investigation of csf1-csf1r signaling in aml-bone marrow ......mem) in particular led to significant...

201
Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow Stromal Cell Interactions By Ayesha Rashid A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Medical Biophysics, University of Toronto © Copyright by Ayesha Rashid, 2019

Upload: others

Post on 03-Mar-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow Stromal

Cell Interactions

By

Ayesha Rashid

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Department of Medical Biophysics, University of Toronto

© Copyright by Ayesha Rashid, 2019

Page 2: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

ii

ABSTRACT

CSF1-CSF1R Signaling in AML-Bone Marrow Stromal Cell Interactions

Ayesha Rashid

Department of Medical Biophysics

University of Toronto

2019

Acute myeloid leukemia (AML) is a highly heterogeneous and growth factor-dependent

disease. Colony stimulating factor 1 (CSF1) is a cytokine produced by bone marrow (BM)

stromal cells that directs the differentiation and growth of myeloid cell precursors into

monocytes and macrophages. Human CSF1 (hCSF1) exists as a soluble/secreted (hCSF1-sol),

and a transmembrane (hCSF1-mem) isoform. CSF1 acts on the CSF1 receptor (CSF1R), which

is expressed on mononuclear phagocytic cells. In this study, the CSF1-CSF1R ligand-receptor

pair was investigated in the context of AML-stromal cell interactions.

Analysis of cell surface CSF1R protein levels revealed that a subset of AML patient

samples express high levels of CSF1R (CSF1Rhigh), and that these patients have shorter overall

survival (OS) times compared to patients with low CSF1R expression (CSF1Rlow). In examining

the CSF1-CSF1R interaction experimentally, the long-term growth and survival of

CSF1Rhigh AML cells was supported by MS-5 stromal cells expressing human CSF1 (hCSF1) in

co-culture experiments. MS-5 cells expressing the membrane-bound form of CSF1 (hCSF1-

mem) in particular led to significant increases in AML cell numbers and adhesion to stroma,

compared to cells co-cultured with MS-5 empty vector (EV) or hCSF1-sol cells. Co-culture of

AML cells with MS-5 hCSF1-mem cells led to activation of mTOR signaling in AML cells as

observed through increases in pS6, increased expression of the hematopoietic stem cell markers

CD34 and c-Kit on AML cells, and distinct changes in the cytokine profiles of both AML and

Page 3: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

iii

stromal cells. Further to this, deletion of the PDZ domain binding motif (PDBM) at the C-

terminal end of CSF1 ligand in MS-5 stromal cells led to decreased stromal support of AML

cells, and reduced mTOR pathway activation.

The interaction between CSF1 and CSF1R mediates supportive effects between AML

and stromal cells by fostering bilateral intracellular and intercellular signaling changes. As such,

targeting the CSF1-CSF1R interaction may prove to be an effective therapeutic strategy to

overcome and compromise the protective and supportive role of the BM niche in AML disease

pathogenesis.

Page 4: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

iv

ACKNOWLEDGEMENTS

I would firstly like to thank my graduate supervisor, Dr. Mark Minden, for giving me the

opportunity to pursue research in his lab under his guidance and supervision. I am extremely

grateful to Dr. Minden for allowing me the flexibility in exploring and working through my

project, and for always lending his unconditional support. His passion and dedication to his work

have served as great inspiration and motivation for me throughout my graduate studies.

I would also like to thank my committee members Drs Vuk Stambolic, Aaron Schimmer and

Dwayne Barber for their feedback, suggestions and engaging discussions at meetings. Thank you

for your continued support all throughout. I would also like to thank Chau Dang, Daphne Sears

and Annette Chan from the Department of Medical Biophysics for their support and guidance

throughout the program.

I would like to express my gratitude to members of the Minden lab (past and present) for their

support, guidance and friendship. I am especially thankful to Dr. Ruijuan He who has been a

wonderful mentor, teacher and friend at all times. I would also like to thank Xiu-Zhi Yang, Jian

Liu, Dr. Michael Jain, Dr. John Woolley, Dr. Irakli Dzneladze, Dr. Narmin Ibrahimova, Dr.

Mohammed Fateen, Dr. Rob Laister, Dr. Jenny Jun, Dr. Youqi Han and Francesca Pulice for

their support and assistance throughout. Thank you especially to Drs John Woolley, George Ren

and Michael Jain for their invaluable scientific advice and mentorship.

This journey would not have been possible without the unwavering support of my family. I

would like to thank my parents and my sister Sarah who have supported me throughout the

journey with constant love, support and patience. Thank you to my parents for instilling in me

values of honesty, compassion, genuine hard work and the will to succeed.

Page 5: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

v

TABLE OF CONTENTS

ABSTRACT .................................................................................................................................... ii

ACKNOWLEDGEMENTS ........................................................................................................... iv

TABLE OF CONTENTS ................................................................................................................ v

LIST OF FIGURES ...................................................................................................................... vii

LIST OF TABLES ......................................................................................................................... ix

ABBREVIATIONS ........................................................................................................................ x

CHAPTER 1: Introduction .......................................................................................................... 1

1.1. HEMATOPOIESIS ...................................................................................................... 2

1.1.1. Hematopoietic growth factors and cytokines ............................................................ 5

1.2. ACUTE MYELOID LEUKEMIA.............................................................................. 10 1.2.1. AML Classification ................................................................................................. 12 1.2.2. AML Risk Groups................................................................................................... 15

1.2.3. Genetic Mutations in AML ..................................................................................... 18 1.2.4. AML Treatment ...................................................................................................... 25

1.3. HEMATOPOIETIC AND LEUKEMIC STEM CELLS............................................ 28 1.3.1. In-Vitro Hematopoietic and Leukemic Cell Colony Formation Assays ................. 28

1.3.2. In-Vivo Hematopoietic and Leukemic Cell Assays ................................................ 29

1.3.3. Hematopoietic Cell Surface Markers ...................................................................... 33

1.4. THE BONE MARROW NICHE ................................................................................ 35

1.4.1. Cells and Factors in the Bone Marrow Niche ......................................................... 36 1.4.2. Hematopoietic and Leukemic Cells in the Bone Marrow Niche ............................ 40

1.5. BONE MARROW STROMAL CELL ASSAYS ...................................................... 43

1.6. COLONY STIMULATING FACTOR 1 RECEPTOR (CSF1R) .......................................... 45 1.6.1. CSF1R Structure ..................................................................................................... 45 1.6.2. CSF1R Intracellular Signaling ................................................................................ 50

1.6.3. CSF1R in Disease ................................................................................................... 55

1.7. COLONY STIMULATING FACTOR 1 (CSF1) .................................................................. 56 1.7.1. CSF1 Gene Structure and Isoforms ........................................................................ 57 1.7.2. CSF1 in Pathology .................................................................................................. 62

1.8. INTERLEUKIN-34 (IL-34) ................................................................................................... 63

1.9. SOLUBLE VS. TRANSMEMBRANE LIGANDS ............................................................... 64

Page 6: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

vi

1.10 PDZ DOMAINS AND PDZ DOMAIN BINDING MOTIFS .................................... 64 1.11. HTRA SERINE PROTEASES ................................................................................... 68 1.12. EXPERIMENTAL MODELING ............................................................................... 71 1.13. THESIS FOCUS ......................................................................................................... 72

1.14. THESIS RATIONALE, HYPOTHESES AND OBJECTIVES ................................. 74

CHAPTER 2: CSF1R is associated with poor clinical outcome and promotes a leukemic

cell phenotype in AML-stromal cell interactions ..................................................................... 76 2.1. ABSTRACT ........................................................................................................................... 77

2.2. INTRODUCTION ................................................................................................................. 78 2.3. MATERIALS AND METHODS ........................................................................................... 81 2.4. RESULTS .............................................................................................................................. 87 2.5. DISCUSSION ...................................................................................................................... 125

CHAPTER 3: CSF1 ligand signaling in AML-bone marrow stromal cell interactions ..... 132

3.1. ABSTRACT ......................................................................................................................... 133

3.2. INTRODUCTION ............................................................................................................... 134 3.3. MATERIALS AND METHODS ......................................................................................... 138 3.4. RESULTS ............................................................................................................................ 142

3.5. DISCUSSION ...................................................................................................................... 156

CHAPTER 4: Thesis discussion and significance of findings ............................................... 161

4.1. OVERALL DISCUSSION & SIGNIFICANCE OF FINDINGS ........................................ 162

4.2. FUTURE DIRECTIONS ..................................................................................................... 167

4.3. CONCLUSIONS.................................................................................................................. 169

REFERENCES .......................................................................................................................... 171

Page 7: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

vii

LIST OF FIGURES

Figure 1.1. Hematopoiesis is organized as a cellular hierarchy. ..................................................... 4

Figure 1.2. Architecture of the bone marrow niche. ..................................................................... 39

Figure 1.3. Structure of CSF1R. ................................................................................................... 47

Figure 1.4. CSF1 ligand binding to CSF1R. ................................................................................. 49

Figure 1.5. CSF1R activation of intracellular signal transduction pathways. .............................. 54

Figure 1.6. Alternative splicing of CSF1 leads to different isoforms. .......................................... 60

Figure 1.7. Model of the two major variants of monomeric CSF1. .............................................. 61

Figure 1.8. Structure and function of HtrA serine proteases. ....................................................... 70

Figure 1.9. Modeling an AML-bone marrow microenvironment. ................................................ 73

Figure 2.1. CSF1R gene expression levels are high in certain prognostically significant

subgroups of AML patients. ......................................................................................................... 88

Figure 2.2. High CSF1R high levels are associated with shorter OS in AML patients. ............... 89

Figure 2.3. CSF1R cell surface protein levels are associated with poor AML clinical outcome. 93

Figure 2.4. CSF1R and CD34 co-expression patterns are associated with OS............................. 98

Figure 2.5. CSF1R expression in AML cell lines and patient samples. CSF1R cell surface ..... 100

Figure 2.6. AML cells show minimal growth responses to exogenous soluble CSF1. .............. 102

Figure 2.7. CSF1 overexpression in MS-5 stromal cell lines. .................................................... 104

Figure 2.8. MS-5 stromal cells expressing hCSF1 support the proliferation and survival of factor-

dependent CSF1R-expressing AML cell lines. ........................................................................... 107

Figure 2.9. MS-5 stromal cells that express hCSF1 support the proliferation and survival of

primary AML cells that express CSF1R. .................................................................................... 110

Figure 2.10. Blockade of CSF1R on AML cells inhibits their proliferation in co-culture with MS-

5 hCSF1 cells. ............................................................................................................................. 112

Figure 2.11. AML cells exhibit changes in cytokine production and mTOR pathway activation

after co-culture with hCSF1-expressing MS-5 cells. .................................................................. 115

Page 8: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

viii

Figure 2.12. OCI/AML-5 cells exhibit mTOR pathway activation after co-culture with hCSF1-

expressing MS-5 cells. ................................................................................................................ 116

Figure 2.13. AML cells grown in co-culture with hCSF1-expressing MS-5 cells exhibit changes

in cell surface marker expression. ............................................................................................... 119

Figure 2.14. AML cells exhibit engraftment in humanized CSF1 (huCSF1) mice, with changes in

cell surface marker expression. ................................................................................................... 121

Figure 2.15. BM stromal cells derived from huCSF1 mice support AML cell proliferation. .... 123

Figure 3.1. The intracellular domain of CSF1 possesses a conserved C-terminal PDZ domain

binding motif (PDBM). ............................................................................................................... 143

Figure 3.2. CSF1-expressing MS-5 stromal cells demonstrate differential phenotypic and

signaling behaviour. .................................................................................................................... 146

Figure 3.3. CSF1 expression in MS-5 Tet-off inducible cells. ................................................... 149

Figure 3.4. CSF1-induced mTOR signaling is mediated by HtrA1 in MS-5 cells. .................... 151

Figure 3.5. Support of AML cells by hCSF1-expressing MS-5 cells. ........................................ 153

Figure 3.6. Model of CSF1-induced multi-directional signaling in AML-stromal cell interactions.

..................................................................................................................................................... 155

Page 9: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

ix

LIST OF TABLES

Table 1-1. FAB-based classification system for AML based on morphology and cytogenetics. . 13

Table 1-2. WHO classification of myeloid neoplasms and acute leukemia, 2008. ...................... 14

Table 1-3. NCCN risk groups classification of AML. Adapted from National Comprehensive

Cancer Network (NCCN) Guidelines: Acute Myeloid Leukemia. Version 2.2019. .................... 17

Table 1-4. Types of frequently mutated genes in AML based on function .................................. 24

Table 1-5. Cross-reactivity of hematopoietic and stromal cell growth factors and cytokines

between mouse and human. .......................................................................................................... 31

Table 1-6. Classification of PDZ domain binding recognition sequences.................................... 67

Table 2-1. Table of AML patient characteristics from the PMCC/UHN dataset. ........................ 92

Page 10: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

x

ABBREVIATIONS

AML Acute Myeloid Leukemia

α-MEM Alpha Modification Minimal Essential Medium Eagle

BM Bone Marrow

CAFC Cobblestone-Area-Forming Cell

CFC Colony Forming Cell

c-KIT KIT Proto-Oncogene Receptor Tyrosine Kinase

CR Complete Remission

CSF1 Colony-Stimulating Factor 1

CSF1R Colony-Stimulating Factor 1 Receptor

CXCL12 C-X-C Motif Chemokine Ligand 12

CXCR4 C-X-C Motif Chemokine Receptor 4

FAB French-American-British

FBS Fetal Bovine Serum

FLT3 Fms Related Tyrosine Kinase 3

FMS McDonough Feline Sarcoma Viral (V-Fms) Oncogene Homolog

GM-CSF Granulocyte-Macrophage Colony-Stimulating Factor

G-CSF Granulocyte Colony-Stimulating Factor

HSC Hematopoietic Stem Cell

HTRA1 High-Temperature Requirement A Serine Peptidase 1

IL-3 Interleukin-3

IL-6 Interleukin-6

LTC-IC Long Term Culture Initiating Cell

LSC Leukemia Stem Cell

MAPK Mitogen-Activated Protein Kinase

MSC Mesenchymal Stem Cell

mTOR Mammalian/Mechanistic Target of Rapamycin

NPM1 Nucleophosmin 1

PCR Polymerase Chain Reaction

PDBM PDZ Domain Binding Motif

PI3K Phosphoinositide 3-Kinase

PKB Protein Kinase B (AKT)

MS-5 Murine Stromal (cell line) 5

RBC Red Blood Cell

rhCSF1 Recombinant Human CSF1

(RP)S6 Ribosomal Protein S6

RTK Receptor Tyrosine Kinase

SCF Stem Cell Factor

SCID Severe Combined Immunodeficient

siRNA Short Interference Ribonucleic Acid

STAT Signal Transducers and Activators of Transcription

TNF Tumor Necrosis Factor

PVDF Polyvinylidene Difluoride

WBC White Blood Cell

WHO World Health Organization

Page 11: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

1

CHAPTER 1

Introduction

Page 12: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

2

1.1. HEMATOPOIESIS

Hematopoiesis is a highly regulated process that gives rise to the over 10 distinct cell types

of the blood. The hematopoietic stem cell (HSC) is the most primitive hematopoietic cell from

which all blood cells are derived. As a stem cell, the HSC is defined by properties of sustained

proliferation, multi-lineage differentiation and long-term self-renewal (McCulloch, 1983). The

HSC is found at a frequency of only 1 in 10,000 cells (or 0.01% of total BM cells) within the BM

(Rossi, Challen, Sirin, Lin, & Goodell, 2011). Despite its rarity, a single HSC transplanted into

immune deficient mice irradiated to ablate their BM, has the ability to re-constitute the entire

hematopoietic system.

Hematopoiesis is organized in a cellular hierarchy maintained by a pool of self-renewing

pluripotent HSCs. HSCs can have both long and short life spans: long-term HSC (LT-HSC) and

short-term HSC (ST-HSC). The HSC gives rise to progressively more differentiated progenitor

cell intermediates, leading to the formation of distinct terminally differentiated blood cells

(Figure 1.1).

There are two main lineages of hematopoietic blood cells: myeloid and lymphoid. The

pluripotent HSC is stimulated by hematopoietic growth factors and cytokines, which direct its

differentiation and maturation into hematopoietic multipotent progenitor (MPP) cells, namely the

common myeloid progenitor (CMP) and the common lymphoid progenitor (CLP) (Tsiftsoglou,

Vizirianakis, & Strouboulis, 2009). These can differentiate into more lineage-restricted

oligopotent progenitors i.e. myeloblasts and lymphoblasts, respectively, which then differentiate

into unilineage, unipotent progenitor intermediates (i.e. promyelocytes, prolymphocytes) as they

finally mature into unipotent lineage committed cells. Through myelopoiesis, myeloid

progenitors terminally differentiate into mature erythrocytes (RBCs), thrombocytes (platelets),

Page 13: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

3

monocytes and granulocytes. Monocytes can continue to differentiate into macrophages

(osteoclasts and tissue resident macrophages) and dendritic cells (Morrison & Weissman, 1994).

Granulocytes are also known as polymorphonuclear leukocytes (PML or PMNL) due to their

variably shaped multi-lobed nuclei. They also contain granular sacs throughout their cytoplasm,

which contain various toxic substances such as antimicrobial agents, enzymes, oxygen-based

free radicals and low pH vesicles that are released to fight and kill pathogens (Hickey & Kubes,

2009). There are four main types of granulocytes: neutrophils, eosinophils, basophils and mast

cells, with neutrophils being the most abundant of these (Breedveld, Groot Kormelink, van

Egmond, & de Jong, 2017). Cells of the innate immune response include monocytes, neutrophils,

basophils and eosinophils.

On the other hand, lymphoid progenitors differentiate into natural killer (NK) cells, and the

T and B lymphocytes (T cells and B cells), which are part of the adaptive immune response.

Upon activation, B cells further differentiate into specific antibody-producing plasma cells.

Leukocytes (or WBCs) include both myeloid (granulocytes, monocytes) and lymphoid

(lymphocytes) lineage-derived cells. Fully differentiated, mature cells have limited proliferative

potential and lack self-renewal capacity. Fate determination in hematopoietic precursors is

signaled by specific growth factors and cytokines that direct cell differentiation, growth and

maturation (Laurenti & Göttgens, 2018).

Page 14: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

4

Figure 1.1. Hematopoiesis is organized as a cellular hierarchy.

The hematopoietic stem cell (HSC) resides at the apex of a hematopoietic cellular hierarchy

which gives rise to progressively more differentiated cells induced by specific hematopoietic

cytokines at each stage of development. There are two main lineages of cells that the HSC gives

rise to: myeloid and lymphoid. Each differentiate into increasingly lineage-committed progenitor

cells to give rise to the different mature blood cell types. Figure adapted from: Molecular Cell

Biology (textbook). Lodish, Harvey F. 5. ed.: – New York : W. H. Freeman and Co., 2003, 973 s.

b ill. ISBN 0-7167-4366-3.

Page 15: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

5

1.1.1. Hematopoietic growth factors and cytokines

Hematopoiesis is regulated by hematopoietic growth factors and cytokines that induce the

differentiation and growth of hematopoietic cell precursors into mature, lineage-committed cells.

Specific factors govern the lineage-specific differentiation into different cell types (Figure 1.1).

Among these are the colony stimulating factors (G-CSF, GM-CSF, MCSF), stem cell factor

(SCF) erythropoietin (EPO), thrombopoietin (TPO), interleukin-3 (IL-3), interleukin-6 (IL-6)

and interleukin-11 (IL-11) (Norris, Magwood, & Bennett, 2014; Orkin & Zon, 2008).

Colony-stimulating factors (CSF) are predominantly secreted glycoprotein hematopoietic

growth factors that bind to receptors on hematopoietic cells to induce their growth,

differentiation and survival (Donald Metcalf, 2013). They were first identified based on their

ability to induce the expansion of single cells into morphologically and functionally distinct

hematopoietic cell colonies in colony forming assays (CFA). There are three major CSFs:

macrophage colony-stimulating factor (MCSF or CSF1), granulocyte-macrophage colony-

stimulating factor (GM-CSF or CSF2) and granulocyte colony-stimulating factor (G-CSF or

CSF3). Each factor is named based on the type of cell colony that it is able to give rise to i.e.

MCSF stimulates the formation of colonies of macrophages while G-CSF leads to formation of

colonies of granulocytes (Norris et al., 2014). It should be noted that the other hematopoietic

cytokines should also technically be classified as colony stimulating factors as they also have the

ability to give rise to lineage-specific hematopoietic cell colonies. While most of the CSFs are

secreted, soluble proteins, MCSF can also exist as a membrane-bound ligand, predominantly in

the BM (MCSF will be discussed in detail later on). The CSFs act in autocrine, endocrine and

paracrine signaling to elicit their effects.

Page 16: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

6

GM-CSF

GM-CSF is a monomeric glycoprotein produced and secreted by a wide array of different

cells in the BM including macrophages, mast cells, T cells, NK cells, endothelial cells and

fibroblasts (Gasson, 1991). It acts on HSCs, by binding to the GM-CSF receptor, to induce their

differentiation into granulocytes (neutrophils, basophils, eosinophils) and monocytes, which

eventually mature into macrophages and dendritic cells. It is thus involved in

immune/inflammatory responses as it leads to the expansion of core cells, particularly

macrophages, in the innate immune response. GM-CSF binds to the GM-CSF receptor (also

known as CD116) and elicits its intracellular effects through activation of STAT proteins,

namely STAT5 as well as STAT3 in some cases (Faderl et al., 2003). It is highly expressed in

rheumatoid arthritis and neoplasias, and GM-CSF inhibitors have been developed for the

potential treatment of rheumatoid arthritis in particular. GM-CSF and related derivatives are used

clinically to increase WBC counts after chemotherapy or bone marrow transplantation

(Armitage, 1998).

G-CSF

G-CSF is a cytokine and hormone that stimulates the differentiation of myeloid cells into

granulocytes. It is a soluble glycoprotein that is produced by endothelial cells, macrophages and

other immune cells within the BM. G-CSF acts on the G-CSF receptor expressed on

hematopoietic precursors to induce their proliferation, growth, survival and differentiation into

mature granulocytes, as well as neutrophils (D Metcalf, 1985; Souza et al., 1986). G-CSF elicits

these effects through activation of the JAK/STAT, MAPK/ERK and PI3K/Akt pathways which

are involved in growth, survival and differentiation (these will be discussed later). In addition, G-

Page 17: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

7

CSF also exerts potent effects on HSCs by inducing their mobilization from the BM. This has

important clinical applications as it is used to mobilize HSCs into the bloodstream of donors,

which are then collected via leukapheresis for use in allogeneic stem cell transplantation

(Schwella, Braun, Ahrens, Rick, & Salama, 2003). G-CSF is also used to treat neutropenia and

bolster WBC counts after chemotherapy (Bendall & Bradstock, 2014; Morstyn et al., 1988).

IL-6

Interleukin-6 (IL-6) is both a pro-inflammatory cytokine, and an anti-inflammatory

myokine (a cytokine produced by muscle cells), produced in inflammation, infection and

cancers. It is produced by T cells and monocytes/macrophages, in response to binding of

lipopolysaccharide (LPS) to toll-like receptor 4 (TLR4), to activate immune responses after

injury or infection, particularly those involving an inflammatory response such as burns or other

tissue damage (Richards, 1998). It is also secreted by fibroblasts, keratinocytes and endothelial

cells in response to IL-1, and by osteoblasts to activate osteoclast production (Ishimi et al.,

1990). It can also act as an anti-inflammatory cytokine by exerting inhibitory effects on pro-

inflammatory cytokines such as TNF-α, IL-1, and activating effects on IL-1Ra and IL-10

(Kishimoto, 2010). In hematopoiesis, IL-6 may play a role in the survival, self-renewal and

differentiation of HSCs and in B-cell differentiation (Bernad et al., 1994; Patchen, MacVittie,

Williams, Schwartz, & Souza, 1991). Importantly, it synergizes with other hematopoietic

cytokines, namely the GM-CSF, SCF (stem cell factor) and IL-3, to induce the survival and

differentiation of hematopoietic progenitors in-vitro and in-vivo (K. Ikebuchi et al., 1987).

In infection and inflammation, IL-6 is largely involved in mediating fever in the acute

phase response (an inflammatory response involving plasma proteins). IL-6 acts on the IL-6

Page 18: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

8

receptor (IL-6R) complex which is composed of the IL-6Rα (CD126) ligand-binding chain and

the signal transducing unit glycoprotein 130 (gp130 or CD130). Upon activation, the IL-6

ligand-receptor complex activates intracellular pathways including JAK/STAT to activate STAT

proteins (namely STAT3) and other transcription factors (Heinrich et al., 1998). There also exists

a soluble form of the IL-6R, sIL-6R, which can be isolated from human serum and urine. IL-6

stimulates inflammatory and auto-immune responses in several diseases and disorders including

diabetes, atherosclerosis, rheumatoid arthritis, Alzheimer’s disease, multiple myeloma and

prostate cancer among others (Barton, 2005; Gado, Domjan, Hegyesi, & Falus, 2000; Kristiansen

& Mandrup-Poulsen, 2005; Nishimoto, 2006; Smith, Hobisch, Lin, Culig, & Keller, 2001;

Swardfager et al., 2010). An anti-IL-6 agent (tocilizumab) has been approved for treatment of

rheumatoid arthritis and there is great interest in the use and further development of anti-IL-6

therapies for other diseases including cancer (Emery et al., 2008). Blockade of IL-6 may be

useful in advanced prostate and pancreatic cancers in which there are elevated levels of IL-6,

which is associated with poor survival outcomes (Bellone et al., 2006).

IL-3

IL-3 is a glycoprotein that binds to the IL-3 receptor (IL-3R). It is involved in the body’s

immune responses in injury and disease and has been shown to mediate inflammation in sepsis

(Weber et al., 2015). In hematopoiesis, IL-3 directs the differentiation of multipotent HSCs into

myeloid progenitor cells, as well as lymphoid progenitor cells with the addition of IL-7. In

conjunction with cytokines such as IL-6, erythropoietin (EPO) and GM-CSF, IL-3 promotes the

growth and proliferation of cells within the myeloid lineage, namely, granulocytes, monocytes

and dendritic cells (Y. C. Yang et al., 1986). It is produced and secreted by basophils and T-cells.

Page 19: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

9

Kit ligand/Stem Cell Factor (SCF)/Steel Factor

Kit ligand (KitL), also known as steel factor or stem cell factor (SCF), is a cytokine that

binds to the class III tyrosine kinase receptor c-Kit. It exists as both soluble and transmembrane

proteins, and is involved in hematopoiesis, spermatogenesis and melanogenesis (Kent et al.,

2008; Wehrle-Haller, 2003). In the BM, SCF (both soluble and transmembrane forms) is

produced by endothelial cells and fibroblasts. SCF is seen to play an important role in HSC

survival, maintenance and homing to HSC BM niches as it is involved in adhesion (Keller, Ortiz,

& Ruscetti, 1995). Moreover, HSCs and hematopoietic progenitors are seen to preferentially

localize and migrate to areas with high SCF levels, indicating its chemotactic role. During

embryogenesis, SCF is expressed in the fetal liver and BM where it is critical for hematopoiesis

as SCF null mice die in utero due to severe anemia (Geissler et al., 1991). Upon binding to its

receptor, c-Kit, SCF induces receptor dimerization and activation, leading to stimulation of

intracellular pathways that regulate growth, survival and differentiation such as the JAK/STAT,

MAPK and PI3K/Akt pathways.

Page 20: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

10

1.2. ACUTE MYELOID LEUKEMIA

Acute Myeloid Leukemia (AML) is a highly heterogeneous and aggressive hematological

malignancy of the bone marrow (BM) characterized by the clonal expansion of hematopoietic

cell precursors that are blocked in their ability to differentiate. Its incidence increases with age

and with current therapy, overall 5-year survival ranges from 30-70%, based on disease subtype

and associated risk factors (Liersch, Müller-Tidow, Berdel, & Krug, 2014). AML affects more

males than females and age is a strong prognostic indicator, with older adults having poorer

survival outcomes compared to younger individuals (Juliusson et al., 2009). Despite achieving an

initial complete remission to treatment, more than half of all AML patients relapse within 1-2

years, likely due to the outgrowth of leukemic cells that were therapy resistant and hence evaded

and/or survived chemotherapy. The symptoms of AML are ambiguous and non-specific as they

often mimic those of the common flu, particularly fatigue and fever, but can be accompanied by

weight loss, dyspnea (shortness of breath), frequent or increased risk of infection and easy

bleeding and/or bruising (Hoffman et al., 2017). As an acute disease, AML can progress quite

rapidly and can be fatal in a span of days or weeks if left untreated. The risk factors for AML are

not entirely clear but include smoking, myelodysplastic syndrome (MDS), previous

chemotherapy or radiation therapy and exposure to the chemical benzene (Arber, 2017).

Page 21: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

11

AML can be described as a BM failure state, characterized by decreased production of

normal hematopoietic cells and increased production of immature progenitor cells that are

blocked in their ability to differentiate; these latter cells are able to proliferate extensively in a

clonal fashion, as well as self-renew, a property essential for the maintenance of the disease

(Betz & Hess, 2010b; Ferrara & Schiffer, 2013a). AML is a malignancy of myeloid lineage cells

that arises due to acquired mutations that occur in the HSC or in early myeloid progenitor cells,

most typically in the myeloblast cell, also known as ‘blast’ cells. A major consequence of these

mutations is the disruption of differentiation signals, leading to the arrest of cells in an immature,

undifferentiated state. At the same time, growth and cell cycle checkpoints are lost, as is typical

in most cancers, resulting in uncontrolled proliferation. This leads to an accumulation of

abnormal, undifferentiated blasts, i.e. leukemia cells, in the BM which interferes with the

production of normal hematopoietic cells as they compete for space and resources. This creates a

cellularly and biochemically competitive environment where leukemic cells may out-populate

and replace normal blood cells. This is evident through reductions in numbers of normal white

blood cells (WBC) (i.e. neutrophils), red blood cells and platelets, with WBC counts that can be

either abnormally high or low (Betz & Hess, 2010b).

Page 22: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

12

1.2.1. AML Classification

AML is diagnosed based on the finding of at least 20% blasts in the peripheral blood

and/or BM, along with the presence of recurrent cytogenetic abnormalities which may include

t(15;17), t(8;21), t(16;16) or inv(16) (Betz & Hess, 2010b; Tallman et al., 2019). Diagnosis

begins with abnormal findings in a complete blood count (CBC) test, in which blasts may or may

not be detected. The CBC may show low platelet and neutrophil counts, and total WBC counts

that are higher (leukocytosis) or lower (leukopenia) than normal. A BM aspiration or biopsy is

performed to assess BM involvement i.e. the presence of blasts and other cellular abnormalities

in the BM. Blood and BM samples are assessed using cytological and molecular approaches that

involve examination of morphology, cell surface marker expression, cytogenetics and molecular

profile to make the final diagnosis and classify the subtype of disease (O’Donnell et al., 2017).

The two main classification systems for AML are the French-American-British (FAB)

system and the World Health Organization (WHO). The FAB system was first introduced in

1976 and relies largely on cell morphological characteristics and cytogenetics. It also has more

stringent requirements such as a higher blast percentage of at least 20-30% in the peripheral

blood/BM (Bennett et al., 1976). The FAB system classifies AML into eight different subtypes

that range from M0 to M7, based on the type of cell involved and its degree of maturation, which

are evaluated using light microscopy. Cytogenetics is also included in the classification, with

certain abnormalities being commonly associated with specific subtypes. Table 1-1 outlines the

eight different FAB classifications of AML. The WHO system was introduced in 2008 and is a

relatively more thorough evaluation that integrates cytogenetics, morphology, genetic markers

and immunologic makers, with the aim of providing a more clinically and prognostically relevant

assessment than the FAB system (Table 1-2) (Weinberg et al., 2009).

Page 23: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

13

Table 1-1. FAB-based classification system for AML based on morphology and

cytogenetics.

There are 8 different subtypes of AML according to the FAB system (Bennett et al., 1976b).

Page 24: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

14

Table 1-2. WHO classification of myeloid neoplasms and acute leukemia, 2008.

Page 25: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

15

1.2.2. AML Risk Groups

AML is characterized by a great degree of cellular and genetic heterogeneity owing to the

diversity of molecular drivers in the disease. These include recurrently occurring driver

mutations and genomic alterations, which lead to a cellular block in differentiation and increased

proliferation. Driver mutations cause the clonal expansion of specific sub-clones, while

passenger mutations may be associated with the clonal expansion but do not confer any

functional effect. Overall, differences in leukemogenic drivers manifest in the differences seen in

clinical characteristics and treatment responses.

Large-scale genomic studies have identified and characterized driver mutations that

underlie AML evolution (Baldus & Bullinger, 2008; Cancer & Atlas, 2013; Papaemmanuil et al.,

2016b; Welch et al., 2012) For example, one study involving 1540 patients identified 5234

potential driver mutations across 76 genes and genomic regions (Papaemmanuil et al., 2016). At

least one driver mutation was identified in 96% of all AML patients, with two or more driver

mutations seen in 86% of patients. Clinically, the authors demonstrate a significant difference in

disease presentation and patient survival across the genomic subgroups.

Common genetic alterations include chromosomal rearrangements, amplifications,

deletions and point mutations. Approximately 45% of AML cases present with one or more

cytogenetic alterations, many of which are prognostically significant. 15% of patients within this

group harbour 3 or more chromosomal abnormalities which identifies a group designated

‘complex karyotype’ AML (Mrózek, Heerema, & Bloomfield, 2004). This leaves a group of 40-

50% of AMLs that do not carry any grossly obvious cytogenetic lesions and are hence deemed

cytogenetically normal (CN), although a normal karyotype does not generally predict for a

favourable outcome as there is considerable variation across individual cases.

Page 26: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

16

Cytogenetic and molecular profiles provide important prognostic value in the prediction

of clinical outcome and guiding of treatment strategies, and are thus used to help stratify patients

based on risk. The National Comprehensive Cancer Network (NCCN) outlines three major risk

groups according to the presence (or absence) of a combination of genetic lesions: adverse,

intermediate and favourable (O’Donnell et al., 2017; Tallman et al., 2019). The adverse risk

group includes inv(3) and complex karyotype, the intermediate group includes t(9;11) and CN-

AML, while the favourable group includes t(8;21), inv(16) and t(15;17), the latter being the M3,

APL subtype that is characterized by the PML-RARA (PML = promeylocytic leukemia gene;

RARA = retinoic acid protein alpha) fusion protein. The risk groups differ with respect to

response to therapy, likelihood of relapse, and predicted long-term patient survival. The three

major risk groups are summarized in Table 1-3.

Page 27: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

17

Table 1-3. NCCN risk groups classification of AML. Adapted from National

Comprehensive Cancer Network (NCCN) Guidelines: Acute Myeloid Leukemia. Version

2.2019.

Page 28: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

18

1.2.3. Genetic Mutations in AML

In addition to cytogenetics, the molecular profiling of AML has becoming increasingly

important with demonstrated value in AML diagnosis and prognosis. There are a number of

recurrent molecular abnormalities that have aided in the prognostic stratification of the highly

heterogeneous group of CN-AML. Among frequently occurring molecular abnormalities are

internal tandem duplications (ITD) in FLT3 (FLT3-ITD) (25 – 30% of AML cases), mutations in

the transcriptional regulator nucleophosmin 1 (NPM1) that results in its cytoplasmic (c)

mislocalization (NPM1c) (30% of cases), MLL partial tandem duplications and a variety of

nucleotide substitutions and/or short insertions or deletions within the coding regions of the

genes NPM1, CEBPA, NRAS and WT1 (Betz & Hess, 2010a). Mutations in the tyrosine kinase

domain (TKD) of FLT3 may also occur in CN-AML but they are less common, occurring in

approximately 15% of all AML cases (Patnaik, 2018). Other recurrent mutations include

mutations in the isocitrate dehydrogenase (IDH) genes IDH1 and IDH2, TP53, RUNX1 (runt-

related transcription factor 1), TET2 (Tet methylcytosine dioxygenase 2), and DNMT3A (DNA

Methyltransferase 3 Alpha). In the prognostic stratification of AML,

DNMT3A and RUNX1 mutations have been shown to be predictors of shorter overall survival

(OS) in AML patients that are < 60 years old and with intermediate-risk cytogenetics. In

addition, NPM1 mutations in the absence of FLT3-ITD, mutated TP53, and biallelic CEBPA

mutations (discussed later) are significant molecular prognostic indicators associated with OS,

regardless of patient age (Metzeler et al., 2016). As such, FLT3-ITD, NPM1c and CEBPA are

recommended to be routinely screened for in CN-AML patients (Papaemmanuil et al., 2016b;

Roloff & Griffiths, 2018; Tallman et al., 2019).

Page 29: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

19

FLT3-ITD

The FLT3-ITD is a recurrent mutation found in approximately 25-30% of AML cases

(Daver, Schlenk, Russell, & Levis, 2019). FLT3 is a transmembrane class III receptor tyrosine

kinase (RTK). Class III RTKs are characterized by an insert region in the intracellular tyrosine

kinase domain and a cytoplasmic auto-inhibitory juxtamembrane domain adjacent to the

transmembrane domain. Mutations in the juxtamembrane domain result in ITDs that lead to

ligand-independent auto-phosphorylation of tyrosine residues in the kinase domain, resulting in

constitutive activation of the receptor (Gary Gilliland & Griffin, 2002). The ITDs arise from the

duplication and tandem insertion of a small variably sized (3 – 400 nucleotides) fragment of the

gene. The ITDs can be readily identified using PCR based on a larger product size, indicating

presence of the duplication, as compared to the wild-type gene. FLT3 activation leads to

stimulation of intracellular signal transduction pathways involved in growth, proliferation,

survival and differentiation, namely the JAK/STAT, MAPK/ERK and PI3K/Akt pathways.

FLT3-ITD mutations have been shown to induce the aberrant activation of STAT5 while

inhibiting the activity of transcription factors involved in myeloid differentiation (Choudhary et

al., 2007, 2005; Mizuki et al., 2000). STAT5 has been shown to play an important role in HSC

self-renewal, and is also found to be constitutively activated in human leukemias including AML

(Benekli, Baer, Baumann, & Wetzler, 2003).

NPM1

NPM1 is a phosphoprotein that shuttles between the nucleus and cytoplasm to regulate a

wide range of cellular functions. It predominantly resides in the nucleolus where it is involved in

ribosomal assembly and regulation of the tumor suppressors p53 and ARF. Mutations in NPM1

Page 30: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

20

are amongst the most common genetic aberrations in AML, occurring in 50-60% of CN-AML,

and 30% of all de novo AML cases (Welch et al., 2012). Over 40 mutations in exon 12 of the

NPM1 gene have been identified, with nearly all of them resulting in a 4-nucleotide insertion.

