kelsey, et al. (2002). "Liposomal encapsulation diminishes daunorubicin-

induced generation of reactive oxygen species, depletion of ATP and necrotic cell death

in human leukaemic cells." British Journal of Haematology 117(2): 333-342.

Liu, H., E. H. Y. Cheng, et al. (2009). "MLL fusions Pathways to leukemia." Cancer Biology

& Therapy 8(13): 1204-1211.

Liu, H., H. Nishitoh, et al. (2000). "Activation of apoptosis signal-regulating kinase 1 (ASK1)

by tumor necrosis factor receptor-associated factor 2 requires prior dissociation of the

ASK1 inhibitor thioredoxin." Molecular and Cellular Biology 20(6): 2198-2208.

Liu, L., D. Xing, et al. (2009). "mu-Calpain regulates caspase-dependent and apoptosis

inducing factor-mediated caspase-independent apoptotic pathways in cisplatin-induced

apoptosis." International Journal of Cancer 125(12): 2757-2766.

Liu, X. L., T. Van Vleet, et al. (2004). "The role of calpain in oncotic cell death." Annual

Review of Pharmacology and Toxicology 44: 349-370.

Liu, Z. M. and H. S. Huang (2006). "As2O3-induced c-Src/EGFR/ERK signaling is via Sp1

binding sites to stimulate p21(WAF1/CIP1) expression in human epidermoid

carcinoma A431 cells." Cellular Signalling 18(2): 244-255.

Liu, Z. M. and H. S. Huang (2008). "Arsenic trioxide phosphorylates c-Fos to transactivate

p21(WAF1/CIP1) expression." Toxicology and Applied Pharmacology 233(2): 297-

307.

Livak, K. J. and T. D. Schmittgen (2001). "Analysis of Relative Gene Expression Data Using

Real-Time Quantitative PCR and the 2−ΔΔCT Method." Methods 25(4): 402-408.

Lonic, A., J. A. Powell, et al. (2013). "Phosphorylation of Serine 779 in Fibroblast Growth

Factor Receptor 1 and 2 by Protein Kinase C epsilon Regulates Ras/Mitogen-activated

Protein Kinase Signaling and Neuronal Differentiation." Journal of Biological

Chemistry 288(21): 14874-14885.

Lopez, A. V., M. N. V. Garcia, et al. (2014). "Mesenchymal Stromal Cells Derived from the

Bone Marrow of Acute Lymphoblastic Leukemia Patients Show Altered BMP4

Production: Correlations with the Course of Disease." PLoS ONE 9(1).

Los, M., M. Mozoluk, et al. (2002). "Activation and caspase-mediated inhibition of PARP: A

molecular switch between fibroblast necrosis and apoptosis in death receptor

signaling." Molecular Biology of the Cell 13(3): 978-988.

Lozzio, C. B. and B. B. Lozzio (1975). "HUMAN CHRONIC MYELOGENOUS LEUKEMIA

CELL-LINE WITH POSITIVE PHILADELPHIA CHROMOSOME." Blood 45(3):

321-334.

Luo, Y., P. Zou, et al. (2011). "Autophagy regulates ROS-induced cellular senescence via p21

in a p38 MAPK alpha dependent manner." Experimental Gerontology 46(11): 860-867.

Lutzny, G., T. Kocher, et al. (2013). "Protein Kinase C-β-Dependent Activation of NF-κB in

Stromal Cells Is Indispensable for the Survival of Chronic Lymphocytic Leukemia B

Cells In Vivo." Cancer Cell 23(1): 77-92.

Magnusson, M., A. C. M. Brun, et al. (2007). "Hoxa9/hoxb3/hoxb4 compound null mice

display severe hematopoietic defects." Experimental Hematology 35(9): 1421.e1421-

1421.e1429.

Magnusson, M., A. C. M. Brun, et al. (2007). "HOXA10 is a critical regulator for hematopoietic

stem cells and erythroid/megakaryocyte development." Blood 109(9): 3687-3696.

Mahner, S., C. Baasch, et al. (2008). "C-Fos expression is a molecular predictor of progression

and survival in epithelial ovarian carcinoma." British Journal of Cancer 99(8): 1269-

1275.

Page 248: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

234

Mahoney, B. P., N. Raghunand, et al. (2003). "Tumor acidity, ion trapping and

chemotherapeutics I. Acid pH affects the distribution of chemotherapeutic agents in

vitro." Biochemical Pharmacology 66(7): 1207-1218.

Mandal, D., P. K. Moitra, et al. (2002). "Caspase 3 regulates phosphatidylserine externalization

and phagocytosis of oxidatively stressed erythrocytes." FEBS letters 513(2–3): 184-

188.

Mandic, A., K. Viktorsson, et al. (2002). "Calpain-mediated bid cleavage and calpain-

independent Bak modulation: Two separate pathways in cisplatin-induced apoptosis."

Molecular and Cellular Biology 22(9): 3003-3013.

Mann, R. S., K. M. Lelli, et al. (2009). Chapter 3 Hox Specificity: Unique Roles for Cofactors

and Collaborators. Current Topics in Developmental Biology, Academic Press.

Volume 88: 63-101.

Marcelli, M., G. R. Cunningham, et al. (1999). "Signaling Pathway Activated during Apoptosis

of the Prostate Cancer Cell Line LNCaP: Overexpression of Caspase-7 as a New Gene

Therapy Strategy for Prostate Cancer." Cancer Research 59(2): 382-390.

Marcucci, G., K. Mrozek, et al. (2005). "Molecular heterogeneity and prognostic biomarkers in

adults with acute myeloid leukemia and normal cytogenetics." Current Opinion in

Hematology 12(1): 68-75.

Marone, M., S. Mozzetti, et al. (2001). "Semiquantitative RT-PCR analysis to assess the

expression levels of multiple transcripts from the same sample." Biological Procedures

Online 3(1 Cited Dec 17, 2001): 19-25.

Marone, M., G. Scambia, et al. (2002). "Transforming growth factor-beta 1 transcriptionally

activates CD34 and prevents induced differentiation of TF-1 cells in the absence of any

cell-cycle effects." Leukemia 16(1): 94-105.

Marschalek, R. (2011). "Mechanisms of leukemogenesis by MLL fusion proteins." British

Journal of Haematology 152(2): 141-154.

Marshall, K. and C. Baines (2014). "Necroptosis: Is There a Role for Mitochondria?" Frontiers

in Physiology 5.

Martin, L. J. (2010). "Mitochondrial and Cell Death Mechanisms in Neurodegenerative

Diseases." Pharmaceuticals 3(4): 839-915.

Martin, P. and T. Papayannopoulou (1982). "HEL CELLS - A NEW HUMAN

ERYTHROLEUKEMIA CELL-LINE WITH SPONTANEOUS AND INDUCED

GLOBIN EXPRESSION." Science 216(4551): 1233-1235.

Mas, V. M.-D., H. Hernandez, et al. (2003). "Protein kinase Cζ mediated Raf-1/extracellular-

regulated kinase activation by daunorubicin." Blood 101(4): 1543-1550.

Masgras, I., S. Carrera, et al. (2012). "Reactive Oxygen Species and Mitochondrial Sensitivity

to Oxidative Stress Determine Induction of Cancer Cell Death by p21." Journal of

Biological Chemistry 287(13): 9845-9854.

Mathiasen, I. S., I. N. Sergeev, et al. (2002). "Calcium and calpain as key mediators of

apoptosis-like death induced by vitamin D compounds in breast cancer cells." Journal

of Biological Chemistry 277(34): 30738-30745.

Mayotte, N., D. C. Roy, et al. (2002). "Oncogenic interaction between BCR-ABL and NUP98-

HOXA9 demonstrated by the use of an in vitro purging culture system." Blood 100(12):

4177-4184.

McClung, J. M., A. R. Judge, et al. (2009). "Calpain-1 is required for hydrogen peroxide-

induced myotube atrophy." American Journal of Physiology-Cell Physiology 296(2):

C363-C371.

Medina-Martinez, O. and R. Ramirez-Solis (2003). "In vivo mutagenesis of the Hoxb8

hexapeptide domain leads to dominant homeotic transformations that mimic the loss-

of-function mutations in genes of the Hoxb cluster." Developmental Biology 264(1):

77-90.

Meng, J., H. H. Zhang, et al. (2012). "The histone deacetylase inhibitor trichostatin A induces

cell cycle arrest and apoptosis in colorectal cancer cells via p53-dependent and -

independent pathways." Oncology Reports 28(1): 384-388.

Page 249: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

235

Meyer, C., E. Kowarz, et al. (2009). "New insights to the MLL recombinome of acute

leukemias." Leukemia 23(8): 1490-1499.

Miao, B. C. and A. Degterev (2009). "Methods to Analyze Cellular Necroptosis." Apoptosis:

Methods and Protocols, Second Edition 559: 79-93.

Mielcarek, M., B. Torok-Storb, et al. (2011). "Pharmacological Immunosuppression Reduces

But Does Not Eliminate the Need for Total-Body Irradiation in Nonmyeloablative

Conditioning Regimens for Hematopoietic Cell Transplantation." Biology of Blood

and Marrow Transplantation 17(8): 1255-1260.

Mihara, M., S. Erster, et al. (2003). "p53 Has a Direct Apoptogenic Role at the Mitochondria."

Molecular Cell 11(3): 577-590.

Mikula, M., J. Gotzmann, et al. (2003). "The proto-oncoprotein c-Fos negatively regulates

hepatocellular tumorigenesis." Oncogene 22(43): 6725-6738.

Miller, G. M., B. A. Ogunnaike, et al. (2010). "Robust dynamic balance of AP-1 transcription

factors in a neuronal gene regulatory network." Bmc Systems Biology 4.

Milne, T. A., S. D. Briggs, et al. (2002). "MLL Targets SET Domain Methyltransferase Activity

to Hox Gene Promoters." Molecular Cell 10(5): 1107-1117.

Minotti, G., P. Menna, et al. (2004). "Anthracyclines: Molecular advances and pharmacologic

developments in antitumor activity and cardiotoxicity." Pharmacological Reviews

56(2): 185-229.

Mitchell, D. J., L. Steinman, et al. (2000). "Polyarginine enters cells more efficiently than other

polycationic homopolymers." The Journal of Peptide Research 56(5): 318-325.

Mochizuki, T., S. Furuta, et al. (2006). "Inhibition of NADPH oxidase 4 activates apoptosis via

the AKT/apoptosis signal-regulating kinase 1 pathway in pancreatic cancer PANC-1

cells." Oncogene 25(26): 3699-3707.

Moens, C. B. and L. Selleri (2006). "Hox cofactors in vertebrate development." Developmental

Biology 291(2): 193-206.

Mohle, R., M. Schittenhelm, et al. (2000). "Functional response of leukaemic blasts to stromal

cell-derived factor-1 correlates with preferential expression of the chemokine receptor

CXCR4 in acute myelomonocytic and lymphoblastic leukaemia." British Journal of

Haematology 110(3): 563-572.

Montero, J., C. Dutta, et al. (2013). "p53 regulates a non-apoptotic death induced by ROS."

Cell Death and Differentiation 20(11): 1465-1474.

Morgan, M. J., Y.-S. Kim, et al. (2008). "TNF[alpha] and reactive oxygen species in necrotic

cell death." Cell Research 18(3): 343-349.

Morgan, R., A. Boxall, et al. (2012). "Targeting the HOX/PBX dimer in breast cancer." Breast

Cancer Research and Treatment 136(2): 389-398.

Morgan, R., A. Boxall, et al. (2014). "Targeting HOX transcription factors in prostate cancer."

BMC urology 14: 17-17.

Morgan, R., P. M. Pirard, et al. (2007). "Antagonism of HOX/PBX dimer formation blocks the

in vivo proliferation of melanoma." Cancer Research 67(12): 5806-5813.

Morgan, R., L. Plowright, et al. (2010). "Targeting HOX and PBX transcription factors in

ovarian cancer." BMC Cancer 10(1): 89.

Motiwala, T., S. Majumder, et al. (2009). "PTPROt Inactivates the Oncogenic Fusion Protein

BCR/ABL and Suppresses Transformation of K562 Cells." Journal of Biological

Chemistry 284(1): 455-464.

Mrozek, K. (2008). "Cytogenetic, molecular genetic, and clinical characteristics of acute

myeloid leukemia with a complex karyotype." Seminars in Oncology 35(4): 365-377.

Mrozek, K., G. Marcucci, et al. (2012). "Prognostic Significance of the European LeukemiaNet

Standardized System for Reporting Cytogenetic and Molecular Alterations in Adults

With Acute Myeloid Leukemia." Journal of Clinical Oncology 30(36): 4515-4523.

Mrózek, K., G. Marcucci, et al. (2007). "Clinical relevance of mutations and gene-expression

changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a

prognostically prioritized molecular classification?" Blood 109(2): 431-448.

Page 250: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

236

Mrózek, K., S. M. Tanner, et al. (2003). "Molecular cytogenetic characterization of the KG-1

and KG-1a acute myeloid leukemia cell lines by use of spectral karyotyping and

fluorescence in situ hybridization." Genes, Chromosomes and Cancer 38(3): 249-252.

Mullighan, C. G., A. Kennedy, et al. (2007). "Pediatric acute myeloid leukemia with NPM1

mutations is characterized by a gene expression profile with dysregulated HOX gene

expression distinct from MLL-rearranged leukemias." Leukemia 21(9): 2000-2009.

Muscella, A., L. Urso, et al. (2009). "Anti-apoptotic effects of protein kinase C-delta and c-fos

in cisplatin-treated thyroid cells." British Journal of Pharmacology 156(5): 751-763.

Mut, M., S. Amos, et al. (2010). "PKC Alpha Phosphorylates Cytosolic NF-kappaB/p65 and

PKC Delta Delays Nuclear Trans location of NF-kappaB/p65 in U1242 Glioblastoma

Cells." Turkish Neurosurgery 20(3): 277-285.

Nagel, S., L. Venturini, et al. (2010). "Polycomb repressor complex 2 regulates HOXA9 and

HOXA10, activating ID2 in NK/T-cell lines." Molecular Cancer 9(1): 151.

Nakagawa, T., S. Shimizu, et al. (2005). "Cyclophilin D-dependent mitochondrial permeability

transition regulates some necrotic but not apoptotic cell death." Nature 434(7033): 652-

658.

Nakamura, T., D. A. Largaespada, et al. (1996). "Fusion of the nucleoporin gene NUP98 to

HOXA9 by the chromosome translocation t(7;11)(p15;p15) in human myeloid

leukaemia." Nat Genet 12(2): 154-158.

Nakayama, I., M. Shibazaki, et al. (2013). "Loss of HOXD10 expression induced by

upregulation of miR-10b accelerates the migration and invasion activities of ovarian cancer

cells." International Journal of Oncology 43(1): 63-71.