Mutations in NPM1 were first identified in AML based on the mislocalization of the protein in

the cytoplasm (Falini et al., 2009; Verhaak et al., 2005). The mutant (NPM1mut) form is denoted

NPM1c and was found to be associated with a CD34- immunophenotype. Clinically, NPM1c is

associated with a good response to induction therapy, lower rates of relapse and favourable

prognosis with good overall survival. Importantly, the prognostic value of NPM1c is highly

dependent on FLT3-ITD status, with NPM1c having a favourable prognosis only in FLT3-ITD

negative cases (Gale et al., 2008). For this reason, NPM1c and FLT3-ITD are now routinely

tested for clinically, in order to make more informed treatment plans and evaluate prognostic

outcomes. This highlights the significance of the contextual molecular interplay between

multiple disease markers, lending support to the notion of AML being a heterogeneous, multi-

factorial disease.

CEBPA

CCAAT enhancer binding protein alpha (CEBPA) is a transcription factor that regulates

the differentiation and development of granulocytes from hematopoietic progenitors. In

leukemogenesis, mutations in CEBPA lead to a loss in its function, which results in a block in

granulocytic differentiation (Ohlsson, Schuster, Hasemann, & Porse, 2016). CEBPA mutations

can occur either as frameshift mutations in the N-terminal region of the coding region of the

protein, leading to a truncated form of the full length p42 CEBPA protein, or C-terminal in-

frame mutations that result in defects in the dimerization and DNA binding activities of both the

Page 31: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

21

full-length p42 CEBPA protein and a shorter p30 isoform (Pabst et al., 2001). Most AML cases

with mutations in CEBPA harbour both mutations, occurring on different alleles, and are hence

biallelic, although about 1/3 of CN-AML cases only display one of the mutations (monoallelic).

It has been reported that biallelic mutations are associated with a favourable prognosis in CN-

AML, making it important to distinguish between biallelic and monoallelic cases (Dufour et al.,

2010). CEBPA mutations are most effectively detected using DNA sequencing, which is both

time and labour intensive, thereby limiting its widespread clinical testing. CEBPA mutations

occur at a frequency of 5-10% in de novo AML and are present in nearly 15% of CN-AML

cases. CEBPA is an independent prognostic marker and is associated with good overall survival

when both alleles are affected, and lower rates of relapse, similar to what is seen for

NPM1mut/FLT3-ITDneg AML (Nerlov, 2004).

Types of AML gene mutations

AML gene mutations can be broadly divided into several categories with respect to the

protein factors that they effect. These include epigenetic modifiers, components of signal

transduction pathways, transcription factors and gene fusion protein products. These are listed in

Table 1-4.

Mutations in genes encoding epigenetic modifiers such as DNMT3A, DNMT1, IDH1/2,

ASXL1, TET2 and EZH2 are often found in the ‘founder’ clone as they occur early on in

leukemogenesis (Döhner, 2007; Papaemmanuil et al., 2016a). Mutations in these genes are found

in elderly patients with clonal hematopoiesis, which puts them at higher risk for developing

hematological malignancies (Papaemmanuil et al., 2016b). Although seen as driver mutations,

Page 32: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

22

they are often not sufficient to drive disease, and require the acquisition of subsequent

mutation(s) to give rise to full-fledged disease.

Genes involved in signaling pathways are the most commonly mutated group of genes in

AML, representing up to 50% of all AML cases (The Cancer Genome Atlas Research Network,

2013). The most common mutations are found in genes encoding the class III receptor tyrosine

kinases FLT3 and KIT (or c-Kit), and the GTPase family of signaling mediators RAS (namely

KRAS and NRAS) (Fröhling, Scholl, Gilliland, & Levine, 2005). Activating mutations in RAS

proteins lead to their constitutive activation, contributing to the enhanced growth and

proliferation of AML cells.

Common chromosomal abnormalities in AML include t(8;21), inv(16)/t(16;16) and

t(15;17), which give rise to the RUNX1-RUNX1T1 (or RUNX1-ETO), Core-Binding Factor

(CBF)B-MYH11 and PML-RARA protein fusion products, respectively (Arber et al., 2016;

Ishikawa et al., 2009). RUNX1 and CBFB are subunits of the CBF, which is a transcription

factor involved in regulating normal hematopoiesis. As such, their gene fusion products,

resulting from the t(8;21) and inv(16)/t(16;16) rearrangements, cytogenetically define the class

of core-binding factor AMLs (Sangle & Perkins, 2011). Inv(16)/t(16;16) causes juxtaposition of

the CBF (core binding factor) gene located in 16q22 and the myosin, heavy chain 11, smooth

muscle (MYH11) gene located in 16p13, resulting in the fusion protein product (Delaunay et al.,

2003). The inv(16) rearrangement is usually associated with good prognosis (Patel et al., 2012).

AML and leukemogenesis can be seen to follow the classic ‘two-hit’ hypothesis model

wherein leukemia progenitor or initiating cells acquire two different mutations that lead to full

transformative potential and disease onset. This is highlighted by the fact that mutations in genes

from a similar group i.e. epigenetic regulators, do not occur concurrently in AML. For example,

Page 33: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

23

mutations in epigenetic regulators such as DNMT3a occur early on in leukemogenesis, followed

by mutations in NPM1 and CEBPA, while mutations in pro-growth and -survival signaling

factors involved in disease progression such as FLT3, KIT, NRAS/KRAS and PTPN11 occur later.

Support for this is apparent through the co-expression of FLT3-ITD and NPM1 mutations in

AML.

Page 34: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

24

Table 1-4. Types of frequently mutated genes in AML based on function

Page 35: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

25

1.2.4. AML Treatment

The treatment for AML has remained largely unchanged for the last 40 years, however

despite this, patient outcomes have generally improved over the past few decades. This is largely

owed to advancements in supportive care, including the enhanced use and development of novel

anti-bacterial, anti-fungal and anti-emetic agents, as well as improved management and support

for therapy-related myelosuppression and platelet transfusion (Ferrara & Schiffer, 2013b).

First-line treatment for AML involves chemotherapy administered in two different

phases: induction therapy and consolidation (post-remission) therapy. The goal of induction

therapy is to achieve complete remission (CR) while consolidation therapy aims to eliminate any

residual disease in order to affect a cure. CR is defined as BM blasts less than 5%, absence of

extramedullary leukemia, neutrophil count greater than 1x109/L, platelet count greater than

100x109/L and no dependency on transfusion (Cheson et al., 2003).

The typical induction chemotherapy regimen for all of the AML FAB subtypes except for

M3 or APL, is the ‘3+7’ (or ‘7+3)’ regimen, which consists of 7 consecutive days of continuous

intravenous (IV) administration of cytarabine (also known as cytosine arabinoside or Ara-C) and

3 days administration of an anthracycline such as daunorubicin (Betz & Hess, 2010b).

Structurally cytarabine is cytosine combined with arabinose which is similar in structure to the

nitrogenous DNA base cytosine deoxyribose, and is hence incorporated into DNA during cell

replication and interferes with DNA synthesis, that ultimately results in the formation of DNA

breaks (Löwenberg et al., 2011; Rider, 2011). Daunorubicin is a DNA intercalator and can

displace histones in chromatin, leading to conformational DNA instability (Pang et al., 2013). It

also functions to stabilize topoisomerase II, which is involved in relieving DNA supercoils

formed during the unwinding of DNA, preventing it from being resealed. Like cytarabine,

Page 36: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

26

daunorubicin produces DNA breaks, triggering an apoptotic response in the cell (Al-Aamri et al.,

2019). The M3 subtype (APL) is treated exclusively with all-trans retinoic acid (ATRA) in

conjunction with arsenic trioxide (ATO). ATRA degrades the PML-RARA fusion protein that

drives APL, which restores normal retinoic acid-mediated differentiation, leading to CR rates of

97%, and cure rates of over 90% (Khanna-Gupta & Berliner, 2007). ATO effectively targets

PML-RARA positive leukemic stem cells, leading to enhanced curative effects (Abaza et al.,

2017; Betz & Hess, 2010b; Tallman et al., 2019; Zheng et al., 2007).

Post-remission consolidation therapy is aimed at eliminating residual leukemic cells that

may lead to disease recurrence in the form of refractory or relapsed AML. Refractory disease

exists when the initial induction therapy fails to produce a state of remission with <5% blast cells

in the bone marrow. More common is AML relapse where although patients may achieve CR

from induction therapy, the disease reappears in a few months or after several years. The

mechanisms of relapse remain unclear, however there is evidence to suggest that specific

leukemic clones or sub-clones that are present at diagnosis, persist and undergo expansion post

initial therapy (Shlush et al., 2017). The extent of “consolidation therapy” is largely guided by a

patient’s prognostic factors including cytogenetics and molecular markers. For example, for

good prognosis disease i.e. inv(16), t(8;21) and t(15;17) (APL), consolidation therapy involves 3-

5 rounds of intensive high dose cytarabine containing chemotherapy. Cases with adverse

prognosis and/or having a high risk for relapse (i.e. high-risk cytogenetics, MDS, prior

chemotherapy) are recommended to undergo hematopoietic stem cell transplantation (HSCT)

(Sellar, Goldstone, & Lazarus, 2011). HSCT involves the transplantation of multipotent HSCs,

typically isolated from PB, BM or umbilical cord blood, into the patient. For patients with AML,

cells typically come from an unrelated donor or a matched sibling (allogeneic) (Felfly & Haddad,

Page 37: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

27

2014). The procedure involves first ablating the patient’s BM and immune system using high-

dose chemotherapy and/or irradiation. While stem cell transplantation can prove to be curative, it

is a relatively high-risk procedure. Major complications of allogeneic transplants include

infection and graft-versus-host disease (GVHD) due to transplanted cells recognizing normal

host tissues (i.e. of the skin, gut and liver) as being foreign, causing significant tissue damage,

which in some cases can lead to death (Goker, Haznedaroglu, & Chao, 2001)

.

Page 38: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

28

1.3. HEMATOPOIETIC AND LEUKEMIC STEM CELLS

Mechanisms underlying hematopoiesis and leukemogenesis have largely been elucidated

using in-vitro and in-vivo experimental models. Hematopoietic stem and progenitor cell

populations can be readily identified in in-vitro cell culture assays which assess the self-renewal

and multi-lineage differentiation properties of progenitor cells through their colony formation

ability.

1.3.1. In-Vitro Hematopoietic and Leukemic Cell Colony Formation Assays

The two main in-vitro assays used in the functional interrogation of HSCs are the colony

forming cell (CFC) and the long-term culture (LTC) assays (Dexter, Allen, & Lajtha, 1977;

Gartner & Kaplan, 1980a). The CFC assay involves plating single cells in a semi-solid medium

such as soft agar or methylcellulose with cytokine supplementation, and observing colony

formation over a period of 9-14 days. A single colony is scored when it has approximately 50

cells or more; the differentiation potential of the cell is inferred by the observed nature of the

cells in the colony. In these assays, the most primitive hematopoietic progenitor cell is the colony

forming unit (CFU), with different identifiable CFUs that each give rise to a distinct

hematopoietic lineage i.e. CFU-E (colony forming unit-erythroid), CFU-G (colony forming unit-

granulocyte, CFU-GM (colony forming unit-granulocyte-macrophage), BFU-E (burst forming

unit-erythroid), CFU-M (colony forming unit-macrophage) and CFU-GEMM (colony forming

unit-granulocyte, erythrocyte, macrophage, megakaryocyte) (Carow, Hangoc, & Broxmeyer,

1993).

LTC assays are co-culture assays that involve the use of stromal cells as a feeder layer,

along with growth factor/cytokine supplementation to help support the growth, differentiation

Page 39: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

29

and long-term maintenance of primitive hematopoietic progenitors in culture (Gartner & Kaplan,

1980b; M. Liu, Miller, & Eaves, 2013). The assays assess the ability of hematopoietic

progenitors to give rise to clonogenic myeloid progenitors and mature granulocytes and

macrophages that can be sustained for a period of 5-7 weeks or longer, under optimal culture

conditions. LTC assays to assess lymphoid and NK cell development have also been developed

(Miller, Verfaillie, & McGlave, 1992). The hematopoietic progenitors capable of initiating and

sustaining myelopoiesis over long-term periods are called long-term culture-initiating cells

(LTC-IC) and can be identified and quantified in the LTC assay (Sutherland, Lansdorp,

Henkelman, Eaves, & Eaves, 1990). CFUs are the progeny of LTC-ICs and can be detected at

approximately from a week or so to 5 weeks in semisolid assays.

1.3.2. In-Vivo Hematopoietic and Leukemic Cell Assays

The murine model has served as a useful tool for transplantation and re-population assays

in which single cells or single populations of hematopoietic or leukemic cells can be examined

for long-term hematopoietic re-constitution ability. In the 1960s, Drs. Till and McCulloch first

reported the ability of a specific population of cells from blood-forming tissue to form colonies

in the spleen of irradiated mice (Becker, McCulloch, & Till, 1963). These macroscopic colonies

were observed to contain cells along the granulocytic, erythroid and megakaryocytic lineages,

indicating clonal differentiation and expansion from a single cell population.

While the syngeneic system was very useful for studying the nature of murine

hematopoiesis and leukemia, it was not possible to evaluate human cells due to rapid rejection by

the mouse immune system. The non-obese diabetic/severe combined immunodeficient

(NOD/SCID) mouse model overcomes this problem, in part, and has been used extensively in

Page 40: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

30

human xenograft models. NOD/SCID mice harbour defects in both adaptive and innate immune

responses, with deficiencies in functional T, B and NK cells, which facilitates engraftment of

human cells in the murine background (Greiner et al., 1995; Larochelle et al., 1996; Shultz et al.,

2010). Through the use of such animals, the structure of the hematopoietic system first identified

for murine cells, has been confirmed for human hematopoiesis.

It is important to note that mouse models are in fact just that, models, in the study of

human cellular and molecular interactions. This is because there are significantly limited cross-

species reactivities between many human and mouse proteins. Many human growth factors do

not act on counterpart mouse receptors and vice versa, despite high protein homology among

some of the factors. Table 1-5 outlines a compiled list of protein ligands with known cross-

species receptor reactivity, along with % homology between the mouse and human forms of the

proteins. Of the dozen or so ligands listed, the overwhelming majority of mouse ligands (all

except SCF) do not act on the counterpart human receptor, despite sequence similarities of up to

91% (i.e. for HGF) between some of the human and mouse ligands. Interestingly in comparison,

about half a dozen of the listed human factors are actually active on mouse receptors, which may

suggest that placement of human cells in mice may actually induce changes in the mouse as

opposed to the other way around, which could have implications for the study model. These

considerations must be taken into account when working with animal models in general.

Page 41: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

31

Table 1-5. Cross-reactivity of hematopoietic and stromal cell growth

factors and cytokines between mouse and human.

Courtesy Laurie Ailles - A.L. Allan (ed.), Cancer Stem Cells in Solid Tumors,

Stem Cell Biology and Regenerative Medicine. DOI 10.1007/978-1-61779-

246-5_24

Page 42: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

32

Similar to hematopoiesis, leukemias like AML are also clonal in nature and organized into

a hierarchy with a leukemic stem cell (LSC) residing at its apex (Bonnet & Dick, 1997; Buick,

Minden, & McCulloch, 1979). McCulloch and colleagues first postulated that as with normal

hematopoiesis, leukemogenesis also consisted of multi-lineage differentiation and clonal

expansion stemming from discrete pools of stem and progenitor cells (McCulloch, 1983). This

was demonstrated by the fact that AML blasts have limited proliferative capacity, with only

about 1% of leukemic cells being capable of forming colonies in methylcellulose i.e. AML-

CFUs. Moreover, leukemias also appeared to retain many of the same growth and differentiation

properties evident in hematopoiesis. Moreover, John Dick’s group identified a rare population of

cells (1 per 1X106 leukemic blasts), termed human SCID leukemia-initiating cells (SL-IC), that

had the ability to re-populate NOD/SCID mice to propagate and re-capitulate AML in xenograft

transplants (Lapidot et al., 1994). These SL-ICs produced leukemic grafts that were

representative of the original patient’s disease with identical blast morphology and dissemination

patterns, and were thus deemed to be the cell of origin/LSC in the model. Furthermore, it has

been shown that there are several classes of LSCs, indicating the existence of a cellular hierarchy

in the disease (Hope, Jin, & Dick, 2004). For example, SL-ICs have been found to be enriched in

the CD34+/CD38- fraction, while AML-CFUs are found in the CD34+/CD38+ fraction, indicating

that the leukemic clone is organized as a hierarchy with the SL-IC as the cell of origin that gives

rise to AML-CFUs and blasts.

Page 43: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

33

1.3.3. Hematopoietic Cell Surface Markers

Expression of cell surface markers has played an important role in characterizing the cells

of the hematopoietic system, and in particular, the identification and isolation of HSCs, LSCs

and their progenitors. These, in conjunction with functional assays, helps to confirm the

biological identity and activity of isolated cell fractions containing putative HSCs or progenitor

cells.

CD34

Cluster of differentiation 34 (CD34) is a transmembrane phosphoglycoprotein that was

originally discovered as a cell surface antigen on primitive hematopoietic cells (Batinić et al.,

1989; Civin et al., 1984; Tindle et al., 1985). Although largely expressed on hematopoietic cells,

CD34 is also expressed on several other cell types such as endothelial cells (in the BM and in

vascular tissue), mesenchymal stem cells, interstitial cells, muscle satellite cells and cells of the

placenta (Sidney, Branch, Dunphy, Dua, & Hopkinson, 2014). While its exact function is not

clearly known, CD34 is an important adhesion molecule as it helps HSCs to home to the BM,

and also aids T-cell entry into lymph nodes. It may also be involved in chemokine-mediated

migration of eosinophils and dendritic cells. In disease, CD34 is found to be expressed on the

malignant cells of pre B-ALL, AML, gastrointestinal stromal tumors, Kaposi’s sarcoma,

liposarcoma, neurofibromas, meningiomas, papillary thyroid carcinomas and alveolar soft part

sarcoma among others. In most of these cancers including AML, high CD34 levels are generally

associated with poor clinical outcome (Vergez et al., 2011).

Page 44: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

34

c-Kit

c-Kit (CD117) is a receptor tyrosine kinase that is the cellular homolog of the feline

sarcoma viral oncogene v-kit (Yarden et al., 1987). It is also known as the proto-oncogene Kit or

the Stem Cell Growth Factor Receptor (SCFR). c-Kit is mainly expressed on the surface of

primitive hematopoietic cells i.e. stem and progenitor cells, with predominant expression on

myeloid progenitors. It is a type III receptor tyrosine kinase that binds to its ligand, Kit ligand,

which is also known as stem cell factor (SCF) (Edling & Hallberg, 2007). c-Kit is found to be

overexpressed in some cancers and activating mutations (typically in exon 11 or exon 17) are

commonly found in gastrointestinal stromal tumors, testicular seminoma, melanoma and AML

(Ashman & Griffith, 2013).

CD38

CD38 is a cell surface glycoprotein found to be mainly expressed on the surface of immune

cells including T-cells, B-cells and natural killer cells. It is also known as cyclic ADP ribose

hydrolase, a multifunctional ectoenzyme that catalyzes the synthesis and hydrolysis of cyclic

ADP ribose (Jackson & Bell, 1990). It is involved in signal transduction, calcium signaling and

cell adhesion. CD38 holds prognostic value in leukemias and is associated with shorter time to

progression in chronic lymphocytic leukemia (CLL) (Burgler, 2015). CD38 expression may be

related to cell activation, as expression levels increase when HSCs are activated. Case in point,

CD38 levels have been shown to fluctuate on CD34+ LT-HSCs, seemingly in accordance to cell

cycle state (McKenzie, Gan, Doedens, & Dick, 2007).

Page 45: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

35

1.4. THE BONE MARROW NICHE

The bone marrow (BM) is the principal site of hematopoiesis as well as osteogenesis, or

bone formation. Sites of hematopoiesis in the BM are niche microenvironments composed of

complexes of cells, distinct spatial/anatomical regions, temperature and oxygen gradients,

extracellular matrix and signaling molecules. The oxygen content in the BM is quite low, ranging

from 0.2 – 1%, making it a relatively hypoxic environment. There are a multitude of different

cell types within the BM which include hematopoietic, osteogenic (i.e. osteocytes, osteoblasts,

osteoclasts), adipocytes, perivascular, reticular fibroblasts, neuronal-associated glial cells and

BM stromal cells (Chan et al., 2013; Gimble, Robinson, Wu, & Kelly, 1996; Méndez-Ferrer,

Michurina, Ferraro, Mazloom, MacArthur, Lira, Scadden, Mag’Ayan, et al., 2010; Sacchetti et

al., 2007). Osteocytes and osteoblasts, the latter of which are involved in bone formation and

mineral deposition, are derived from osteoprogenitor cells (which arise from mesenchymal stem

cells), while osteoclasts which resorb and remodel bone, derive from hematopoietic myeloid

cells (Nijweide, Burger, & Feyen, 2017; Teitelbaum, 2000).

In the human adult, hematopoiesis specifically occurs in the BM of the femur, ribs,

vertebrae and pelvic bones as these bones contain ‘red marrow’ or ‘myeloid tissue’. BM tissue is

composed of three main types of cells: hematopoietic, supporting stromal and adipose tissue

cells. In humans, marrow tissue exists as either ‘red’ or ‘yellow’ marrow, depending on the

proportion of hematopoietic (red) vs. adipocytes or fat cells (yellow) (Poulton, Murphy, Duerk,

Chapek, & Feiglin, 1993).

Page 46: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

36

1.4.1. Cells and Factors in the Bone Marrow Niche

The BM is divided into two main compartments: the hematopoietic (or parenchymal) cell

component called the parenchyma, and a non-hematopoietic component called the stroma. By

definition, ‘stroma’ means ‘covering’, ‘bed’ or ‘layer’ in Greek, which points to a supportive and

protective role for it. The stroma, in biological systems, is largely made up of connective tissue

to provide structural and connective supports in tissues and organs. The stroma is composed of

osteogenic cells, adipocytes, nerves, vasculature, ducts and stromal cells such as fibroblasts,

mesenchymal cells (MSC) and endothelial cells. Fibroblasts are large, flat, thin (spindle-shaped)

cells that help establish and maintain the structural integrity and framework of tissues, and also

play critical roles in wound healing and tissue repair (Hans-Georg Kopp, Avecilla, Hooper, &

Rafii, 2005; Silzle, Randolph, Kreutz, & Kunz-Schughart, 2004). They produce collagen and

extracellular matrix protein precursors, namely glycoproteins and reticular and elastic fibres, in

addition to a number of important cytokines and growth factors. Fibroblasts have typically been

difficult to identify definitively, with identification largely based on their morphological spindle

shape, along with positive staining for the mesenchymal marker vimentin, combined with

negative staining for epithelial or other mesenchymal cell types such as muscle cells, astrocytes

or hematopoietic cells (Botstein et al., 2002). MSCs, also known as marrow stromal cells, are

multipotent stromal cells derived from the mesoderm that have the capacity to differentiate into

three main cell types: osteoblasts, adipocytes and chondrocytes which make up bone, adipose

and connective tissue, respectively (Méndez-Ferrer, Michurina, Ferraro, Mazloom, MacArthur,

Lira, Scadden, Ma’ayan, et al., 2010; Schroeder, Geyh, Germing, & Haas, 2016). Collectively,

BM stromal cells produce important adhesion, growth and signaling molecules that regulate key

BM processes including hematopoiesis.

Page 47: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

37

The physical architecture of the BM is organized into spatial areas or zones with

differences in cell and tissue composition. Long bones primarily contain two types of bone

tissue: spongy and compact. Spongy, also known as trabecular or cancellous, bone is soft,

loosely organized, porous, highly vascular and contains red BM i.e. hematopoietic cells. It is

located in the trabecular regions, or the ends, of long bone, often near joints. Compact, or

cortical, bone tissue is more dense and forms the hard outer layers of the bone (except at joints).

Bones have an outer membranous layer called the periosteum and within the bone, there is a

layer called the endosteum that separates compact bone from spongy bone (Morrison & Scadden,

2014a; Scadden, 2006). The vasculature within the BM is sinusoidal in that the endothelial cells

covering the vessels are not covered by a layer of connective tissue and are in direct contact with

parenchymal cells (H.-G. Kopp, 2005). HSCs have been shown to preferentially localize to two

predominant niches within the BM: an ‘endosteal or osteoblastic’ niche where cells localize to

endosteal regions lined with osteoblasts within trabecular bone and a ‘perivascular niche’ where

cells localize adjacent to sinusoidal blood vessels (Figure 1.2) (Colmone et al., 2008; Guezguez

et al., 2013; Morrison & Scadden, 2014b). As HSCs differentiate, the niches that developing

cells occupy can change. For example it has been shown that while some HSCs predominantly

reside in perivascular niches, lymphoid progenitors appear to occupy endosteal niches (Ding &

Morrison, 2013).

Osteoblasts produce growth factors and cytokines involved in cellular growth and adhesion

such as G-CSF, GM-CSF, IL-1, IL-6 and TGF-β. These cells also produce the key adhesion

factor and chemokine, stromal-derived factor-1 (SDF-1), also known as CXC-chemokine ligand

12 (CXCL12), which acts on the CXCR4 receptor found on HSCs (Calvi et al., 2003; Galán-

Díez & Kousteni, 2018; Sugiyama, Kohara, Noda, & Nagasawa, 2006). CXCR4 antagonists

Page 48: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

38

induce the mobilization of HSCs into the bloodstream, as a result of detachment and release from

the BM space (Broxmeyer et al., 2005). Osteoblasts also produce vascular and angiogenic

growth factors such as Angiopoietin-1 (Ang-1) which is thought to maintain HSC quiescence by

acting on the Tie2 receptor expressed on HSCs; activation of this pathway is involved in cell

cycle arrest and in maintaining stemness (Arai et al., 2004).

Page 49: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

39

Figure 1.2. Architecture of the bone marrow niche.

The BM contains various zones that contain distinct supporting BM stromal cells. HSCs as well

as LSCs typically reside within the perivascular and endosteal regions. Figure adapted from

(Morrison & Scadden, 2014a).

Page 50: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

40

1.4.2. Hematopoietic and Leukemic Cells in the Bone Marrow Niche

The BM niche is also important for the proliferation and maintenance of leukemic cells and

provides protection against toxic agents including chemotherapeutic drugs. It also maintains the

dormancy and quiescence of both HSCs and LSCs. This niche is a dynamically evolving

microenvironment as it responds to biochemical perturbations, and can become altered itself. For

example, it has been shown that osteoblasts with activating mutations in β-catenin lead to

enhanced HSC proliferation and leukemogenesis in mouse models (Kode et al., 2014).

Moreover, impaired activity of the hematopoietic transcription factor Ikaros in pre-B-cells has

been shown to lead to a stroma-dependent leukemia (Joshi et al., 2014). This underscores the

importance of components of the niche environment as functionally dynamic players in BM

homeostasis. Moreover, leukemic cells often share/compete for the same niche environments as

their normal hematopoietic cell counterparts. It has been shown that in competitive repopulation

assays, normal HSPCs in some instances can actually out-compete LSCs (Boyd et al., 2014).

This highlights the dynamic relationship that exists between normal hematopoiesis and

leukemogenesis which reside in the same microenvironments, competing for dominance.

Leukemic stem/initiating cells

The notion that stem cells may exist in cancers and be central to carcinogenic processes

emerged at around the same time normal stem cells were being studied. Central to the theory was

that similar to hematopoiesis, leukemias, are also clonal in nature and organized into a hierarchy

with a leukemic stem cell (LSC) residing at its apex (Bonnet & Dick, 1997). McCulloch and

colleagues first postulated that like normal hematopoiesis, leukemogenesis also consisted of

multi-lineage differentiation and clonal expansions stemming from discrete pools of stem and

Page 51: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

41

progenitor cells (McCulloch, 1983). This is supported by the fact that the majority of AML blasts

have limited proliferative capacity, with only about 1% of leukemic cells being clonogenic

progenitors i.e. AML-CFUs. John Dick’s group identified in a subset of AML patients, a rare

population of cells (1 per 1X10^6 leukemic blasts), termed human SCID leukemia-initiating cells

(SL-IC). These cells had the ability to re-populate NOD/SCID mice to propagate and re-

capitulate AML in xenograft transplants (Lapidot et al., 1994). The leukemic grafts developing

from SL-IC were representative of the original patient’s disease, having identical blast

morphology and dissemination patterns, and were thus deemed to be the cell of origin/LSC in the

model. Moreover, there are distinct LSC classes, demonstrated by the fact that SL-ICs are found

to be enriched in the CD34+/CD38- fraction, while AML-CFUs are found in the CD34+/CD38+

fraction (Hope et al., 2004). This indicates that the leukemic clone is organized as a hierarchy

with the SL-IC as the cell of origin that gives rise to AML-CFUs and blasts. Furthermore,

through clonal tracking studies, distinct classes of SL-ICs were observed: short-term, long-term

and quiescent.

Clonal hematopoiesis refers to the process whereby distinct pools of stem or progenitor

cells acquire one or more somatic mutations, and undergo clonal expansion to become a

dominant clone that can give rise to disease (Bowman, Busque, & Levine, 2018; Greaves &

Maley, 2012). In essence, somatic mutations acquired throughout an individual’s life may be

predictive of disease onset later on in life. In what is termed age-related clonal hematopoiesis,

healthy individuals are seen to carry recurrent somatic mutations in specific genes associated

with hematological malignancies, namely mutations in the key epigenetic regulators DNMT3A,

TET2 and AXSL1. The frequency of these mutations increases with age and is associated with an

Page 52: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

42

increased risk for hematological cancer and possibly cardiovascular disease as well (Jaiswal et

al., 2014). The mechanism for the latter is an area of intense investigation.

The clonal nature of hematopoiesis and leukemia begs the question as to how these clones

evolve and potentially interact with each other. Moreover, in leukemia, the question is which

clones acquire transforming mutations to become malignant, and how and when they evolve in

time and space to become the dominant clone. Tracing the evolutionary origins of distinct

disease-driving hematopoietic clones led to the identification of pre-leukemic HSCs in AML

patients. Employing deep-sequencing methods and lineage tracing, the investigators found that

HSCs from AML patients harbouring a DNMT3A mutation displayed enhanced multi-lineage

repopulation in xenografts compared to HSCs without the mutation (Jan et al., 2012; Liran I.

Shlush, Sasan Zandi, Amanda Mitchell, Weihsu Claire Chen, 2014). This DNMT3Amut

harbouring clone was deemed to be a pre-leukemic HSC clone. Pre-leukemic HSCs were also

found in remission samples, suggesting that they are able to survive chemotherapy. It therefore

appears that DNMT3A mutations occur early on in leukemogenesis, likely in HSCs, leading to

the clonal expansion of this pool of pre-leukemic HSCs that may eventually give rise to AML.

LSC clones thus evolve early on in the route to malignancy, survive therapy, and persist to give

rise to relapse. To support the latter, Shlush et al. reported that there are distinct LSC clones and

subclones that are present both at diagnosis and also found later in AML relapse (Shlush et al.,

2017). This suggests that pre-leukemic and leukemic stem cells are highly potent and persistent,

as they drive and maintain disease throughout all stages of leukemogenesis.

Page 53: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

43

1.5. BONE MARROW STROMAL CELL ASSAYS

Classical assays used to characterize hematopoietic and leukemic cells in-vitro and ex-

vivo such as the LTC-IC assay, largely make use of BM stromal cells as a supportive feeder layer

in a co-culture system for the long-term growth of cells in culture. The murine MS-5 stromal cell

line has been widely used in hematopoietic cell co-culture assays due to the ease in their growth

and maintenance (Itoh et al., 1989). MS-5 cells were initially derived from adherent layers of

Dexter-type murine long-term marrow cultures (used to study murine hematopoiesis) as one of 7

cell lines. From these 7 cell lines, MS-5 cells were found to be the most active in supporting the

growth of HSCs (CFU-S and CFU-GM) for more than 2 months in-vitro, without the addition of

any exogenous factors (Itoh et al., 1989; Varma, el-Awar, Palsson, Emerson, & Clarke, 1992).

MS-5 cells grow as fibroblast-like cells, adhere well to plastic surfaces, typically grow in a

monolayer and produce a number of hematopoietic growth factors, making them ideal for the

support of hematopoiesis. They were also found to support the proliferation and survival of

primitive CD34+/CD38- HSCs in both short-term (5-8 weeks) and long-term (8-10 weeks) co-

cultures (Issaad, Croisille, Katz, Vainchenker, & Coulombel, 1993). MS-5 cells synthesize

extracellular matrix proteins such as fibronectin, laminin and collagen type I. In addition, these

cells produce important hematopoietic growth factors including IL-6, IL-3, GM-CSF and SCF.

Interestingly, despite the fact that many murine growth factors and cytokines do not act on their

counterpart human receptors, murine MS-5 cells are still able to effectively support human

hematopoiesis without any human growth factor supplementation. While there are a few factors

that display species cross-reactivity, namely SCF, CXCL12 and GM-CSF, their involvement is

not enough to explain the robust support that MS-5 cells provide human cells in long-term

cultures (Kobari, Dubart, Le Pesteur, Vainchenker, & Sainteny, 1995). The exact mechanisms

Page 54: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

44

underlying the efficacy of MS-5 stromal cells in the support of human hematopoietic cells

remains unclear, but are likely attributable to unidentified factors that MS-5 cells produce in

small amounts, or to extracellular matrix protein products that may lend both structural and

biochemical support. Human stromal cell lines such as HS-5 and HS-27 can also be used in co-

culture assays but they are not as efficient in supporting hematopoietic cells in long-term assays.

Long-term hematopoietic and leukemic cell assays have certain defining features

associated with the growth of specific cell populations. For example, the culture of CD34

positive AML cells on MS-5 stromal cells leads to their long-term expansion, which can be

maintained for up to 24 weeks (van Gosliga et al., 2007) for some samples. After a period of 1-2

weeks, these cells form ‘cobblestone-area forming cells’ (CAFC) which are clusters of cells that

burrow and settle between stromal cells and the substratum (i.e. the plastic dish). CAFCs can be

visualized using phase contrast microscopy under which they appear as dull, ‘cobblestone’-like

areas (Ploemacher, van der Sluijs, Voerman, & Brons, 1989). CAFCs arise from a CD34+

progenitor pool that is present in both normal hematopoietic and leukemic cell populations.

CAFCs can be re-plated to give secondary and tertiary CAFCs, providing evidence for the

existence of a pool of pluripotent progenitor cells with self-renewal capacity and long-term

marrow repopulating ability (Ploemacher, van der Sluijs, van Beurden, Baert, & Chan, 1991).

Page 55: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

45

1.6. COLONY STIMULATING FACTOR 1 RECEPTOR (CSF1R)

The colony stimulating factor 1 receptor (CSF1R, c-FMS, CD115) is a class III tyrosine

kinase receptor, belonging to the same family as the Platelet-Derived Growth Factor (PDGF), c-

Kit and Fms-like tyrosine kinase 3 (FLT-3) receptors (Françoise Birg, Carbuccia, Rosnet, &

Birnbaumt, 1994; Casteran, Beslu, Lecocq, Gomez, & Dubreuil, 1998). It is involved in

modulating innate immunity, inflammation as well as diseases including cancers. CSF1R is

expressed on the surface of mononuclear phagocytes, namely monocytes, macrophages and

dendritic cells and their precursors to direct their growth, survival and differentiation (Auffray,

Sieweke, & Geissmann, 2009; Sasmono et al., 2003). CSF1R is involved in a number of

physiological processes including osteoclastogenesis as related to bone and tooth development,

immune responses, mammary gland and placental development during pregnancy and microglial

cell development in the brain. During immune responses and inflammation, CSF1R is involved

in the activation of monocytes and macrophages to promote their cytotoxic, phagocytotic and

chemotactic functions through release of cytokines and chemokines (Raivich et al., 1998; Y.

Wang et al., 2012).

1.6.1. CSF1R Structure

In humans, CSF1R is encoded by the c-fms proto-oncogene, the human homologue of the

v-fms oncogene from the Susan McDonough strain of the feline sarcoma virus (Garceau et al.,

2010; Carl W. Rettenmier, Chen, Roussel, & Sherr, 1985; Charles J. Sherr et al., 1985). The gene

is located on the long arm of chromosome 5 (5q32) and is 60 kb in length with 22 exons, 21 of

which are coding exons. The mature protein product is heavily glycosylated, 972 amino acids in

length and has a molecular weight of approximately 150-165 kDa. It is a single type I pass

Page 56: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

46

membrane protein comprised of a 512 amino acid extracellular portion containing five

immunoglobulin (Ig)-like repeat domains (D1 - D5), a single transmembrane segment, a

juxtamembrane domain that lies between the transmembrane domain and cytoplasmic domain,

and a cytoplasmic domain containing an ATP binding/activation loop, kinase insert region

(resulting in a split kinase domain), the main kinase domain and an auto-inhibitory C-terminal

tail (Figure 1.3) (Hamilton & Achuthan, 2013; Charles J. Sherr et al., 1985). In its inactive auto-

inhibited state, CSF1R is stabilized by its juxtamembrane domain, which binds to a site adjacent

to the ATP binding pocket in the kinase domain. This inhibits the activation loop from adopting

an active conformation, maintaining it in an inactive, locked state to prevent binding of ATP and

substrates (Walter et al., 2007).

Page 57: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

47

Figure 1.3. Structure of CSF1R.

CSF1R contains an extracellular domain with five immunoglobulin loops (D1 – D5), a

juxtamembrane domain, a transmembrane domain, a split kinase domain with a kinase insert,

and a cytoplasmic tail that can be autoinhibitory.

Page 58: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

48

Colony stimulating factor 1 (CSF1) and IL-34 are the two known ligands of CSF1R.

CSF1 binds to the first three extracellular domains (D1 - D3) of CSF1R, resulting in receptor

dimerization through interaction of the D4 and D5 domains (Figure 1.4). Upon ligand binding,

oligomerization of the receptor chains brings the kinase domains of the two chains into close

proximity, allowing for the auto and cross phosphorylation of several tyrosine residues within the

domains. These residues serve as docking sites for various cytoplasmic proteins, namely SH2

domain-containing proteins such as Grb2, phosphatidylinositide 3-kinase (PI3K) and Src family

kinases. Binding of these proteins triggers activation of signal transduction pathways involved in

cellular growth, proliferation and survival. Following CSF1 stimulation, activated CSF1R can

lead to various cellular events including actin skeleton reorganization, membrane ruffling,

immediate cytoplasmic alkalinization, increased hexose transport and induction of early response

genes such as c-fos and c-jun that participate in regulation of cellular metabolism and growth

(Yu et al., 2008); discussed in more detail below.