Neuhaus, O., B. C. Kieseier, et al. (2004). "Mechanisms of mitoxantrone in multiple sclerosis–

what is known?" Journal of the Neurological Sciences 223(1): 25-27.

Nguyen, C. N., H. E. Kim, et al. (2013). "Caffeoylserotonin Protects Human Keratinocyte

HaCaT Cells against H2O2-Induced Oxidative Stress and Apoptosis through

Upregulation of HO-1 Expression via Activation of the PI3K/Akt/Nrf2 Pathway."

Phytotherapy Research 27(12): 1810-1818.

Nieminen, A. L., A. K. Saylor, et al. (1994). "ATP depletion rather than mitochondrial

depolarization mediates hepatocyte killing after metabolic inhibition." American

Journal of Physiology - Cell Physiology 267(1): C67-C74.

Nithianandarajah-Jones, G. N., B. Wilm, et al. (2012). "ERK5: Structure, regulation and

function." Cellular Signalling 24(11): 2187-2196.

Nör, J. E., J. Christensen, et al. (1999). "Vascular Endothelial Growth Factor (VEGF)-Mediated

Angiogenesis Is Associated with Enhanced Endothelial Cell Survival and Induction of

Bcl-2 Expression." The American Journal of Pathology 154(2): 375-384.

O'Donnell, M. R., C. N. Abboud, et al. (2012). "Acute Myeloid Leukemia." Journal of the

National Comprehensive Cancer Network 10(8): 984-1021.

O'Hare, T., M. S. Zabriskie, et al. (2012). "Pushing the limits of targeted therapy in chronic

myeloid leukaemia." Nat Rev Cancer 12(8): 513-526.

Okada, S., T. Fukuda, et al. (1999). "Prolonged expression of c-fos suppresses cell cycle entry

of dormant hematopoietic stem cells." Blood 93(3): 816-825.

Olesen, L. H., C. G. Nyvold, et al. (2005). "Delineation and molecular characterization of acute

myeloid leukemia patients with coduplication of FLT3 and MLL." European Journal

of Haematology 75(3): 185-192.

Oliveira-Ferrer, L., K. Rossler, et al. (2014). "c-FOS suppresses ovarian cancer progression by

changing adhesion." British Journal of Cancer 110(3): 753-763.

Ono, R., H. Kumagai, et al. (2009). "Mixed-lineage-leukemia (MLL) fusion protein

collaborates with Ras to induce acute leukemia through aberrant Hox expression and

Raf activation." Leukemia 23(12): 2197-2209.

Orkin, S. H. and L. I. Zon (2008). "Hematopoiesis: An evolving paradigm for stem cell

biology." Cell 132(4): 631-644.

Page 251: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

237

Oshima, M., M. Endoh, et al. (2011). "Genome-wide analysis of target genes regulated by

HoxB4 in hematopoietic stem and progenitor cells developing from embryonic stem

cells." Blood 117(15): e142-e150.

Ossenkoppele, G. and G. J. Schuurhuis (2013). "MRD in AML: time for redefinition of CR?"

Blood 121(12): 2166-2168.

Ostergaard, M., L. H. Olesen, et al. (2004). "WT1 gene expression: an excellent tool for

monitoring minimal residual disease in 70% of acute myeloid leukaemia patients -

results from a single-centre study." British Journal of Haematology 125(5): 590-600.

Ouyang, L., Z. Shi, et al. (2012). "Programmed cell death pathways in cancer: a review of

apoptosis, autophagy and programmed necrosis." Cell Proliferation 45(6): 487-498.

Pabla, N., G. Dong, et al. (2011). "Inhibition of PKCδ reduces cisplatin-induced nephrotoxicity

without blocking chemotherapeutic efficacy in mouse models of cancer." The Journal

of Clinical Investigation 121(7): 2709-2722.

Palandri, F., F. Castagnetti, et al. (2008). "Chronic myeloid leukemia in blast crisis treated with

imatinib 600 mg: outcome of the patients alive after a 6-year follow-up."

Haematologica-the Hematology Journal 93(12): 1792-1796.

Pallepati, P. and D. A. Averill-Bates (2011). "Activation of ER stress and apoptosis by

hydrogen peroxide in HeLa cells: Protective role of mild heat preconditioning at

40&#xa0;°C." Biochimica et Biophysica Acta (BBA) - Molecular Cell Research

1813(12): 1987-1999.

Palmqvist, L., B. Argiropoulos, et al. (2006). "The Flt3 receptor tyrosine kinase collaborates

with NUP98-HOX fusions in acute myeloid leukemia." Blood 108(3): 1030-1036.

Palmqvist, L., N. Pineault, et al. (2007). "Candidate Genes for Expansion and Transformation

of Hematopoietic Stem Cells by NUP98-HOX Fusion Genes." PLoS ONE 2(8): e768.

Pan, Q. and R. Simpson (2001). "Antisense knockout of HOXB4 blocks 1,25-dihydroxyvitamin

D3 inhibition of c-myc expression." Journal of Endocrinology 169(1): 153-159.

Pandey, D., J.-P. Gratton, et al. (2011). "Calcium/Calmodulin-Dependent Kinase II Mediates

the Phosphorylation and Activation of NADPH Oxidase 5." Molecular Pharmacology

80(3): 407-415.

Paneni, F., J. A. Beckman, et al. (2013). "Diabetes and vascular disease: pathophysiology,

clinical consequences, and medical therapy: part I." European Heart Journal 34(31):

2436-U2434.

Park, I. K., A. Mishra, et al. (2013). "Inhibition of the receptor tyrosine kinase Axl impedes

activation of the FLT3 internal tandem duplication in human acute myeloid leukemia:

implications for Axl as a potential therapeutic target." Blood 121(11): 2064-2073.

Park, J. S., J. S. Jung, et al. (2011). "Antioxidant mechanism of isoflavone metabolites in

hydrogen peroxide-stimulated rat primary astrocytes: critical role of hemeoxygenase-

1 and NQO1 expression." Journal of Neurochemistry 119(5): 909-919.

Park, S. E., J. D. Song, et al. (2007). "Diphenyleneiodonium induces ROS-independent p53

expression and apoptosis in human RPE cells." FEBS letters 581(2): 180-186.

Parker, P. J., V. Justilien, et al. (2014). "Atypical Protein Kinase Cι as a human oncogene and

therapeutic target." Biochemical Pharmacology 88(1): 1-11.

Patel, J. P. and R. L. Levine (2012). "How do novel molecular genetic markers influence

treatment decisions in acute myeloid leukemia?" Hematology-American Society

Hematology Education Program: 28-34.

Pawar, P., L. P. Ma, et al. (2009). "Molecular Mechanisms of Tamoxifen Therapy for

Cholangiocarcinoma: Role of Calmodulin." Clinical Cancer Research 15(4): 1288-

1296.

Pei, L., Y. Shang, et al. (2014). "DAPK1–p53 Interaction Converges Necrotic and Apoptotic

Pathways of Ischemic Neuronal Death." The Journal of Neuroscience 34(19): 6546-

6556.

Pereira, L., A. Igea, et al. (2013). "Inhibition of p38 MAPK sensitizes tumour cells to cisplatin-

induced apoptosis mediated by reactive oxygen species and JNK." Embo Molecular

Medicine 5(11): 1759-1774.

Page 252: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

238

Persaud, S. D., V. Hoang, et al. (2005). "Involvement of proteolytic activation of PKC delta in

cisplatin-induced apoptosis in human small cell lung cancer H69 cells." International

Journal of Oncology 27(1): 149-154.

Phelan, M., I. Rambaldi, et al. (1995). "Cooperative interactions between HOX and PBX

proteins mediated by a conserved peptide motif." Mol. Cell. Biol. 15(8): 3989-3997.

Philpott, N., A. Turner, et al. (1996). "The use of 7-amino actinomycin D in identifying

apoptosis: simplicity of use and broad spectrum of application compared with other

techniques." Blood 87(6): 2244-2251.

Piccaluga, P. P., G. Visani, et al. (2002). "Liposomal daunorubicin (DaunoXome) for treatment

of relapsed meningeal acute myeloid leukemia." Leukemia 16(9): 1880-1881.

Pietsch, E. C., J. I. J. Leu, et al. (2007). "The tetramerization domain of p53 is required for

efficient BAK oligomerization." Cancer Biology & Therapy 6(10): 1576-1583.

Pillai, P., S. Desai, et al. (2011). "A novel PKC-iota inhibitor abrogates cell proliferation and

induces apoptosis in neuroblastoma (vol 43, pg 784, 2011)." International Journal of

Biochemistry & Cell Biology 43(9): 1412-1412.

Pillay, L. M., A. M. Forrester, et al. (2010). "The Hox cofactors Meis1 and Pbx act upstream

of gata1 to regulate primitive hematopoiesis." Developmental Biology 340(2): 306-

317.

Pineault, N., C. Abramovich, et al. (2004). "Differential and Common Leukemogenic Potentials

of Multiple NUP98-Hox Fusion Proteins Alone or with Meis1." Mol. Cell. Biol. 24(5):

1907-1917.

Pineault, N., C. Buske, et al. (2003). "Induction of acute myeloid leukemia in mice by the

human leukemia-specific fusion gene NUP98-HOXD13 in concert with Meis1." Blood

101(11): 4529-4538.

Pineault, N., C. D. Helgason, et al. (2002). "Differential expression of Hox, Meis1, and Pbx1

genes in primitive cells throughout murine hematopoietic ontogeny." Experimental

Hematology 30(1): 49-57.

Pinkus, R., L. M. Weiner, et al. (1996). "Role of Oxidants and Antioxidants in the Induction of

AP-1, NF-κB, and Glutathione S-Transferase Gene Expression." Journal of Biological

Chemistry 271(23): 13422-13429.

Plowright, L., K. J. Harrington, et al. (2009). "HOX transcription factors are potential

therapeutic targets in non-small-cell lung cancer (targeting HOX genes in lung

cancer)." Br J Cancer 100(3): 470-475.

Potapova, O., S. Basu, et al. (2001). "Protective role for c-Jun in the cellular response to DNA

damage." Journal of Biological Chemistry 276(30): 28546-28553.

Pouyssegur, J., F. Dayan, et al. (2006). "Hypoxia signalling in cancer and approaches to enforce

tumour regression." Nature 441(7092): 437-443.

Raab, M. S., I. Breitkreutz, et al. (2009). "Targeting PKC: a novel role for beta-catenin in ER

stress and apoptotic signaling." Blood 113(7): 1513-1521.

Raghunand, N., B. P. Mahoney, et al. (2003). "Tumor acidity, ion trapping and

chemotherapeutics I. pH-dependent partition coefficients predict importance of ion

trapping on pharmacokinetics of weakly basic chemotherapeutic agents." Biochemical

Pharmacology 66(7): 1219-1229.

Rau, R. and P. Brown (2009). "Nucleophosmin (NPM1) mutations in adult and childhood acute

myeloid leukaemia: towards definition of a new leukaemia entity." Hematological

Oncology 27(4): 171-181.

Ravindranath, Y., C. P. Steuber, et al. (1991). "HIGH-DOSE CYTARABINE FOR

INTENSIFICATION OF EARLY THERAPY OF CHILDHOOD ACUTE

MYELOID-LEUKEMIA - A PEDIATRIC ONCOLOGY GROUP-STUDY." Journal

of Clinical Oncology 9(4): 572-580.

Rawat, V. P. S., S. Thoene, et al. (2008). "Overexpression of CDX2 perturbs HOX gene

expression in murine progenitors depending on its N-terminal domain and is closely

correlated with deregulated HOX gene expression in human acute myeloid leukemia."

Blood 111(1): 309-319.

Page 253: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

239

Ray, N., M. Kuwahara, et al. (2006). "c-Fos suppresses systemic inflammatory response to

endotoxin." International Immunology 18(5): 671-677.

Ray, P. D., B. W. Huang, et al. (2012). "Reactive oxygen species (ROS) homeostasis and redox

regulation in cellular signaling." Cellular Signalling 24(5): 981-990.

Raza-Egilmez, S. Z., S. N. Jani-Sait, et al. (1998). "NUP98-HOXD13 Gene Fusion in Therapy-

related Acute Myelogenous Leukemia." Cancer Research 58(19): 4269-4273.

Redaelli, S., R. Piazza, et al. (2009). "Activity of Bosutinib, Dasatinib, and Nilotinib Against

18 Imatinib-Resistant BCR/ABL Mutants." Journal of Clinical Oncology 27(3): 469-

471.

Regberg, J., A. Srimanee, et al. (2012). "Applications of Cell-Penetrating Peptides for Tumor

Targeting and Future Cancer Therapies." Pharmaceuticals 5(9): 991-1007.

Renneville, A., C. Roumier, et al. (2008). "Cooperating gene mutations in acute myeloid

leukemia: a review of the literature." Leukemia 22(5): 915-931.

Repp, R., U. Schaekel, et al. (2003). "Immunophenotyping is an independent factor for risk

stratification in AML." Cytometry Part B: Clinical Cytometry 53B(1): 11-19.

Reyland, M. (2007). Protein Kinase C and Apoptosis. Apoptosis, Cell Signaling, and Human

Diseases. R. Srivastava, Humana Press: 31-55.

Rice, K. L. and J. D. Licht (2007). "HOX deregulation in acute myeloid leukemia." The Journal

of Clinical Investigation 117(4): 865-868.

Roche, J., C. Zeng, et al. (2004). "Hox expression in AML identifies a distinct subset of patients

with intermediate cytogenetics." Leukemia 18(6): 1059-1063.

Rozovskaia, T., E. Feinstein, et al. (2001). "Upregulation of Meis1 and HoxA9 in acute

lymphocytic leukemias with the t(4 : 11) abnormality." Oncogene 20(7): 874-878.

Rubnitz, J. E., H. Inaba, et al. (2010). "Minimal residual disease-directed therapy for childhood

acute myeloid leukaemia: results of the AML02 multicentre trial." The Lancet

Oncology 11(6): 543-552.

Rubnitz, J. E., S. C. Raimondi, et al. (2002). "Characteristics and outcome of t(8;21)-positive

childhood acute myeloid leukemia: a single institution's experience." Leukemia 16(10):

2072-2077.

Ruchaud, S., N. Korfali, et al. (2002). Caspase‐6 gene disruption reveals a requirement for

lamin A cleavage in apoptotic chromatin condensation.

Rucker, F. G., S. Sander, et al. (2006). "Molecular profiling reveals myeloid leukemia cell lines

to be faithful model systems characterized by distinct genomic aberrations." Leukemia

20(6): 994-1001.