Page 59: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

49

Figure 1.4. CSF1 ligand binding to CSF1R.

Homodimeric CSF1 binds to the D1 – D3 loops of the extracellular domain of CSF1R. This

leads to receptor dimerization, resulting in the autophosphorylation of tyrosine residues in

the juxtamembrane, kinase insert and kinase domains of the receptor.

Page 60: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

50

1.6.2. CSF1R Intracellular Signaling

Interaction and binding of signaling molecules to phosphorylated tyrosine residues on

CSF1R leads to activation of key growth and survival pathways, the predominant of which

include the PI3K/Akt, MAPK/ERK and JAK/STAT pathways (Figure 1.5). CSF1R activation

leads to tyrosine phosphorylation of PI3K which is a class I PI3K heterodimer containing p85

regulatory and p110 catalytic (PIK3CA) subunits (Bunney and Katan, 2010). PI3K converts

phosphatidylinositol (4,5) bisphosphate (PI(4,5)P2) into phosphatidylinositol (3,4,5)

trisphosphate (PI(3,4,5)P3), which recruits Akt and phosphoinositol-dependent kinase 1 (PDK1)

to the cell membrane via interaction with their pleckstrin homology (PH) domains. PDK1

phosphorylates Akt on one of its residues, Thr308, with full activation requiring phosphorylation

of another residue, Ser473, which can be mediated by other kinases including PDK2, DNA

protein-dependent kinase (DNA-PK), mammalian or mechanistic target of rapamycin complex

(mTORC) and integrin-linked kinase (ILK) (Cantley, 2002; Hemmings & Restuccia, 2012;

Osaki, Oshimura, & Ito, 2004). Akt functions to regulate pathways involved in cell proliferation,

differentiation, apoptosis and survival through transcriptional level control of gene expression, or

through direct phosphorylation of effector molecules including transcription factors. Akt

activates mTOR through phosphorylation-mediated inhibition of its negative regulator, the

tuberous sclerosis (TSC) protein complex, TSC1/2, which is made up of hamartin (TSC1) and

tuberin (TSC2). TSC2 specifically has a GTPase Activating Protein (GAP) domain which

downregulates the GTPase activity of the small GTPase Rheb, which when active in its GTP-

bound form, is an activator of the mTORC1 complex (which consists of mTOR, which is the

catalytic subunit of mTORC complexes, regulatory-associated protein of TOR (Raptor), DEP

domain-containing mTOR-interacting protein (DEPTOR), mammalian lethal with SEC13 protein

Page 61: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

51

8 (MLST8) and PRAS40). Activated mTOR phosphorylates target effectors involved in protein

synthesis, namely p70S6 Kinase (p70S6K), which phosphorylates the S6 ribosomal protein, and

the eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) (Manning &

Cantley, 2007). Akt also inhibits Glycogen Synthase Kinase-3 (GSK-3), leading to inhibition of

glycogen synthesis. In its pro-survival function, Akt both activates anti-apoptotic, and inhibits

apoptotic effectors. Some of the transcriptional targets of Akt include the Forkhead Box (FOX)O

family of transcription factors (through phosphorylation) and pro-survival genes. Akt can cause

degradation of IκB which binds to NF-κB, causing NF-κB translocation to the nucleus where it

can promote expression of caspase inhibitors such as Bcl-xL (Vanhaesebroeck & Alessi,

2000). Akt has been shown to up-regulate expression of the anti-apoptotic protein Bcl-2 through

phosphorylation of the cAMP response element binding protein (CREB), which leads to

recruitment of CREB-binding protein (CBP) to the promoter of Bcl-2 (Pugazhenthit et al., 2000;

Ross & Teitelbaum, 2005). Akt itself can translocate from the cytoplasm to the nucleus in

various cell types to elicit its transcriptional level control through interaction with various

nuclear proteins including RNA polymerase II, as well as β-actin (Carling, 2017; Coa et al.,

2019).

CSF1R also induces phosphorylation of the cytoplasmic Tyk2 tyrosine kinase belonging

to the family of JAK kinases, leading to induction of the signal transducers and activators of

transcription (STAT) protein transcription factors. CSF1 induced CSF1R activation can also lead

to Ras activation. The Ras family of GTPases are signaling proteins anchored within the cell

membrane from where they transduce signals into the cell. Ras GTPase activity is activated by

Ras guanine nucleotide exchange factors (GEF) that exchange GDP for GTP to activate the

protein. SOS is a GEF that interacts with the adaptor proteins Grb2 and Shc. Grb2 has been

Page 62: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

52

shown to bind to the phosphorylated Y699 residue in the kinase domain of activated CSF1R.

Activated Ras can then activate MAPK/ERK signaling. ERK1 can also bind directly to several

phosphotyrosine residues in activated CSF1R. In macrophages, it has been shown that the

binding and activation of multiple signal transducers and pathways to CSF1R including Src

kinases, Akt, ERK and PLC is required for macrophage proliferation (Takeshita et al., 2007).

The autophosphorylation of specific tyrosine residues (of which there are 6-8) in the

cytoplasmic kinase domain, as well as the kinase insert region (containing Y699, Y708 and

Y723), of CSF1R leads to the binding of cytoplasmic proteins to these residues resulting in their

activation, leading to the regulation of key cellular events. The binding proteins targeted to a

number of these tyrosine sites have been elucidated. The Y809 site in the activation loop of the

receptor (Y807 in mouse) has been shown to promote differentiation in FDC-P1 myeloid cells by

acting as a key regulator of a critical switch between growth and differentiation signals

(Rohrschneider et al., 1997). Moreover ectopic expression of a substitution mutant of Y809,

Y809F, resulted in a G1 cell cycle arrest and impaired production of c-myc mRNA in ligand-

stimulated NIH3T3 murine fibroblasts, with cells being mitogenically inactive (Roussel & Sherr,

1989; Roussel, Shurtleff, Downing, & Sherr, 1990). The mitogenic activity of cells could be

rescued with c-Myc overexpression in the cells, demonstrating that c-Myc is a downstream target

of CSF1R induced in response to CSF1 ligand binding. In a CSF1R-deficient murine BM

macrophage cell line (MacCsf1r-/-), ectopic expression of wild-type CSF1R with mutations

induced in the eight different tyrosine residues revealed that the Y559F mutation in the

juxtamembrane domain and the Y807F mutation in the activation loop led to severe impairments

in proliferation and differentiation, while the Y706, Y721F, and Y974F mutations led to changes

in morphological responses, with Y706F increasing differentiation in the cells (Yu et al., 2008).

Page 63: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

53

This demonstrates the specific phenotypic functionalities of individual tyrosine phosphorylated

residues within the kinase domain of CSF1R.

Tyrosine autophosphorylation allows for the binding of cytoplasmic proteins with SH2

domains. In Rat-2 fibroblasts transduced with murine c-fms, Y697 (Y699 in human) was

identified as a binding site for the adaptor protein Grb2 upon CSF1 stimulation. Activation of

PI3K activity in response to CSF1 has been reported, with the Y721 having been shown to be the

binding site for the p85 regulatory subunit in Rat-2 fibroblasts. In the juxtamembrane domain of

activated human CSF1R, phosphorylated Y561 (Y559 murine) may be a binding site for Src

family members. The E3 ubiquitin-protein ligase and proto-oncogene c-Cbl (murine), which is

found to be mutated in some AML cases, is also a CSF1R binding target that induces

ubiquitination of activated CSF1R by associating with tyrosine-phosphorylated Shc at the plasma

membrane. This may also involve association with PI3K to form a complex leading to receptor

ubiquitination and degradation. STAT transcription factors are also targets of CSF1-induced

CSF1R activation. Murine Y706, and human 708 of CSF1R, has been shown to be necessary for

the binding of STAT1. STAT3, STAT5 and STAT5a are also induced in response to CSF1 but

do not bind to the same residues as bound by STAT1 (Price, Choi, Rosenberg, & Stanley, 1992;

Reedijk et al., 1992).

CSF1R activity is downregulated through dephosphorylation of the receptor and its

downstream effectors by protein phosphatases such as INPP5D/SHIP-1, as well as through rapid

internalization of the activated receptor (E. Richard Stanley & Chitu, 2014).

Page 64: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

54

Figure 1.5. CSF1R activation of intracellular signal transduction pathways.

Upon activation, CSF1R stimulates the activation of a number of intracellular signal transduction

pathways involved in regulation of cell growth, proliferation, survival, apoptosis, differentiation

and metabolism.

Page 65: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

55

1.6.3. CSF1R in Disease

In disease, CSF1R is found to be over-expressed or mutated in several neurodegenerative

disorders. High CSF1R expression is found on microglia i.e. the resident macrophages in the

brain, in Alzheimer’s disease and amyotrophic lateral sclerosis (ALS), (Akiyama et al., 1994;

Knight, Brill, Queen, Tarwater, & Mankowski, 2018). CSF1R mutations are found in the kinase

domain of the receptor in hereditary diffuse leukoencephalopathy with spheroids (HDLS), an

autosomal dominant neurodegenerative disorder (Rademakers et al., 2012). CSF1R has been

shown to specifically regulate the density and distribution of microglia in adult-onset

leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) (Oosterhof et al., 2018).

In cancers, CSF1R has been found to be highly expressed in breast, ovarian, endometrial,

prostate and renal cancers, as well as in melanoma and classical Hodgkin lymphoma (Espinosa et

al., 2009; Ide et al., 2002; B M Kacinski, 1991; Barry M. Kacinski, 1997; Kirma et al., 2007;

Morandi, Barbetti, Riverso, Dello Sbarba, & Rovida, 2011; Tang et al., 1992). High CSF1R

expression in these cancers is usually associated with co-expression of CSF1 ligand. In most of

these solid tumors, increased CSF1R expression is found on tumor-associated macrophages

(TAM) (Edginton-White et al., 2018; Martín-Moreno et al., 2015; Q. Wang et al., 2018).

Macrophages are classically classified based on an activation state polarity of either being pro-

inflammatory (anti-tumorigenic) or anti-inflammatory (pro-tumorigenic), being termed M1 vs.

M2 macrophages, respectively. There are 2 main pathways that lead to divergent routes of

macrophage activation, with activation of distinct T cell immune responses. Classical

macrophage activation leads to production of M1 macrophages through the action of interferon

gamma (IFN-γ); these are involved in the killing of microbial pathogens. In contrast, an

alternative activation leads to production of M2 macrophages in response to IL-4 or IL-13 which

Page 66: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

56

have been shown to fight against parasites and allergens (Gordon & Martinez, 2010; Schroder,

Hertzog, Ravasi, & Hume, 2004).

In most cancers, the predominant TAMs have the M2 phenotype and are anti-

inflammatory, immune-suppressant and hence tumor-promoting (Ley, 2017; Noy & Pollard,

2014). Myeloid cells are an important component of the tumor microenvironment as they play

key roles in tumorigenesis. These cells include TAMs, polymorphonuclear myeloid-derived

suppressor cells, (PMN-MDSC), monocytic MDSC (M-MDSC) and dendritic cells (DC) (Kumar

et al., 2017). TAMs and MDSCs have been shown to promote tumorigenesis by suppressing T-

cell functions and promoting tumor angiogenesis, proliferation, survival and metastasis

(Coussens & Pollard, 2011; Talmadge & Gabrilovich, 2013). CSF1R functions to regulate TAM

differentiation, growth and survival and is often overexpressed by the cells. As such, CSF1R has

become a therapeutic target of interest with the development of small molecule inhibitors and

antibodies, some of which are currently in clinical trials for classical Hodgkin lymphoma,

melanoma and tenosynovial giant-cell tumor (Tap et al., 2015; Von Tresckow et al., 2015).

1.7. COLONY STIMULATING FACTOR 1 (CSF1)

Colony stimulating factor 1 (CSF1, also known as macrophage colony stimulating factor,

MCSF) is a cytokine that regulates the differentiation and proliferation of monocytes and

macrophages from myeloid cell precursors (E. R. Stanley, Guilbert, Tushinski, & Bartelmez,

1983). It is highly expressed in the bone marrow, brain, liver and female reproductive tissues

(The Human Protein Atlas). CSF1 is a homodimeric glycoprotein and in the BM, it is produced

by BM stromal cells such as fibroblasts, mesenchymal cells and osteoblasts. During pregnancy,

CSF1 is involved in mammary gland and placental development, with serum and tissue

Page 67: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

57

concentrations rising by 2-fold and levels in the uterus rising by 1000-fold (Cecchini et al.,

1994). CSF1 has pleiotropic effects as CSF1-deficient op/op mice, which have a natural

inactivating mutation in the CSF1 protein, exhibit osteopetrosis, severe growth retardation, low

fertility and decreased numbers of tissue macrophages (Begg et al., 1993; Wiktor-Jedrzejczak et

al., 1990). CSF1 acts on CSF1R, which is the only known receptor that it binds to.

1.7.1. CSF1 Gene Structure and Isoforms

CSF1 is encoded by the human CSF1 gene located on chromosome 1 (1p13.3) and

consists of 9 exons spanning approximately 21 kb. Four different human CSF1 cDNAs have

been identified with lengths of, 4.0, 2.5, 2.3 and 1.6 kb. The 4.0- and the 2.3-kb cDNAs are full-

length cDNA clones that differ in the 3’-untranslated region due to alternative use of two exons

(9 and 10). The 2.5- and 1.6 kb species are truncated due to alternative splicing in the 5’ end of

exon 6, which leads to large segments being spliced out of the full length species (Cerretti et al.,

1988; Ladner et al., 1987) (Figure 1.6).

The 4.0 kb full length mRNA species gives rise to a putative 554 amino acid, 86-150 kDa

homodimeric transmembrane-spanning precursor protein termed variant 1 (V1) or protein

isoform a (Price et al., 1992). It consists of a 32 amino acid N-terminus leader sequence, an

extracellular region with a 149 amino acid receptor binding domain, a transmembrane domain

with 24 amino acids (amino acids 464-488) and a short 37 amino acid cytoplasmic tail (Douglass

et al., 2008; E. Richard Stanley et al., 1997). Proteolytic cleavage occurs at several sites in the

extracellular domain, including those at amino acids 251, 298, and 423, to release a soluble

protein that may be secreted from the cell (Stanley et al., 1997). The full length protein is co-

translationally glycosylated in the endoplasmic reticulum, after which N-linked and O-linked

Page 68: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

58

sugars are added to it in the Golgi apparatus. In addition to N-glycosylation and O-glycosylation

sites, the soluble/secreted isoform also has a site for the addition of one glycosaminoglycan

carbohydrate chain, to which a chondroitin sulfate molecule can be added. Addition of the

glycosaminoglycan sugar leads it to being secreted as a proteoglycan, whereas a lack of it results

in it being a glycoprotein. Both forms are secreted and accumulate rapidly with a doubling time

of approximately 40 minutes (E. Richard Stanley et al., 1997).

The secreted glycoprotein form is homodimeric and is 85 kDa, whereas the proteoglycan

can be up to 150 kDa, due to the addition of the chondroitin sulfate (Fixe et al., 1998). Mature

CSF1 protein can be as small as 150 amino acids and still function, with the first seven cysteines

being necessary for its function (Kawasaki et al., 1990). Mouse CSF1 is a 520 amino acid

membrane-spanning precursor that has significant homology to human CSF1. Despite the

homology, murine CSF1 is unable to induce activation of the human CSF1 receptor (CSF1R)

(Rettenmier at al., 1988; Price at al., 1992).

The 1.6 kb variant that results from alternate splicing of the full length mRNA species

leads to the deletion of a 298 amino acid region from the extracellular domain, which contains

proteolytic cleavage site(s) at which the biologically active, mature protein is cleaved

(Rettenmier and Roussel, 1988) (Figure 1.7). This alternatively spliced form produces a shorter

256 amino acid protein product termed variant 3 (V3) or protein isoform c that is membrane-

bound (Rettenmier et al., 1987). Expression of this species in NIH3T3 cells leads to a cell

surface-bound, membrane-spanning dimeric CSF1 protein that is 68 kDa in size. This precursor

is expressed as a relatively stable membrane-bound glycoprotein as it is not cleaved in secretory

transport vesicles. As such, it is stably expressed at the cell surface, although it can be slowly

Page 69: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

59

released as a smaller 44 kDa homodimer through proteolysis (Rettenmier et al., 1987).

Membrane-bound CSF1 is exclusively N-glycosylated as it lacks sites for O-glycosylation.

The soluble/secreted glycoprotein can only act humorally as it is released into the

circulation, whereas the soluble/secreted proteoglycan can act both humorally and locally; the

latter due to the glycosaminoglycan chains that allow it to be sequestered within the bone

marrow stroma (Stanley et al., 1997). CSF1 is found as a proteoglycan in the bloodstream at

biologically active concentrations of 10 ng/mL (Hume & MacDonald, 2012).

Taken together, there are 3 major versions of CSF1 protein that can be produced (with

two forms of the soluble/secreted version): a secreted/soluble proteoglycan, a secreted/soluble

glycoprotein and a cell surface membrane-bound glycoprotein. Homodimeric CSF1 binds to the

D1 - D3 domains of the extracellular portion of CSF1R (Chen, Liu, Focia, Shim, & He, 2008;

Felix et al., 2015; E. R. Stanley et al., 1983). The N-terminal region of CSF1 consists of an

active 149-amino acid fragment that forms a 4 α-helix bundle; for the homodimer, two bundles

of the 4 α-helices are laid end-to-end, linked by a disulfide bond (Pandit et al., 1992).

Little is known about how CSF1 expression is transcriptionally regulated but it has been

reported that angiotensin II may stimulate CSF1 transcription in vascular endothelial cells

through the action of Brahma related gene 1 (BRG1), which is a core component of the

SWI/SNF chromatin remodeling complex (Shao et al., 2019).

Page 70: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

60

Figure 1.6. Alternative splicing of CSF1 leads to different isoforms.

Alternative splicing in exon 6 of full length genomic CSF1 DNA yields the shorter membrane-

bound isoform of CSF1, termed V3 or isoform c.

Page 71: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

61

Figure 1.7. Model of the two major variants of monomeric CSF1.

The full length CSF1 cDNA precursor (4.0 kb) is processed to yield a 1.6 kb cDNA that gives

rise to a shorter, membrane-bound isoform (bottom schematic). While the full length precursor

contains both N- and O-linked glycosylation sites, along with a glycosaminoglycan site to which

chondroitin sulfate can be added, the shorter membrane-bound isoform only contains sites for O-

glycosylation.

Page 72: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

62

1.7.2. CSF1 in Pathology

Circulating levels of CSF1 increase in various pathologies including infections, chronic

inflammatory disease and cancer. In inflammation, CSF1 may be produced by macrophages

themselves, although in the mouse this does not appear to be the case as macrophages actually

fail to survive if there is no CSF1 present. CSF1 levels are regulated by tissue macrophages

through receptor-mediated endocytosis of its receptor, CSF1R, leading to intracellular uptake

and destruction in lysosomes (Hume & MacDonald, 2012). CSF1 is found to be highly expressed

in breast, ovarian, lung, prostate and renal cancers, and high circulating levels are found in

melanoma. The malignant cells in melanoma and breast cancer have been shown to produce

increased amounts of CSF1, which likely impacts disease behaviour.

Interestingly, compared to other hematopoietic myeloid growth factors such as G-CSF

and GM-CSF, CSF1 administration has not demonstrated efficacy in bone marrow

transplantation or in recovery from myelosuppressive chemotherapy (Dale, 2006; Heuser,

Ganser, & Bokemeyer, 2007). However, its potential therapeutic use is being investigated in

conjunction with other factors, particularly with GM-CSF, as both have been shown to generate

large macrophage colonies in-vivo. Moreover, a role for CSF1-stimulated macrophages is also

being explored in the treatment of fungal infections. In phase I clinical trials, soluble CSF1 has

demonstrated an ability to increase monocyte numbers (Bukowski et al., 1994). Further to this,

CD8+ T cells have been shown to induce increased production of CSF1 by melanoma cells,

which can be blocked with anti-CSF1R neutralizing antibodies, in conjunction with blockade of

programmed cell death protein 1 (PD-1) (Neubert et al., 2018).

Page 73: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

63

1.8. INTERLEUKIN-34 (IL-34)

Interleukin-34 (IL-34) is a relatively newly identified cytokine that was discovered in

2008 in a functional screen of the extracellular proteome; it was found to be a second ligand for

CSF1R (H. Lin et al., 2008)(Wei et al., 2010). Interestingly, such alternative binding by a second

ligand has not been found for other structurally similar receptors, namely c-Kit or Flt3 (Chihara

et al., 2010). While IL-34 has no structural similarity to CSF1, it has been found to function

similarly to it, in terms of promoting the differentiation, growth and survival of monocytes,

macrophages and osteoclasts, as well as the development of microglia (Baud’Huin et al.,

2010; Guillonneau, Bézie, & Anegon, 2017). However, unlike CSF1 which can only bind to

CSF1R, IL-34 was discovered to bind to two other receptors: the receptor type protein-tyrosine

phosphatase zeta (PTP-ζ) and CD138 (Syndecan 1 or SDC1) (Nandi et al., 2013; Segaliny et

al., 2015). The existence of an alternate CSF1R ligand was suggested by the observation that in

comparison to CSF1-deficient CSF1op/op mice that have reduced macrophage numbers,

osteopetrosis and other abnormalities in bone formation, CSF1R-/- mice exhibit more severe

adverse effects. This suggested that in the absence of CSF1, there may be another ligand that acts

on CSF1R to prevent the severe osteogenic phenotypes that occur when CSF1R is lost (Wei et

al., 2010). CSF1 and IL-34 have both been shown to be expressed in human and mouse brain,

but in distinct spatial regions. While CSF1 has been found to be highly expressed in the

cerebellum to mediate cerebellar microglia development, IL-34 is expressed in forebrain areas

including the cerebral cortex and the CA3 region of the hippocampus (Kana et al., 2019).

Moreover, the cells producing the ligands also differed, with glial cells producing CSF1, and

neurons producing IL-34. CSF1R was expressed on microglia throughout the brain. This

Page 74: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

64

highlights that the two CSF1R binding proteins are not redundant, but have functionally

distinctive roles in the brain.

1.9. SOLUBLE VS. TRANSMEMBRANE LIGANDS

Evidence suggests that the soluble and membrane-bound forms of growth factor ligands

differ in their functional activity. For example, it has been shown that the soluble and membrane-

bound forms of Kit ligand signal through distinct phosphotyrosine residues in the c-Kit receptor,

with the Y728 residue being required for mitogenic stimulation and leukemic growth induced by

the membrane-bound, but not soluble, ligand (Jennifer L Gommerman, Sittaro, Klebasz,

Williams, & Berger, 2000). Similarly, soluble recombinant CSF1 has been shown to fail to

promote hematopoietic and leukemic cell survival, and may actually induce cell differentiation

and cell death, whereas the membrane-bound form of the ligand supports long-term growth of

the cells (Tsuboi, Revol, Blanchet, & Mouchiroud, 2000b). This suggests that transmembrane

ligands may have distinctive functional properties, most likely involving mediation of cell

adhesion to foster enhanced cell-cell contact and bidirectional intracellular signaling.

1.10 PDZ DOMAINS AND PDZ DOMAIN BINDING MOTIFS

PDZ (Postsynaptic density 95, PSD-85; Discs large, Dlg; Zonula occludens-1, ZO-1)

domains mediate protein-protein interactions to serve as central organizers of multi-dynamic

signaling complexes and represent the most common protein-protein interaction domain (Lee &

Zheng, 2010). PDZ domains are typically 80-90 amino acids in length and found in

approximately 180 different human proteins, with multiple copies present in many proteins.

Although there are hundreds of unique PDZ domain sequences, they all contain a highly

Page 75: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

65

conserved glycine-leucine-glycine-phenylalanine sequence that gives rise to the domain’s folded,

globular, cup-like structure. This structure has the ability to recognize and bind to short, finger-

like hydrophobic peptides (Harris & Lim, 2001). The structural modules are comprised of 5-6

beta strands and 2-3 alpha helices (Doyle et al., 1996; Lee & Zheng, 2010).

PDZ domain recognition sequences

Because of their modular structure, PDZ domains usually bind to specific C-terminal sequence

motifs on protein ligands. These sequence motifs are known as PDZ domain binding motifs

(PDBM) which are typically 4-6 amino acids in length. The C-terminus residue is designated as

the P0 residue while subsequent residues towards the N-terminus are referred to as P−1, P−2, P−3,

etc. (Harris & Lim, 2001). It is suggested that the P0 and P−2 residues are the most important for

PDZ domain binding recognition (Songyang et al., 1997). In fact, studies have shown that PDZ

domains can be divided into three main classes based on their binding preferences for residues at

the P0 and P−2 sites: Class I PDZ domains recognize the motif S/T-X-Φ-COOH (where Φ is a

hydrophobic amino acid at the P0 site and X is any amino acid); class II PDZ domains recognize

the motif Φ-X-Φ-COOH; and class III PDZ domains recognize the motif X-X-C-COOH (Table

1-6) (Costa et al., 2018). There are a few other PDZ domains that do not fall in any of the three

classes. PDZ domains recognize these motifs usually only if they are located at the C-terminus of

the protein ligand. This is because the end of the peptide-binding groove of PDZ domains have a

specific carboxylate-binding loop which binds the peptide ligand terminal carboxylate to position

it in the binding groove. This specific positioning results in the side chains of the P0 and P−2

ligand residues to protrude into the base of the peptide-binding groove, with the P0 residue

pointing into a large hydrophobic pocket (Costa et al., 2018). This specific docking position

Page 76: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

66

helps explain why the P0 residue is hydrophobic. In PSD-95 (postsynaptic density protein 95),

which has a preference for valine at P0 of its binding ligand, the pocket is formed by Phe325,

Leu379 and other hydrophobic residues, while a slightly larger hydrophobic pocket in the first

PDZ domain from NHERF/EBP50 causes a preference for leucine at the P0 position (Doyle et

al., 1996; Karthikeyan, Leung, Birrane, Webster, & Ladias, 2001). The side chain of the P−2

ligand residue points into a separate pocket, which in class I PDZ domains, contains a histidine

residue that hydrogen bonds with the side chains of a serine or threonine residue. Class II

domains recognize a hydrophobic residue at position P−2, typically leucine or methionine

(Daniels, Cohen, Anderson, & Brünger, 1998).

The functional importance of the PDZ binding domains of several transmembrane

proteins have been characterized. Work by Tony Pawson’s group has shown that the PDZ

domain binding motif of the ephrin B1 ligand interacts with the PDZ domain of proteins such as

syntenin (involved in cytoskeletal organization and protein trafficking) and the tyrosine

phosphatase FAP-1 (Holland et al., 1996; D. Lin, Gish, Songyang, & Pawson, 1999). Similarly,

the C-terminal PDZ binding domain of transmembrane transforming growth factor α (TGF-α)

also interacts with syntenin, as well as the Golgi protein p59/GRASP55 in the Golgi apparatus,

both of which mediate targeting of the ligand to the cell membrane (Fernández-Larrea, Merlos-

Suárez, Ureña, Baselga, & Arribas, 1999; Kuo, 2002). Interestingly, p59 also interacts with the

transmembrane form of kit ligand (Flanagan, Chan, & Leder, 1991).

Page 77: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

67

Table 1-6. Classification of PDZ domain binding recognition sequences

PDZ domain binding motifs are divided into three main classes based on the specific residue

binding preference of interacting PDZ domains. Adapted from Lim et al., 2001.

Page 78: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

68

1.11. HTRA SERINE PROTEASES

The mammalian high temperature requirement A (HtrA) is a family of trypsin-like serine

proteases, originally identified in bacteria for their heat shock activity (Clausen, Kaiser, Huber,

& Ehrmann, 2011; Krojer, Garrido-Franco, Huber, Ehrmann, & Clausen, 2002; Lipinska, Zylicz,

& Georgopoulos, 1990). HtrA proteases also exhibit molecular chaperone and serine protease

activity, the latter of which fosters degradation of misfolded proteins (Clausen, Southan, &

Ehrmann, 2002a; Krojer et al., 2002). The human HtrA family consists of at least four members,

HtrA 1 - 4, of which HtrA2 is largely localized to the mitochondria where it regulates apoptosis

(Clausen et al., 2011; Hegde et al., 2002; Suzuki et al., 2001). The proteases contain a highly

conserved trypsin-like serine protease domain and at least one PDZ domain (Clausen et al.,

2002a; MURWANTOKO et al., 2004). HtrA1, 3 and 4 also contain an insulin-like growth factor-

binding domain as well as a Kazal-type serine protease inhibitor domain and signal peptide in the

N-terminal domain. HtrA1 is largely a secreted protease, but it can also be found in the cytosol

as a soluble peptide (Clawson, Bui, Xin, Wang, & Pan, 2008). HtrA1 is seen to play roles in cell-

cell adhesion as it degrades components of the extracellular matrix such as type III collagen and

fibronectin, and can also cleave the epithelial cell surface adhesion protein E-cadherin. HtrA1 is

expressed in the BM where it is involved in osteogenesis, bone remodeling and musculoskeletal

development (Tiaden et al., 2012a). In cancers, HtrA1 is down-regulated in ovarian cancer and

melanoma, suggesting a role for it in carcinogeneis through potential aberrant remodeling of the

tumor microenvironment (Baldi et al., 2002; Chien et al., 2004).

Structurally, HtrA1 exists as a trimer where the PDZ domains hinder access of substrates

to the inner catalytic core to mediate auto-inhibition of the protein complex. Binding of

substrates to the PDZ domain opens up the trimeric structure to expose the catalytic core, thereby

Page 79: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

69

relieving the auto-inhibition (Figure 1.8). Recent evidence has demonstrated a role for

cytoplasmic HtrA1 in regulating intracellular signaling as it has been shown to cleave

tuberin/tuberous sclerosis 2 (TSC2), a negative regulator of the mammalian target of rapamcyin

(mTOR), accompanied by activation of the mTOR targets 4E-BP1 and S6K (Campioni et al.,

2010). The identification of TSC2 as a substrate of HtrA1 proteolytic activity makes the mTOR

pathway a relevant signaling target of HtrA1.

Page 80: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

70

Figure 1.8. Structure and function of HtrA serine proteases.

HtrA1 exists as a trimer where the PDZ domains of each unit are auto-inhibitory as they block

access to the inner catalytic core. Binding of substrates to the PDZ domains opens up the

conformation of the trimeric structure to expose the active site.

Page 81: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

71

1.12. EXPERIMENTAL MODELING

Modelling human disease in experimental in-vitro and in-vivo models often presents

biological and technical challenges. Mouse model systems are limited in their recapitulation of

the human disease owing to genetic differences between organisms. In-vivo xenograft mouse

models rely on the successful engraftment of human cells in mice, which is a challenge as only

40-50 % of primary AML samples engraft in conventional immune deficient mouse models such

as the NOD/SCID mouse and its newer and improved prototypes NOD/SCIDγ (or NSG), with

large variations in actual degree of engraftment, ranging from as low as 0.1 – 50 % (Sanchez et

al., 2009). Given this, there is growing interest in the development of humanized mouse models

wherein human cells, tissues, organs and/or genes are introduced into the mouse to support the

transplantation of human cells. Mice expressing human growth factors and cytokines have been

shown to model and recapitulate a more human-based genetic and molecular background and/or

tissue microenvironment to improve engraftment and disease modeling. For example, expression

of human cytokines such as SCF, GM-CSF, M-CSF (CSF-1) and IL-3 in mouse models show

improved engraftment of human cells (Ito et al., 2013; Rathinam et al., 2011a; Walsh et al.,

2017). A humanized CSF1 knock-in mouse, where human CSF1 was inserted into the

corresponding mouse locus of Balb/c Rag2-/- γc-/- mice, shows improved frequency and function

of myeloid cells such as increased monocyte/macrophage activation and migration (Rathinam et

al., 2011b).

Page 82: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

72

1.13. THESIS FOCUS

My work in this thesis centered on the study of the CSF1-CSF1R ligand-receptor pair

because 1) the significance of CSF1R expression levels are largely unknown in AML and 2)

CSF1 is a hematopoietic ligand that is constitutively expressed in the BM stroma. CSF1-CSF1R

signaling was assessed between CSF1R-expressing AML cells and BM stromal cells engineered

to express different isoforms of the human CSF1 ligand (Figure 1.9). Moreover, potential

differences in the function of soluble vs. membrane-bound/transmembrane CSF1 were also

assessed with respect to the adhesion and long-term support of AML cells on stroma in co-

culture assays. While the binding of CSF1 to CSF1R stimulates well-described growth and

proliferative intracellular signal transduction pathways downstream of the receptor, the impact of

CSF1R binding to CSF1 ligand in stromal cells is less understood. This is of particular relevance,

as many transmembrane ligands contain signaling domains which have demonstrated potential to

reverse signal within cells. Given this, the potential phenotypic and signaling changes induced by

CSF1-CSF1R were investigated in both interacting AML, and BM stromal cells. This would

allow for a better understanding of the role of this ligand-receptor pair on leukemogenesis within

the BM microenvironment.

Page 83: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

73

Figure 1.9. Modeling an AML-bone marrow microenvironment.

Investigation of CSF1-CSF1R signaling between interacting AML and BM stromal cells

expressing the receptor and ligand, respectively.

Page 84: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

74

1.14. THESIS RATIONALE, HYPOTHESES AND OBJECTIVES

RATIONALE

Given that the CSF1-CSF1R interaction is important in regulating normal myelopoiesis, and that

CSF1R may be found to be expressed in subsets of AML patients, it is of importance and

relevance to investigate the function of this ligand-receptor interaction in the myeloid

malignancy of AML. In this project, the prognostic significance of CSF1R gene and protein

expression levels was evaluated in a cohort of AML patients, and the CSF1-CSF1R ligand-

receptor interaction was functionally interrogated in in-vitro co-culture, as well as in-vivo models

of AML.

HYPOTHESES

1) CSF1R levels are associated with clinical patient survival

2) The CSF1-CSF1R ligand-receptor pair helps mediate interactions between AML and stromal

cells to elicit phenotypic (enhanced growth, survival, adhesion) and signaling changes in

AML cells.

3) Transmembrane CSF1 can engage in bi-directional or reverse intracellular signaling in

stromal cells: CSF1 can signal within stromal cells to alter stromal cell signaling, which has

implications for its support of AML cells. AML cells can also impact the behaviour and

phenotype of stromal cells via the CSF1-CSF1R interaction between the cells.

Page 85: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

75

OBJECTIVES

1) Evaluate CSF1R expression levels in AML patient samples and examine the association

between expression levels and clinical patient outcome.

a. Interrogate CSF1R gene expression levels in publicly available gene expression

datasets and correlate it with clinical data, including patient survival (when treated

with curative intent).

b. Examine CSF1R cell surface protein expression levels on blasts of AML patient

samples using flow cytometry; use clinical patient information to evaluate

potential associations of CSF1R levels and patient survival.

2) Investigate the functional role of CSF1-CSF1R in AML-stromal cell interactions

a. Overexpress human CSF1 ligand (hCSF1) in MS-5 stromal cells: compare effects

of soluble vs. membrane-bound/transmembrane isoforms

b. Perform co-culture experiments using hCSF1-expressing MS-5 stromal cells and

CSF1R-expressing AML cells

c. Evaluate the growth, proliferation, survival, signaling, cytokine and gene

expression changes in AML cells upon exposure to CSF1 in co-cultures.

d. Utilize a humanized CSF1 mouse model to examine whether it provides improved

engraftment of primary human AML cells as well as AML cells lines.

3) Examine signaling potential of CSF1 ligand: reverse intracellular signaling within BM

stromal cells

a. In MS-5 stromal cells, delete the last five residues, which constitute a putative

PDZ domain binding motif (PDBM), at the C-terminal end of transmembrane

CSF1. Evaluate signaling and phenotypic changes occurring as a result. Examine

the role of HtrA1 as a potential signaling mediator of CSF1.

Page 86: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

76

CHAPTER 2

CSF1R is associated with poor clinical outcome and promotes a

leukemic cell phenotype in AML-stromal cell interactions

This chapter is in preparation for a manuscript to be submitted for publication

Page 87: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

77

2.1. ABSTRACT

The colony stimulating factor 1 receptor (CSF1R) is important in the regulation of normal

myelopoiesis, but its status and function remains largely unknown in Acute Myeloid Leukemia

(AML). In this work, the clinical significance of CSF1R was investigated, along with its

functionality in AML- bone marrow (BM) stromal cell interactions. It was found that a subset of

AML patient samples express high levels of CSF1R (CSF1Rhigh) and that patients with

CSF1Rhigh AML cells have shorter overall survival (OS) compared to patients with low CSF1R

(CSF1Rlow) expression. In in-vitro co-cultures, it was demonstrated that MS-5 stromal cells

expressing either the soluble (sol) or transmembrane (mem) variants of human CSF1 (hCSF1),

enhanced the long-term growth and survival of CSF1Rhigh AML cells (patient samples and

OCI/AML-4 and OCI/AML-5 cell lines); this growth advantage was not observed for CSF1Rlow

AML cells. MS-5 cells expressing the transmembrane variant of CSF1 provided greater

supportive effects compared to the soluble variant. Co-culture of CSF1Rhigh AML cells with MS-

5 hCSF1-mem cells led to increased cell surface expression of the hematopoietic stem/progenitor

cell markers CD34 and c-Kit, along with increased phosphorylation of the mTOR target S6 in

CSF1Rhigh AML cells. It also led to distinct changes in the cytokine profile of AML. Application

of a CSF1R antibody mitigated much of these effects, with decreased cell proliferation and

inhibition of mTOR signaling in AML cells. In humanized CSF1 mice, CSF1Rhigh AML cells

exhibited significantly improved engraftment compared to NOD-SCID mice. These results

suggest that CSF1-CSF1R signaling has important clinical prognostic value and biological

function to support the growth and maintenance of AML cells in AML-stromal cell interactions.

This makes CSF1-CSF1R an attractive therapeutic target within the BM microenvironment of

AML.