Rushworth, S. A. and D. J. MacEwan (2008). "HO-1 underlies resistance of AML cells to TNF-

induced apoptosis." Blood 111(7): 3793-3801.

Rushworth, S. A., L. Zaitseva, et al. (2010). "FLIP regulation of HO-1 and TNF signalling in

human acute myeloid leukemia provides a unique secondary anti-apoptotic

mechanism." Oncotarget 1(5): 359-366.

Saha, S., P. Bhattacharjee, et al. (2014). "Contribution of the ROS-p53 feedback loop in thuja-

induced apoptosis of mammary epithelial carcinoma cells." Oncology Reports 31(4):

1589-1598.

Sanchez-Gomez, M. V., E. Alberdi, et al. (2011). "Bax and Calpain Mediate Excitotoxic

Oligodendrocyte Death Induced by Activation of Both AMPA and Kainate Receptors."

Journal of Neuroscience 31(8): 2996-3006.

Sanchez, P. V., R. L. Perry, et al. (2009). "A robust xenotransplantation model for acute

myeloid leukemia." Leukemia 23(11): 2109-2117.

Saraste, A. and K. Pulkki (2000). "Morphologic and biochemical hallmarks of apoptosis."

Cardiovascular Research 45(3): 528-537.

Sarko, D., B. Beijer, et al. (2010). "The Pharmacokinetics of Cell-Penetrating Peptides."

Molecular Pharmaceutics 7(6): 2224-2231.

Satoh, Y., I. Matsumura, et al. (2004). "Roles for c-Myc in Self-renewal of Hematopoietic Stem

Cells." Journal of Biological Chemistry 279(24): 24986-24993.

Sauvageau, G., P. M. Lansdorp, et al. (1994). "Differential Expression of Homeobox Genes in

Functionally Distinct CD34+ Subpopulations of Human Bone Marrow Cells."

Page 254: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

240

Proceedings of the National Academy of Sciences of the United States of America

91(25): 12223-12227.

Sauvageau, G., U. Thorsteinsdottir, et al. (1995). "Overexpression of HOXB4 in hematopoietic

cells causes the selective expansion of more primitive populations in vitro and in vivo."

Genes & Development 9(14): 1753-1765.

Sauvageau, G., U. Thorsteinsdottir, et al. (1997). "Overexpression of HOXB3 in hematopoietic

cells causes defective lymphoid development and progressive myeloproliferation."

Immunity 6(1): 13-22.

Saw, J., D. J. Curtis, et al. (2013). "The fusion partner specifies the oncogenic potential of

NUP98 fusion proteins." Leukemia Research 37(12): 1668-1673.

Schinzel, A. C., O. Takeuchi, et al. (2005). "Cyclophilin D is a component of mitochondrial

permeability transition and mediates neuronal cell death after focal cerebral ischemia."

Proceedings of the National Academy of Sciences of the United States of America

102(34): 12005-12010.

Schlenk, R. F., K. Döhner, et al. (2008). "Mutations and Treatment Outcome in Cytogenetically

Normal Acute Myeloid Leukemia." New England Journal of Medicine 358(18): 1909-

1918.

Schmidmaier, R., M. Oellerich, et al. (2004). "Treosulfan-induced apoptosis in acute myeloid

leukemia cells is accompanied by translocation of protein kinase C delta and enhanced

by bryostatin-1." Experimental Hematology 32(1): 76-86.

Schnittger, S., W. Kern, et al. (2009). "Minimal residual disease levels assessed by NPM1

mutation-specific RQ-PCR provide important prognostic information in AML." Blood

114(11): 2220-2231.

Scholl, C., D. Bansal, et al. (2007). "The homeobox gene CDX2 is aberrantly expressed in most

cases of acute myeloid leukemia and promotes leukemogenesis." Journal of Clinical

Investigation 117(4): 1037-1048.

Schroder, K., M. Zhang, et al. (2012). "Nox4 Is a Protective Reactive Oxygen Species

Generating Vascular NADPH Oxidase." Circulation Research 110(9): 1217-+.

Schuringa, J. J. and H. Schepers (2009). Ex Vivo Assays to Study Self-Renewal and Long-

Term Expansion of Genetically Modified Primary Human Acute Myeloid Leukemia

Stem Cells. Leukemia:METHODS AND PROTOCOLS. C. W. E. So. 538: 287-300.

Schwabedissen, H., M. Grube, et al. (2006). "Epidermal growth factor-mediated activation of

the map kinase cascade results in altered expression and function of ABCG2 (BCRP)."

Drug Metabolism and Disposition 34(4): 524-533.

Shah, C. A., L. Bei, et al. (2012). "HoxA10 Protein Regulates Transcription of Gene Encoding

Fibroblast Growth Factor 2 (FGF2) in Myeloid Cells." Journal of Biological Chemistry

287(22): 18230-18248.

Shah, C. A., H. Wang, et al. (2011). "HoxA10 Regulates Transcription of the Gene Encoding

Transforming Growth Factor β2 (TGFβ2) in Myeloid Cells." Journal of Biological

Chemistry 286(4): 3161-3176.

Shah, N. and S. Sukumar (2010). "The Hox genes and their roles in oncogenesis." Nat Rev

Cancer 10(5): 361-371.

Shan, W.-L. and X.-L. Ma (2013). "How to Establish Acute Myeloid Leukemia Xenograft

Models Using Immunodeficient Mice." Asian Pacific Journal of Cancer Prevention

14(12): 7057-7063.

Shears, L., L. Plowright, et al. (2008). "Disrupting the interaction between HOX and PBX

causes necrotic and apoptotic cell death in the renal cancer lines CaKi-2 and 769-P." J

Urol 180(5): N2196 - 2201.

Shen, L., S. H. Kim, et al. (2012). "Sensitization of Human Pancreatic Cancer Cells Harboring

Mutated K-ras to Apoptosis." PLoS ONE 7(7).

Shen, W.-F., C.-P. Chang, et al. (1996). "Hox Homeodomain Proteins Exhibit Selective

Complex Stabilities with Pbx and DNA." Nucleic Acids Research 24(5): 898-906.

Shen, W., J. Montgomery, et al. (1997). "AbdB-like Hox proteins stabilize DNA binding by the

Meis1 homeodomain proteins." Mol. Cell. Biol. 17(11): 6448-6458.

Page 255: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

241

Shi, N.-Q., W. Gao, et al. (2012). "Enhancing cellular uptake of activable cell-penetrating

peptide-doxorubicin conjugate by enzymatic cleavage." International Journal of

Nanomedicine 7: 1613-1621.

Shi, Y. (2002). "Mechanisms of Caspase Activation and Inhibition during Apoptosis."

Molecular Cell 9(3): 459-470.

Shimamoto, T., Y. Tang, et al. (1999). "Hematopoietic progenitor cell abnormalities in Hoxc-

8 null mutant mice." Journal of Experimental Zoology 283(2): 186-193.

Shimizu, T., T. Kayamori, et al. (2012). "Bone morphogenetic protein (BMP)-4 and BMP-7

suppress granulosa cell apoptosis via different pathways: BMP-4 via PI3K/PDK-1/Akt

and BMP-7 via PI3K/PDK-1/PKC." Biochemical and Biophysical Research

Communications 417(2): 869-873.

Shin, S. Y., C. G. Kim, et al. (2011). "The ETS Family Transcription Factor ELK-1 Regulates

Induction of the Cell Cycle-regulatory Gene p21(Waf1)Cip1 and the BAX Gene in

Sodium Arsenite-exposed Human Keratinocyte HaCaT Cells." Journal of Biological

Chemistry 286(30): 26860-26872.