Page 88: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

78

2.2. INTRODUCTION

Acute Myeloid Leukemia (AML) is a highly aggressive and fatal hematological

malignancy characterized by significant heterogeneity and growth-factor dependency. Curative

intent treatment involves induction therapy, often using the ‘3+7’ regimen of

doxorubicin/daunorubicin and Ara-C (Murphy & Yee, 2017). While most patients achieve a

complete remission with such a regimen, despite further consolidation therapy, more than half of

all patients relapse with high mortality rates thereafter (Betz & Hess, 2010b; Liersch et al.,

2014). AML arises due to a block in differentiation of myeloid cell precursors known as

myeloblasts (or blasts), occurring largely as a result of molecular and cytogenetic aberrations.

The clonal expansion and accumulation of these abnormal undifferentiated precursors interferes

with normal hematopoiesis, leading to a bone marrow (BM) failure state, characterized by

anemia, thrombocytopenia and neutropenia in most patients.

The BM microenvironment is a complex niche of interconnected cells and factors that

provides supportive conditions not only for normal hematopoiesis, but also leukemic cells.

Colony stimulating factor 1 (CSF1) is a cytokine produced by BM stromal cells such as

fibroblasts, endothelial and mesenchymal cells (Hamilton & Achuthan, 2013). CSF1 exists in

several isoforms including a secreted proteoglycan or glycoprotein, and a cell surface

transmembrane glycoprotein, with the latter being the predominant form in the BM. (Horiuchi et

al., 2007; C W Rettenmier & Roussel, 1988). CSF1 binds to the CSF1 receptor (CSF1R, c-FMS,

CD115), which is a membrane-spanning class III receptor tyrosine kinase (RTK) expressed on

mononuclear phagocytic myeloid-lineage cells (F Birg, Rosnet, Carbuccia, & Birnbaum, 1994;

Rosnet & Birnbaum, 1993; C J Sherr & Rettenmier, 1986). Binding of CSF1 ligand to CSF1R

induces the differentiation and growth of myeloid cell precursors into mature monocytes and

Page 89: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

79

macrophages (Rohrschneider et al., 1997). Activation of the receptor through binding CSF1

leads to receptor oligomerization and autophosphorylation of tyrosine residues in the

cytoplasmic domain of CSF1R. Subsequently, this results in the activation of intracellular growth

and survival pathways including the PI3K/Akt, MAPK/Erk and Jak/STAT pathways (Huynh,

Kwa, Cook, Hamilton, & Scholz, 2012; Pixley & Stanley, 2004; Reedijk et al., 2018).

In cancers, CSF1R is found to be highly expressed in ovarian cancer, glioma, giant

tenosynovial giant-cell tumor, renal and pancreatic cancers, with expression being localized to

tumor-associated macrophages (TAM) in solid tumors (De et al., 2016; Swierczak et al., 2014).

CSF1R is also associated with poor clinical outcomes in renal and pancreatic cancers. In AML,

the clinical prognostic significance of CSF1R remains unknown. Moreover, while the CSF1-

CSF1R ligand-receptor pair is well established to instruct macrophage development in normal

myelopoeisis, its role and function in myeloid malignancy remains rudimentary.

In this study, we found that CSF1R is highly expressed by leukemic cells in subsets of

AML patients, with high expression (both cell surface protein levels, as well as transcript levels)

being associated with poor overall and event-free clinical survival. Furthermore, CSF1R

demonstrated significance in the subgroup stratification of FLT3 and NPM1 normal AML

patients. In the interrogation of CSF1-CSF1R signaling between AML and stromal cells in in-

vitro co-culture assays, it was found that MS-5 stromal cells expressing the transmembrane form

of human CSF1 (hCSF1-mem) provided the best support for the long-term growth and

maintenance of CSF1R-expressing AML cells compared to MS-5 control cells, and MS-5 cells

expressing the soluble hCSF1 variant. Co-culture of CSF1R-expressing AML cells with hCSF1-

expressing MS-5 cells led to distinct changes in intracellular signaling, namely activation of

mTOR targets, in addition to changes in cytokine levels in AML cells. Furthermore, increased

Page 90: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

80

cell surface expression of the hematopoietic stem cell-associated markers CD34 and c-Kit were

observed in AML cells cultured with hCSF1-expressing MS-5 cells, as well as AML cells grown

in humanized CSF1 mice. Application of a CSF1R antibody abrogated these effects, resulting in

inhibition of AML cell proliferation, and associated signaling and cytokine changes. These

findings demonstrate CSF1R to be a potentially substantive marker in predicting AML clinical

outcome, and highlights an important role for CSF1-CSF1R as a signaling mediator between

leukemic and stromal cells that may be therapeutically targeted within the AML

microenvironment.

Page 91: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

81

2.3. MATERIALS AND METHODS

AML Dataset Analyses

Normalized microarray data was downloaded from GEO (http://www.ncbi.nlm.nih.gov/geo/) and

TCGA (http://cancergenome.nih.gov/) databases. Cross-dataset frequency distribution of CSF1R

expression was visualized by transforming and mode centering CSF1R expression data in R

(http://www.r-project.org). Clinical data analysis was performed on non-transformed data.

Clinical Definitions

Complete remission (CR) was defined as the presence of normal erythropoiesis, granulopoiesis

and megakaryopoiesis, PB absolute neutrophil count ≥1 × 109 /L and platelet count ≥100 × 109

/L, and <5% leukemic blast cells in BM. Relapse was defined as recurrence of >5% blasts in the

BM. Overall survival (OS) was defined as time from diagnosis to death. Event free survival

(EFS) was defined as time from disease diagnosis until relapse or death. OS and EFS were not

calculated for patients with <28 day survival. Patients lost to follow-up were censored at the date

of last follow-up. Risk stratification of patients into good, intermediate and adverse was done in

accordance to NCCN guidelines (O’Donnell M et al., 2017).

Survival Analysis

Analysis of clinical endpoints and response to therapy in the gene expression datasets (Valk,

Metzler) was done exclusively for patients who underwent induction therapy. The univariate

association between CSF1R expression and other clinical features was assessed using the

Fisher’s exact or Wilcoxon rank-sum tests, as appropriate. Kaplan-Meier survival analysis was

used to estimate unadjusted survival curves and the log-rank test was used to compare survival

Page 92: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

82

rates. Cox proportional hazards models were used to estimate the adjusted survival curves,

hazard ratios (HRs) and 95% confidence intervals (CIs) associated with CSF1R expression.

Flow Analysis on AML Patient Samples

Flow cytometry was performed on 146 AML patient samples obtained from the leukemia tissue

bank at the Princess Margaret Cancer Centre (PMCC) at the University Health Network (UHN)

in Toronto, Ontario, Canada. The patients had all received standard curative intent clinical

treatment for AML. The flow cytometry data was analyzed using FlowJo V10 software. AML

blasts were identified based on a CD45dim/SSClow gating parameter. CSF1R expression was

evaluated on the gated AML blast population and the percentage of positive cells was recorded.

DNA Plasmids and Gene Transduction

Human CSF1 cDNAs were purchased from Origene and cloned into a retroviral pBabe-GFP

vector. Empty vector (EV) pBabe-GFP, pBabe-GFP-hCSF1-sol and pBabe-GFP-hCSF1-mem

retroviruses were produced by calcium phosphate transfection of 293-T cells with psPAX2 and

VSVG as described elsewhere (Life Technologies). Virus rich medium was collected 48 and 72

hours post-transfection and concentrated with Lenti-X Concentrator (Clontech). MS-5 cells were

infected with virus rich medium for 24 hours with 8 μg/mL protamine sulfate. Infected cells were

selected by fluorescence activated cell sorting (FACS) for GFP fluorescence.

Cell Culture

AML cell lines were cultured in α-MEM complete medium with 10% fetal bovine serum (FBS),

100 units/mL penicillin and 100 μg/mL streptomycin at 37°C in 5% CO2 . The OCI/AML-4 and

Page 93: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

83

OCI/AML-5 cell lines were cultured with the addition of 10% conditioned medium (CM) from

5637 bladder carcinoma cells, which they require for their growth in cell culture (Meyer &

Drexler, 1999). MS-5 stromal cells were cultured in α-MEM complete medium. 293-T cells used

for lentivirus production were cultured in DMEM complete medium.

Co-culture Assays

For co-culture assays, a stromal cell feeder layer was first established by seeding 1.0 -1.5 x 105

MS-5 cells in 6-well plates and allowing them to adhere overnight. The following day, the MS-5

stromal cells were irradiated to induce growth arrest of the cells. Following this, culture medium

was removed and 1.5 x 105 AML cells (AML cell lines) were added to the MS-5 cells with

addition of fresh medium (total volume of 3 mL). The OCI/AML-4 and OCI/AML-5 cell lines

were grown in the absence of 5637-CM for the co-culture experiments. For AML patient

samples, 1X106 cells were seeded. Co-cultures were maintained by demi-depopulation of AML

cells every 2 - 3 days as described previously. Briefly, half the culture medium was removed by

gently washing medium over the stromal cells to detach any loosely attached AML cells. The

same volume of fresh medium was then added to the cultures. Cultures were maintained for

several days to several weeks, depending on the experimental assay to be conducted.

Cell Growth Curves

AML growth curves were performed by seeding 5 x 104 - 1 x 105 cells in 6-well plates. For drug

assays, the compound was added at time of seeding. Both suspension and adherent AML cells in

co-culture were retrieved for counting. Adherent cells were obtained by gently washing them off

the stromal layer. Cells were stained with trypan blue and counted using a haemocytometer. For

Page 94: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

84

final endpoint counts of AML cells in co-culture, the cultures (including stromal cells with

adherent AML cells) were trypsinized and subjected to flow cytometry analysis where ratios of

GFP-positive MS-5 stromal cells to AML cells were obtained for calculating the counts of each.

Drug Treatment

CSF1R monoclonal antibody (Amgen) was used to treat AML and MS-5 cell co-cultures. Co-

cultures assays were set up as described above. The CSF1R antibody was added to co-cultures at

the time of AML cell seeding. Cell viability was assessed daily with trypan blue or

AlamarBlue® (Life Technologies) as described.

Quantitative RT-PCR

RNA was extracted using RNeasy Mini kit (Qiagen) and first strand cDNA synthesis was

performed using the M-MLV systems (Life Technologies). qPCR reactions were conducted

using the SYBR Green PCR Master Mix kit (Applied Biosystems) on a 7900 HT Real-Time

PCR system with SDS v2.3 software (Applied Biosystems) using standard settings: 95°C (10

min) and 40 cycles of 95°C (20 sec) and 60°C (1 min). mRNA levels were normalized to

GAPDH as a housekeeping gene. Primers used: CSF1R 5’-TGGCCACAGCTTGGCATGGT- 3’

(sense) and 5’-TGCCCACACATCGCAAGGTCA-3’ (anti-sense); sol hCSF1 5’-

CAGCAAAGGGCCAACAGC-3’ (sense) and 5’-GGTCCTCCTTCTGGCTCTG- 3’ (anti-

sense); mem hCSF1 5’-ACAAGGCCTGCGTCCGAACT-3’ (sense) and 5’-

CTCTCATGGCCTTGGCTGGAGCAT-3’ (anti-sense); GAPDH 5’-

AACAGCGACACCCACTCCTC- 3’ (sense) and 5’-CATACCAGGAAATGAGCTTGACAA-

3’ (anti-sense).

Page 95: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

85

Immunoblotting

Protein was extracted by lysing cells in 50 - 100 μl of RIPA lysis buffer (500 mM Tris, pH 7.4,

150 mM NaCl, 0.5% Na deoxycholate, 1% NP-40, 0.1% SDS and complete protease inhibitor

preparation containing leupeptin and aprotinin), incubated on ice for 30 min and centrifuged at

14,000 X g for 15 mins. Cell lysate supernatant was collected and protein levels were quantitated

using the Bradford assay. SDS loading buffer (2% SDS, 10% glycerol, 360 mM 2-

mercaptoethanol, 100 mM Tris) was added to cell lysates and boiled for 5 min. 40 - 100 μg of

total protein was separated using SDS-polyacrylamide gel electrophoresis (PAGE) and

transferred onto PVDF membranes. Membranes were blocked with 5% milk in TBS-tween

(0.1%) wash buffer, followed by incubation with primary antibodies overnight at 4°C. The next

day, membranes were washed and incubated with HRP-conjugated secondary antibodies for 1

hour at room temperature with gentle agitation. Proteins were detected using ECL detection

reagent (GE Healthcare, Amersham, UK). Primary antibodies used: phospho-Akt (Ser 473;

rabbit), Akt rabbit, phospho S6 (Ser 235/236; rabbit), S6 rabbit, phospho CSF1R rabbit and

GAPDH rabbit from Cell Signaling Technology (CST; Danvers, MA, USA); CSF1 mouse was

purchased from Santa Cruz Biotechnology (SCB; Dallas, TX, USA).

Flow Cytometry

Expression of select surface markers on AML cells was evaluated using flow cytometry, before

and after co-culture. A surface marker panel consisting of the following antibodies was set up

and utilized: CSF1R-PE/Cy7, CD34-APC/Cy7, CD38-APC, CD4-FITC, CD11b-BV421 and

CD117-PerCP/Cy5.5 (BioLegend, San Diego, CA, USA) and CD45-PE (Beckman Coulter).

Surface staining was performed by incubating cells with a cocktail of the antibodies in a total

Page 96: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

86

volume of 100 μL wash buffer solution (2% FBS in 1 X PBS) for 30 minutes in the dark at room

temperature. Cells were then washed and re-suspended in 0.5 mL of wash buffer solution and

analyzed on a Beckman Coulter CANTO II flow cytometer (Beckman Coulter).

Cytokine Analysis

The Luminex MAGPIX system (Toronto, Ontario, Canada) as well as the services of Eve

Technologies (Calgary, Alberta, Canada) were used to assess cytokine levels in cell culture

supernatants. Supernatant from co-cultures of AML and MS-5 stromal cells were collected and

spun down (14,000 X g for 15 minutes) to remove debris and other cellular particulates.

Supernatants could then be stored at -80°C for a prolonged period of time or until analysis.

Supernatants were incubated with a cocktail of antibodies conjugated to metal beads (purchased

in the kit) as per the manufacturer’s protocol (Luminex, Toronto, Canada). Briefly, a cocktail of

antibody conjugated magnetic beads were added to a 96-well plate (purchased from Luminex).

The beads were washed twice with wash buffer after which cell culture supernatant was added to

each well, along with standards and controls. The plate was incubated in the dark at 4°C

overnight with gentle agitation. The next day, the beads were washed and detection antibody was

added and incubated for 1 hour with gentle agitation. After several washes, the plate was read on

a MAGPIX machine and data was analyzed using Bio-Plex software (Bio-Rad).

Statistical Analysis

Unless otherwise stated, mean ± S.E.M. values are given and P values were calculated using the

two-tailed unpaired Student’s t-test.

Page 97: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

87

2.4. RESULTS

High CSF1R expression is associated with poor clinical outcome in AML

CSF1R gene expression levels were evaluated in several publically available AML gene

expression datasets including The Cancer Genome Atlas (TCGA), Valk et al. and Metzler et al

(Metzeler et al., 2008; Valk et al., 2004). Valk et al. had identified several prognostically

significant AML subgroups or clusters based on gene expression signatures (Valk et al., 2004).

Using this, CSF1R gene expression levels were evaluated in these subgroups. It was found that

CSF1R was highly expressed in several of the groups, namely groups 1, 5 and 9, with variable

levels of expression across the 285 AML cases overall (Figure 2.1A). Each of the identified

clusters or subgroups were shown to be associated with overall survival (OS). Of the groups with

high CSF1R expression, patients in group 5 had poor OS rates (Figure 2.1B). Group 9 actually

had a relatively high OS due to the presence of inv(16) in this subgroup of patients, which is

associated with favourable clinical outcome when treated with “3+7” and consolidation therapy

(Delaunay et al., 2003).

In the TCGA, Valk and Metzler gene expression datasets, CSF1R gene expression levels

were evaluated and survival analysis was conducted based on the expression levels. AML cases

were dichotomized into ‘low’ and ‘high’ subgroups (CSF1Rlow vs. CSF1Rhigh, respectively)

based on cut-offs established for CSF1R gene expression levels using decision-tree analysis. To

determine whether CSF1R expression status is associated with clinical outcome, Kaplan-Meier

survival analysis was performed to compare overall survival (OS) between the CSF1Rlow vs.

CSF1Rhigh subgroups. Survival analysis revealed that CSF1Rhigh patients have shorter OS

compared to CSF1Rlow patients. The results from the different datasets are as follows: Valk HR =

1.5, P = 0.02; Metzeler: HR = 1.357, P = 0.02; TCGA: HR = 1.490, P = 0.04 (Figure 2.2 A-C).

Page 98: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

88

Figure 2.1. CSF1R gene expression levels are high in certain prognostically significant

subgroups of AML patients.

(A) CSF1R gene expression levels were evaluated in the subgroups/clusters identified in the

Valk et al. gene expression dataset based on molecular signatures and prognostic significance.

(B) Overall survival (OS) probabilities of the identified subgroups (panel B adapted from Valk

et al.)

A

B

Page 99: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

89

Figure 2.2. High CSF1R high levels are associated with shorter OS in AML patients.

Univariate association between CSF1R expression and clinical features was assessed using the

Fisher’s exact or Wilcoxon rank-sum tests and the log-rank test was used to compare survival

rates in Kaplan-Meier survival analysis. Shown are Kaplan-Meier survival plots comparing OS

between CSF1Rlow

and CSF1Rhigh

AML patients in to Valk (A), Metzler (B) and TCGA (C)

gene expression datasets.

A

B

C

Page 100: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

90

CSF1R cell surface protein levels were also evaluated in clinical AML samples. CSF1R

cell surface protein expression was evaluated by flow cytometry for 146 AML patient samples

retrieved from the leukemia tissue bank at the Princess Margaret Cancer Centre (PMCC) at the

University Health Network (UHN) in Toronto, Canada. CSF1R expression was specifically

evaluated on the surface of AML blast cells. In the flow analysis, the AML blast population was

identified and gated based on a side scatter (SSC) ‘low’ and CD45 ‘dim’ gating strategy

(Lacombe et al., 1997). AML blasts are primitive, undifferentiated progenitor cells and are hence

less granular, thereby having low SSC properties. Moreover, blasts also express low levels of the

hematopoietic cell surface marker CD45, the expression of which increases as cells differentiate.

After the blast population was identified, expression levels of CSF1R were evaluated in each of

the AML cases. Figure 2.3A outlines the gating strategy and general workflow of the analysis.

Decision tree analysis was conducted to determine an optimal cut-off for CSF1R protein

expression levels in order to dichotomize patients into clinically relevant CSF1Rhigh vs.

CSF1Rlow subgroups (Figure 2.3B). Analysis of the flow cytometry cell surface protein

expression data in the context of clinical patient information, allowed for the compilation of a

protein-based expression dataset. Kaplan-Meier survival analysis was then conducted to compare

OS in the established CSF1Rhigh and CSF1Rlow subgroups. The survival analysis showed that

CSF1Rhigh patients had shorter OS times compared to CSF1Rlow patients (Figure 2.3C). These

results were consistent with the results of the survival analysis conducted in the gene expression

datasets, indicating that CSF1R RNA transcript levels, and CSF1R cell surface protein

expression levels both identify patients with a worse outcome using standard induction therapy.

Next, in the cell surface protein expression dataset, the AML patients were stratified into

different subgroups based on molecular profiles, namely patients with and without the presence

Page 101: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

91

of FLT3-ITD and NPM1c, which are common mutations found in approximately 25-30% and

30% of all AML cases, respectively. CSF1R expression was evaluated within these groups and it

was observed that there was a significant difference in OS between CSF1Rlow and CSF1Rhigh

patients in the FLT3-ITD negative and NPM1c negative subgroups (Figure 2.3D, E). There was

no significant difference in OS between CSF1Rlow and CSF1Rhigh patients in the counterpart

positive subgroups for both FLT3-ITD and NPM1c. This demonstrates that CSF1R levels may

be prognostically significant in the absence of core AML-associated mutations. CSF1R cell

surface protein expression levels may therefore help stratify groups of patients that may

represent a ‘molecularly normal’ AML subset.

Page 102: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

92

Table 2-1. Table of AML patient characteristics from the PMCC/UHN dataset.

Patients were dichotomized into ‘low’ vs. ‘high’ CSF1R subgroups (CSF1Rlow

vs. CSF1Rhigh

)

based on decision tree analysis techniques.

Page 103: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

93

Figure 2.3. CSF1R cell surface protein levels are associated with poor AML clinical

outcome.

(A) Flow cytometry gating strategy in the identification of the AML blast population from the

bulk AML cell population. (B) Decision-tree analysis to determine an optimal expression value

cut-off for CSF1R expression levels.

A

B

Gating strategy of AML blasts

Decision tree analysis of CSF1R expression

Page 104: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

94

D

E

C

Figure 2.3. CSF1R cell surface protein levels are associated with poor AML clinical

outcome.

Continued from previous page. (C-E) Kaplan-Meier survival plots comparing overall survival

(OS) between CSF1Rlow

vs. CSF1Rhigh

in AML patients and in FLT3-ITD and NPM1c

negative vs. positive AML patient subgroups.

Page 105: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

95

Co-expression of CSF1R and CD34 exhibits heterogeneous patterns and association with

clinical OS in AML

Cell surface co-expression protein levels of CSF1R and CD34 were evaluated using flow

cytometry. CD34 is an important hematopoietic stem and progenitor cell marker and is an

adverse prognostic marker in AML. In the analyzed flow cytometry plots, CSF1R expression is

depicted on the y-axis while CD34 expression is on the x-axis, dividing the plot into 4 different

quadrants based on the co-expression of the two markers: CSF1Rlow/CD34low,

CSF1Rhigh/CD34low, CSF1Rhigh/CD34high, CSF1Rlow/CD34high. Across the same cohort of AML

cases as in the preceding analysis, varying co-expression patterns of CSF1R and CD34 were

observed, with the existence of multiple sub-populations based on CSF1R/CD34 levels (Figure

2.4A). For example, in a given sample that could be classified as being double positive for

CSF1R and CD34 (CSF1Rhigh/CD34high), one would likely expect to see a single cell population

residing in the CSF1Rhigh/CD34high fraction or quadrant. However, it was observed that while a

sample such as 090077 (Figure 2.4A) could be CSF1Rhigh/CD34high overall, it appears to have

two distinct cell subpopulations, each being positive for a single marker, and thus the two

markers are not co-expressed on the same cells. To capture this information, the quadrant

distributions can be represented as star or radar plots where each of the four arms in the plot

corresponds to a given co-expression pattern corresponding to the four different quadrants of the

flow plots (Figure 2.4B).

To evaluate the significance of the different CSF1R/CD34 distributions, unsupervised

cluster analysis was performed, yielding 3 distinct clusters: Cluster A: CSF1Rhigh/CD34low,

Cluster B: CSF1Rlow/CD34high and Cluster C: CSF1Rlow/CD34low (Figure 2.4C). A fourth cluster,

CSF1Rhigh/CD34high, would have been generated however it only contained two patients and thus

could not be characterized as a defined cluster. To evaluate the clinical significance of these

Page 106: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

96

subgroups or clusters, Kaplan-Meier survival analysis was conducted to evaluate whether the

identified CSF1R/CD34 subgroups have an association with clinical patient outcome. Using a

cut-off finder tool, the optimal cut-off for CSF1R was found to be 75% so that CSF1R levels

>75% were defined to be a CSF1Rhigh subgroup, while levels <75% a CSF1Rlow subgroup. A

median cut-off (50%) was established for CD34 where patients with CD34 levels >50% were

classified to be a CD34high subgroup, and <50% a CD34low subgroup. Survival analysis revealed

that patients in the CSF1Rhigh/CD34low cluster had shorter OS times compared to the

CSF1Rlow/CD34high and CSF1Rlow/CD34low clusters. The CSF1Rlow/CD34low cluster or subgroup

had the best OS outcome followed by the CSF1Rlow/CD34high subgroup (Figure 2.4D). Therefore

high levels of either CSF1R or CD34 are associated with poor OS. In CD34low patients, there was

a significant difference between OS between the CSF1Rlow/CD34low and CSF1Rhigh/CD34low

subgroups, with the patients in the latter having a worse prognosis. Therefore CSF1R is

prognostically significant in CD34low patients, identifying a CD34 low group with worse long-

term survival.

Page 107: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

97

A B

C

Page 108: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

98

D

Figure 2.4. CSF1R and CD34 co-expression patterns are associated with OS.

(A) Shown are plots of 6 different AML patient samples depicting co-expression patterns of

CSF1R and CD34. Each quadrant is numbered according to expression levels: 1-

CSF1Rlow

/CD34low

, 2- CSF1Rhigh

/CD34low

3- CSF1Rhigh

/CD34high

4- CSF1Rlow

/CD34high

. (B)

Co-expression patterns in AML patients (n = 146) represented in radar plot format. (C) Radar

plots of the identified clusters. (D) Kaplan-Meier survival plots

of the different clusters.

Page 109: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

99

CSF1R expression in AML cell lines

In order to interrogate CSF1-CSF1R ligand-receptor signaling within AML-BM stromal

cell interactions, AML cell lines and patient samples were examined for CSF1R expression to

determine which cell types would be useful and relevant in the studies. For this, CSF1R cell

surface protein levels were evaluated by flow cytometry in a panel of AML cell lines. Of the

AML cell lines interrogated (OCI/AML-2, OCI/AML-4, OCI/AML-5, HL-60 and U937), all of

the lines showed positive CSF1R staining (Figure 2.5A). CSF1R cell surface expression levels in

AML patient samples were also evaluated by flow cytometry (Figure 2.5B). There was variable

expression of CSF1R in the patient samples evaluated, with some samples having high

expression, and some with little to no expression of CSF1R. For the studies in this work, samples

with exclusively high CSF1R expression vs. low CSF1R expression were used.

Page 110: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

100

A

B

Figure 2.5. CSF1R expression in AML cell lines and patient samples. CSF1R cell surface

protein expression were evaluated by flow cytometry. Shown are a panel of (A) AML cell lines

and (B) select AML patient samples.

Page 111: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

101

AML cells do not show significant growth responses to exogenously presented soluble CSF1

AML cells were first tested for their response to exogenously presented human

recombinant CSF1 (rhCSF1) and conditioned medium (CM) from the engineered MS-5 stromal

cell lines. The OCI/AML-4 and OCI/AMl-5 cell lines are dependent on growth factors derived

from the CM of 5637 bladder carcinoma cells. 5637-CM is enriched in factors including GM-

CSF, G-CSF, as well as CSF1 and SCF in smaller amounts (Quentmeier, Zaborski, & Drexler,

1997). For the experiments, OCI/AML-4 and OCI/AML-5 cells were grown in the absence of

5637-CM to evaluate the efficacy of CSF1 as a single factor. OCI/AML-4, OCI/AML-5 and a

CSF1Rhigh patient sample (090017) were treated with soluble recombinant human CSF1

(rhCSF1), as well as CM from the different MS-5 stromal cell lines (EV, hCSF1-sol and hCSF1-

mem) for a period of 72 hours. It was observed that treatment of AML cells with variable levels

of rhCSF1 or 10% CM from any of the MS-5 cell lines did not induce significant growth or

proliferation of AML cells in culture (Figure 2.6A). Moreover, cells did not survive in culture for

more than 72 hours with the addition of rhCSF1, and thus longer time points could not be

assessed. Higher percent concentrations of the different MS-5 CM were also tested but they

yielded results similar to the use of 10% MS-5 CM. This suggests that as an independent, soluble

factor, CSF1 is unable to induce the growth and proliferation of AML cells in culture. Moreover,

the fact that CM from any of the MS-5 cells, including hCSF1-expressing cells (particularly

hCSF1-sol MS-5 as it produces soluble CSF1 ligand) did not support AML growth, provides

further indication that the context or manner of CSF1 presentation may be of major importance

for AML cell growth.

B

Page 112: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

102

A

B

C

Figure 2.6. AML cells show minimal growth responses to exogenous soluble CSF1.

(A-C) AML cells (OCI/AML-4, OCI/AML-5 and patient sample 090017) were treated with

recombinant human CSF1 (rhCSF1; 10 ng/mL) and 10% conditioned medium (CM) from the

different MS-5 stromal cell lines for 72 hours (initial cell number: 1x10^5). Cell counts were

performed using trypan blue.

Page 113: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

103

CSF1 transduction in the MS-5 stromal cell line

Since soluble recombinant CSF1 protein was unable to exert any significant effects on

the proliferation and survival of AML cells in culture, the effects of stromal cell-presented CSF1

were evaluated through AML-MS-5 cell co-cultures. For this, the murine MS-5 stromal cell line

was transduced with either soluble human CSF1 (hCSF1-sol) or membrane-

bound/transmembrane hCSF1 (hCSF1-mem). The system was designed as such because as noted

in Chapter 1 (section 1.3.2. and Table 1-5), many factors, such as ligand-receptor pairs, do not

show cross-species compatibility, including mouse CSF1, which does not act on human CSF1R

(Garceau et al., 2010). Given this, expressing the human ligand in mouse cells would allow for

the specific interrogation of the human CSF1-CSF1R interaction, with limited interference from

other factors because of the species incompatibility between the cells.

Despite being a murine stromal cell line, the MS-5 cell line has been shown to be able to

support both the short- and long-term growth and maintenance of human hematopoietic and

leukemic cells in-vitro (Berthier et al., 1997; Nishi et al., 1997). The MS-5 cell lines generated

were: MS-5-EV (empty vector), MS-5 hCSF1-sol and MS-5 hCSF1-mem. The overexpression of

CSF1 was confirmed by qPCR and western blotting (Figure 2.7A, B).

Page 114: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

104

A

B

Figure 2.7. CSF1 overexpression in MS-5 stromal cell lines.

MS-5 cells were transduced with a retroviral vector containing soluble human CSF1 (hCSF1-sol)

and membrane-bound human CSF1 (hCSF1-mem). CSF1 gene and protein expression were

evaluated by (A) qPCR and (B) western blot.

Page 115: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

105

CSF1 promotes the growth, survival and adhesion of AML cells in AML-stromal cell co-

culture

The goal of the co-culture studies was to observe the effect of continuously exposing

AML cells to CSF1 ligand directly produced, and presented by stromal cells. CSF1R positive

AML cell lines (OCI/AML-4, OCI/AML-5, OCI-AML-2 and U937) were tested for growth and

survival in the stromal cell co-culture assays. 5637-CM was not added to the co-cultures of

OCI/AML-4 and OCI/AML-5 cells in order to create minimal growth conditions for the

evaluation of CSF1 as a single factor. The growth of AML primary patient samples in stromal

cell co-cultures was also evaluated.

For the co-culture assays, MS-5 cells were plated in 6-well plates and allowed to adhere

overnight, creating a feeder layer for the AML cells. AML cells were then directly plated on the

stromal cells to form a co-culture system (see Materials & Methods) with each of the three

different MS-5 cell lines. The long-term growth and proliferation of AML cells was assessed

over an average period of 2 weeks. It was found that the OCI/AML-2 and U937 cell lines

exhibited no significant difference in their growth and proliferation in co-cultures with hCSF1-

expressing MS-5 cells (both hCSF1-sol and hCSF1-mem) vs. non hCSF1-expressing MS-5-EV

control cells (Figure 2.8A). Both cell lines grew effectively on the three types of MS-5 stromal

cells, and in fact grew at such a rapid rate that they caused the co-cultures to ‘crash’, with

overgrowth of the AML cells causing the stromal cell layer to detach from the bottom of the dish

and die out. The growth of OCI/AML-2 and U937 cells was actually comparable to their growth

in normal cultures (without stromal cells) because these cell lines are growth-factor independent

and grow well in cultures without any added growth factor supplementation. Therefore,

providing any additional support, in the form of stromal cells and CSF1 in this case, did not

confer any additional advantage to the cells. On the other hand, hCSF1-expressing MS-5 cells

Page 116: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

106

(both hCSF1-sol and hCSF1-mem) provided a significant proliferative advantage to OCI/AML-4

and OCI/AML-5 cell lines (grown in the absence of 5637-CM) compared to MS-5-EV control

cells (Figure 2.8B). The growth advantage conferred by hCSF1-expressing MS-5 cells was

readily apparent in the short term (i.e. 72 hours), and was sustained and enhanced in longer term

cultures of 2 weeks. These cells fared poorly in co-cultures with MS-5 EV cells, with little to no

cells surviving beyond a period of 2 weeks. In particular, MS-5 hCSF1-mem cells were better

able to support their growth compared to MS-5 hCSF1-sol cells, with a 3-fold increase in

OCI/AML-4 and OCI/AML-5 cells co-cultured with MS-5 hCSF1-mem cells compared to MS-

5-EV cells, and a 1.5-fold increase compared to MS-5 hCSF1-sol cells over a 2-week co-culture

period (P < 0.05). This demonstrates that CSF1 presented by MS-5 stromal cells supports the

proliferation and survival of growth factor-dependent AML cells (as opposed to cells that are

growth factor-independent), highlighting a CSF1-stromal cell dependency for these cells.

OCI/AML-4 and OCI/AML-5 cells demonstrated a significant amount of adhesion

(approximately 50-60%) to MS-5 hCSF1-mem cells compared to MS-5-EV and hCSF1-sol cells.

AML cells exhibited very little adhesion to the MS-5-EV cells and significantly less adhesion to

MS-5 hCSF1-sol cells (approximately 10%). In addition, the proliferation and growth of

OCI/AML-4 and OCI/AML-5 cells, particularly on MS-5 hCSF1-mem cells, was found to be

comparable to their growth under native 5637-CM supplemented conditions, indicating that

hCSF1-expressing MS-5 cells can compensate for, and act in lieu of 5637-CM supplementation

to provide the conditions necessary for cell growth. Taken together, these results highlight the

fact that there may potentially be key differences in how AML cells expressing CSF1R interact

with, and respond to the soluble vs. transmembrane forms of CSF1 ligand.

Page 117: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

107

Figure 2.8. MS-5 stromal cells expressing hCSF1 support the proliferation and survival

of factor-dependent CSF1R-expressing AML cell lines.

(A) OCI/AML-2 and U937 AML cells were co-cultured with MS-5 EV, hCSF1-sol and

hCSF1-mem stromal cells for a period of 2 weeks. Images were taken of the co-cultures (left)

and AML cell numbers were quantified (right). (B) The factor-dependent OCI/AML-4 and

OCI/AML-5 AML cell lines were also co-cultured with the MS-5 stromal cell lines. Shown are

quantitations from n = 2 - 3 independent experiments ± SEM. *P < 0.001, **P < 0.0001.

A

B

Page 118: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

108

AML primary samples are also highly growth factor-dependent ex vivo. Despite

supplementation with a host of growth factors, they cannot survive for more than a few days in

in-vitro cell cultures, which is the same case for OCI/AML-4 and OCI/AML-5 cells without

5637-CM supplementation. In this way, OCI/AML-4 and OCI/AML-5 cells are quite reflective

of primary samples in terms of their growth-factor dependency, as well as cellular characteristics

including morphology and adhesion.

The growth, proliferation and survival of primary AML samples with high CSF1R levels

(CSF1Rhigh) were compared to those with low CSF1R levels (CSF1Rlow) in co-cultures with the

MS-5 stromal cells. All of the primary AML samples chosen were selected to have low CD34

levels because it is a strong stem cell and adhesion marker, and clinically associated with poor

patient prognosis; being a strong factor, it may confound the effects of other factors, including

CSF1, and was thus excluded. CSF1Rhigh samples exhibited significantly higher proliferation,

with higher cell numbers in 5-week long-term co-cultures with both MS-5 hCSF1-sol and MS-5

hCSF1-mem cells compared to MS-5 EV cells (Figure 2.9A,B). AML cell numbers were 2-fold

higher after co-culture with MS-5 hCSF-1 mem cells compared to MS-5 hCSF1-sol cells.

CSF1Rhigh cells displayed a significant growth advantage particularly with MS-5 hCSF-1 mem

cells, with an average 11.5-fold difference in proliferation of cells cultured with MS-5 hCSF-1

mem cells compared to MS-5 EV cells (Figure 2.9C). On the other hand CSF1Rlow samples

displayed no growth and survival advantage on hCSF1-expressing MS-5 cells, with little to no

cells surviving in the co-cultures. Moreover, CSF1Rhigh cells also demonstrated significantly

more adhesion to MS-5 hCSF1-mem cells compared to MS-5 EV and hCSF1-sol cells.

MS-5 hCSF1-mem cells supported the long-term survival and maintenance of CSF1Rhigh

patient cells, with average survival times of 80 days, compared to MS-5 hCSF1-sol cells that

Page 119: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

109

were able to maintain AML cells for 72 days and MS-5-EV cells for 50 days (Figure 2.9D). In

fact one sample (090077) was sustained on MS-5 hCSF1-mem cells for as long as 6 months.

This indicates that MS-5 hCSF1-mem cells in particular, not only enhance the growth and

proliferation of AML cells, but also the long-term survival of AML cells.

Page 120: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

110

A

C

D

B

Figure 2.9. MS-5 stromal cells that express hCSF1 support the proliferation and survival of

primary AML cells that express CSF1R.

(A) CSF1Rhigh vs. CSF1Rlow AML patient samples were grown with the different MS-5

stromal cells (EV, hCSF1-sol, hCSF1-mem). Shown are images of co-cultures of representative

samples. (B) AML cell numbers (of 6 different primary samples) after co-culture with the

different MS-5 cells for a period of 2 weeks. (D) Days survival of AML patient samples co-

cultured with MS-5 cells.

Page 121: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

111

Blockade of CSF1R inhibits AML growth in co-cultures

To show the direct and specific involvement of CSF1R in the observed support of AML

cells in CSF1-expressing MS-5 co-cultures, a CSF1R monoclonal antibody (Amgen) was used to

block the interaction between CSF1R and hCSF1. It was observed that addition of the CSF1R

antibody to AML-MS-5 co-cultures led to the reduced growth and proliferation of both

OCI/AML-4 and OCI/AML-5 cells particularly cultured with MS-5 hCSF1-expressing cells

(Figure 2.10A-D). Significantly marked reductions in AML cell numbers were observed in co-

cultures of MS-5 hCSF1-sol and hCSF1-mem cells, but not MS-5-EV cells. This indicates that

the CSF1R antibody is specific to blocking the CSF1-CSF1R interaction, which is why AML

cells in the MS-5 hCSF1 co-cultures were sensitive to the drug. In OCI/AML-5 cells there was a

dose-dependent effect of the CSF1R antibody up to a concentration of 0.001 µg/mL, above

which there was no significant difference in the activity of the antibody in terms of inhibiting cell

growth. At concentrations of 0.001 µg/mL, there was a 2-fold decrease in the number of cells

compared to in untreated co-cultures (co-cultures of both MS-5 hCSF1-sol and hCSF1-mem).

This indicates that blocking the CSF1-CSF1R interaction between stromal and AML cells has a

significant impact on AML cell proliferation, providing further evidence that this interaction is

important in promoting and supporting the growth and survival of AML cells.

Page 122: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

112

Figure 2.10. Blockade of CSF1R on AML cells inhibits their proliferation in co-culture

with MS-5 hCSF1 cells.

(A-D) OCI/AML-4 and OCI/AML-5 cell proliferation after addition of an anti-CSF1R

antibody (Ab) from Amgen for 72 hours to 1 week in co-cultures of MS-5 cells. Shown are

results from n = 2 independent experiments ± SEM.

A

C

B

D

Page 123: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

113

Co-culture of AML and hCSF1-expressing MS-5 stromal cells leads to changes in signaling

and cytokine levels in AML cells

Given the significant effects of stromal cell-presented hCSF1 on AML cell growth and

survival, it was postulated that the binding of CSF1R to CSF1 might result in the production of

signaling molecules and cytokines that may in part, drive the enhanced growth of the cells. To

explore this, multiplexed cytokine platforms (Luminex MAGPIX, Eve Technologies) were used

to evaluate a panel of nearly 30 different secreted human cytokines relevant in cell growth,

survival and hematopoietic processes and pathways. For this, cell culture supernatants from

AML (090017 patient sample)-MS-5 cell co-cultures were collected after 48 hours of culture and

subjected to analysis. It was observed that co-culture of AML cells with MS-5 hCSF1-mem led

to increases in levels of GM-CSF, PDGF-AA, IL-8, IL-12p40, MCP-1, MIP-1α and IL-1RA

(Figure 2.11A). The cytokines observed to be upregulated in AML cells are largely those

involved in inflammatory and growth processes, with most produced and secreted by

macrophages (i.e. IL-8, IL-1RA, IL-12, MIP-1α). At the same time, levels of some cytokines,

such as MDC (macrophage-derived chemokine, or CCL22), appeared to decrease. Addition of

the CSF1R antibody to the co-cultures led to a reduction in levels of the elevated cytokines,

indicating that their regulation is dependent on a CSF1-CSF1R interaction between AML and

MS-5 stromal cells.

Potential changes in signaling effectors downstream of CSF1R were also examined in

AML cells after co-culture. For this, OCI/AML-5 cells were co-cultured with MS-5-EV and MS-

5 hCSF1-mem cells for 24 hours, after which intracellular signaling proteins were assessed using

flow cytometry. Signaling molecules in growth and survival pathways downstream of CSF1R

were included in the panel of flow markers assessed, namely pAkt, pS6 and pERK. It was

observed that co-culture of OCI/AML-5 cells with MS-5 hCSF1-mem cells led to increased

Page 124: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

114

levels of pS6 as compared to co-culture with MS-5-EV cells (Figure 2.12A). On the other hand,

there were no changes in levels of pAkt and pERK in AML cells following co-culture. Addition

of the CSF1R antibody to the co-cultures abrogated the observed increase in pS6 in AML cells.

Given the observed increase in pS6 in OCI/AML-5 cells following co-culture with MS-5

hCSF1-mem cells, the effects of inhibition of mTOR was evaluated in the co-cultures. It was

observed that the addition of the mTOR inhibitor MK-8996, led to decreased proliferation of

OCI/AML-5 cells grown with MS5 hCSF1-mem cells (Figure 2.12B). The mTOR inhibitor had

no effect on the growth of OCI/AML-5 cells co-cultured with MS-5 EV control cells. This

indicates that mTOR signaling is activated in AML cells exposed to CSF1, via stromal cells,

helping to promote AML cell growth.

Page 125: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

115

A

Figure 2.11. AML cells exhibit changes in cytokine production and mTOR pathway

activation after co-culture with hCSF1-expressing MS-5 cells.

(A) Multiplexed cytokine array analysis reveals changes in cytokines between MS-5-EV and

MS-5 hCSF1-mem cells which are reversed with a CSF1R blocking antibody (Ab).

Page 126: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

116

A

B

Figure 2.12. OCI/AML-5 cells exhibit mTOR pathway activation after co-culture with

hCSF1-expressing MS-5 cells.

(A) Intracellular flow cytometry to measure pS6 levels which are higher in AML cells after

co-culture with MS-5 hCSF1-mem cells; levels are reduced with a CSF1R Ab. (B) Addition

of an mTOR inhibitor (MK-8996) to co-cultures of OCI/AML-5 and MS-5 hCSF1-mem

cells. Shown are results from n = 3 independent experiments.

Page 127: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

117

The CSF1-CSF1R interaction induces changes in AML cell surface markers

Since culture of AML cells with MS-5 stromal cells led to changes in growth and

signaling, it was investigated whether the CSF1-CSF1R interaction could also lead to changes in

cell surface markers. A panel of human cell surface antigens was devised containing markers

involved in growth, survival and differentiation pathways relevant to hematopoietic and/or

leukemic cells. The markers included in the panel were CD34, CD38, CD4, c-Kit and CD11b.

The cell surface proteins CD34, CD38 and c-Kit are associated with hematopoietic stem cells

and hematopoietic cell differentiation, while CD11b is a marker found on mature monocytes and

macrophages. After co-culture of OCI/AML-5 cells with MS-5 cells for 2 weeks, it was found

that there were significant increases in levels of the hematopoietic stem cell markers CD34 and

c-Kit after culture with hCSF1-expressing MS-5 cells, particularly hCSF1-mem cells (Figure

2.13A,B). There was also an observed decrease in levels of CSF1R. Reduction of cell surface

levels of CSF1R may indicate receptor internalization and downregulation resulting from

regulation of receptor engagement and signaling. While there were some changes in these

markers after culture with MS-5-EV cells, they were not as significant when compared to culture

with MS-5 hCSF1-mem cells. Addition of the blocking CSF1R antibody (Ab) reversed these

effects with reductions in cell surface levels of CD34 and c-Kit on OCI/AML-5 cells (Figure

2.13C). The increased expression of hematopoietic stem cell markers (CD34 and c-Kit) may

indicate a cellular re-programming of cells to a more primitive, immature phenotype. This could

seemingly help create a pool of self-renewing progenitor cells in the leukemia.

Page 128: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

118

A

Page 129: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

119

Figure 2.13. AML cells grown in co-culture with hCSF1-expressing MS-5 cells exhibit

changes in cell surface marker expression.

(A) OCI/AML-5 cells were grown in co-culture with MS-5 EV, hCSF1-sol and hCSF1-mem

cells for 2 weeks. Surface marker expression was assessed using flow cytometry. (B) Addition

of a CSF1R blocking antibody (for 48 hours). Shown are representative results from n = 3

independent experiments.

B

Page 130: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

120

Human AML cells exhibit enhanced engraftment in humanized CSF1 mice

As discussed in the introduction, over 50% of AML primary samples fail to engraft in

conventional mouse models such as the NOD/SCID mouse. OCI/AML-5 cells also do not engraft

in NOD/SCID mice based on work in our lab. Given this, humanized knock-in mouse models

can offer an advantage where mice are ‘humanized’ through transgenic introduction of human

factors such as human cytokines and growth factors. In this work, a humanized CSF1 (huCSF1)

transgenic knock-in mouse from Jackson Laboratories/Regeneron was used to investigate

whether the presence of human CSF1 in the mouse may help the engraftment of CSF1R positive

AML cells that do not engraft in conventional mouse models. For this, OCI/AML-5 cells, as well

as several patient samples that were found to be non-engrafting in NOD/SCID mice, were

separately injected into huCSF1 mice and engraftment was assessed after a period of 5 -7 weeks.

It was observed that there was significant engraftment of the AML cells in the BM of the mice,

as assessed by cell surface staining of the human (h) hematopoietic cell marker CD45 (hCD45)

(Figure 2.14A). OCI/AML-5 cells demonstrated engraftment of up to 70%, while for primary

samples, it was over 35 – 40 %. This suggests that hCSF1 is an important factor in the homing

and engraftment of AML cells within the BM. OCI/AML-5 cells were isolated from the BM and

evaluated for cell surface marker expression. It was observed that there were increases in the

levels of the hematopoietic stem/progenitor markers CD34 and c-Kit on the surface of

OCI/AML-5 cells after growth in the huCSF1 mice (Figure 2.14B). This was similar to the

observed increases in these markers in the AML-MS-5 co-cultures, where OCI/AML-5 cells

displayed increases after co-culture with MS-5 hCSF1-mem cells. This indicates that both the

hCSF1 stromal cell co-culture and the humanized CSF1 mouse models are able to mimic and re-

capitulate each other’s characteristics for the growth of AML cells as driven by CSF1.

Page 131: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

121

A B

A M L ce lls

% h

CD

45

OC

I/A

ML

-5

09

00

17

09

04

81

10

00

19

10

00

20

10

01

12

10

02

86

0

2 0

4 0

6 0

8 0

Figure 2.14. AML cells exhibit engraftment in humanized CSF1 (huCSF1) mice, with

changes in cell surface marker expression.

(A) Plot of engraftment levels of OCI/AML-5 cells in huCSF1 mice as determined by percent

cell surface human CD45 (hCD45) expression evaluated by flow cytometry. (B) Cell surface

marker expression on OCI/AML-5 cells was evaluated by flow cytometry. The first column of

histograms on the left shows the cell surface marker expression profile of OCI/AML-5 cells

grown under endogenous conditions in culture. Shown are results from n = 3 independent

experiments (B).

Page 132: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

122

Further to this, BM stromal cells were isolated from the huCSF1 mice and grown in co-

culture with OCI/AML-5 cells. hCSF1 expression in the BM stromal cells was first confirmed by

western blot analysis (with use of a human specific antibody), which showed expression of

hCSF1 in the cells (Figure 2.15A). In particular, the cells appeared to express hCSF1-mem and

not hCSF1-sol. Co-culture of OCI/AML-5 cells with the huCSF1 mouse BM stromal cells led to

significant proliferation of the cells on the stromal cells compared to MS-5 EV control cells

(Figure 2.15B). The growth and proliferation of the OCI/AML-5 cells in culture with the

huCSF1 mouse BM stromal cells was in fact comparable to their growth on MS-5 hCSF1-mem

cells, indicating that the huCSF1 mouse-derived BM cells are likely similar in nature and

function to the MS-5 hCSF1-mem stromal cell line used in these studies. Furthermore, addition

of the CSF1R blocking antibody led to the reduced proliferation of OCI/AML-5 cells (Figure

2.15C). Finally, cell surface marker expression was also altered on OCI/AML-5 cells after co-

culture with the huCSF1 BM stromal cells, with significant increases in levels of CD34 and c-Kit

which were reversed with addition of the CSF1R antibody (Figure 2.15D). This is consistent

with the results of co-culturing OCI/AML-5 cells with MS-5 hCSF1-mem cells, as well as

injecting them into the huCSF1 mouse itself. Taken together, this demonstrates that the huCSF1

mouse is an improved mouse model for AML, as it provides the improved engraftment of human

AML cells that express CSF1R. Moreover, AML cells grown in huCSF1 mice re-capitulate

much of the cellular growth and phenotypic features seen in the in-vitro MS-5 hCSF1 stromal

cell co-culture model.

Taken together, the data presented in this chapter supports a role for CSF1, particularly

transmembrane CSF1, in promoting the growth, signaling and leukemic phenotype of AML

cells.

Page 133: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

123

A

B

C

Figure 2.15. BM stromal cells derived from huCSF1 mice support AML cell proliferation.

(A) Western blot of hCSF1 expression in BM stromal cells derived from huCSF1 mice. (B)

OCI/AML-5 cells co-cultured with BM stromal cells from huCSF1 mice, along with the MS-5

EV and hCSF1-mem cell lines, for a period of 2 weeks. (C) Addition of CSF1R Ab to co-

cultures of OCI/AML-5 and huCSF1 mouse-derived BM stromal cells. (D) Changes in cell (48

hours culture). Surface marker expression by flow cytometry after culture of OCI/AML-5 cells

with huCSF1 mouse-derived BM stromal cells for a period of 2 weeks. Significance at *P <

0.001, **P < 0.0001, ***P < 0.002 compared to MS-5 EV control. Results from n = 3

independent experiments.

Page 134: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

124

D

Page 135: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

125

2.5. DISCUSSION

As an important factor in the differentiation of myeloid cells, CSF1R has been classically

studied in the context of monocyte/macrophage development in normal individuals, where

normal cell immune cells infiltrate sites of inflammation, cancer, etc. In contrast, its role in

myeloid malignancy has not been deeply explored, likely to its consideration as a marker of

differentiation and more mature type cells. Herein, we have shown that in AML: 1) CSF1R is a

clinically significant marker that shows an association with clinical patient outcome, and 2) the

CSF1-CSF1R ligand-receptor pair supports and promotes leukemogenesis in the context of

AML-BM stromal cell interactions.

In this work, CSF1R was shown to be a clinically relevant marker that may help

prognostically stratify AML patients at both the gene and protein expression levels. Interrogation

of AML gene expression datasets and protein expression data revealed significant associations

between CSF1R levels and overall survival (OS). In the Valk et al. gene expression dataset,

CSF1R gene expression levels were found to be high in specific, previously identified subgroups

or clusters.(Figure 2.1A). Overall, in the Valk dataset, as well as two other gene expression

datasets (Metzler and TCGA), the dichotomization of AML patients into CSF1Rhigh and

CSF1Rlow subgroups revealed that CSF1Rhigh patients had poorer OS survival compared to

CSF1Rlow patients (Figure 2.2), indicating that CSF1R may aid in the stratification of AML

patients receiving induction style chemotherapy. This was further validated at the protein level

where high levels of cell surface protein levels of CSF1R on the blast cells of AML patients were

associated with poor OS, compared to low CSF1R levels (Figure 2.3C). The results obtained

from the gene expression datasets and cell surface flow cytometry data, were consistent with

each other, highlighting the relevance and potential usefulness of both RNA and protein-based

Page 136: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

126

approaches in the evaluation of CSF1R as a potential clinical marker in prognostic and/or

predictive clinical evaluations. Overall, CSF1R was demonstrated to be a significant clinical

marker across the cohorts of AML patients evaluated with its high expression levels in certain

AML cases, and its significant association with clinical survival. In addition, CSF1R was also

found to predict poor clinical outcome in subgroups of AML patients that do not harbour

common AML mutations such as FLT3-ITD and NPM1c (Figure 2.3D, E). This indicates that

CSF1R may help stratify subsets of AML that are ‘molecularly’ normal with respect to some of

the most common AML gene mutations. In this way, CSF1R could present as a potential

prognostic clinical marker, as it may help improve the stratification and treatment of AML

patients in less adverse risk groups.

CSF1R is highly expressed during monocytic differentiation and is thus typically

regarded as a marker of more differentiated cells in the monocytic-myeloid lineage. However,

we observed high levels of CSF1R expression on the blasts of nearly 70% of patients in our

AML cohort (Figure 2.3A and Table 2-1), suggesting that CSF1R may be aberrantly expressed

on more undifferentiated cells in a disease setting like AML. In line with this, CSF1R was

observed to be co-expressed with the stem cell marker CD34 on the blast cells of some AML

samples (Figure 2.4). Different patterns of CSF1R and CD34 co-expression were also found to

be associated with OS, with CSF1Rhigh/CD34low patients exhibiting poorer OS compared to

CSF1Rlow/CD34low patients. This again highlights the ability of CSF1R to be predictive of

clinical patient outcome in patients with less aggressive clinical characteristics such as low CD34

expression and/or the absence of gross molecular abnormalities like the presence of FLT3-ITD

or NPM1c.

Page 137: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

127

The clinical analyses highlighted CSF1R to be a prognostically significant clinical

marker in AML. This provided the rationale and basis for studying CSF1R functionally and in

the context of its ligand, CSF1, within a simulated BM microenvironment. Classic ligand-

receptor interactions are at the heart of juxtacrine signaling between cells, giving rise to distinct

microenvironments. To study the specific CSF1-CSF1R interaction, CSF1R-positive AML cells

and MS-5 stromal cells expressing hCSF1 were grown together in co-culture assays. The

expression of hCSF1 by mouse cells allowed the opportunity to determine what was happening

in the human AML cells, and the modified mouse stromal cells (Chapter 3). It was observed that

MS-5 hCSF1-mem and -sol cells enhanced and supported the long-term growth of the growth-

factor dependent cell lines OCI/AML-4 and OCI/AML-5, as well as AML patient samples

(Figure 2.8B). The proliferation and survival of CSF1R-expressing AML cells was almost

exclusively dependent on the CSF1-CSF1R interaction as MS-5-EV cells failed to support long-

term AML cell growth, primary AML cells expressing low levels of CSF1R (CSF1Rlow) failed to

grow on hCSF1 stroma, and addition of an anti-CSF1R antibody inhibited the proliferation of

OCI/AML-4 and OCI/AML-5 cells (Figure 2.10).

The proliferation and survival of OCI/AML-4 and OCI/AML-5 cells were significantly

supported by MS-5 hCSF1-mem cells in particular, with proliferation rates and numbers

comparable to growth under native conditions involving 5637-CM supplementation. Substituting

5637-CM with stromal cell-presented CSF1 therefore provided the same optimal growth

conditions for the growth of the cells. This highlights a role for CSF1 as an important context-

dependent factor (i.e. presentation by stromal cells) in supporting the proliferation and survival

of highly growth factor-dependent AML cells such as OCI/AML-4 and -5, and primary AML

Page 138: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

128

samples. This helps define more specified conditions (stroma) and factors (CSF1) for the support

of a subset of AML that expresses CSF1R.

AML cells exhibited significant adhesion to MS-5 hCSF1-mem cells compared to MS-5-

EV and hCSF1-sol cells, to which cells demonstrated little to no adhesion. The potent growth-

promoting properties of the membrane-bound/transmembrane form of CSF1 found in this study

is consistent with other studies. One study showed the membrane-bound form of CSF1 to

provide enhanced support of CSF1R-expressing myeloid cells compared to soluble CSF1, with

enhanced cell-cell adhesion and increased cellular growth (Tsuboi, Revol, Blanchet, &

Mouchiroud, 2000a). This suggests that the membrane-anchored form of the ligand may

effectively engage and support AML cells not only by directly binding to CSF1R on the surface

of AML cells, but also by potentially promoting additional adhesive connections between AML

and stromal cells; this provides added layers of support in addition to canonical ligand-receptor

binding. In this way, CSF1 may potentially cooperate with other stromal cell factors in the

physical, structural and biochemical support of AML cells.

Within stromal cells, transmembrane CSF1 may exert more robust effects compared to

soluble CSF1 because of its physical anchorage within the cell, which may provide it increased

stability and function. Moreover, the immobilization and anchorage of CSF1 ligand within the

stromal cell would seemingly provide AML cells continuous unhindered/uninterrupted exposure

to CSF1 compared to soluble CSF1, which could be readily depleted or undergo spatio-temporal

changes in terms of localization. Therefore, both the manner of growth factor ligand presentation

(i.e. by stromal cells) and isoform specificity are critical in dictating the nature of CSF1’s effects

on target AML cells that express CSF1R. Different forms of a ligand can indeed elicit distinct

effects as it has been shown that the soluble and membrane-bound forms of Kit ligand induce

Page 139: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

129

phosphorylation of different intracellular tyrosine residues on the c-Kit receptor to activate

different intracellular signaling pathways. This leads to varying effects on leukemic cell growth

and survival, with the soluble ligand failing to support leukemogenesis in-vivo (J L Gommerman,

Sittaro, Klebasz, Williams, & Berger, 2000). It therefore appears that membrane-anchored

proteins may harbour uniquely different functional properties compared to their soluble

counterparts.

It was observed that AML cells co-cultured with hCSF1-expressing MS-5 stromal cells

developed a LSC-like phenotype, with increased cell surface levels of CD34 and c-Kit (Figure

2.13). This may be suggestive of a re-programming of distinct AML cell populations to render a

more primitive, immature stem-cell phenotype. It could also be that a pre-existing sub-population

of CD34+/c-Kit+ cells are selected for by CSF1-expressing MS-5 stromal cells. This suggests

that the CSF1-CSF1R interaction between AML and stromal cells may drive the expansion and

maintenance of primitive pools of progenitor leukemic cells.

Notable changes in cytokine production were also observed after exposure of AML cells

to hCSF1-mem ligand. Primary AML cells were observed to have increased production of

inflammatory cytokines such as GM-CSF, IL-8 and MIP-1α among others (Figure 2.11A).

Levels of these were reduced with the addition of a CSF1R blocking antibody, indicating that the

CSF1-CSF1R interaction between AML and stromal cells directly induces the observed changes

in cytokine levels in AML cells. IL-8 and MIP-1α are largely pro-inflammatory cytokines that

are involved in chemotactic recruitment of inflammatory cell mediators including neutrophils

(Harada et al., 1994). Extracellular secretion of these cytokines in response to the CSF1-CSF1R

interaction has implications for added communication between AML and stromal cells. A recent

study reported that a CSF1R-expressing AML ‘support’ cell population exposed to CSF1 and

Page 140: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

130

Hepatic Growth Factor (HGF) led to further upregulation of HGF and other cytokines in the cells

(in a positive feedback loop), and that these paracrine signaling cytokines could be blocked by a

CSF1R inhibitor (GW-2580) (Edwards et al., 2019). Interestingly, the authors were unable to

identify or characterize the CSF1R-expressing ‘support’ cells, but it appeared that the cells were

a part of the bulk cell population of the primary AML samples assessed. Signaling cytokines can

therefore mediate cross-talk between cells to elicit and sustain production of cytokine factors

through feedback loops. Moreover, transfer of materials, including exosomes and entire

organelles such as mitochondria, has been shown to occur between stromal and AML cells

(Moschoi et al., 2016; H. Yang et al., 2018). This highlights how potentially different, and

secondary, modes of cell-cell communication can arise and be fostered by primary ligand-

receptor interactions.

mTOR signaling appeared to be activated in OCI/AML-5 cells exposed to MS-5 hCSF1-

mem cells, as observed through increases in pS6 (Figure 2.12A). This was blockable with a

CSF1R inhibitor. Moreover, use of an mTOR inhibitor in OCI/AML-5 – MS-5 hCSF1-mem cell

co-cultures was able to reduce the proliferation of OCI/AML-5 cells, indicating that mTOR is an

important target activated in AML cells as a result of the CSF1-CSF1R interaction. It therefore

appears that interacting AML and BM stromal cells display a dependency on a CSF1-CSF1R

axis which can potentially be therapeutically targeted at various levels.

The employment of a humanized CSF1 mouse model provided predicted and promising

in-vivo results, with demonstrated engraftment of CSF1R+ AML cells (OCI/AMl-5 and primary

AML samples) that do not engraft in traditional NOD-SCID mouse models (Figure 2.14A). This

highlighted a CSF1 dependency for the engraftment of CSF1R-expressing AML cells, indicating

a potential role for the ligand-receptor pair in the adhesion and homing of AML cells within the

Page 141: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

131

BM. BM stromal cells obtained and propagated from the huCSF1 mice were able to support

OCI/AML-5 cell growth in co-culture assays (Figure 2.15B,C); this was accompanied by the

same cell surface marker changes observed in cells directly engrafted in the huCSF1 mice

(Figure 2.14 B and 2.15D). These cell surface marker changes were also consistent with those

observed in OCI/AML-5 cells co-cultured with MS-5 hCSF1-mem cells, not only confirming the

results, but also demonstrating that the in-vitro and in-vivo systems are similar to each other.

These models can therefore serve as effective experimental tools for the study of the CSF1-

CSF1R interaction within the BM microenvironment of AML.

Taken together, CSF1R appears to be a biologically and clinically relevant marker in

AML, as it demonstrates a significant association with clinical patient outcome, and plays an

important role in AML-stromal cell interactions. The CSF1-CSF1R interaction enhances AML

cell growth, promotes an undifferentiated LSC-like phenotype, and induces changes in

intracellular and cytokine signaling factors, within a BM stromal cell context. Moreover, the

transmembrane form of CSF1 elicits significantly more potent effects compared to soluble CSF1,

highlighting the significance of structural and functional differences between CSF1 isoforms on

the support of AML cells. The CSF1-CSF1R axis thus holds considerable biological and clinical

value in AML and its microenvironment, thereby presenting as an attractive target for future

AML therapies.

Page 142: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

132

CHAPTER 3

CSF1 ligand signaling in AML-bone marrow stromal cell interactions

This chapter is in preparation for a manuscript to be submitted for publication

Page 143: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

133

3.1. ABSTRACT

In this study, the potential signaling role of the hematopoietic ligand colony stimulating

factor 1 (CSF1) was investigated in AML in the context of a BM stromal cell microenvironment.

It was found that CSF1 contains a potential, highly conserved PDZ domain binding motif

(PDBM) in its C-terminus that may interact with the serine protease HtrA1 to activate mTOR

signaling in MS-5 stromal cells, which in turn appears to impact on the paracrine support of

AML cells. While overexpression of transmembrane human CSF1 (hCSF1-mem) in MS-5 cells

was found to enhance the long-term growth and survival of AML cells, engineered deletion of

CSF1’s PDBM (hCSF1 ΔPDBM) resulted in reduced AML cell proliferation in in-vitro co-

culture assays. hCSF1-expressing MS-5 cells were observed to have a higher proliferation rate,

changes in cytokine production, and activation of mTOR signaling, as observed through

decreased TSC2 and increased pS6 levels, compared to non-hCSF1 expressing MS-5 cells. These

effects were lost and/or reversed in MS-5 hCSF1 ΔPDBM cells. Moreover, siRNA-mediated

knockdown of HtrA1 in MS-5 hCSF1-mem cells led to increased TSC2 and decreased pS6

levels, indicating inhibition of mTOR signaling. Taken together, these results demonstrate that

CSF1 has the ability to signal within MS-5 stromal cells to activate a CSF1-HtrA1-S6 axis.

Modulation of the stroma by CSF1 in turn has implications for its support of AML cells. As a

stromal cell ligand, CSF1 may therefore participate in bi-directional/multi-component signaling,

including PDZ-based intracellular signaling, to promote supportive interactions between AML

and BM stromal cells.

Page 144: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

134

3.2. INTRODUCTION

Acute Myeloid Leukemia (AML) is a highly complex and heterogeneous clonal disorder

of the bone marrow (BM). Its incidence increases with age and despite showing an initial

response to treatment, more than half of all AML patients relapse within 1-2 years of remission

and subsequently die of their disease (Liersch et al., 2014). AML can be described as a BM

failure state, characterized by decreased production of normal cells and increased production of

immature cells that are blocked in their ability to differentiate; these latter cells are able to

proliferate extensively as well as self-renew, a property essential for the maintenance of the

disease (Betz & Hess, 2010b; Ferrara & Schiffer, 2013b).

The BM is the principal site of hematopoiesis. The BM is a niche microenvironment

comprised of cells, growth factors and other structural elements, collectively known as the

stroma, which is important in regulating hematopoiesis (Morrison & Scadden, 2014b).

Alterations in components of the BM niche may occur to support malignant processes. For

example, it has been shown that osteoblasts with activating mutations in β-catenin leads to

enhanced HSC proliferation and development of leukemia (Kode et al., 2014). On the other

hand, malignant cells may effectively usurp their microenvironment to make it conducive for

tumorigenesis (Colmone et al., 2008). Both scenarios underscore the importance of the BM

microenvironment as a functionally important player in maintaining hematopoietic homeostasis.

Colony-stimulating factor 1 (CSF1) is a cytokine produced by stromal cells in the BM

that directs the differentiation, growth and maturation of myeloid-lineage cell precursors into

mature monocytes, macrophages and dendritic cells via binding to the cell surface receptor,

CSF1R, on target cells. CSF1 is structurally related to other hematopoietic ligands including Kit

ligand and Flt3 ligand. CSF1 regulates a number of other important biological processes

Page 145: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

135

including development of the placenta, uterine contractions and microglial cell development in

the brain, and serves as an unfavourable prognostic marker in some cancers such as renal and

liver cancers. In its active form, CSF1 exists as a disulfide-linked homodimer. There are two

predominant isoforms of CSF1, the first a heavily glycosylated membrane-bound or

transmembrane glycoprotein, and the second as either a soluble secreted glycoprotein or

proteoglycan. Transmembrane CSF1 can actually be slowly cleaved to release a soluble version

of the ligand (Horiuchi et al., 2014). In the BM, the transmembrane form of CSF1 predominates

and is largely produced by BM stromal cells such as fibroblasts and endothelial cells. It has been

shown that different ligand isoforms can elicit distinctly different phenotypic and signaling

effects. For example, the soluble and membrane forms of Kit ligand induce phosphorylation of

different intracellular tyrosine residues on the c-Kit receptor and subsequently activate different

intracellular signaling pathways (J L Gommerman et al., 2000).

Intracellular signal transduction pathways are mediated by interactions between proteins

possessing unique signaling domains, and proteins containing specific recognition binding sites

for these domains. PDZ (Postsynaptic density 95, PSD-85; Discs large, Dlg; Zonula occludens-1,

ZO-1) domains are amongst the most common protein-protein interaction domains found in

mammalian species. PDZ domains are typically 80-90 amino acids in length and found in

approximately 180 different human proteins (Doyle et al., 1996; Lee & Zheng, 2010). PDZ

domains bind to short structural sequences or motifs known as PDZ domain binding motifs

(PDBM). PDBMs are typically 4-6 amino acids in length and located at the extreme C-terminus

of target proteins where they serve as docking sites for the recruitment and activation of PDZ

domain-containing proteins.

Page 146: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

136

Amongst transmembrane proteins, the Ephrin family of ligands has been well described,

largely through the work of Tony Pawson, to participate in reverse intracellular, or bi-directional

signaling through PDZ-based protein-protein interactions. The Ephrin B1 ligand in particular has

been shown to interact with the PDZ domain of intracellular proteins such as syntenin and the

tyrosine phosphatase FAP-1 which are involved in cytoskeletal organization and protein

trafficking in neurons (Holland et al., 1996; D. Lin et al., 1999). Similarly, the C-terminal PDBM

of transmembrane transforming growth factor α (TGF-α) has been demonstrated to interact with

syntenin as well as the Golgi protein p59/GRASP55 in the Golgi apparatus, both of which

mediate targeting of the ligand to the cell membrane (Fernández-Larrea et al., 1999; Kuo, 2002).

Moreover, in cells of the BM, it has been shown that RANK secreted from osteoclasts (via

vesicles) can bind to transmembrane RANK ligand (RANKL) expressed on osteoblasts to

activate reverse signaling to promote bone formation (Y. Ikebuchi et al., 2018).

Previous unpublished work by our group revealed that the specific PDBM sequence of

Kit ligand (KitL) binds to intracellular HtrA1 (data not shown). The mammalian high

temperature requirement A (HtrA) is a family of trypsin-like serine proteases that contains at

least one C-terminal PDZ domain in its structure (Clausen, Southan, & Ehrmann, 2002b;

MURWANTOKO et al., 2004). HtrA proteases function as molecular chaperones and help

mediate the degradation of misfolded proteins (Clausen et al., 2002b; Krojer et al., 2002). The

human HtrA family consists of at least four members, HtrA 1 - 4, of which HtrA1 is primarily

soluble/secreted and expressed in the BM (Clausen et al., 2011). In the BM, HtrA1 plays key

roles in osteogenesis, bone remodelling and musculoskeletal development (Tiaden et al., 2012b).

Cytoplasmic HtrA1 has been shown to cleave tuberin/tuberous sclerosis 2 (TSC2), a negative

regulator of the mammalian target of rapamcyin (mTOR), followed by activation of the mTOR

Page 147: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

137

targets 4E-BP1 and S6K (Campioni et al., 2010). Because CSF1 shares high sequence and

PDBM similarity to KitL, it was postulated that HtrA1 may also be a CSF1 binding partner.

Furthermore, since HtrA1 has been shown to be a regulator of TSC2, this provided rationale for

examining mTOR signaling in the proposed setting.

Based on the hypothesis that the C-terminus of CSF1 contains a PDZ binding motif we

specifically set out to test the functionality of the PDBM of transmembrane CSF1 (hCSF1-mem)

using stromal cells engineered to express natural or mutant forms of hCSF1. Based on this it was

evident that CSF1 can function as a signaling ligand in both intracellular and intercellular

communication through multi-directional pathways (i.e. forward and reverse signaling).

Page 148: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

138

3.3. MATERIALS AND METHODS

DNA Plasmids and Gene Transduction

The full length membrane-bound/transmembrane variant of human CSF1 (hCSF1) cDNA was

purchased from OriGene (Rockville, MD, USA). It was used as a template to generate a PDBM

deletion mutant (ΔPDBM) by PCR reaction using the following PCR primers: 5’-

GAAGATCTACCATGACCGCGCCGGGCGCC-3’ (forward) and 5’

GGAATTCCTACTGTCTGTCATCCTGAGTC -3’ (reverse). Full length hCSF1-mem and

hCSF1-mem ΔPDBM were cloned into the Lenti-X Tet-off Advanced Inducible Expression

System (Clontech, Mountain View, CA, USA). These constructs were transduced into murine

MS-5 cells by first producing lentivirus through calcium phosphate transfection of 293-T cells

with psPAX2 and VSVG as described previously (Life Technologies). Virus rich medium was

collected 48 and 72 hours post-transfection. MS-5 cells were infected with the lentivirus for 24

hours with addition of 8 μg/mL protamine sulfate. Infected cells were obtained by puromycin

selection. The MS-5 Tet-off cell lines generated were: EV, hCSF1-mem and hCSF1-mem

ΔPDBM.

Cell Culture

Murine MS-5 stromal cells were cultured in α-MEM complete medium with 10% fetal bovine

serum (FBS), 100 units/mL penicillin and 100 μg/mL streptomycin at 37°C in 5% CO2. Human

OCI/AML-4 and OCI/AML-5 cell lines were cultured in the same conditions as the MS-5 cells,

but with the addition of 10% conditioned medium (CM) from 5637 bladder carcinoma cells.

Page 149: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

139

Co-culture Assays

For co-culture assays, a stromal cell feeder layer was first established by seeding 1.5 x 105 MS-5

cells in 6-well plates and allowing them to adhere overnight. The following day, the culture

medium was removed and 1.5 x 105 AML cells (OCI/AML-4 and OCI/AML-5 cell lines) were

added to the MS-5 cells with addition of fresh medium. Co-cultures were maintained by demi-

depopulation of AML cells every 2-3 days as described previously. Briefly, half the culture

medium was removed by gently washing medium over the stroma to detach some of the adherent

AML cells in order to remove them. The same volume of fresh medium was then added to the

cultures.

Growth Curves

MS-5 growth curves were performed by seeding 1 x 105 cells in 6-well plates (separate plates for

each different time point). Cells were counted daily by trypsinizing the cells and staining them

with trypan blue. Cells were counted manually using a haemocytometer.

Immunoblotting

Protein was extracted by lysing cells in 50-100 μL of RIPA lysis buffer (500 mM Tris, pH 7.4,

150 mM NaCl, 0.5% Na deoxycholate, 1% NP-40, 0.1% SDS and complete protease inhibitor

preparation containing leupeptin and aprotinin), incubated on ice for 30 min and centrifuged at

14,000 X g for 15 mins. Cell lysate supernatant was collected, and protein levels quantified using

the Bradford assay. SDS loading buffer (2% SDS, 10% glycerol, 360 mM 2-mercaptoethanol,

100 mM Tris) was added to cell lysates and boiled for 5 min. 40 - 100 μg of total protein was

separated using SDS-polyacrylamide gel electrophoresis (PAGE) and transferred onto PVDF

Page 150: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

140

membranes. Membranes were blocked with 5% milk in TBS-tween (0.1%) wash buffer,

followed by incubation with primary antibodies overnight at 4°C. The next day, membranes were

washed, and incubated with HRP-conjugated secondary antibodies for 1 hour at room

temperature with gentle agitation. Proteins were detected using ECL detection reagent (GE

Healthcare, Amersham, UK). The primary antibodies phospho-Akt (Ser 473), Akt, phospho S6

(Ser 235/236), S6 and GAPDH were purchased from Cell Signaling Technology (CST, Danvers,

MA, USA). CSF1 and HtrA1 primary antibodies were purchased from Santa Cruz Biotechnology

(SCB, Dallas, TX, USA).

Cytokine Analysis

The Luminex MAGPIX system was used to measure cytokine levels in cell culture supernatants.

Supernatant from co-cultures of AML and MS-5 stromal cells were collected and centrifuged

(14,000 X g for 15 minutes) to remove debris and other cellular particulates. Supernatants could

then be stored at -80°C for a prolonged period of time, or until analysis. Supernatants were

incubated with a cocktail of antibodies conjugated to metal beads (purchased in the kit) as per the

manufacturer’s protocol (Luminex, Toronto, Canada). Briefly, a cocktail of antibody conjugated

magnetic beads were added to a 96-well plate (purchased from Luminex). The beads were

washed twice with wash buffer after which cell culture supernatant was added to each well,

along with standards and controls. The plate was incubated in the dark at 4°C overnight with

gentle agitation. The next day, the beads were washed, and detection antibody was added and

incubated for 1 hour with gentle agitation. After several washes, the plate was read on a

MAGPIX machine and data was analyzed using Bio-Plex software (Bio-Rad).

Page 151: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

141

HtrA1 knockdown

HtrA1 and non-targeting control luciferase and GFP siRNA oligonucleotides were purchased

from Sigma-Aldrich (Oakville, ON, Canada). MS-5 cells were transfected with the siRNAs using

RNAiMax transfection reagent purchased from Thermo Fisher. Cells were transfected with 50 –

100 nM of siRNA according to the manufacturer’s protocol. 48 hours post-transfection, cells

were harvested, and subjected to western blotting analysis to evaluate levels of HtrA1 protein.

Statistical Analysis

Unless otherwise stated, mean ± S.E.M. values are given and P values were calculated using the

two-tailed unpaired Student’s t-test.

Page 152: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

142

3.4. RESULTS

Identification of a PDZ binding motif (PDBM) in CSF1

Based on sequence analysis and previous work by our group showing that KitL contains a

C-terminus PDBM, we wondered whether two other important hematopoietic transmembrane

ligands, CSF1 and Flt3L, might also contain a PDBM. PDBMs can vary in length from between

5-20 amino acids. Sequence analysis confirmed that KitL and CSF1 contain PDBMs in their

cytoplasmic regions, with the last amino acid (typically a valine residue in PDBMs) being

incompatible with binding to a PDZ domain (Figure 3.1A, top panel). Flt3L is unlikely to have a

PDBM as it lacks a terminal valine and shows considerable species variation; the latter was not

the case for KitL or CSF1.

While both the PDBMs of CSF1 and KitL contain 5 amino acids, their sequences differ.