Shiraki, A., J. Oyama, et al. (2012). "The glucagon-like peptide 1 analog liraglutide reduces

TNF-alpha-induced oxidative stress and inflammation in endothelial cells."

Atherosclerosis 221(2): 375-382.

Shultz, L. D., P. A. Schweitzer, et al. (1995). "MULTIPLE DEFECTS IN INNATE AND

ADAPTIVE IMMUNOLOGICAL FUNCTION IN NOD/LTSZ-SCID MICE." Journal

of Immunology 154(1): 180-191.

Shumway, S. D., C. M. Berchtold, et al. (2002). "Evidence for unique calmodulin-dependent

nuclear factor-kappa B regulation in WEHI-231 B cells." Molecular Pharmacology

61(1): 177-185.

Siegler, U., C. P. Kalberer, et al. (2005). "Activated natural killer cells from patients with acute

myeloid leukemia are cytotoxic against autologous leukemic blasts in NOD/SCID

mice." Leukemia 19(12): 2215-2222.

Silvestre, D. C., G. A. Gil, et al. (2010). "Growth of Peripheral and Central Nervous System

Tumors Is Supported by Cytoplasmic c-Fos in Humans and Mice." PLoS ONE 5(3).

Slany, R. K. (2009). "The molecular biology of mixed lineage leukemia." Haematologica-the

Hematology Journal 94(7): 984-993.

Smith, A., D. Howell, et al. (2011). "Incidence of haematological malignancy by sub-type: a

report from the Haematological Malignancy Research Network." Br J Cancer 105(11):

1684-1692.

Snyder, E. L., C. C. Saenz, et al. (2005). "Enhanced Targeting and Killing of Tumor Cells

Expressing the CXC Chemokine Receptor 4 by Transducible Anticancer Peptides."

Cancer Research 65(23): 10646-10650.

So, C. W., H. Karsunky, et al. (2004). "Leukemic transformation of hematopoietic progenitors

by MLL-GAS7 in the absence of Hoxa7 or Hoxa9." Blood 103(8): 3192-3199.

Son, Y.-O., J.-C. Lee, et al. (2010). "Cadmium Induces Intracellular Ca2+- and H2O2-

Dependent Apoptosis through JNK- and p53-Mediated Pathways in Skin Epidermal

Cell line." Toxicological Sciences 113(1): 127-137.

Sosna, J., S. Voigt, et al. (2014). "TNF-induced necroptosis and PARP-1-mediated necrosis

represent distinct routes to programmed necrotic cell death." Cellular and Molecular

Life Sciences 71(2): 331-348.

Soulier, J., E. Clappier, et al. (2005). "HOXA genes are included in genetic and biologic

networks defining human acute T-cell leukemia (T-ALL)." Blood 106(1): 274-286.

Speicher, T., U. A. Köhler, et al. (2012). "Fructose Protects Murine Hepatocytes from Tumor

Necrosis Factor-induced Apoptosis by Modulating JNK Signaling." Journal of

Biological Chemistry 287(3): 1837-1846.

Speleman, F., B. Cauwelier, et al. (2005). "A new recurrent inversion, inv(7)(p15q34), leads to

transcriptional activation of HOXA10 and HOXA11 in a subset of T-cell acute

lymphoblastic leukemias." Leukemia 19(3): 358-366.

Staber, P. B., W. Linkesch, et al. (2004). "Common alterations in gene expression and increased

proliferation in recurrent acute myeloid leukemia." Oncogene 23(4): 894-904.

Page 256: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

242

Stockmann, C., A. Doedens, et al. (2008). "Deletion of vascular endothelial growth factor in

myeloid cells accelerates tumorigenesis." Nature 456(7223): 814-818.

Storr, S. J., N. O. Carragher, et al. (2011). "The calpain system and cancer." Nature Reviews

Cancer 11(5): 364-374.

Sukhai, M. A., S. Prabha, et al. (2013). "Lysosomal disruption preferentially targets acute

myeloid leukemia cells and progenitors." Journal of Clinical Investigation 123(1): 315-

328.

Sun (2008). "Photoreceptor protection against light damage by AAV-mediated overexpression

of heme oxygenase-1 (vol 48, pg 5699, 2007)." Investigative Ophthalmology & Visual

Science 49(3): 893-893.

Sun, L. M., H. Y. Wang, et al. (2012). "Mixed Lineage Kinase Domain-like Protein Mediates

Necrosis Signaling Downstream of RIP3 Kinase." Cell 148(1-2): 213-227.

Suzuki, A., Y. Ito, et al. (2002). "t(7;11)(p15;p15) chronic myeloid leukaemia developed into

blastic transformation showing a novel NUP98/HOXA11 fusion." British Journal of

Haematology 116(1): 170-172.

Sykes, S. M., S. W. Lane, et al. (2011). "AKT/FOXO Signaling Enforces Reversible

Differentiation Blockade in Myeloid Leukemias." Cell 146(5): 697-708.

Szydlowska, K. and M. Tymianski (2010). "Calcium, ischemia and excitotoxicity." Cell

Calcium 47(2): 122-129.

Taghon, T., K. Thys, et al. (2003). "Homeobox gene expression profile in human hematopoietic

multipotent stem cells and T-cell progenitors: implications for human T-cell

development." Leukemia 17(6): 1157-1163.

Tait, Stephen W. G., A. Oberst, et al. (2013). "Widespread Mitochondrial Depletion via

Mitophagy Does Not Compromise Necroptosis." Cell Reports 5(4): 878-885.

Takada, Y., N. Ray, et al. (2010). "Fos Proteins Suppress Dextran Sulfate Sodium-Induced

Colitis through Inhibition of NF-kappa B." Journal of Immunology 184(2): 1014-1021.

Takeda, A., C. Goolsby, et al. (2006). "NUP98-HOXA9 Induces Long-term Proliferation and

Blocks Differentiation of Primary Human CD34+ Hematopoietic Cells." Cancer

Research 66(13): 6628-6637.

Takeshita, K., J. A. Bollekens, et al. (1993). "A HOMEOBOX GENE OF THE

ANTENNAPEDIA CLASS IS REQUIRED FOR HUMAN ADULT

ERYTHROPOIESIS." Proceedings of the National Academy of Sciences of the United

States of America 90(8): 3535-3538.

Taketani, T., T. Taki, et al. (2002). "The HOXD11 gene is fused to the NUP98 gene in acute

myeloid leukemia with t(2;11)(q31;p15)." Cancer Research 62(1): 33-37.

Taketani, T., T. Taki, et al. (2002). "Novel NUP98-HOXC11 fusion gene resulted from a

chromosomal break within exon 1 of HOXC11 in acute myeloid leukemia with

t(11;12)(p15;q13)." Cancer Research 62(16): 4571-4574.

Tallman, M. S. and J. K. Altman (2009). How I treat acute promyelocytic leukemia.

Tam, Wai L., H. Lu, et al. (2013). "Protein Kinase C α Is a Central Signaling Node and

Therapeutic Target for Breast Cancer Stem Cells." Cancer Cell 24(3): 347-364.

Tano, J. Y. and G. Vazquez (2011). "Requirement for non-regulated, constitutive calcium

influx in macrophage survival signaling." Biochemical and Biophysical Research

Communications 407(2): 432-437.