The PDBM sequence of CSF1 is VELPV, which consists of largely hydrophobic amino acids

whereas the PDBM sequence of KitL is EFQEV, which is more varied with both hydrophilic and

acidic amino acids. The hydrophobic terminal amino acid, valine, however is common to both

CSF1 and KitL and most known PDBMs. The intracellular domain of CSF1 is highly conserved

across species, with high sequence similarity between human, mouse, rat and cow (Figure 3.1A,

bottom panel). The PDBM of CSF1 shows 100% sequence similarity between human, mouse

and rat. This indicates that the PDBM is likely an important functional domain in CSF1.

Results from a yeast two-hybrid screen revealed that KitL binds to the serine protease

HtrA1 via its PDBM. HtrA1 contains one PDZ domain at the end of its C-terminus (Figure

3.1B). Because KitL is structurally related to CSF1, particularly with respect to its cytoplasmic

domain, HtrA1 was also posited to be a potential binding partner of CSF1 based on its potential

for participating in PDZ-based interactions.

Page 153: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

143

Figure 3.1. The intracellular domain of CSF1 possesses a conserved C-terminal PDZ

domain binding motif (PDBM).

(A) The type III RTK hematopoietic ligands CSF1, Kit and Flt3 contain a conserved

intracellular domain with a C-terminal recognition sequence (last 5 amino acids) for proteins

with PDZ domains. This is referred to as the PDZ domain binding motif (PDBM). (B) The

intracellular domain and PDBM of CSF1 shows high conservation across species.

A

B

Page 154: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

144

Phenotypic and signaling profiles of hCSF1-expressing MS-5 cells

Given that CSF1 may harbour an intracellular signaling potential through its C-terminal

PDBM, the behaviour of cells expressing hCSF1 were examined. The growth and signaling of

murine MS-5 stromal cells overexpressing transmembrane hCSF1 (MS-5 hCSF1-mem)

engineered in our previous work, were compared to MS-5 control empty vector cells (MS-5-EV).

It was found that MS-5 hCSF1-mem cells grew at a faster rate compared to MS-5-EV cells

(Figure 3.2A). After 96 - 120 hours, MS-5 hCSF1-mem cells were almost double in number

compared to the number of MS-5-EV cells.

Based on the hypothesis of a plausible CSF1-HtrA1 binding interaction, levels of TSC2,

a reported substrate of HtrA1, were examined. It was observed that TSC2 levels were indeed

lower in MS-5 hCSF1-mem cells compared to MS-5-EV control cells, indicating a possible

activation of HtrA1 proteolytic activity in CSF1-expressing cells (Figure 3.2B). Since TSC2 is a

negative regulator of mTOR activity, lower TSC2 levels should relieve its inhibition of mTOR.

To address this, phosphorylated (p), and hence active, levels of the mTOR target S6 were

examined. In keeping with my hypothesis there were indeed higher levels of pS6 (S235/236) in

MS-5 hCSF1-mem cells compared to MS-5-EV cells (Figure 3.2B). There were no detectable

differences in levels of pAkt between MS-5 hCSF1-mem cells and MS-5-EV cells, suggesting

that the increased levels of pS6 observed are likely due to Akt-independent mechanisms

involving HtrA1-mediated degradation of TSC2. Lower TSC2 levels along with concomitantly

higher pS6 levels helped to confirm mTOR pathway activation in the hCSF1-expressing MS-5

cells. Activation of a pro-growth mTOR pathway downstream of hCSF1 helps to explain the

observed differences in cell growth rates between MS-5 hCSF1-mem and MS-5-EV cells.

Page 155: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

145

In addition to differences in growth and signaling differences, it was observed that MS-5

hCSF1-mem cells had a distinctly different cytokine profile than MS-5-EV cells, with higher

levels of G-CSF, IL-6, MCP-1, KC (keratinocyte chemoattractant, also known as CXCL1), and

lower levels of MIP-1a and VEGF (Figure 3.2C). Levels of these cytokines were reversed with

the addition of a blocking CSF1R antibody (Ab), demonstrating the direct implication of CSF1

in inducing the production of these signaling cytokines.

Page 156: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

146

A B

C

Figure 3.2. CSF1-expressing MS-5 stromal cells demonstrate differential phenotypic and

signaling behaviour.

(A) The growth rates of MS-5 Tet-off EV vs. hCSF1-mem cells were compared over a period

of five days. Cells were plated and counted each day using trypan blue. (B) Signaling proteins

relevant to the mTOR pathway were evaluated using western blot. (C) Cytokine production

was evaluated using multiplexed cytokine arrays. Shown are results from n = 3 independent

experiments.

Page 157: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

147

Generation of a ΔPDBM mutant to interrogate the function of CSF1’s PDBM

To investigate the potential role of CSF1’s PDBM in the observed phenotypes and

signaling events in hCSF1-expressing MS-5 cells, the PDBM of hCSF1 was deleted (terminal 5

amino acids) to create a deletion mutant (hCSF1 ΔPDBM); this was transduced into MS-5 cells.

An inducible tetracycline-based ‘OFF’ (Tet-off) vector system was used so that hCSF1

expression could be regulated i.e. ‘turned off’ or inhibited with the addition of doxycycline

(Dox), which is a more stable analog of tetracycline, in order to assess phenotypic and signaling

changes associated with hCSF1 expression. The MS-5 Tet-off cell lines generated upon

transduction of the different constructs were: MS-5 Tet-off 1) Empty Vector (EV), 2) hCSF1-

mem and 3) hCSF1-mem ΔPDBM. To assess and confirm expression of hCSF1 in the

engineered MS-5 cells, western blotting was performed to evaluate hCSF1 protein levels. It was

observed that there was robust expression of hCSF1 in both MS-5 Tet-off hCSF1-mem and

hCSF1-mem ΔPDBM cells in the non-induced (silenced) cells (Figure 3.3A). Interestingly, the

molecular weight of hCSF1-mem ΔPDBM appeared to be slightly less than that of full length

hCSF1-mem. While the removal of 5 amino acids may lend to the observed smaller size, there

may be potential differences in post-translational modifications (i.e. glycosylation, prenylation

etc.) between the full length and deletion mutant that may account for the size difference.

To confirm and test the functionality of the Tet-off system, increasing concentrations of

doxycycline were added to the MS-5 Tet-off cells in order to inhibit hCSF1 expression.

According to the manufacturer’s protocol, protein expression levels can be regulated in a

doxycycline concentration-dependent manner. Upon addition of Dox, it was observed that there

was a significant reduction in hCSF1 protein levels at comparable amounts (approximately 90%)

in both hCSF1-mem and hCSF1-mem ΔPDBM cells (Figure 3.3B). The regulatable system is

Page 158: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

148

quite sensitive, as concentrations as low as 0.001 µg/mL led to almost complete elimination of

hCSF1 expression in both cell lines. This shows that the system is quite robust and not ‘leaky’ in

terms of rendering optimal inhibition of protein expression levels. Moreover, higher Dox

concentrations did not achieve greater inhibition of expression. A Dox concentration of 1 µg/mL

was chosen and used for subsequent experiments as this was found to be an optimal

concentration according to our tests, in addition to being a commonly used concentration in such

inducible systems.

To investigate whether the observed activation of mTOR signaling in MS-5 hCSF1-mem

cells (Figure 3.2B) was directly linked to CSF1 expression and activity, MS-5 Tet-off cells were

treated with Dox to ‘turn off’ and inhibit the expression of CSF1. It was observed that inhibition

of CSF1 in MS-5 Tet-off hCSF1-mem cells led to decreased levels of pS6 compared to untreated

cells (Figure 3.3C). Levels of pS6 were almost entirely diminished and comparable to those in

the MS-5 Tet-off EV cells.

Page 159: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

149

Figure 3.3. CSF1 expression in MS-5 Tet-off inducible cells.

(A) MS-5 cells were transduced with full length hCSF1-mem, hCSF1-mem ΔPDBM or empty

vector (EV). hCSF1 expression was confirmed by western blotting. (B) Increasing

concentrations of doxycycline (Dox) were added to the MS-5 Tet-off cells to ‘turn off’ or

inhibit hCSF1 expression. CSF1 levels were evaluated by western blotting. (C) MS-5 Tet-off

cells were treated with Dox (1 µg/mL) and levels of pS6 were examined using western

blotting.

A

B

C

Page 160: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

150

CSF1 activates mTOR signaling via HtrA1

To examine the role of hCSF1’s PDBM in the observed activation of mTOR signaling,

pS6 levels were examined in MS-5 Tet-off cells expressing full length hCSF1 vs. the PDBM

deletion mutant. It was observed that pS6 levels were lower, and in fact completely abolished, in

MS-5 Tet-off hCSF1-mem ΔPDBM cells compared to hCSF1-mem cells, at levels comparable to

those in MS-5-EV cells (Figure 3.4A). These results indicate that CSF1 induces activation of

mTOR signaling, as observed through increased levels of pS6 in hCSF1-expressing MS-5 cells,

specifically through its PDBM. There were also observed differences in cell growth between

MS-5 Tet-off cells with the hCSF1-mem exhibiting greater cell numbers after 72 hours

compared to EV and hCSF1-mem ΔPDBM cells (Figure 3.4B). The growth rates of EV and

hCSF1-mem ΔPDBM were comparable, indicating that loss of the PDBM from hCSF1

significantly impacts the growth of the stromal cell. In addition, cytokine analysis revealed that

levels of cytokines such as IL-6 and KC were lower in MS-5 Tet-off hCSF1-mem ΔPDBM cells

compared to hCSF1-mem cells (Figure 3.4C). This indicates that the production of these

cytokines is dependent on the presence of CSF1’s PDBM, which fosters signaling mechanisms

that lead to control of cytokine production.

To examine the role of HtrA1 as a potential mediator of CSF1-induced mTOR pathway

activation in MS-5 cells, siRNA-mediated knockdown of HtrA1 was performed. It was observed

that knockdown of HtrA1 in MS-5 Tet-off hCSF1-mem cells led to an increase in levels of

TSC2, with a concomitant decrease in levels of pS6 compared to cells transfected with control

GFP siRNA (Figure 3.4D). This demonstrates that HtrA1 is a mediator of mTOR pathway

activation induced by CSF1 in MS-5 cells.

Page 161: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

151

Figure 3.4. CSF1-induced mTOR signaling is mediated by HtrA1 in MS-5 cells.

(A) pS6 levels were evaluated in MS-5 Tet-off cells expressing full length hCSF1 (hCSF1-

mem) and the hCSF1-mem PDBM deletion mutant (ΔPDBM). (B) Growth curves of the MS-5

Tet-off cells (EV, hCSF1-mem, hCSF1-mem ΔPDBM). (C) Cytokine levels were evaluated in

the MS-5 Tet-off cells using a multiplexed mouse-specific cytokine array. (D) Knockdown of

HtrA1 in MS-5 Tet-off cells was performed using siRNA transfection.

A

C

B

D

Page 162: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

152

The PDBM of CSF1 impacts the support of AML cells in AML-MS-5 cell co-cultures

To assess the impact of CSF1’s cytoplasmic PDBM on the support of AML cells by MS-

5-AML cells expressing the ligand, the MS-5 Tet-off cells (EV, hCSF1-mem and hCSF1-mem

ΔPDBM) were co-cultured with the OCI/AML-4 and OCI/AML-5 cell lines. After a two-week

co-culture period, it was observed that the MS-5 Tet-off hCSF1-mem ΔPDBM cells conferred a

reduced growth advantage to the AML cells compared to MS-5 Tet-off hCSF1-mem cells

(Figure 3.5A, B). While MS-5 Tet-off hCSF1-mem cells supported a greater than 2-fold increase

in AML cell number compared to MS-5-EV cells, AML cells grown with MS-5 Tet-off hCSF1-

mem ΔPDBM cells only showed a 1.3-fold increase in cell number. This demonstrates that the

growth-promoting properties of hCSF1-mem are significantly reduced with loss of the PDBM.

To further examine the growth-promoting properties of CSF1, the clonogenic potential of

OCI/AML-5 cells was assessed following co-culture with the different MS-5 Tet-off cells for a

period of 48 hours. After co-culture, AML cells were collected and plated into methylcellulose

with growth factors to determine the number of clonogenic cells remaining. It was observed that

OCI/AML-5 cells that had been pre-cultured with MS-5 Tet-off hCSF1-mem cells gave rise to a

larger number of colonies in the colony assays compared to cells that had been pre-cultured with

MS-5 Tet-off EV cells (Figure 3.5C). AML cells that had been co-cultured with MS-5 Tet-off

hCSF1-mem ΔPDBM cells gave rise to colony numbers that were comparable to AML cells that

had been pre-cultured with MS-5 Tet-off EV cells. This suggests that pre-culture of AML cells

with hCSF1-expressing MS-5 cells can potentiate the clonogenic ability of AML cells, and that

the PDBM of hCSF1 appears to play a role.

Page 163: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

153

Figure 3.5. Support of AML cells by hCSF1-expressing MS-5 cells.

A, B) OCI/AML-4 and OCI/AML-5 cells were co-cultured with the different MS-5 Tet-off

cells. Shown are cell numbers after a 2-week co-culture period. (C) OCI/AML-5 cells were

pre-cultured with the MS-5 Tet-off cells for 48 hours and then plated into methylcellulose.

Colonies (a colony was scored as having more than 30 cells) were counted after 10 days.

Significance at * P < 0.001. Shown are results from n = 2 independent experiments.

A

B

C

Page 164: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

154

Model of multi-directional CSF1 ligand signaling

The results here demonstrate that hCSF1 has the ability to activate the mTOR pathway, in

a reverse manner via its PDBM. Specifically, a CSF1-HtrA1-pS6 signaling axis appears to be

activated in which hCSF1 and HtrA1 interact via PDZ interactions. This is demonstrated by the

fact that both deletion of hCSF1’s PDBM and knockdown of HtrA1 lead to abolishment of pS6

levels in MS-5 cells. Therefore a proposed model for hCSF1 intracellular signaling within MS-5

cells involves hCSF1-induced activation of HtrA1’s proteolytic activity, leading to degradation

of TSC2 and hence activation of S6, which can then lead to changes in gene expression and

production of secondary signaling cytokines (such as IL-6, KC and TNFα). In this way, as a

signaling ligand, hCSF1 can elicit multiple signals in multiple directions through various

mechanisms to ultimately impact the support of AML cells: 1) CSF1-CSF1R receptor-ligand

engagement (‘forward’ signal), 2) reverse intracellular signaling via PDZ-mediated protein-

protein interactions 3) production of secondary/tertiary signaling molecules resulting from

hCSF1 reverse signaling (Figure 3.6B).

Page 165: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

155

Figure 3.6. Model of CSF1-induced multi-directional signaling in AML-stromal cell

interactions.

(A) CSF1 interacts with HtrA1 via its C-terminal PDBM to activate mTOR signaling, which

can lead to changes in cytokine production and gene expression. (B) CSF1 can elicit multiple

signals in multiple directions to impact the support of AML in the BM.

A

B

Page 166: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

156

3.5. DISCUSSION

The results from this work indicate that hCSF1 possesses intrinsic signaling function by

means of its C-terminal PDBM, which allows it to reverse signal within MS-5 stromal cells to

activate a hCSF1-HtrA1-S6 signaling axis. Activation of this axis may modulate the phenotype

and behaviour of MS-5 stromal cells, which in turn, appears to positively support AML cell

growth and survival. The existence of a putative PDBM in the structure of CSF1 was first

identified through sequence analysis which confirmed that CSF1 contains one PDBM located at

its C-terminus (Figure 3.1A). The PDBMs of CSF1, and the related KitL, both have a C-terminal

hydrophobic valine, which is almost invariably found as the last residue, i.e. in the P0 position, of

most PDBMs. The intracellular domain of CSF1 is highly conserved across species, with 100%

conservation of the PDBM in particular, highlighting its potential physiological importance

(Figure 3.1B).

Phenotypic modulation of MS-5 stromal cells by the presence of CSF1 was highlighted

by observed differences in growth rates and signaling between MS-5 cells expressing the ligand

vs. cells that do not. MS-5 hCSF1-mem cells were found to proliferate faster than MS-5-EV

cells, with almost a two-fold difference in growth rates (Figure 3.2A). Loss of the PDBM of

CSF1 led to reduced proliferation to levels comparable to MS-5 Tet-off EV cells (Figure 3.5D).

Potential mechanisms underlying these phenotypic differences were examined with respect to

potential CSF1 binding partners and signaling mediators. Previous work from our group has

shown that the serine protease HtrA1 is a binding partner of the related KitL. Because HtrA1 has

been shown to degrade the mTOR regulator TSC2 (Campioni et al., 2010), it was hypothesized

that a CSF1-HtrA1 interaction would lead to activation of mTOR signaling in MS-5 stromal cells

expressing CSF1. To support this hypothesis, it was found that MS-5 hCSF1-mem cells indeed

Page 167: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

157

had lower levels of TSC2, and concomitantly higher levels of pS6 ribosomal protein levels

compared to MS-5-EV cells (Figure 3.2B). CSF1 was found to be directly implicated in the

observed mTOR pathway activation, as inhibition of CSF1 expression through addition of Dox

to the MS-5 Tet-off hCSF1-mem cells caused a reduction in levels of pS6 (Figure 3.3C). The

enhanced growth that CSF1-expressing MS-5 cells experience, compared to cells that don’t

express CSF1, could potentially be explained by the activation of mTOR signaling in the cells, as

activated S6 (as part of the ribosomal complex) leads to increased protein production through

stimulation of cap-dependent mRNA translation via activation of eukaryotic translation initiation

factor 4E (eIF4E).

A direct implication for CSF1’s PDBM role in eliciting reverse intracellular signaling in

MS-5 stromal cells was demonstrated by the fact that the deletion of the PDBM led to lower

levels of pS6, indicating the inhibition/de-activation of mTOR signaling in the cells (Figure

3.4A). The absence of CSF1’s PDBM also led to changes in levels of cytokines such as IL-6

(Figure 3.4C). Moreover, downstream of CSF1, HtrA1 was demonstrated to be a mediator of the

observed mTOR pathway activation, as knockdown of HtrA1 led to increased levels of TSC2

and decreased levels of pS6 in MS-5 hCSF1-mem cells (Figure 3.4D). This mimicked the effects

of CSF1 PDBM deletion, which showed that a loss of function of either CSF1 or HtrA1 results in

inhibition of mTOR signaling in CSF1-expressing MS-5 cells. This work was therefore able to

identify a novel CSF1-HtrA1-mTOR signaling axis in BM stromal cells.

There were also observable differences in the cytokine profiles of hCSF1-expressing vs.

non-CSF1-expressing MS-5 cells (Figure 3.2C) with higher levels of cytokines such as IL-6, KC

(CXCL1) and MCP-1, which are factors associated with growth and inflammatory pathways. Co-

culture of AML cells with the MS-5 cells led to further changes in these cytokines, with addition

Page 168: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

158

of a CSF1R antibody leading to a reversal in the changes, as did deletion of the PDBM of CSF1

(Figure 3.2C, 3.5D). This indicates that intracellular CSF1 ligand signaling can directly induce

changes in cytokine secretion. Interestingly, the observed activation of mTOR signaling in

hCSF1-expressing cells could potentially induce IL-6 production in the cells. This is likely due

to the known ability of activated mTOR to stimulate the transcriptional regulator NF-κB, which

can induce transcription of IL-6 and other signaling cytokines (Brasier, 2010; Dan et al., 2008; T.

Liu, Zhang, Joo, & Sun, 2017). IL-6 is an important inflammatory and hematopoietic cytokine.

When produced by BM stromal cells, it can contribute to the protective properties of the tumor

microenvironment as it can promote inflammation, immune suppression, osteolysis (leading to

release of additional inflammatory cytokines including IL-6 itself) and tumorigenesis (Harmer,

Falank, & Reagan, 2019). In fact, IL-6 levels are found to be upregulated in multiple myeloma

and some solid tumors. In a paediatric AML study, high IL-6 levels in the BM (assessed in BM

aspirates) were shown to be associated with poor clinical outcome (Sanchez-Correa et al., 2013;

Stevens, Miller, Munoz, Gaikwad, & Redell, 2017). This highlights a role for IL-6 in the

microenvironment of AML, production of which could be driven by CSF1-CSF1R signaling.

CSF1-induced activation of pro-growth signaling pathways such as mTOR-S6, along

with changes in the production of signaling cytokines, could help explain the increased growth

and survival of MS-5 cells expressing hCSF1. These phenotypic and signaling differences

indicate that the presence of hCSF1 in MS-5 stromal cells can intrinsically alter their

characteristics and behaviour. Similarly, it has been shown that Ephrin B ligand reverse signaling

within neuronal cells induces morphological changes such as membrane blebbing and cell

retraction through engagement with proteins such as ROCK (Rho-associated protein kinase)

(Bochenek, Dickinson, Astin, Adams, & Nobes, 2010). This highlights a role for reverse ligand

Page 169: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

159

signaling in the functionality of the ligand-expressing host cell. Interestingly, CSF1 demonstrates

activity within the MS-5 stromal cell without any required binding or engagement by an external

protein or receptor. In other words, it does not appear that CSF1 needs to be ‘activated’ by

binding to its receptor (i.e. CSF1R) in order to elicit signaling within the cell. This could be

explained in two ways: 1) CSF1 is normally expressed as a homodimer and as such, it may be

constitutively active in its ability to reverse signal without binding to its receptor; 2) in the

engineered CSF1 inducible overexpression system, high CSF1 levels may alter its distribution on

the cell surface, i.e. forming signaling complexes in lipid rafts for example, resulting in its

constitutive activation and signaling.

In previous work (Chapter 2), the CSF1-CSF1R ligand-receptor pair was investigated

within the context of AML-stromal cell interactions. Data from that work showed that hCSF1-

expressing MS-5 cells effectively support the long-term growth and survival of AML cells in co-

culture assays. To investigate whether CSF1’s cytoplasmic PDBM plays a role in this,

OCI/AML-4 and OCI/AML-5 cell were co-cultured with MS-5 Tet-off hCSF1-mem vs. hCSF1-

mem ΔPDBM, and EV control cells. It was observed that MS-5 hCSF1-mem ΔPDBM cells

provided reduced support (approximately 1.5 fold lower) for the OCI/AML-5 and OCI/AML-4

cells in co-culture compared to MS-5 hCSF1-mem cells (Figure 3.5A, B). The reduced support

of AML cells by MS-5 cells expressing PDBM-deleted hCSF1 suggests that the PDBM lends

important functionality and structure to CSF1, as its absence fails in mediating the full

supportive capacity of stromal cells for growth factor/cytokine dependent AML cells. Because

CSF1 can reverse signal within MS-5 stromal cells via its PDBM, this may allow it to interact

with various intracellular proteins involved in structure and adhesion. For example, the

scaffolding protein NHERF1, which contains two PDZ domains, has been shown to mediate the

Page 170: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

160

binding of the cystic fibrosis transmembrane conductance regulator (CFTR) to the actin

cytoskeleton (Li, Callaway, & Bu, 2009). Such mechanisms may increase the stability and

functionality of transmembrane CSF1 within the stromal cell that it is anchored in, which may

enhance its binding to AML cells via CSF1R engagement and/or other adhesive mechanisms.

Taken together, these results demonstrate that CSF1 is capable of reverse signaling

within BM stromal cells to activate a CSF1-HtrA1-S6 axis via PDZ-based interactions (Figure

3.6A). CSF1 ligand appears to be a dynamic functional player in AML-stromal cell interactions

as it may participate in bi-directional, reverse signaling to elicit multi-directional and multi-

faceted effects in the support of AML cells (Figure 3.6B). In this model, CSF1 not only binds to

its receptor on target leukemic cells in the ‘forward’ direction, but it also has the ability to alter

the behaviour of host stromal cells through reverse signaling, inducing the production of

signaling cytokines that could act in both autocrine and juxtacrine manners to influence the

stromal cell, residual normal cells and surrounding leukemic cells. Cell-cell communication

through the exchange of factors and materials between leukemic and stromal cells can help

create a transformed BM niche conducive for leukomogenesis.

Identifying elements and factors within the BM that may be involved in promoting a

malignant microenvironment is critical to enhancing our understanding of BM malignancies like

AML. In this work, a novel CSF1-HtrA1-S6 axis was identified in BM stromal cells that may be

therapeutically targeted to reduce support for subpopulations of CSF1R positive AML cells. This

highlights an important role for ligand signaling within BM stromal cells, which can actively

participate in cell-cell communication within the tumor microenvironment. Targeting these

cellular interactions within the BM niche is thus critical in the design of future therapeutic

approaches for AML.

Page 171: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

161

CHAPTER 4

Thesis discussion and significance of findings

Page 172: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

162

4.1. OVERALL DISCUSSION & SIGNIFICANCE OF FINDINGS

The BM niche is a complex milieu of cells and molecules that establish symbiotic

relationships to help to sustain and propagate, life essential cells. The BM microenvironment is

therefore a dynamic infrastructure susceptible to perturbations and adaptations. In BM disease

settings where leukemic cells often outcompete normal hematopoietic cells for space and

resources, supporting BM cells likely play a pivotal role in the transformative and malignant

process by offering leukemia supportive and normal hematopoietic disruptive forces. The

sequence of events in a ‘microenvironmental’ transformation remains unclear i.e. do malignant

cells usurp and exploit their microenvironments in a parasitic fashion to alter it, or is the

microenvironment first transformed in order to support carcinogenesis and malignancy?

Regardless of the exact sequence of events, it is clear that BM stromal cells lend important

supportive functions for the maintenance and sustenance of both normal hematopoietic and

leukemic cells within the BM.

AML is a highly aggressive malignancy of the BM with dismal long-term clinical

outcomes. While patients may benefit from initial induction chemotherapy, the vast majority of

patients relapse and succumb to their disease. In recent years, there has been growing interest in

investigating mechanisms underlying AML relapse, with focus on the investigation of sub-clones

that may resist or evade chemotherapy altogether, as well as tracing the evolution of leukemias

from pre-leukemia through to the full blown leukemic state. Cells that lie at the heart of relapse

evade initial chemotherapy or, as is becoming more evident, exist as smaller sub-populations of

drug resistant progenitor-like cells that may later evolve and undergo expansion to give rise to

the dominant relapse clone. It is suggested that the tumor microenvironment serves to protect

these types of quiescent evolving leukemic clones and thus targeting BM niches for the treatment

Page 173: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

163

of leukemias is a topic of great research and clinical interest. In this work, I identified a

functional supportive role for the CSF1-CSF1R interaction where the growth and signaling of

AML cells expressing CSF1R were positively supported by CSF1-expressing stromal cells.

Inhibition of this interaction through the use of a CSF1R antibody was shown to have efficacy in

abrogating these supportive effects, leading to reduced AML cell proliferation and mTOR/S6

growth signaling. This highlights a role for the targeting of CSF1-CSF1R as a means to inhibit

signals from BM stromal cells to specifically sever the supportive effects of CSF1-producing

stromal cells.

CSF1R as a Significant Prognostic Indicator in AML

In this thesis, the outcomes of the CSF1-CSF1R interaction, as mediated between

interacting AML and stromal cells, were explored for their effects on both AML and stromal cell

phenotypes and signaling. High level CSF1R expression at the level of RNA and protein was

demonstrated to be a significant prognostic marker in the prediction of patient outcome in

subsets of AML patients, and an important functional receptor that when activated by CSF1

ligand, promoted AML cell proliferation and production of signaling molecules associated with

cell growth and inflammation. CSF1R could also effectively stratify subsets of AML cases that

do not bear the common AML mutations FLT3-ITD and NPM1c, which highlights a potential

role for it in sub-classifying AML. This is promising because CSF1R could serve as a functional,

actionable target in this subgroup of AML patients; this is important as we move from ‘one size

fits all’ therapy, to targeted therapy driven by knowledge of specific mutations and protein/gene

expression.

Page 174: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

164

The Significance of Transmembrane CSF1 in AML Growth and Adhesion

In this thesis I have found that the CSF1-CSF1R interaction is specifically significant

within the context of signaling between AML and stromal cells. I have found that the manner of

ligand presentation is of major importance in the responses of CSF1R-expressing AML cells to

stimulation by CSF1 ligand, with membrane bound ligand being more active than secreted

ligand. This may be due to the enhanced cell-cell contact that transmembrane CSF1 may

promote due to its physical anchorage within stromal cells, which when binding to CSF1R on

AML cells, fosters the cells to be brought into close physical proximity with each other. While

the binding of CSF1 to CSF1R may in itself serve as an adhesive function between the cells,

interacting CSF1-CSF1R may potentially promote the upregulation of adhesion molecules such

as integrins and other components of the extracellular matrix to further enhance adhesion.

The isoform specificity and other specifics associated with the CSF1-CSF1R interaction

underscores the relevance of ‘Baserga’s caveats’, attributed to Dr. Renato Basergo, which were

introduced to me by my supervisor at the onset of this project. These caveats speak to the

importance of the specificity of conditions for any given experimental concept, trial or clinical

treatment. The three main caveats are: 1) These cells or these patients (i.e. cellular, molecular,

clinical characteristics), 2) These conditions or treatment 3) At this time. These caveats highlight

the importance of determining the precise characteristics and context of any experimental design.

This could not hold any more true for CSF1, as it was found to function optimally as a

transmembrane ligand vs. soluble ligand in the specific context of interacting AML and BM

stromal cells. Changing the conditions of the experimental design such as using different cell

types or altering the properties of the signaling ligand (as was done with the PDBM deletion)

would impact the outcome of results, which in essence would highlight the significance of the

Page 175: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

165

initial components and context. In this case, CSF1 was found to operate at its best as a

transmembrane ligand anchored within BM stromal cells.

CSF1 as a Signaling Ligand within BM Stromal Cells: Reverse, Bi-Directional Signaling

While the binding of CSF1 to CSF1R on AML cells is a more canonical ligand-receptor

interaction that mediates signaling and cell-cell contact between AML and stromal cells, the role

of CSF1 as a functional intracellular signaling ligand, and not just a static binding partner

ofCSF1R, was also explored. This work showed that CSF1 has a PDZ binding motif (PDBM) in

its cytoplasmic tail that is capable of modifying downstream signaling proteins, resulting in

reverse intracellular signaling within the stromal cell. A specific CSF1-HtrA1-S6 signaling axis,

dependent on PDZ-mediated interactions between CSF1 and HtrA1, was suggested in MS-5

hCSF1-mem cells, as knockdown of HtrA1 abrogated the effect of CSF1-mem. Activation of this

pathway may underlie the phenotypic effects for hCSF1-expressing MS-5 stromal cells, namely

increased growth and changes in cytokine production. Identification of this pathway also

revealed potential actionable targets such as CSF1-CSF1R, HtrA1 and components of the mTOR

pathway, including S6 within the stroma, and secreted cytokines such as IL-6. Interestingly,

increased phosphorylation of S6 was observed in both AML and stromal cells, indicating that

mTOR signaling is potentially activated in both cell types. This has promising implications for

the dual targeting of both CSF1R+ AML cells and CSF1+ stromal cells given that they share a

common target.

The observed stromal cell changes induced by AML cells may in turn impact the manner

in which they feed back to support AML cells, demonstrating a functionally dynamic role for

Page 176: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

166

supporting BM stromal cells. This lends support to the idea that the stroma is not a static entity in

the BM, but rather that it has active function, and is modifiable.

The interaction between AML and stromal cells yields the dynamic modulation of both

cell types. Co-culture of AML and stromal cells induced changes in signaling and in the cytokine

profiles of both cell types. Production of secondary signaling molecules such as cytokines and

other growth factors by both AML and stromal cells as induced by the CSF1-CSF1R interaction

could function in autocrine and juxtacrine manners on the cells. For example, molecules

produced in response to CSF1R activation by CSF1 on AML cells, could act on stromal cells and

vice versa, adding additional pathways of interaction and support between AML and stromal

cells. In this way, leukemic cells may actively support stromal cells to foster the genesis of its

own supportive microenvironment.

Taken together, this work demonstrates the importance of studying AML in the context

of its BM microenvironment. By employing an AML-stromal cell co-culture system, the role of

CSF1-CSF1R signaling was investigated under more ‘native’ conditions in aims of capturing and

re-capitulating features of the BM as pertaining to the supporting stroma. This helped identify

the CSF1-CSF1R axis as an actionable target that has a dual impact on interacting AML and

stromal cells. Dually targeting AML and supporting stromal cells could be achieved by blocking

the bi-directional support between the cells (CSF1R inhibitor), or by targeting common targets

(mTOR/pS6) in the cells, or the combination of the two. The significance of tumor cell niches as

protective microenvironments has important implications for drug resistance, evasion of immune

responses and other defense mechanisms that potentially give rise to more aggressive, sustained

tumorigenesis. Therapeutic targeting of elements within the microenvironment in BM

Page 177: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

167

malignancies like AML may thus be critical in achieving more favourable clinical responses, and

perhaps even potential disease eradication.

4.2. FUTURE DIRECTIONS

In the continuation of this work, future studies could involve further investigation of

CSF1R as a clinical prognostic marker in AML. While the studies herein identified an

association between CSF1R expression levels and patient survival, further analyses in more gene

expression and protein expression datasets could be employed to verify and lend greater

robustness to the findings. Clinical analyses are often limited by sample numbers and in this

study, there were 146 AML patient cases for which cell surface protein expression profiling, of

CSF1R was performed by flow cytometry. Future work could involve obtaining more patient

samples for CSF1R expression profiling, so as to increase the number of samples in the compiled

protein expression dataset. In addition, for the clinical analyses, more detailed analysis on

disease subtypes, and molecular profiles could be performed, particularly as molecular profiling

is now becoming more mainstream in the clinical diagnosis of AML.

While a CSF1R antibody was employed in this study to block the CSF1-CSF1R

interaction, other identified targets downstream of CSF1R such as the PI3k/Akt pathway, as well

as newly identified targets induced by CSF1 including HtrA1 and S6 could be targeted by

specific inhibitors. Moreover, it appears that S6 is activated in both AML and hCSF1-expressing

stromal cells, making it a common target that could be inhibited with added advantage, as the

pathway would be blocked both in the AML cell, and the stroma cells that contribute to the

growth and survival of the AML cells. While HtrA1 was identified as a downstream target of

CSF1 in MS-5 stromal cells, other binding partners of CSF1 could be identified using a CSF1-

Page 178: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

168

specific yeast-two-hybrid screen that incorporates the PDBM, or through BioID approaches or

pull down-mass spectometry studies; in any case it will be critical to maintain the last 5 amino

acids of CSF1.

In addition, immune-based therapies such as CAR (chimeric antigen receptor)-T cell

therapy could be employed where cells could be engineered to express CSF1 ligand to target

CSF1R positive AML cells; such a CAR-T would not only eliminate CSF1R expressing AML

cells but also infiltrating macrophage derived from residual cells and that may be providing an

immune suppressive effect (M2-macrophage). At the same time, CAR-T cells could also be

made to express the domain of CSF1R that binds CSF1. These T cells would be targeted to

CSF1-expressing stromal cells within the BM to target cases of CSF1-driven disease. This could

help prevent the adhesion and homing of CSF1R positive AML cells to CSF1-expressing BM

stromal cells, thereby severing supportive interactions between the cells. This could also help

mobilize CSF1R positive AML cells from the bone marrow, making them more accessible to be

targeted by chemotherapeutic agents. Finally the elimination of CSF1-expressing stroma would

reduce the production of cytokines and adhesion molecules that can contribute to the growth of

the AML cells.

While levels of CSF1R were assessed on the peripheral blood blasts of AML patients,

future work should also involve examining CSF1 levels within the BM cells of AML patients.

This work is actually being currently carried out with the assistance of collaborators. For this

work, bone marrow biopsies are being obtained which will be stained with CSF1 antibodies, as

well as antibodies against pS6, to evaluate CSF1 levels and downstream activation of mTOR

signaling inBM stromal cells. This would lend further support and clinical relevance to the

findings in this work. The potential of finding that there is variable expression of CSF1 in the

Page 179: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

169

stroma of AML cases would provide the impetus to identify the factors that enhance the

production of CSF1, and the identification of means to mitigate that expression.

Finally, more in-vivo studies should be performed including engraftment studies with

more patient samples in the humanized CSF1 mouse model. The CSF1R antibody should also be

tested in these mice to evaluate whether it can block engraftment of CSF1R positive AML cells

within the BM. Engrafted AML cells should be interrogated for cell surface markers as well as

intracellular signaling markers; these latter studies could be performed using CyTOF technology

for multiplexed ‘discovery’ screening of novel and/or relevant markers. Much of this work is

currently underway and will be continued given that the preliminary in-vivo findings from the

humanized CSF1 mouse model have proven to be quite promising.

4.3. CONCLUSIONS

The work herein has identified significant roles for CSF1 and its receptor CSF1R in AML

clinical prognosis, and in the AML BM niche. Signaling between CSF1 and CSF1R was shown

to foster supportive connections between the stromal and AML cells expressing them,

respectively. The CSF1-CSF1R-dependent interaction between AML and BM stromal cells

improved the proliferation, survival and adhesion of AML cells, through changes in signaling

and cell surface immunophenotypes, in in-vitro and in-vivo models. The CSF1-CSF1R

interaction was not only supportive of AML cells, but also drove signaling changes within MS-5

stromal cells through the bi-directional, reverse signaling action of CSF1 ligand. CSF1 could

signal within the stromal cell via its cytoplasmic PDBM to activate mTOR signaling, and induce

changes in cytokine secretion. CSF1-CSF1R may promote cellular cross-talk between AML and

supporting stromal cells through an interplay of multi-directional forward/reverse signaling,

Page 180: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

170

producing a symbiotic relationship that propagates and sustains leukemogenesis. A CSF1R-

driven AML is proposed to be an aggressive disease subset, supported by the fact that high

CSF1R levels were shown to be associated with poor clinical survival. Taken together, CSF1-

CSF1R is a biologically and clinically relevant signaling axis that may be therapeutically

targeted in AML and its supportive microenvironment.

Page 181: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

171

REFERENCES

Abaza, Y., Kantarjian, H., Garcia-Manero, G., Estey, E., Borthakur, G., Jabbour, E., …

Ravandil, F. (2017). Long-term outcome of acute promyelocytic leukemia treated with all-

trans-retinoic acid, arsenic trioxide, and gemtuzumab. Blood. https://doi.org/10.1182/blood-

2016-09-736686

Akiyama, H., Nishimura, T., Kondo, H., Ikeda, K., Hayashi, Y., & McGeer, P. L. (1994).

Expression of the receptor for macrophage colony stimulating factor by brain microglia and

its upregulation in brains of patients with Alzheimer’s disease and amyotrophic lateral

sclerosis. Brain Research. https://doi.org/10.1016/0006-8993(94)91779-5

Al-Aamri, H. M., Ku, H., Irving, H. R., Tucci, J., Meehan-Andrews, T., & Bradley, C. (2019).

Time dependent response of daunorubicin on cytotoxicity, cell cycle and DNA repair in

acute lymphoblastic leukaemia. BMC Cancer. https://doi.org/10.1186/s12885-019-5377-y

Arai, F., Hirao, A., Ohmura, M., Sato, H., Matsuoka, S., Takubo, K., … Suda, T. (2004).

Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone

marrow niche. Cell, 118, 149–161. https://doi.org/10.1016/j.cell.2004.07.004

Arber, D. A. (2017). Acute Myeloid Leukemia. In Hematopathology: A Volume in the Series:

Foundations in Diagnostic Pathology. https://doi.org/10.1016/B978-0-323-47913-4.00014-

8

Arber, D. A., Orazi, A., Hasserjian, R., Thiele, J., Borowitz, M. J., Le Beau, M. M., …

Vardiman, J. W. (2016). The 2016 revision to the World Health Organization classification

of myeloid neoplasms and acute leukemia. Blood. https://doi.org/10.1182/blood-2016-03-

643544

Armitage, J. O. (1998). Emerging applications of recombinant human granulocyte-macrophage

colony-stimulating factor. BLOOD, 92, 4491–4508.

Ashman, L. K., & Griffith, R. (2013). Therapeutic targeting of c-KIT in cancer. Expert Opinion

on Investigational Drugs. https://doi.org/10.1517/13543784.2013.740010

Auffray, C., Sieweke, M. H., & Geissmann, F. (2009). Blood Monocytes: Development,

Heterogeneity, and Relationship with Dendritic Cells. Annual Review of Immunology.

https://doi.org/10.1146/annurev.immunol.021908.132557

Baldi, A., De Luca, A., Morini, M., Battista, T., Felsani, A., Baldi, F., … Paggi, M. G. (2002).

The HtrA1 serine protease is down-regulated during human melanoma progression and

represses growth of metastatic melanoma cells. Oncogene.

https://doi.org/10.1038/sj.onc.1205911

Baldus, C. D., & Bullinger, L. (2008). Gene Expression With Prognostic Implications in

Cytogenetically Normal Acute Myeloid Leukemia. Seminars in Oncology.

https://doi.org/10.1053/j.seminoncol.2008.04.006

Barton, B. E. (2005). Interleukin-6 and new strategies for the treatment of cancer,

hyperproliferative diseases and paraneoplastic syndromes. Expert Opinion on Therapeutic

Targets, 9, 737–752. https://doi.org/10.1517/14728222.9.4.737

Batinić, D., Tindle, R., Boban, D., Tiefenbach, A., Rajić, L., Labar, B., … Boranić, M. (1989).

Expression of haematopoietic progenitor cell-associated antigen BI-3C5/CD34 in

leukaemia. Leukemia Research, 13, 83–85. https://doi.org/10.1016/0145-2126(89)90035-0

Baud’Huin, M., Renault, R., Charrier, C., Riet, A., Moreau, A., Brion, R., … Heymann, D.

(2010). Interleukin-34 is expressed by giant cell tumours of bone and plays a key role in

RANKL-induced osteoclastogenesis. Journal of Pathology.

Page 182: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

172

https://doi.org/10.1002/path.2684

Becker, A. J., McCulloch, E. A., & Till, J. E. (1963). Cytological demonstration of the clonal

nature of spleen colonies derived from transplanted mouse marrow cells. Nature, 197, 452–

454. https://doi.org/10.1038/197452a0

Begg, S. K., Radley, J. M., Pollard, J. W., Chisholm, O. T., Stanley, E. R., & Bertoncello, I.

(1993). Delayed hematopoietic development in osteopetrotic (op/op) mice. The Journal of

Experimental Medicine.

Bellone, G., Smirne, C., Mauri, F. A., Tonel, E., Carbone, A., Buffolino, A., … Emanuelli, G.

(2006). Cytokine expression profile in human pancreatic carcinoma cells and in surgical

specimens: implications for survival. Cancer Immunology, Immunotherapy : CII, 55, 684–

698. https://doi.org/10.1007/s00262-005-0047-0

Bendall, L. J., & Bradstock, K. F. (2014). G-CSF: From granulopoietic stimulant to bone marrow

stem cell mobilizing agent. Cytokine and Growth Factor Reviews.

https://doi.org/10.1016/j.cytogfr.2014.07.011

Benekli, M., Baer, M. R., Baumann, H., & Wetzler, M. (2003). Signal transducer and activator of

transcription proteins in leukemias. Blood. https://doi.org/10.1182/blood-2002-04-1204

Bennett, J. M., Catovsky, D., Daniel, M. ‐T, Flandrin, G., Galton, D. A. G., Gralnick, H. R., &

Sultan, C. (1976). Proposals for the Classification of the Acute Leukaemias

French‐American‐British (FAB) Co‐operative Group. British Journal of Haematology, 33,

451–458. https://doi.org/10.1111/j.1365-2141.1976.tb03563.x

Bernad, A., Kopf, M., Kulbacki, R., Weich, N., Koehler, G., & Gutierrez-Ramos, J. C. (1994).

Interleukin-6 is required in vivo for the regulation of stem cells and committed progenitors

of the hematopoietic system. Immunity, 1, 725–731. https://doi.org/10.1016/S1074-

7613(94)80014-6

Berthier, R., Prandini, M. H., Schweitzer, A., Thevenon, D., Martin-Sisteron, H., & Uzan, G.

(1997). The MS-5 murine stromal cell line and hematopoietic growth factors synergize to

support the megakaryocytic differentiation of embryonic stem cells. Exp Hematol, 25(6),

481–490.

Betz, B. L., & Hess, J. L. (2010a). Acute myeloid leukemia diagnosis in the 21st century.

Archives of Pathology & Laboratory Medicine, 134, 1427–1433.

https://doi.org/10.1043/2010-0245-RA.1

Betz, B. L., & Hess, J. L. (2010b). Acute myeloid leukemia diagnosis in the 21st century.

Archives of Pathology and Laboratory Medicine.

Birg, F, Rosnet, O., Carbuccia, N., & Birnbaum, D. (1994). The expression of FMS, KIT and

FLT3 in hematopoietic malignancies. Leuk.Lymphoma, 13, 223–227.

https://doi.org/10.3109/10428199409056285

Birg, Françoise, Carbuccia, N., Rosnet, O., & Birnbaumt, D. (1994). The expression of FMS,

KIT and FLT3 in hematopoietic malignancies. Leukemia and Lymphoma.

https://doi.org/10.3109/10428199409056285

Bochenek, M. L., Dickinson, S., Astin, J. W., Adams, R. H., & Nobes, C. D. (2010). Ephrin-B2

regulates endothelial cell morphology and motility independently of Eph-receptor binding.

Journal of Cell Science. https://doi.org/10.1242/jcs.061903

Bonnet, D., & Dick, J. E. (1997). Human acute myeloid leukemia is organized as a hierarchy that

originates from a primitive hematopoietic cell. Nature Medicine, 3, 730–737.

https://doi.org/10.1038/nm0797-730

Botstein, D., Chang, H. Y., Brown, P. O., van de Rijn, M., Dudoit, S., Bondre, C., & Chi, J.-T.

Page 183: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

173

(2002). Diversity, topographic differentiation, and positional memory in human fibroblasts.

Proceedings of the National Academy of Sciences. https://doi.org/10.1073/pnas.162488599

Bowman, R. L., Busque, L., & Levine, R. L. (2018). Clonal Hematopoiesis and Evolution to

Hematopoietic Malignancies. Cell Stem Cell, 22, 157–170.

https://doi.org/10.1016/j.stem.2018.01.011

Boyd, A. L., Campbell, C. J. V., Hopkins, C. I., Fiebig-Comyn, A., Russell, J., Ulemek, J., …

Bhatia, M. (2014). Niche displacement of human leukemic stem cells uniquely allows their

competitive replacement with healthy HSPCs. The Journal of Experimental Medicine, 211,

1925–1935. https://doi.org/10.1084/jem.20140131

Brasier, A. R. (2010). The nuclear factor-B-interleukin-6 signalling pathway mediating vascular

inflammation. Cardiovascular Research. https://doi.org/10.1093/cvr/cvq076

Breedveld, A., Groot Kormelink, T., van Egmond, M., & de Jong, E. C. (2017). Granulocytes as

modulators of dendritic cell function. Journal of Leukocyte Biology.

https://doi.org/10.1189/jlb.4mr0217-048rr

Broxmeyer, H. E., Orschell, C. M., Clapp, D. W., Hangoc, G., Cooper, S., Plett, P. A., … Srour,

E. F. (2005). Rapid mobilization of murine and human hematopoietic stem and progenitor

cells with AMD3100, a CXCR4 antagonist. The Journal of Experimental Medicine, 201,

1307–1318. https://doi.org/10.1084/jem.20041385

Buick, R. N., Minden, M. D., & McCulloch, E. A. (1979). Self-renewal in culture of proliferative

blast progenitor cells in acute myeloblastic leukemia. Blood.

Bukowski, R. M., Budd, G. T., Gibbons, J. A., Bauer, R. J., Childs, A., Antal, J., … Thomassen,

M. J. (1994). Phase I trial of subcutaneous recombinant macrophage colony-stimulating

factor: Clinical and immunomodulatory effects. Journal of Clinical Oncology.

https://doi.org/10.1200/JCO.1994.12.1.97

Burgler, S. (2015). Role of CD38 Expression in Diagnosis and Pathogenesis of Chronic

Lymphocytic Leukemia and Its Potential as Therapeutic Target. Critical Reviews in

Immunology, 35, 417–432. https://doi.org/10.1615/CritRevImmunol.v35.i5.50

Calvi, L. M., Adams, G. B., Weibrecht, K. W., Weber, J. M., Olson, D. P., Knight, M. C., …

Scadden, D. T. (2003). Osteoblastic cells regulate the haematopoietic stem cell niche.

Nature. https://doi.org/10.1038/nature02040

Campioni, M., Severino, A., Manente, L., Tuduce, I. L., Toldo, S., Caraglia, M., … Baldi, A.

(2010). The Serine Protease HtrA1 Specifically Interacts and Degrades the Tuberous

Sclerosis Complex 2 Protein. Molecular Cancer Research. https://doi.org/10.1158/1541-

7786.mcr-09-0473

Cancer, T., & Atlas, G. (2013). Genomic and Epigenomic Landscapes of Adult De Novo Acute

Myeloid Leukemia The Cancer Genome Atlas Research Network. The New England

Journal of Medicine. https://doi.org/10.1056/NEJMoa1301689

Cantley, L. C. (2002). The phosphoinositide 3-kinase pathway. Science.

https://doi.org/10.1126/science.296.5573.1655

Carling, D. (2017). AMPK signalling in health and disease. Current Opinion in Cell Biology.

https://doi.org/10.1016/j.ceb.2017.01.005

Carow, C. E., Hangoc, G., & Broxmeyer, H. E. (1993). Human Multipotential Progenitor Cells

(CFU-GEMM) Have Extensive Replating Capacity for Secondary CFU-GEMM: An Effect

Enhanced by Cord Blood Plasma. BLOOD.

Casteran, N., Beslu, N., Lecocq, E., Gomez, S., & Dubreuil, P. (1998). Specific and common

activities of the FLT3 and KIT tyrosine kinase receptors revealed by the use of cultured

Page 184: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

174

mast cells. Leukemia. https://doi.org/10.1038/sj.leu.2401057

Cecchini, M. G., Dominguez, M. G., Mocci, S., Wetterwald, A., Felix, R., Fleisch, H., …

Stanley, E. R. (1994). Role of colony stimulating factor-1 in the establishment and

regulation of tissue macrophages during postnatal development of the mouse. Development

(Cambridge, England).

Chan, C. K. F., Lindau, P., Jiang, W., Chen, J. Y., Zhang, L. F., Chen, C.-C., … Weissman, I. L.

(2013). Clonal precursor of bone, cartilage, and hematopoietic niche stromal cells.

Proceedings of the National Academy of Sciences, 110, 12643–12648.

https://doi.org/10.1073/pnas.1310212110

Chen, X., Liu, H., Focia, P. J., Shim, A. H.-R., & He, X. (2008). Structure of macrophage colony

stimulating factor bound to FMS: Diverse signaling assemblies of class III receptor tyrosine

kinases. Proceedings of the National Academy of Sciences.

https://doi.org/10.1073/pnas.0807762105

Cheson, B. D., Bennett, J. M., Kopecky, K. J., Büchner, T., Willman, C. L., Estey, E. H., … and

Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia, I. W. G. for D. S.

of R. C. T. O. (2003). Revised recommendations of the International Working Group for

Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting

Standards for Therapeutic Trials in Acute Myeloid Leukemia. Journal of Clinical

Oncology : Official Journal of the American Society of Clinical Oncology, 21, 4642–4649.

https://doi.org/10.1200/JCO.2003.04.036

Chien, J., Staub, J., Hu, S. I., Erickson-Johnson, M. R., Couch, F. J., Smith, D. I., … Shridhar, V.

(2004). A candidate tumor suppressor HtrA1 is downregulated in ovarian cancer. Oncogene.

https://doi.org/10.1038/sj.onc.1207271

Choudhary, C., Brandts, C., Schwable, J., Tickenbrock, L., Sargin, B., Ueker, A., … Serve, H.

(2007). Activation mechanisms of STAT5 by oncogenic Flt3-ITD. Blood, 110, 370–374.

https://doi.org/10.1182/blood-2006-05-024018

Choudhary, C., Schwäble, J., Brandts, C., Tickenbrock, L., Sargin, B., Kindler, T., … Serve, H.

(2005). AML-associated Flt3 kinase domain mutations show signal transduction differences

compared with Flt3 ITD mutations. Blood, 106, 265–273. https://doi.org/10.1182/blood-

2004-07-2942

Civin, C. I., Strauss, L. C., Brovall, C., Fackler, M. J., Schwartz, J. F., & Shaper, J. H. (1984).

Antigenic analysis of hematopoiesis. III. A hematopoietic progenitor cell surface antigen

defined by a monoclonal antibody raised against KG-1a cells. Journal of Immunology, 133,

157–165.

Clausen, T., Kaiser, M., Huber, R., & Ehrmann, M. (2011). HTRA proteases: Regulated

proteolysis in protein quality control. Nature Reviews Molecular Cell Biology.

https://doi.org/10.1038/nrm3065

Clausen, T., Southan, C., & Ehrmann, M. (2002a). The HtrA family of proteases: implications

for protein composition and cell fate. Molecular Cell.

Clausen, T., Southan, C., & Ehrmann, M. (2002b). The HtrA family of proteases: Implications

for protein composition and cell fate. Molecular Cell. https://doi.org/10.1016/S1097-

2765(02)00658-5

Clawson, G. A., Bui, V., Xin, P., Wang, N., & Pan, W. (2008). Intracellular localization of the

tumor suppressor HtrA1/Prss11 and its association with HPV16 E6 and E7 proteins.

Journal of Cellular Biochemistry. https://doi.org/10.1002/jcb.21804

Coa, L. L., Abreu, T. F., Tashima, A. K., Green, J., Pascon, R. C., Vallim, M. A., & Machado-Jr,

Page 185: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

175

J. (2019). AKT/protein kinase B associates with β-actin in the nucleus of melanoma cells.

Bioscience Reports. https://doi.org/10.1042/bsr20181312

Colmone, A., Amorim, M., Pontier, A. L., Wang, S., Jablonski, E., & Sipkins, D. A. (2008).

Leukemic cells create bone marrow niches that disrupt the behavior of normal

hematopoietic progenitor cells. Science, 322, 1861–1865.

https://doi.org/10.1126/science.1164390

Costa, M. J., Kudaravalli, J., Liu, W. H., Stock, J., Kong, S., & Liu, S. H. (2018). A mouse

model for evaluation of efficacy and concomitant toxicity of anti-human CXCR4

therapeutics. PLoS ONE. https://doi.org/10.1371/journal.pone.0194688

Coussens, L. M., & Pollard, J. W. (2011). Leukocytes in mammary development and cancer.

Cold Spring Harbor Perspectives in Biology. https://doi.org/10.1101/cshperspect.a003285

Dale, D. C. (2006). Colony-Stimulating Factors for the Management of Neutropenia in Cancer

Patients. Drugs. https://doi.org/10.2165/00003495-200262001-00001

Dan, H. C., Cooper, M. J., Cogswell, P. C., Duncan, J. A., Ting, J. P. Y., & Baldwin, A. S.

(2008). Akt-dependent regulation of NF-κB is controlled by mTOR and Raptor in

association with IKK. Genes and Development. https://doi.org/10.1101/gad.1662308

Daniels, D. L., Cohen, A. R., Anderson, J. M., & Brünger, A. T. (1998). Crystal structure of the

hCASK PDZ domain reveals the structural basis of class II PDZ domain target recognition.

Nature Structural Biology. https://doi.org/10.1038/nsb0498-317

Daver, N., Schlenk, R. F., Russell, N. H., & Levis, M. J. (2019). Targeting FLT3 mutations in

AML: review of current knowledge and evidence. Leukemia.

https://doi.org/10.1038/s41375-018-0357-9

De, I., Steffen, M. D., Clark, P. A., Patros, C. J., Sokn, E., Bishop, S. M., … Collier, L. S.

(2016). CSF1 overexpression promotes high-grade glioma formation without impacting the

polarization status of glioma-associated microglia and macrophages. Cancer Research, 76,

2552–2560. https://doi.org/10.1158/0008-5472.CAN-15-2386

Delaunay, J., Vey, N., Leblanc, T., Fenaux, P., Rigal-Huguet, F., Witz, F., … Dombret, H.

(2003). Prognosis of inv(16)/t(16;16) acute myeloid leukemia (AML): A survey of 110

cases from the French AML intergroup. Blood. https://doi.org/10.1182/blood-2002-11-3527

Dexter, T. M., Allen, T. D., & Lajtha, L. G. (1977). Conditions controlling the proliferation of

haemopoietic stem cells in vitro. Journal of Cellular Physiology.

https://doi.org/10.1002/jcp.1040910303

Ding, L., & Morrison, S. J. (2013). Haematopoietic stem cells and early lymphoid progenitors

occupy distinct bone marrow niches. Nature, 495, 231–235.

https://doi.org/10.1038/nature11885

Döhner, H. (2007). Implication of the molecular characterization of acute myeloid leukemia.

Hematology / the Education Program of the American Society of Hematology. American

Society of Hematology. Education Program. https://doi.org/10.1182/asheducation-

2007.1.412

Douglass, T. G., Driggers, L., Zhang, J. G., Hoa, N., Delgado, C., Williams, C. C., … Jadus, M.

R. (2008). Macrophage colony stimulating factor: Not just for macrophages anymore! A

gateway into complex biologies. International Immunopharmacology.

https://doi.org/10.1016/j.intimp.2008.04.016

Doyle, D. A., Lee, A., Lewis, J., Kim, E., Sheng, M., & MacKinnon, R. (1996). Crystal

structures of a complexed and peptide-free membrane protein- binding domain: Molecular

basis of peptide recognition by PDZ. Cell. https://doi.org/10.1016/S0092-8674(00)81307-0

Page 186: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

176

Dufour, A., Schneider, F., Metzeler, K. H., Hoster, E., Schneider, S., Zellmeier, E., …

Spiekermann, K. (2010). Acute myeloid leukemia with biallelic CEBPA gene mutations and

normal karyotype represents a distinct genetic entity associated with a favorable clinical

outcome. Journal of Clinical Oncology. https://doi.org/10.1200/JCO.2008.21.6010

Edginton-White, B., Cauchy, P., Assi, S. A., Hartmann, S., Riggs, A. G., Mathas, S., … Bonifer,

C. (2018). Global long terminal repeat activation participates in establishing the unique

gene expression programme of classical Hodgkin lymphoma. Leukemia.

https://doi.org/10.1038/s41375-018-0311-x

Edling, C. E., & Hallberg, B. (2007). c-Kit-A hematopoietic cell essential receptor tyrosine

kinase. International Journal of Biochemistry and Cell Biology.

https://doi.org/10.1016/j.biocel.2006.12.005

Edwards, D. K., Watanabe-Smith, K., Rofelty, A., Damnernsawad, A., Laderas, T., Lamble, A.,

… Tyner, J. W. (2019). CSF1R inhibitors exhibit antitumor activity in acute myeloid

leukemia by blocking paracrine signals from support cells. Blood.

https://doi.org/10.1182/blood-2018-03-838946

Emery, P., Keystone, E., Tony, H. P., Cantagrel, A., Van Vollenhoven, R., Sanchez, A., …

Kremer, J. (2008). IL-6 receptor inhibition with tocilizumab improves treatment outcomes

in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals:

Results from a 24-week multicentre randomised placebo-controlled trial. Annals of the

Rheumatic Diseases, 67, 1516–1523. https://doi.org/10.1136/ard.2008.092932

Espinosa, I., Beck, A. H., Lee, C. H., Zhu, S., Montgomery, K. D., Marinelli, R. J., … Van De

Rijn, M. (2009). Coordinate expression of colony-stimulating factor-1 and colony-

stimulating factor-1-related proteins is associated with poor prognosis in gynecological and

nongynecological leiomyosarcoma. American Journal of Pathology.

https://doi.org/10.2353/ajpath.2009.081037

Faderl, S., Harris, D., Van, Q., Kantarjian, H. M., Talpaz, M., & Estrov, Z. (2003). Granulocyte-

macrophage colony-stimulating factor (GM-CSF) induces antiapoptotic and proapoptotic

signals in acute myeloid leukemia. Blood. https://doi.org/10.1182/blood-2002-06-1890

Falini, B., Bolli, N., Liso, A., Martelli, M. P., Mannucci, R., Pileri, S., & Nicoletti, I. (2009).

Altered nucleophosmin transport in acute myeloid leukaemia with mutated NPM1:

Molecular basis and clinical implications. Leukemia. https://doi.org/10.1038/leu.2009.124

Felfly, H., & Haddad, G. G. (2014). Hematopoietic stem cells: potential new applications for

translational medicine. Journal of Stem Cells. https://doi.org/jsc.2014.9.3.163

Felix, J., De Munck, S., Verstraete, K., Meuris, L., Callewaert, N., Elegheert, J., & Savvides, S.

N. (2015). Structure and Assembly Mechanism of the Signaling Complex Mediated by

Human CSF-1. Structure. https://doi.org/10.1016/j.str.2015.06.019

Fernández-Larrea, J., Merlos-Suárez, A., Ureña, J. M., Baselga, J., & Arribas, J. (1999). A role

for a PDZ protein in the early secretory pathway for the targeting of proTGF-α to the cell

surface. Molecular Cell. https://doi.org/10.1016/S1097-2765(00)80470-0

Ferrara, F., & Schiffer, C. A. (2013a). Acute myeloid leukaemia in adults. In The Lancet.

https://doi.org/10.1016/S0140-6736(12)61727-9

Ferrara, F., & Schiffer, C. A. (2013b). Acute myeloid leukaemia in adults. In The Lancet (Vol.

381, pp. 484–495). https://doi.org/10.1016/S0140-6736(12)61727-9

Flanagan, J. G., Chan, D. C., & Leder, P. (1991). Transmembrane form of the kit ligand growth

factor is determined by alternative splicing and is missing in the SId mutant. Cell.

https://doi.org/10.1016/0092-8674(91)90326-T

Page 187: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

177

Fröhling, S., Scholl, C., Gilliland, D. G., & Levine, R. L. (2005). Genetics of myeloid

malignancies: Pathogenetic and clinical implications. Journal of Clinical Oncology.

https://doi.org/10.1200/JCO.2005.05.010

Gado, K., Domjan, G., Hegyesi, H., & Falus, A. (2000). Role of INTERLEUKIN-6 in the

pathogenesis of multiple myeloma. Cell Biol Int, 24, 195–209.

https://doi.org/10.1006/cbir.2000.0497

Galán-Díez, M., & Kousteni, S. (2018). A bone marrow niche-derived molecular switch between

osteogenesis and hematopoiesis. Genes and Development.

https://doi.org/10.1101/gad.314013.118

Gale, R. E., Green, C., Allen, C., Mead, A. J., Burnett, A. K., Hills, R. K., & Linch, D. C. (2008).

The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction

with NPM1 mutations in a large cohort of young adult patients with acute myeloid

leukemia. Blood. https://doi.org/10.1182/blood-2007-08-109090

Garceau, V., Smith, J., Paton, I. R., Davey, M., Fares, M. A., Sester, D. P., … Hume, D. A.

(2010). Pivotal Advance: Avian colony-stimulating factor 1 ( CSF-1 ), interleukin-34 ( IL-

34 ), and CSF-1 receptor genes and gene products . Journal of Leukocyte Biology.

https://doi.org/10.1189/jlb.0909624

Gartner, S., & Kaplan, H. S. (1980a). Long-term culture of human bone marrow cells.

Proceedings of the National Academy of Sciences. https://doi.org/10.1073/pnas.77.8.4756

Gartner, S., & Kaplan, H. S. (1980b). Long-term culture of human bone marrow cells.

Proceedings of the National Academy of Sciences of the United States of America, 77,

4756–4759. https://doi.org/10.1073/pnas.77.8.4756

Gary Gilliland, D., & Griffin, J. D. (2002). The roles of FLT3 in hematopoiesis and leukemia.

Blood. https://doi.org/10.1182/blood-2002-02-0492

Gasson, J. C. (1991). Molecular Physiology of Granulocyte-Macrophage Colony-Stimulating

Factor. Blood, 77, 1131–1146.

Geissler, E. N., Liao, M., Brook, J. D., Martin, F. H., Zsebo, K. M., Housman, D. E., & Galli, S.

J. (1991). Stem cell factor (SCF), a novel hematopoietic growth factor and ligand for c-kit

tyrosine kinase receptor, maps on human chromosome 12 between 12q14.3 and 12qter.

Somatic Cell and Molecular Genetics, 17, 207–214. https://doi.org/10.1007/BF01232978

Gimble, J. M., Robinson, C. E., Wu, X., & Kelly, K. A. (1996). The function of adipocytes in the

bone marrow stroma: An update. Bone, 19, 421–428. https://doi.org/10.1016/S8756-

3282(96)00258-X

Goker, H., Haznedaroglu, I. C., & Chao, N. J. (2001). Acute graft-vs-host disease: Pathobiology

and management. Experimental Hematology. https://doi.org/10.1016/S0301-

472X(00)00677-9

Gommerman, J L, Sittaro, D., Klebasz, N. Z., Williams, D. a, & Berger, S. a. (2000). Differential

stimulation of c-Kit mutants by membrane-bound and soluble Steel Factor correlates with

leukemic potential. Blood, 96, 3734–3742.

Gommerman, Jennifer L, Sittaro, D., Klebasz, N. Z., Williams, D. A., & Berger, S. A. (2000).

Differential stimulation of c-Kit mutants by membrane-bound and soluble Steel Factor

correlates with leukemic potential. Blood. https://doi.org/10.3719/weed.49.21

Gordon, S., & Martinez, F. O. (2010). Alternative activation of macrophages: mechanism and

functions. Immunity. https://doi.org/10.1016/j.immuni.2010.05.007

Greaves, M., & Maley, C. C. (2012). Clonal evolution in cancer. Nature.

https://doi.org/10.1038/nature10762

Page 188: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

178

Greiner, D. L., Shultz, L. D., Yates, J., Appel, M. C., Perdrizet, G., Hesselton, R. M., … et al.

(1995). Improved engraftment of human spleen cells in NOD/LtSz-scid/scid mice as

compared with C.B-17-scid/scid mice. Am J Pathol, 146, 888–902.

Guezguez, B., Campbell, C. J. V., Boyd, A. L., Karanu, F., Casado, F. L., Di Cresce, C., …

Bhatia, M. (2013). Regional localization within the bone marrow influences the functional

capacity of human HSCs. Cell Stem Cell, 13, 175–189.

https://doi.org/10.1016/j.stem.2013.06.015

Guillonneau, C., Bézie, S., & Anegon, I. (2017). Immunoregulatory properties of the cytokine

IL-34. Cellular and Molecular Life Sciences. https://doi.org/10.1007/s00018-017-2482-4

Hamilton, J. A., & Achuthan, A. (2013). Colony stimulating factors and myeloid cell biology in

health and disease. Trends in Immunology. https://doi.org/10.1016/j.it.2012.08.006

Harada, A., Sekido, N., Akahoshi, T., Wada, T., Mukaida, N., & Matsushima, K. (1994).

Essential involvement of interleukin-8 (IL-8) in acute inflammation. In Journal of

Leukocyte Biology. https://doi.org/10.1002/jlb.56.5.559

Harmer, D., Falank, C., & Reagan, M. R. (2019). Interleukin-6 interweaves the bone marrow

microenvironment, bone loss, and multiple myeloma. Frontiers in Endocrinology.

https://doi.org/10.3389/fendo.2018.00788

Harris, B. Z., & Lim, W. A. (2001). Mechanism and role of PDZ domains in signaling complex

assembly. Journal of Cell Science.

Hegde, R., Srinivasula, S. M., Zhang, Z., Wassell, R., Mukattash, R., Cilenti, L., … Alnemri, E.

S. (2002). Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that

disrupts inhibitor of apoptosis protein-caspase interaction. Journal of Biological Chemistry.

https://doi.org/10.1074/jbc.M109721200

HEINRICH, P. C., BEHRMANN, I., MÜLLER-NEWEN, G., SCHAPER, F., & GRAEVE, L.

(1998). Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway.

Biochemical Journal, 334, 297–314. https://doi.org/10.1042/bj3340297

Hemmings, B. A., & Restuccia, D. F. (2012). PI3K-PKB/Akt pathway. Cold Spring Harbor

Perspectives in Biology. https://doi.org/10.1101/cshperspect.a011189

Heuser, M., Ganser, A., & Bokemeyer, C. (2007). Use of Colony-Stimulating Factors for

Chemotherapy-Associated Neutropenia: Review of Current Guidelines. Seminars in

Hematology. https://doi.org/10.1053/j.seminhematol.2007.04.002

Hickey, M. J., & Kubes, P. (2009). Intravascular immunity: The host-pathogen encounter in

blood vessels. Nature Reviews Immunology. https://doi.org/10.1038/nri2532

Hoffman, R., Benz, E. J., Silberstein, L. E., Heslop, H. E., Weitz, J. I., Anastasi, J., … Abutalib,

S. (2017). Hematology: Basic Principles and Practice. Hematology: Basic Principles and

Practice. https://doi.org/10.1016/C2013-0-23355-9

Holland, S. J., Gale, N. W., Mbamalu, G., Yancopoulos, G. D., Henkemeyer, M., & Pawson, T.

(1996). Bidirectional signalling through the EPH-family receptor Nuk and its

transmembrane ligands. Nature. https://doi.org/10.1038/383722a0

Hope, K. J., Jin, L., & Dick, J. E. (2004). Acute myeloid leukemia originates from a hierarchy of

leukemic stem cell classes that differ in self-renewal capacity. Nature Immunology, 5, 738–

743. https://doi.org/10.1038/ni1080

Horiuchi, K., Miyamoto, T., Takaishi, H., Hakozaki, A., Kosaki, N., Miyauchi, Y., … Toyama,

Y. (2007). Cell surface colony-stimulating factor 1 can be cleaved by TNF-alpha converting

enzyme or endocytosed in a clathrin-dependent manner. J Immunol, 179(10), 6715–6724.

https://doi.org/179/10/6715 [pii]

Page 189: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

179

Horiuchi, K., Miyamoto, T., Takaishi, H., Hakozaki, A., Kosaki, N., Miyauchi, Y., … Toyama,

Y. (2014). Cell Surface Colony-Stimulating Factor 1 Can Be Cleaved by TNF- Converting

Enzyme or Endocytosed in a Clathrin-Dependent Manner. The Journal of Immunology.

https://doi.org/10.4049/jimmunol.179.10.6715

Hume, D. A., & MacDonald, K. P. A. (2012). Therapeutic applications of macrophage colony-

stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood.

https://doi.org/10.1182/blood-2011-09-379214

Huynh, J., Kwa, M. Q., Cook, A. D., Hamilton, J. A., & Scholz, G. M. (2012). CSF-1 receptor

signalling from endosomes mediates the sustained activation of Erk1/2 and Akt in

macrophages. Cellular Signalling. https://doi.org/10.1016/j.cellsig.2012.04.022

Ide, H., Seligson, D. B., Memarzadeh, S., Xin, L., Horvath, S., Dubey, P., … Witte, O. N.

(2002). Expression of colony-stimulating factor 1 receptor during prostate development and

prostate cancer progression. Proceedings of the National Academy of Sciences.

https://doi.org/10.1073/pnas.222537099

Ikebuchi, K., Wong, G. G., Clark, S. C., Ihle, J. N., Hirai, Y., & Ogawa, M. (1987). Interleukin 6

enhancement of interleukin 3-dependent proliferation of multipotential hemopoietic

progenitors. Proceedings of the National Academy of Sciences, 84, 9035–9039.

https://doi.org/10.1073/pnas.84.24.9035

Ikebuchi, Y., Aoki, S., Honma, M., Hayashi, M., Sugamori, Y., Khan, M., … Suzuki, H. (2018).

Coupling of bone resorption and formation by RANKL reverse signalling. Nature.

https://doi.org/10.1038/s41586-018-0482-7

Ishikawa, Y., Kiyoi, H., Tsujimura, A., Miyawaki, S., Miyazaki, Y., Kuriyama, K., … Naoe, T.

(2009). Comprehensive analysis of cooperative gene mutations between class i and class II

in de novo acute myeloid leukemia. European Journal of Haematology.

https://doi.org/10.1111/j.1600-0609.2009.01261.x

Ishimi, Y., Miyaura, C., Jin, C. H., Akatsu, T., Abe, E., Nakamura, Y., … Hirano, T. (1990). IL-

6 is produced by osteoblasts and induces bone resorption. Journal of Immunology

(Baltimore, Md. : 1950).

Issaad, C., Croisille, L., Katz, A., Vainchenker, W., & Coulombel, L. (1993). A Murine Stromal

Cell Line Allows the Proliferation of Very Primitive Human CD34 +. Blood, 81, 2916–

2924.

Ito, R., Takahashi, T., Katano, I., Kawai, K., Kamisako, T., Ogura, T., … Ito, M. (2013).

Establishment of a Human Allergy Model Using Human IL-3/GM-CSF-Transgenic NOG

Mice. The Journal of Immunology. https://doi.org/10.4049/jimmunol.1203543

Itoh, K., Tezuka, H., Sakoda, H., Konno, M., Nagata, K., Uchiyama, T., … Mori, K. J. (1989).

Reproducible establishment of hemopoietic supportive stromal cell lines from murine bone

marrow. Experimental Hematology.

Jackson, D. G., & Bell, J. I. (1990). Isolation of a cDNA encoding the human CD38 (T10)

molecule, a cell surface glycoprotein with an unusual discontinuous pattern of expression

during lymphocyte differentiation. Journal of Immunology (Baltimore, Md. : 1950), 144,

2811–2815.

Jaiswal, S., Fontanillas, P., Flannick, J., Manning, A., Grauman, P. V., Mar, B. G., … Ebert, B.

L. (2014). Age-Related Clonal Hematopoiesis Associated with Adverse Outcomes. New

England Journal of Medicine, 371, 2488–2498. https://doi.org/10.1056/NEJMoa1408617

Jan, M., Snyder, T. M., Corces-Zimmerman, M. R., Vyas, P., Weissman, I. L., Quake, S. R., &

Majeti, R. (2012). Clonal evolution of preleukemic hematopoietic stem cells precedes

Page 190: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

180

human acute myeloid leukemia. Science Translational Medicine, 4.

https://doi.org/10.1126/scitranslmed.3004315

Joshi, I., Yoshida, T., Jena, N., Qi, X., Zhang, J., Van Etten, R. A., & Georgopoulos, K. (2014).

Loss of Ikaros DNA-binding function confers integrin-dependent survival on pre-B cells

and progression to acute lymphoblastic leukemia. Nature Immunology, 15, 294–304.

https://doi.org/10.1038/ni.2821

Juliusson, G., Antunovic, P., Derolf, Å., Lehmann, S., Möllgård, L., Stockelberg, D., …

Höglund, M. (2009). Age and acute myeloid leukemia: Real world data on decision to treat

and outcomes from the Swedish Acute Leukemia Registry. Blood.

https://doi.org/10.1182/blood-2008-07-172007

Kacinski, B M. (1991). FMS (CSF1 receptor) and CSF1 transcripts and protein are expressed by

human breast carcinomas in vivo and in vitro. Oncogene.

Kacinski, Barry M. (1997). CSF-1 and its receptor in breast carcinomas and neoplasms of the

female reproductive tract. Molecular Reproduction and Development.

https://doi.org/10.1002/(SICI)1098-2795(199701)46:1<71::AID-MRD11>3.0.CO;2-6

Karthikeyan, S., Leung, T., Birrane, G., Webster, G., & Ladias, J. A. A. (2001). Crystal structure

of the PDZ1 domain of human Na+/H+ exchanger regulatory factor provides insights into

the mechanism of carboxyl-terminal leucine recognition by class I PDZ domains. Journal of

Molecular Biology. https://doi.org/10.1006/jmbi.2001.4634

Keller, J. R., Ortiz, M., & Ruscetti, F. W. (1995). Steel factor (c-kit ligand) promotes the survival

of hematopoietic stem/progenitor cells in the absence of cell division. Blood, 86, 1757–

1764.

Kent, D., Copley, M., Benz, C., Dykstra, B., Bowie, M., & Eaves, C. (2008). Regulation of

hematopoietic stem cells by the steel factor/KIT signaling pathway. Clinical Cancer

Research. https://doi.org/10.1158/1078-0432.CCR-07-5134

Khanna-Gupta, A., & Berliner, N. (2007). ATRA: Finding targeted APL therapy targets. Blood.

https://doi.org/10.1182/blood-2007-04-086348

Kirma, N., Hommes, L. S., Liu, Y. G., Nair, H. B., Valente, P. T., Kumar, S., … Tekmal, R. R.