Tavor, S., I. Petit, et al. (2004). "CXCR4 Regulates Migration and Development of Human

Acute Myelogenous Leukemia Stem Cells in Transplanted NOD/SCID Mice." Cancer

Research 64(8): 2817-2824.

Thiede, C., S. Koch, et al. (2006). "Prevalence and prognostic impact of NPM1 mutations in

1485 adult patients with acute myeloid leukemia (AML)." Blood 107(10): 4011-4020.

Thiede, C., C. Steudel, 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.

Thoene, S., V. P. S. Rawat, et al. (2009). "The homeobox gene CDX2 is aberrantly expressed

and associated with an inferior prognosis in patients with acute lymphoblastic

leukemia." Leukemia 23(4): 649-655.

Page 257: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

243

Thompson, A., M. F. Quinn, et al. (2003). "Global down-regulation of HOX gene expression

in PML-RAR alpha(+) acute promyelocytic leukemia identified by small-array real-

time PCR." Blood 101(4): 1558-1565.

Thorsteinsdottir, U., E. Kroon, et al. (2001). "Defining Roles for HOX and MEIS1 Genes in

Induction of Acute Myeloid Leukemia." Mol. Cell. Biol. 21(1): 224-234.

Thorsteinsdottir, U., A. Mamo, et al. (2002). "Overexpression of the myeloid leukemia–

associatedHoxa9 gene in bone marrow cells induces stem cell expansion." Blood 99(1):

121-129.

Tian, W., D. Xu, et al. (2013). "Cyclophilin D modulates cell death transition from early

apoptosis to programmed necrosis induced by honokiol." International Journal of

Oncology 42(5): 1654-1663.

Tobiume, K., A. Matsuzawa, et al. (2001). "ASK1 is required for sustained activations of

JNK/p38 MAP kinases and apoptosis." Embo Reports 2(3): 222-228.

Tosic, N., M. Stojiljkovic, et al. (2009). "Acute myeloid leukemia with NUP98-HOXC13

fusion and FLT3 internal tandem duplication mutation: case report and literature

review." Cancer Genetics and Cytogenetics 193(2): 98-103.

Toyota, H., N. Yanase, et al. (2003). "Calpain-induced Bax-cleavage product is a more potent

inducer of apoptotic cell death than wild-type Bax." Cancer Letters 189(2): 221-230.

Tredan, O., C. M. Galmarini, et al. (2007). "Drug resistance and the solid tumor

microenvironment." Journal of the National Cancer Institute 99(19): 1441-1454.

Tsoyi, K., H. J. Jang, et al. (2011). "Stimulation of Alpha7 Nicotinic Acetylcholine Receptor

by Nicotine Attenuates Inflammatory Response in Macrophages and Improves

Survival in Experimental Model of Sepsis Through Heme Oxygenase-1 Induction."

Antioxidants & Redox Signaling 14(11): 2057-2070.

Tsujimoto, Y. (1997). "Apoptosis and necrosis: Intracellular ATP level as a determinant for cell

death modes." Cell Death and Differentiation 4(6): 429-434.

Upton, J. W., W. J. Kaiser, et al. (2010). "Virus Inhibition of RIP3-Dependent Necrosis." Cell

Host & Microbe 7(4): 302-313.

Ussat, S., U.-E. Werner, et al. (2002). "Upregulation of p21WAF1/Cip1 precedes tumor

necrosis factor-induced necrosis-like cell death." Biochemical and Biophysical

Research Communications 294(3): 672-679.

Valencia, A. and I. E. Kochevar (2007). "Nox1-Based NADPH Oxidase Is the Major Source of

UVA-Induced Reactive Oxygen Species in Human Keratinocytes." J Invest Dermatol

128(1): 214-222.

van Gosliga, D., H. Schepers, et al. (2007). "Establishing long-term cultures with self-renewing

acute myeloid leukemia stem/progenitor cells." Experimental Hematology 35(10):

1538-1549.

Vandenabeele, P., S. Grootjans, et al. (2013). "Necrostatin-1 blocks both RIPK1 and IDO:

consequences for the study of cell death in experimental disease models." Cell Death

and Differentiation 20(2): 185-187.

Vanlangenakker, N., T. Vanden Berghe, et al. (2012). "Many stimuli pull the necrotic trigger,

an overview." Cell Death and Differentiation 19(1): 75-86.

Vaseva, A. V., N. D. Marchenko, et al. (2012). "p53 Opens the Mitochondrial Permeability

Transition Pore to Trigger Necrosis." Cell 149(7): 1536-1548.

Vaseva, A. V. and U. M. Moll (2009). "The mitochondrial p53 pathway." Biochimica et

Biophysica Acta (BBA) - Bioenergetics 1787(5): 414-420.

Vassiliou, G. S., J. L. Cooper, et al. (2011). "Mutant nucleophosmin and cooperating pathways

drive leukemia initiation and progression in mice." Nat Genet advance online

publication.

Verheij, M. (2008). "Clinical biomarkers and imaging for radiotherapy-induced cell death."

Cancer and Metastasis Reviews 27(3): 471-480.

Vermes, I., C. Haanen, et al. (1995). "A novel assay for apoptosis Flow cytometric detection of

phosphatidylserine expression on early apoptotic cells using fluorescein labelled

Annexin V." Journal of Immunological Methods 184(1): 39-51.

Page 258: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

244

Vermeulen, K., D. R. Van Bockstaele, et al. (2005). "Apoptosis: mechanisms and relevance in

cancer." Annals of Hematology 84(10): 627-639.

Verrax, J., N. Dejeans, et al. (2011). "Intracellular ATP levels determine cell death fate of

cancer cells exposed to both standard and redox chemotherapeutic agents."

Biochemical Pharmacology 82(11): 1540-1548.

Vinante, F., A. Rigo, et al. (1999). "Interleukin-8 in acute myeloid leukemia." Blood 93(4):

1437-1437.

Vives, E., J. Schmidt, et al. (2008). "Cell-penetrating and cell-targeting peptides in drug

delivery." Biochimica Et Biophysica Acta-Reviews on Cancer 1786(2): 126-138.

Volk, A., J. Li, et al. (2014). "Co-inhibition of NF-kappa B and JNK is synergistic in TNF-

expressing human AML." Journal of Experimental Medicine 211(6): 1093-1108.

Vosler, P. S., D. D. Sun, et al. (2009). "Calcium dysregulation induces apoptosis-inducing

factor release: Cross-talk between PARP-1-and calpain- signaling pathways."

Experimental Neurology 218(2): 213-220.

Wang, H., Y. Lu, et al. (2007). "HoxA10 Activates Transcription of the Gene Encoding

Mitogen-activated Protein Kinase Phosphatase 2 (Mkp2) in Myeloid Cells." Journal of

Biological Chemistry 282(22): 16164-16176.

Wang, H., X. Wang, et al. (2012). "The proteasome inhibitor bortezomib reverses P-

glycoprotein-mediated leukemia multi-drug resistance through the NF-kappa B

pathway." Pharmazie 67(2): 187-192.

Wang, L., F. Du, et al. (2008). "TNF-α Induces Two Distinct Caspase-8 Activation Pathways."

Cell 133(4): 693-703.

Wang, Y., A. Yabuuchi, et al. (2008). "Cdx gene deficiency compromises embryonic

hematopoiesis in the mouse." Proceedings of the National Academy of Sciences

105(22): 7756-7761.

Wang, Z., S. Li, et al. (2010). "Calmodulin antagonists induce platelet apoptosis." Thrombosis

Research 125(4): 340-350.

Wang, Z. G., H. Jiang, et al. (2012). "The Mitochondrial Phosphatase PGAM5 Functions at the

Convergence Point of Multiple Necrotic Death Pathways." Cell 148(1-2): 228-243.

Waring, P., D. Lambert, et al. (1999). "Increased cell surface exposure of phosphatidylserine

on propidium iodide negative thymocytes undergoing death by necrosis." Cell Death

and Differentiation 6(7): 624-637.

Warr, M. R., E. M. Pietras, et al. (2011). "Mechanisms controlling hematopoietic stem cell

functions during normal hematopoiesis and hematological malignancies." Wiley

Interdisciplinary Reviews-Systems Biology and Medicine 3(6): 681-701.

Was, H., M. Sokolowska, et al. (2011). "Effects of heme oxygenase-1 on induction and

development of chemically induced squamous cell carcinoma in mice." Free Radical

Biology and Medicine 51(9): 1717-1726.

Weisberg, E., R. D. Wright, et al. (2008). "Stromal-mediated protection of tyrosine kinase

inhibitor-treated BCR-ABL-expressing leukemia cells." Molecular Cancer

Therapeutics 7(5): 1121-1129.

Weisser, M., W. Kern, et al. (2005). "Prognostic impact of RT-PCR-based quantification of

WT1 gene expression during MRD monitoring of acute myeloid leukemia." Leukemia

19(8): 1416-1423.

Wenzel, M., M. Wunderlich, et al. (2012). "Cytosolic DNA Triggers Mitochondrial Apoptosis

via DNA Damage Signaling Proteins Independently of AIM2 and RNA Polymerase

III." Journal of Immunology 188(1): 394-403.

Whang, Y. M., U. Jo, et al. (2013). "Wnt5a Is Associated with Cigarette Smoke-Related Lung

Carcinogenesis via Protein Kinase C." PLoS ONE 8(1).

Williams, R. T., W. den Besten, et al. (2007). "Cytokine-dependent imatinib resistance in

mouse Bcr-Abl(+), Arf-null lymphoblastic leukemia." Blood 110(11): 775a-775a.

Wilson, T. R., D. B. Longley, et al. (2006). "Chemoresistance in solid tumours." Annals of

Oncology 17: X315-X324.

Page 259: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

245

Win, H. Y. and M. Acevedo-Duncan (2009). "Role of protein kinase C-iota in transformed non-

malignant RWPE-1 cells and androgen-independent prostate carcinoma DU-145 cells."

Cell Proliferation 42(2): 182-194.

Woo, M. G., K. Xue, et al. (2012). "Calpain-mediated Processing of p53-associated Parkin-like

Cytoplasmic Protein (PARC) Affects Chemosensitivity of Human Ovarian Cancer

Cells by Promoting p53 Subcellular Trafficking." Journal of Biological Chemistry

287(6): 3963-3975.

Xu, Y., S. Huang, et al. (2006). "Poly(ADP-ribose) polymerase-1 signaling to mitochondria in

necrotic cell death requires RIP1/TRAF2-mediated JNK1 activation." Journal of

Biological Chemistry 281(13): 8788-8795.

Yanada, M., K. Matsuo, et al. (2005). "Prognostic significance of FLT3 internal tandem

duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-

analysis." Leukemia 19(8): 1345-1349.

Yang, Z., B. Yu, et al. (2014). "A microfluidic method to synthesize transferrin-lipid

nanoparticles loaded with siRNA LOR-1284 for therapy of acute myeloid leukemia."

Nanoscale 6(16): 9742-9751.

Yekta, S., C. J. Tabin, et al. (2008). "MicroRNAs in the Hox network: an apparent link to

posterior prevalence." Nat Rev Genet 9(10): 789-796.

Yoon, S. O., M. M. Kim, et al. (2001). "Selenite suppresses hydrogen peroxide-induced cell

apoptosis through inhibition of ASK1/JNK and activation of PI3-K/Akt pathways."

Faseb Journal 15(13): 111-+.

Yu, C. F., Y. Minemoto, et al. (2004). "JNK suppresses apoptosis via phosphorylation of the

proapoptotic Bcl-2 family protein BAD." Molecular Cell 13(3): 329-340.

Yu, J. H., K. H. Kim, et al. (2006). "Role of NADPH oxidase and calcium in cerulein-induced

apoptosis - Involvement of apoptosis-inducing factor." Signal Transduction Pathways,

Pt A 1090: 292-297.

Yuan, K., G. Jing, et al. (2011). "Calmodulin Mediates Fas-induced FADD-independent

Survival Signaling in Pancreatic Cancer Cells via Activation of Src-Extracellular

Signal-regulated Kinase (ERK)." Journal of Biological Chemistry 286(28): 24776-

24784.

Zangenberg, M., L. Grubach, et al. (2009). "The combined expression of HOXA4 and MEIS1

is an independent prognostic factor in patients with AML." Eur J Haematol 83(5): 439-

448.

Zangrando, A., M. C. Dell'Orto, et al. (2009). "MLL rearrangements in pediatric acute

lymphoblastic and myeloblastic leukemias: MLL specific and lineage specific

signatures." Bmc Medical Genomics 2.

Zhang, D. W., J. Shao, et al. (2009). "RIP3, an Energy Metabolism Regulator That Switches

TNF-Induced Cell Death from Apoptosis to Necrosis." Science 325(5938): 332-336.

Zhang, L., T. F. Gajewski, et al. (2009). "PD-1/PD-L1 interactions inhibit antitumor immune

responses in a murine acute myeloid leukemia model." Blood 114(8): 1545-1552.

Zhang, S. H., Y. Zhang, et al. (2012). "Matrine Induces Apoptosis in Human Acute Myeloid

Leukemia Cells via the Mitochondrial Pathway and Akt Inactivation." PLoS ONE

7(10).

Zhang, X., L. Zhang, et al. (2007). "c-Fos as a proapoptotic agent in TRAIL-induced apoptosis

in prostate cancer cells." Cancer Res 67: 9425 - 9434.

Zhang, Y., S. Patel, et al. (2012). "CXCR4 inhibitors selectively eliminate CXCR4-expressing

human acute myeloid leukemia cells in NOG mouse model." Cell Death Dis 3: e396.

Zhao, M., X.-F. Duan, et al. (2009). "Protein Kinase Cδ Stimulates Proteasome-Dependent

Degradation of C/EBPα during Apoptosis Induction of Leukemic Cells." PLoS ONE

4(8): e6552.

Zhivotovsky, B. and S. Orrenius (2011). "Calcium and cell death mechanisms: A perspective

from the cell death community." Cell Calcium 50(3): 211-221.

Zhou, X. P., L. Hua, et al. (2012). "FRK controls migration and invasion of human glioma cells

by regulating JNK/c-Jun signaling." Journal of Neuro-Oncology 110(1): 9-19.

Page 260: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

246

Zhu, D. M., W. H. Fang, et al. (1999). "A requirement for protein kinase C inhibition for

calcium-triggered apoptosis in acute lymphoblastic leukemia cells." Clinical Cancer

Research 5(2): 355-360.

Ziegler, U. and P. Groscurth (2004). "Morphological features of cell death." News in

Physiological Sciences 19: 124-128.

Page 261: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

247

Publications

Page 262: HOX/PBX interaction as a therapeutic target in Acute ...epubs.surrey.ac.uk/807885/1/thesis after corrections version.pdf · HOX/PBX interaction as a therapeutic target in Acute Myeloid

248