(2007). Elevated expression of the oncogene c-fms and its ligand, the macrophage colony-

stimulating factor-1, in cervical cancer and the role of transforming growth factor-β1 in

inducing c-fms expression. Cancer Research. https://doi.org/10.1158/0008-5472.CAN-06-

1991

Kishimoto, T. (2010). IL-6: From its discovery to clinical applications. International

Immunology, 22, 347–352. https://doi.org/10.1093/intimm/dxq030

Knight, A. C., Brill, S. A., Queen, S. E., Tarwater, P. M., & Mankowski, J. L. (2018). Increased

microglial CSF1R expression in the SIV/macaque model of HIV CNS disease. Journal of

Neuropathology and Experimental Neurology. https://doi.org/10.1093/jnen/nlx115

Kobari, L., Dubart, A., Le Pesteur, F., Vainchenker, W., & Sainteny, F. (1995).

Hematopoietic‐promoting activity of the murine stromal cell line MS‐5 is not related to the

expression of the major hematopoietic cytokines. Journal of Cellular Physiology, 163, 295–

304. https://doi.org/10.1002/jcp.1041630210

Kode, A., Manavalan, J. S., Mosialou, I., Bhagat, G., Rathinam, C. V., Luo, N., … Kousteni, S.

(2014). Leukaemogenesis induced by an activating β-catenin mutation in osteoblasts.

Nature, 506, 240–244. https://doi.org/10.1038/nature12883

Kopp, H.-G. (2005). The Bone Marrow Vascular Niche: Home of HSC Differentiation and

Mobilization. Physiology, 20, 349–356. https://doi.org/10.1152/physiol.00025.2005

Page 191: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

181

Kopp, Hans-Georg, Avecilla, S. T., Hooper, A. T., & Rafii, S. (2005). The Bone Marrow

Vascular Niche: Home of HSC Differentiation and Mobilization. Physiology.

https://doi.org/10.1152/physiol.00025.2005

Kristiansen, O. P., & Mandrup-Poulsen, T. (2005). Interleukin-6 and diabetes: The good, the bad,

or the indifferent? Diabetes. https://doi.org/10.2337/diabetes.54.suppl_2.S114

Krojer, T., Garrido-Franco, M., Huber, R., Ehrmann, M., & Clausen, T. (2002). Crystal structure

of DegP (HtrA) reveals a new protease-chaperone machine. Nature.

https://doi.org/10.1038/416455a

Kumar, V., Donthireddy, L., Marvel, D., Condamine, T., Wang, F., Lavilla-Alonso, S., …

Gabrilovich, D. I. (2017). Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect

of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell.

https://doi.org/10.1016/j.ccell.2017.10.005

Kuo, A. (2002). Transmembrane transforming growth factor-alpha tethers to the PDZ domain-

containing, Golgi membrane-associated protein p59/GRASP55. The EMBO Journal.

https://doi.org/10.1093/emboj/19.23.6427

Lacombe, F., Durrieu, F., Briais, A., Dumain, P., Belloc, F., Bascans, E., … Bernard, P. (1997).

Flow cytometry CD45 gating for immunophenotyping of acute myeloid leukemia.

Leukemia, 11, 1878–1886. https://doi.org/10.1038/sj.leu.2400847

Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes, J., … Dick, J. E.

(1994). A cell initiating human acute myeloid leukaemia after transplantation into SCID

mice. Nature, 367, 645–648. https://doi.org/10.1038/367645a0

Larochelle, A., Vormoor, J., Hanenberg, H., Wang, J. C. Y., Bhatia, M., Lapidot, T., … Dick, J.

E. (1996). Identification of primitive human hematopoietic cells capable of repopulating

NOD/SCID mouse bone marrow: Implications for gene therapy. Nature Medicine, 2, 1329–

1337. https://doi.org/10.1038/nm1296-1329

Laurenti, E., & Göttgens, B. (2018). From haematopoietic stem cells to complex differentiation

landscapes. Nature. https://doi.org/10.1038/nature25022

Lee, H. J., & Zheng, J. J. (2010). PDZ domains and their binding partners: Structure, specificity,

and modification. Cell Communication and Signaling. https://doi.org/10.1186/1478-811X-

8-8

Ley, K. (2017). M1 Means Kill; M2 Means Heal. The Journal of Immunology.

https://doi.org/10.4049/jimmunol.1701135

Li, J., Callaway, D. J. E., & Bu, Z. (2009). Ezrin Induces Long-Range Interdomain Allostery in

the Scaffolding Protein NHERF1. Journal of Molecular Biology.

https://doi.org/10.1016/j.jmb.2009.07.005

Liersch, R., Müller-Tidow, C., Berdel, W. E., & Krug, U. (2014). Prognostic factors for acute

myeloid leukaemia in adults - biological significance and clinical use. British Journal of

Haematology. https://doi.org/10.1111/bjh.12750

Lin, D., Gish, G. D., Songyang, Z., & Pawson, T. (1999). The carboxyl terminus of B class

ephrins constitutes a PDZ domain binding motif. Journal of Biological Chemistry.

https://doi.org/10.1074/jbc.274.6.3726

Lin, H., Lee, E., Hestir, K., Leo, C., Huang, M., Bosch, E., … Williams, L. T. (2008). Discovery

of a cytokine and its receptor by functional screening of the extracellular proteome. Science.

https://doi.org/10.1126/science.1154370

Lipinska, B., Zylicz, M., & Georgopoulos, C. (1990). The HtrA (DegP) protein, essential for

Escherichia coli survival at high temperatures, is an endopeptidase. Journal of Bacteriology.

Page 192: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

182

https://doi.org/10.1128/jb.172.4.1791-1797.1990

Liran I. Shlush, Sasan Zandi, Amanda Mitchell, Weihsu Claire Chen, J. M. B. and V. G. (2014).

Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature, 506,

328. https://doi.org/http://dx.doi.org/10.1038/nature13038

Liu, M., Miller, C. L., & Eaves, C. J. (2013). Human long-term culture initiating cell assay.

Methods in Molecular Biology, 946, 241–256. https://doi.org/10.1007/978-1-62703-128-8-

15

Liu, T., Zhang, L., Joo, D., & Sun, S. C. (2017). NF-κB signaling in inflammation. Signal

Transduction and Targeted Therapy. https://doi.org/10.1038/sigtrans.2017.23

Löwenberg, B., Pabst, T., Vellenga, E., van Putten, W., Schouten, H. C., Graux, C., …

Ossenkoppele, G. J. (2011). Cytarabine Dose for Acute Myeloid Leukemia. New England

Journal of Medicine. https://doi.org/10.1056/nejmoa1010222

Manning, B. D., & Cantley, L. C. (2007). AKT/PKB signaling: navigating downstream. Cell.

https://doi.org/10.1016/j.cell.2007.06.009

Martín-Moreno, A. M., Roncador, G., Maestre, L., Mata, E., Jiménez, S., Martínez-

Torrecuadrada, J. L., … García, J. F. (2015). CSF1R protein expression in reactive

lymphoid tissues and lymphoma: Its relevance in classical Hodgkin lymphoma. PLoS ONE.

https://doi.org/10.1371/journal.pone.0125203

McCulloch, E. A. (1983). Stem cells in normal and leukemic hemopoiesis (Henry Stratton

Lecture, 1982). Blood, 62, 1–13. https://doi.org/10.1017/CBO9781107415324.004

McKenzie, J. L., Gan, O. I., Doedens, M., & Dick, J. E. (2007). Reversible cell surface

expression of CD38 on CD34-positive human hematopoietic repopulating cells.

Experimental Hematology, 35, 1429–1436. https://doi.org/10.1016/j.exphem.2007.05.017

Méndez-Ferrer, S., Michurina, T. V., Ferraro, F., Mazloom, A. R., MacArthur, B. D., Lira, S. A.,

… Frenette, P. S. (2010). Mesenchymal and haematopoietic stem cells form a unique bone

marrow niche. Nature, 466, 829–834. https://doi.org/10.1038/nature09262

Méndez-Ferrer, S., Michurina, T. V., Ferraro, F., Mazloom, A. R., MacArthur, B. D., Lira, S. A.,

… Frenette, P. S. (2010). Mesenchymal and haematopoietic stem cells form a unique bone

marrow niche. Nature. https://doi.org/10.1038/nature09262

Metcalf, D. (1985). The granulocyte-macrophage colony-stimulating factors. Science, 229, 16–

22. https://doi.org/10.1126/science.2990035

Metcalf, Donald. (2013). The colony-stimulating factors and cancer. Cancer Immunology

Research. https://doi.org/10.1158/2326-6066.CIR-13-0151

Metzeler, K. H., Herold, T., Rothenberg-Thurley, M., Amler, S., Sauerland, M. C., Görlich, D.,

… Spiekermann, K. (2016). Spectrum and prognostic relevance of driver gene mutations in

acute myeloid leukemia. In Blood. https://doi.org/10.1182/blood-2016-01-693879

Metzeler, K. H., Hummel, M., Bloomfield, C. D., Spiekermann, K., Braess, J., Sauerland, M. C.,

… Buske, C. (2008). An 86-probe-set gene-expression signature predicts survival in

cytogenetically normal acute myeloid leukemia. Blood. https://doi.org/10.1182/blood-2008-

02-134411

Meyer, C., & Drexler, H. G. (1999). FLT3 ligand inhibits apoptosis and promotes survival of

myeloid leukemia cell lines. Leukemia and Lymphoma.

https://doi.org/10.3109/10428199909058416

Miller, J. S., Verfaillie, C., & McGlave, P. (1992). The generation of human natural killer cells

from CD34+/DR- primitive progenitors in long-term bone marrow culture. Blood, 80,

2182–2187.

Page 193: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

183

Mizuki, M., Fenski, R., Halfter, H., Matsumura, I., Schmidt, R., Müller, C., … Serve, H. (2000).

Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D

cells mediated by the Ras and STAT5 pathways. Blood, 96, 3907–3914.

https://doi.org/10.1016/0092-8674(91)90010-v

Morandi, A., Barbetti, V., Riverso, M., Dello Sbarba, P., & Rovida, E. (2011). The Colony-

Stimulating Factor-1 (CSF-1) receptor sustains ERK1/2 activation and proliferation in

breast cancer cell lines. PLoS ONE. https://doi.org/10.1371/journal.pone.0027450

Morrison, S. J., & Scadden, D. T. (2014a). The bone marrow niche for haematopoietic stem

cells. Nature. https://doi.org/10.1038/nature12984

Morrison, S. J., & Scadden, D. T. (2014b). The bone marrow niche for haematopoietic stem

cells. Nature. https://doi.org/10.1038/nature12984

Morrison, S. J., & Weissman, I. L. (1994). The long-term repopulating subset of hematopoietic

stem cells is deterministic and isolatable by phenotype. Immunity.

https://doi.org/10.1016/1074-7613(94)90037-X

Morstyn, G., Souza, L. M., Keech, J., Sheridan, W., Campbell, L., Alton, N. K., … Fox, R.

(1988). EFFECT OF GRANULOCYTE COLONY STIMULATING FACTOR ON

NEUTROPENIA INDUCED BY CYTOTOXIC CHEMOTHERAPY. The Lancet, 331,

667–672. https://doi.org/10.1016/S0140-6736(88)91475-4

Moschoi, R., Imbert, V., Nebout, M., Chiche, J., Mary, D., Prebet, T., … Griessinger, E. (2016).

Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid

leukemic cells during chemotherapy. Blood. https://doi.org/10.1182/blood-2015-07-655860

Mrózek, K., Heerema, N. A., & Bloomfield, C. D. (2004). Cytogenetics in acute leukemia. Blood

Reviews. https://doi.org/10.1016/S0268-960X(03)00040-7

Murphy, T., & Yee, K. W. L. (2017). Cytarabine and daunorubicin for the treatment of acute

myeloid leukemia. Expert Opinion on Pharmacotherapy.

https://doi.org/10.1080/14656566.2017.1391216

MURWANTOKO, YANO, M., UETA, Y., MURASAKI, A., KANDA, H., OKA, C., &

KAWAICHI, M. (2004). Binding of proteins to the PDZ domain regulates proteolytic

activity of HtrA1 serine protease. Biochemical Journal. https://doi.org/10.1042/BJ20040435

Nandi, S., Cioce, M., Yeung, Y. G., Nieves, E., Tesfa, L., Lin, H., … Stanley, E. R. (2013).

Receptor-type protein-tyrosine phosphatase ζ is a functional receptor for interleukin-34.

Journal of Biological Chemistry. https://doi.org/10.1074/jbc.M112.442731

Nerlov, C. (2004). C/EBPα mutations in acute myeloid leukaemias. Nature Reviews Cancer.

https://doi.org/10.1038/nrc1363

Neubert, N. J., Schmittnaegel, M., Bordry, N., Nassiri, S., Wald, N., Martignier, C., … Speiser,

D. E. (2018). T cell-induced CSF1 promotes melanoma resistance to PD1 blockade. Science

Translational Medicine. https://doi.org/10.1126/scitranslmed.aan3311

Nijweide, P. J., Burger, E. H., & Feyen, J. H. (2017). Cells of bone: proliferation, differentiation,

and hormonal regulation. Physiological Reviews.

https://doi.org/10.1152/physrev.1986.66.4.855

Nishi, N., Ishikawa, R., Inoue, H., Nishikawa, M., Yoneya, T., Kakeda, M., … Mori, K. J.

(1997). In vitro long-term culture of human primitive hematopoietic cells supported by

murine stromal cell line MS-5. Leukemia, 11 Suppl 3, 468–473.

Nishimoto, N. (2006). Interleukin-6 in rheumatoid arthritis. Current Opinion in Rheumatology.

https://doi.org/10.1097/01.bor.0000218949.19860.d1

Norris, L. B., Magwood, J., & Bennett, C. L. (2014). Hematopoietic Growth Factors. In Cancer

Page 194: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

184

Consult: Expertise for Clinical Practice. https://doi.org/10.1002/9781118589199.ch132

Noy, R., & Pollard, J. W. (2014). Tumor-Associated Macrophages: From Mechanisms to

Therapy. Immunity. https://doi.org/10.1016/j.immuni.2014.06.010

O’Donnell M, Tallman M, Abboud C, Altman J, Appelbaum F, Arber D, Bhatt V, Dale Bixby,

MD, PhD; William Blum, MD; Steven E. Coutre, MD; Marcos De Lima, MD; Amir T. Fat,

M. (2017). NCCN Clinical Practice Guidelines in Oncology: Acute Myeloid Leukemia

2017. Journal of the National Comprenhensive Cancer Network (JNCCN).

O’Donnell, M. R., Tallman, M. S., Abboud, C. N., Altman, J. K., Appelbaum, F. R., Arber, D.

A., … Ogba, N. (2017). Acute myeloid leukemia, version 3.2017: Clinical practice

guidelines in oncology. JNCCN Journal of the National Comprehensive Cancer Network.

https://doi.org/10.6004/jnccn.2017.0116

Ohlsson, E., Schuster, M. B., Hasemann, M., & Porse, B. T. (2016). The multifaceted functions

of C/EBPα in normal and malignant haematopoiesis. Leukemia.

https://doi.org/10.1038/leu.2015.324

Oosterhof, N., Kuil, L. E., van der Linde, H. C., Burm, S. M., Berdowski, W., van Ijcken, W. F.

J., … van Ham, T. J. (2018). Colony-Stimulating Factor 1 Receptor (CSF1R) Regulates

Microglia Density and Distribution, but Not Microglia Differentiation In Vivo. Cell

Reports. https://doi.org/10.1016/j.celrep.2018.06.113

Orkin, S. H., & Zon, L. I. (2008). Hematopoiesis: an evolving paradigm for stem cell biology.

Cell. https://doi.org/10.1016/j.cell.2008.01.025

Osaki, M., Oshimura, M., & Ito, H. (2004). PI3K-Akt pathway: Its functions and alterations in

human cancer. Apoptosis. https://doi.org/10.1023/B:APPT.0000045801.15585.dd

Pabst, T., Mueller, B. U., Zhang, P., Radomska, H. S., Narravula, S., Schnittger, S., … Tenen, D.

G. (2001). Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding

protein-α (C/EBPα), in acute myeloid leukemia. Nature Genetics.

https://doi.org/10.1038/85820

Pandit, J., Bohm, A., Jancarik, J., Halenbeck, R., Koths, K., & Kim, S. H. (1992). Three-

dimensional structure of dimeric human recombinant macrophage colony-stimulating

factor. Science. https://doi.org/10.1126/science.1455231

Pang, B., Qiao, X., Janssen, L., Velds, A., Groothuis, T., Kerkhoven, R., … Neefjes, J. (2013).

Drug-induced histone eviction from open chromatin contributes to the chemotherapeutic

effects of doxorubicin. Nature Communications. https://doi.org/10.1038/ncomms2921

Papaemmanuil, E., Gerstung, M., Bullinger, L., Gaidzik, V. I., Paschka, P., Roberts, N. D., …

Campbell, P. J. (2016a). Genomic Classification and Prognosis in Acute Myeloid

Leukemia. The New England Journal of Medicine.

https://doi.org/10.1056/NEJMoa1516192

Papaemmanuil, E., Gerstung, M., Bullinger, L., Gaidzik, V. I., Paschka, P., Roberts, N. D., …

Campbell, P. J. (2016b). Genomic Classification and Prognosis in Acute Myeloid

Leukemia. New England Journal of Medicine. https://doi.org/10.1056/nejmoa1516192

Patchen, M. L., MacVittie, T. J., Williams, J. L., Schwartz, G. N., & Souza, L. M. (1991).

Administration of interleukin-6 stimulates multilineage hematopoiesis and accelerates

recovery from radiation-induced hematopoietic depression. Blood, 77, 472–480.

https://doi.org/papers3://publication/uuid/8F1D389D-C168-4191-A3EB-980FC9228F4A

Patel, J. P., Gönen, M., Figueroa, M. E., Fernandez, H., Sun, Z., Racevskis, J., … Levine, R. L.

(2012). Prognostic Relevance of Integrated Genetic Profiling in Acute Myeloid Leukemia.

New England Journal of Medicine. https://doi.org/10.1056/nejmoa1112304

Page 195: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

185

Patnaik, M. M. (2018). The importance of FLT3 mutational analysis in acute myeloid leukemia.

Leukemia and Lymphoma. https://doi.org/10.1080/10428194.2017.1399312

Pixley, F. J., & Stanley, E. R. (2004). CSF-1 regulation of the wandering macrophage:

Complexity in action. Trends in Cell Biology. https://doi.org/10.1016/j.tcb.2004.09.016

Ploemacher, R. E., van der Sluijs, J. P., van Beurden, C. A., Baert, M. R., & Chan, P. L. (1991).

Use of limiting-dilution type long-term marrow cultures in frequency analysis of marrow-

repopulating and spleen colony-forming hematopoietic stem cells in the mouse. Blood, 78,

2527–2533.

Ploemacher, R. E., van der Sluijs, J. P., Voerman, J. S., & Brons, N. H. (1989). An in vitro

limiting-dilution assay of long-term repopulating hematopoietic stem cells in the mouse.

Blood, 74, 2755–2763.

Poulton, T. B., Murphy, W. D., Duerk, J. L., Chapek, C. C., & Feiglin, D. H. (1993). Bone

marrow reconversion in adults who are smokers: MR imaging findings. American Journal

of Roentgenology, 161, 1217–1221. https://doi.org/10.2214/ajr.161.6.8249729

Price, L. K. H., Choi, H. U., Rosenberg, L., & Stanley, E. R. (1992). The predominant form of

secreted colony stimulating factor-1 is a proteoglycan. Journal of Biological Chemistry.

Pugazhenthit, S., Nesterova, A., Sable, C., Heidenreich, K. A., Boxer, L. M., Heasley, L. E., &

Reusch, J. E. B. (2000). Akt/protein kinase B up-regulates Bcl-2 expression through cAMP-

response element-binding protein. Journal of Biological Chemistry.

https://doi.org/10.1074/jbc.275.15.10761

Quentmeier, H., Zaborski, M., & Drexler, H. G. (1997). The human bladder carcinoma cell line

5637 constitutively secretes functional cytokines. Leukemia Research.

https://doi.org/10.1016/S0145-2126(96)00132-4

Rademakers, R., Baker, M., Nicholson, A. M., Rutherford, N. J., Finch, N., Soto-Ortolaza, A., …

Wszolek, Z. K. (2012). Mutations in the colony stimulating factor 1 receptor (CSF1R) gene

cause hereditary diffuse leukoencephalopathy with spheroids. Nature Genetics.

https://doi.org/10.1038/ng.1027

Raivich, G., Haas, S., Werner, A., Klein, M. A., Kloss, C., & Kreutzberg, G. W. (1998).

Regulation of MCSF receptors on microglia in the normal and injured mouse central

nervous system: A quantitative immunofluorescence study using confocal laser microscopy.

Journal of Comparative Neurology. https://doi.org/10.1002/(SICI)1096-

9861(19980808)395:3<342::AID-CNE6>3.0.CO;2-2

Rathinam, C., Poueymirou, W. T., Rojas, J., Murphy, A. J., Valenzuela, D. M., Yancopoulos, G.

D., … Flavell, R. A. (2011a). Efficient differentiation and function of human macrophages

in humanized CSF-1 mice. Blood. https://doi.org/10.1182/blood-2010-12-326926

Rathinam, C., Poueymirou, W. T., Rojas, J., Murphy, A. J., Valenzuela, D. M., Yancopoulos, G.

D., … Flavell, R. A. (2011b). Efficient differentiation and function of human macrophages

in humanized CSF-1 mice. Blood, 118, 3119–3128. https://doi.org/10.1182/blood-2010-12-

326926

Reedijk, M., Liu, X., van der Geer, P., Letwin, K., Waterfield, M. D., Hunter, T., & Pawson, T.

(1992). Tyr721 regulates specific binding of the CSF-1 receptor kinase insert to PI 3′-kinase

SH2 domains: a model for SH2-mediated receptor-target interactions. The EMBO Journal.

https://doi.org/10.1002/j.1460-2075.1992.tb05181.x

Reedijk, M., Liu, X., van der Geer, P., Letwin, K., Waterfield, M. D., Hunter, T., & Pawson, T.

(2018). Tyr721 regulates specific binding of the CSF-1 receptor kinase insert to PI 3′-kinase

SH2 domains: a model for SH2-mediated receptor-target interactions. The EMBO Journal.

Page 196: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

186

https://doi.org/10.1002/j.1460-2075.1992.tb05181.x

Rettenmier, C W, & Roussel, M. F. (1988). Differential processing of colony-stimulating factor

1 precursors encoded by two human cDNAs. Molecular and Cellular Biology, 8, 5026–

5034.

Rettenmier, Carl W., Chen, J. H., Roussel, M. F., & Sherr, C. J. (1985). The product of the c-fms

proto-oncogene: A glycoprotein with associated tyrosine kinase activity. Science.

https://doi.org/10.1126/science.2580348

Richards, C. D. (1998). Interleukin-6. Cytokines, 6, 87–108. https://doi.org/10.1016/B978-

012498340-3/50007-5

Rider, B. J. (2011). Cytarabine. In xPharm: The Comprehensive Pharmacology Reference.

https://doi.org/10.1016/B978-008055232-3.61536-3

Rohrschneider, L. R., Bourette, R. P., Lioubin, M. N., Algate, P. A., Myles, G. M., & Carlberg,

K. (1997). Growth and differentiation signals regulated by the M-CSF receptor. Molecular

Reproduction and Development. https://doi.org/10.1002/(SICI)1098-

2795(199701)46:1<96::AID-MRD15>3.0.CO;2-1

Roloff, G. W., & Griffiths, E. A. (2018). When to obtain genomic data in acute myeloid

leukemia (AML) and which mutations matter. Blood Advances.

https://doi.org/10.1182/bloodadvances.2018020206

Rosnet, O., & Birnbaum, D. (1993). Hematopoietic receptors of class III receptor-type tyrosine

kinases. Critical Reviews in Oncogenesis, 4, 595–613.

Ross, F. P., & Teitelbaum, S. L. (2005). αvβ3 and macrophage colony-stimulating factor:

Partners in osteoclast biology. Immunological Reviews. https://doi.org/10.1111/j.0105-

2896.2005.00331.x

Rossi, L., Challen, G., Sirin, O., Lin, K., & Goodell, M. (2011). Hematopoietic Stem Cell

Characterization and Isolation. Methods in Molecular Biology, 750, 117–129.

https://doi.org/10.1007/978-1-61779-145-1

Roussel, M. F., & Sherr, C. J. (2006). Mouse NIH 3T3 cells expressing human colony-

stimulating factor 1 (CSF-1) receptors overgrow in serum-free medium containing human

CSF-1 as their only growth factor. Proceedings of the National Academy of Sciences.

https://doi.org/10.1073/pnas.86.20.7924

Roussel, M. F., Shurtleff, S. A., Downing, J. R., & Sherr, C. J. (2006). A point mutation at

tyrosine-809 in the human colony-stimulating factor 1 receptor impairs mitogenesis without

abrogating tyrosine kinase activity, association with phosphatidylinositol 3-kinase, or

induction of c-fos and junB genes. Proceedings of the National Academy of Sciences.

https://doi.org/10.1073/pnas.87.17.6738

Sacchetti, B., Funari, A., Michienzi, S., Di Cesare, S., Piersanti, S., Saggio, I., … Bianco, P.

(2007). Self-Renewing Osteoprogenitors in Bone Marrow Sinusoids Can Organize a

Hematopoietic Microenvironment. Cell, 131, 324–336.

https://doi.org/10.1016/j.cell.2007.08.025

Sanchez-Correa, B., Bergua, J. M., Campos, C., Gayoso, I., Arcos, M. J., Bañas, H., …

Tarazona, R. (2013). Cytokine profiles in acute myeloid leukemia patients at diagnosis:

Survival is inversely correlated with IL-6 and directly correlated with IL-10 levels.

Cytokine. https://doi.org/10.1016/j.cyto.2012.12.023

Sanchez, P. V., Perry, R. L., Sarry, J. E., Perl, A. E., Murphy, K., Swider, C. R., … Carroll, M.

(2009). A robust xenotransplantation model for acute myeloid leukemia. Leukemia, 23,

2109–2117. https://doi.org/10.1038/leu.2009.143

Page 197: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

187

Sangle, N. A., & Perkins, S. L. (2011). Core-binding factor acute myeloid leukemia. Archives of

Pathology and Laboratory Medicine. https://doi.org/10.5858/arpa.2010-0482-RS

Sasmono, R. T., Oceandy, D., Pollard, J. W., Tong, W., Pavli, P., Wainwright, B. J., … Hume,

D. A. (2003). A macrophage colony-stimulating factor receptor-green fluorescent protein

transgene is expressed throughout the mononuclear phagocyte system of the mouse. Blood.

https://doi.org/10.1182/blood-2002-02-0569

Scadden, D. T. (2006). The stem-cell niche as an entity of action. Nature.

https://doi.org/10.1038/nature04957

Schroder, K., Hertzog, P. J., Ravasi, T., & Hume, D. A. (2004). Interferon-gamma: an overview

of signals, mechanisms and functions. Journal of Leukocyte Biology.

https://doi.org/10.1189/jlb.0603252

Schroeder, T., Geyh, S., Germing, U., & Haas, R. (2016). Mesenchymal stromal cells in myeloid

malignancies. Blood Research. https://doi.org/10.5045/br.2016.51.4.225

Schwella, N., Braun, A., Ahrens, N., Rick, O., & Salama, A. (2003). Leukapheresis after high-

dose chemotherapy and autologous peripheral blood progenitor cell transplantation: A novel

approach to harvest a second autograft. Transfusion. https://doi.org/10.1046/j.1537-

2995.2003.00306.x

Segaliny, A. I., Brion, R., Mortier, E., Maillasson, M., Cherel, M., Jacques, Y., … Heymann, D.

(2015). Syndecan-1 regulates the biological activities of interleukin-34. Biochimica et

Biophysica Acta - Molecular Cell Research. https://doi.org/10.1016/j.bbamcr.2015.01.023

Sellar, R., Goldstone, A. H., & Lazarus, H. M. (2011). Redefining transplant in acute leukemia.

Current Treatment Options in Oncology. https://doi.org/10.1007/s11864-011-0166-0

Shao, J., Weng, X., Zhuo, L., Yu, L., Li, Z., Shen, K., … Xu, Y. (2019). Angiotensin II induced

CSF1 transcription is mediated by a crosstalk between different epigenetic factors in

vascular endothelial cells. Biochimica et Biophysica Acta - Gene Regulatory Mechanisms.

https://doi.org/10.1016/j.bbagrm.2018.10.001

Sherr, C J, & Rettenmier, C. W. (1986). The fms gene and the CSF-1 receptor. Cancer Surveys,

5, 221–232.

Sherr, Charles J., Rettenmier, C. W., Sacca, R., Roussel, M. F., Look, A. T., & Stanley, E. R.

(1985). The c-fms proto-oncogene product is related to the receptor for the mononuclear

phagocyte growth factor, CSF 1. Cell. https://doi.org/10.1016/S0092-8674(85)80047-7

Shlush, L. I., Mitchell, A., Heisler, L., Abelson, S., Ng, S. W. K., Trotman-Grant, A., … Dick, J.

E. (2017). Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature,

547, 104–108. https://doi.org/10.1038/nature22993

Shultz, L. D., Schweitzer, P. A., Christianson, S. W., Gott, B., Tennent, B., Mckenna, S., …

Leiter, E. H. (2010). Multiple defects in innate and adaptive immunologic function in NOD

/ LtSz-scid mice. Journal of Immunology, 154, 180–191.

https://doi.org/10.4049/jimmunol.179.1.89

Sidney, L. E., Branch, M. J., Dunphy, S. E., Dua, H. S., & Hopkinson, A. (2014). Concise

review: Evidence for CD34 as a common marker for diverse progenitors. Stem Cells.

https://doi.org/10.1002/stem.1661

Silzle, T., Randolph, G. J., Kreutz, M., & Kunz-Schughart, L. A. (2004). The fibroblast: Sentinel

cell and local immune modulator in tumor tissue. International Journal of Cancer.

https://doi.org/10.1002/ijc.11542

Smith, P. C., Hobisch, a, Lin, D. L., Culig, Z., & Keller, E. T. (2001). Interleukin-6 and prostate

cancer progression. Cytokine & Growth Factor Reviews, 12, 33–40. https://doi.org/doi:

Page 198: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

188

10.1016/S1359-6101(00)00021-6

Songyang, Z., Fanning, A. S., Fu, C., Xu, J., Marfatia, S. M., Chishti, A. H., … Cantley, L. C.

(1997). Recognition of unique carboxyl-terminal motifs by distinct PDZ domains. Science.

https://doi.org/10.1126/science.275.5296.73

Souza, L. M., Boone, T. C., Gabrilove, J., Lai, P. H., Zsebo, K. M., Murdock, D. C., … Chen, K.

K. (1986). Recombinant human granulocyte colony-stimulating factor: effects on normal

and leukemic myeloid cells. Science (New York, N.Y.), 232, 61–65.

https://doi.org/10.1126/science.2420009

Stanley, E. R., Guilbert, L. J., Tushinski, R. J., & Bartelmez, S. H. (1983). CSF-1?A

mononuclear phagocyte lineage-specific hemopoietic growth factor. Journal of Cellular

Biochemistry. https://doi.org/10.1002/jcb.240210206

Stanley, E. Richard, Berg, K. L., Einstein, D. B., Lee, P. S. W., Pixley, F. J., Wang, Y., &

Yeung, Y. G. (1997). Biology and action of colony-stimulating factor-1. Molecular

Reproduction and Development. https://doi.org/10.1002/(SICI)1098-

2795(199701)46:1<4::AID-MRD2>3.0.CO;2-V

Stanley, E. Richard, & Chitu, V. (2014). CSF-1 receptor signaling in myeloid cells. Cold Spring

Harbor Perspectives in Biology. https://doi.org/10.1101/cshperspect.a021857

Stevens, A. M., Miller, J. M., Munoz, J. O., Gaikwad, A. S., & Redell, M. S. (2017). Interleukin-

6 levels predict event-free survival in pediatric AML and suggest a mechanism of

chemotherapy resistance. Blood Advances.

https://doi.org/10.1182/bloodadvances.2017007856

Sugiyama, T., Kohara, H., Noda, M., & Nagasawa, T. (2006). Maintenance of the Hematopoietic

Stem Cell Pool by CXCL12-CXCR4 Chemokine Signaling in Bone Marrow Stromal Cell

Niches. Immunity. https://doi.org/10.1016/j.immuni.2006.10.016

Sutherland, H. J., Lansdorp, P. M., Henkelman, D. H., Eaves, A. C., & Eaves, C. J. (1990).

Functional characterization of individual human hematopoietic stem cells cultured at

limiting dilution on supportive marrow stromal layers. Proceedings of the National

Academy of Sciences of the United States of America, 87, 3584–3588.

https://doi.org/10.1073/pnas.87.9.3584

Suzuki, Y., Imai, Y., Nakayama, H., Takahashi, K., Takio, K., & Takahashi, R. (2001). A serine

protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell

death. Molecular Cell. https://doi.org/10.1016/S1097-2765(01)00341-0

Swardfager, W., Lanctt, K., Rothenburg, L., Wong, A., Cappell, J., & Herrmann, N. (2010). A

meta-analysis of cytokines in Alzheimer’s disease. Biological Psychiatry, 68, 930–941.

https://doi.org/10.1016/j.biopsych.2010.06.012

Swierczak, A., Cook, A. D., Lenzo, J. C., Restall, C. M., Doherty, J. P., Anderson, R. L., &

Hamilton, J. A. (2014). The Promotion of Breast Cancer Metastasis Caused by Inhibition of

CSF-1R/CSF-1 Signaling Is Blocked by Targeting the G-CSF Receptor. Cancer

Immunology Research, 2, 765–776. https://doi.org/10.1158/2326-6066.CIR-13-0190

Takeshita, S., Faccio, R., Chappel, J., Zheng, L., Feng, X., Weber, J. D., … Ross, F. P. (2007).

C-Fms tyrosine 559 is a major mediator of M-CSF-induced proliferation of primary

macrophages. Journal of Biological Chemistry. https://doi.org/10.1074/jbc.M610938200

Tallman, M. S., Wang, E. S., Altman, J. K., Appelbaum, F. R., Bhatt, V. R., Bixby, D., … Ogba,

N. (2019). Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice Guidelines

in Oncology. Journal of the National Comprehensive Cancer Network.

https://doi.org/10.6004/jnccn.2019.0028

Page 199: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

189

Talmadge, J. E., & Gabrilovich, D. I. (2013). History of myeloid-derived suppressor cells.

Nature Reviews Cancer. https://doi.org/10.1038/nrc3581

Tang, R., Ojeda, M., Pouillart, P., Scholl, S., Beuvon, F., & Mosseri, V. (1992). M‐CSF

(monocyte colony stimulating factor) and M‐CSF receptor expression by breast tumour

cells: M‐CSF mediated recruitment of tumour infiltrating monocytes? Journal of Cellular

Biochemistry. https://doi.org/10.1002/jcb.240500403

Tap, W. D., Wainberg, Z. A., Anthony, S. P., Ibrahim, P. N., Zhang, C., Healey, J. H., … Bollag,

G. (2015). Structure-Guided Blockade of CSF1R Kinase in Tenosynovial Giant-Cell

Tumor. New England Journal of Medicine. https://doi.org/10.1056/NEJMoa1411366

Teitelbaum, S. L. (2000). Bone resorption by osteoclasts. Science.

https://doi.org/10.1126/science.289.5484.1504

Tiaden, A. N., Breiden, M., Mirsaidi, A., Weber, F. A., Bahrenberg, G., Glanz, S., … Richards,

P. J. (2012a). Human serine protease HTRA1 positively regulates osteogenesis of human

bone marrow-derived mesenchymal stem cells and mineralization of differentiating bone-

forming cells through the modulation of extracellular matrix protein. Stem Cells.

https://doi.org/10.1002/stem.1190

Tiaden, A. N., Breiden, M., Mirsaidi, A., Weber, F. A., Bahrenberg, G., Glanz, S., … Richards,

P. J. (2012b). Human serine protease HTRA1 positively regulates osteogenesis of human

bone marrow-derived mesenchymal stem cells and mineralization of differentiating bone-

forming cells through the modulation of extracellular matrix protein. Stem Cells.

https://doi.org/10.1002/stem.1190

Tindle, R. W., Nichols, R. A. B., Chan, L., Campana, D., Catovsky, D., & Birnie, G. D. (1985).

A novel monoclonal antibody BI-3C5 recognises myeloblasts and non-B non-T

lymphoblasts in acute leukaemias and CGL blast crises, and reacts with immature cells in

normal bone marrow. Leukemia Research, 9, 1–9. https://doi.org/10.1016/0145-

2126(85)90016-5

Tsiftsoglou, A. S., Vizirianakis, I. S., & Strouboulis, J. (2009). Erythropoiesis: Model systems,

molecular regulators, and developmental programs. IUBMB Life.

https://doi.org/10.1002/iub.226

Tsuboi, I., Revol, V., Blanchet, J. P., & Mouchiroud, G. (2000a). Role of the membrane form of

human colony-stimulating factor-1 (CSF-1) in proliferation of multipotent hematopoietic

FDCP-mix cells expressing human CSF-1 receptor. Leukemia, 14(8), 1460–1466.

Tsuboi, I., Revol, V., Blanchet, J. P., & Mouchiroud, G. (2000b). Role of the membrane form of

human colony-stimulating factor-1 (CSF-1) in proliferation of multipotent hematopoietic

FDCP-Mix cells expressing human CSF-1 receptor. Leukemia.

https://doi.org/10.1038/sj.leu.2401847

Valk, P. J., Verhaak, R. G., Beijen, M. A., Erpelinck, C. A., Barjesteh van Waalwijk van Doorn-

Khosrovani, S., Boer, J. M., … Delwel, R. (2004). Prognostically useful gene-expression

profiles in acute myeloid leukemia. N Engl J Med, 350(16), 1617–1628.

https://doi.org/10.1056/NEJMoa040465350/16/1617 [pii]

van Gosliga, D., Schepers, H., Rizo, A., van der Kolk, D., Vellenga, E., & Schuringa, J. J.

(2007). Establishing long-term cultures with self-renewing acute myeloid leukemia

stem/progenitor cells. Experimental Hematology, 35, 1538–1549.

https://doi.org/10.1016/j.exphem.2007.07.001

Vanhaesebroeck, B., & Alessi, D. R. (2000). The PI3K-PDK1 connection: more than just a road

to PKB. The Biochemical Journal.

Page 200: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

190

Varma, A., el-Awar, F. Y., Palsson, B. O., Emerson, S. G., & Clarke, M. F. (1992). Can Dexter

cultures support stem cell proliferation? Exp Hematol, 20, 87–91.

Vergez, F., Green, A. S., Tamburini, J., Sarry, J. E., Gaillard, B., Cornillet-Lefebvre, P., …

Bardet, V. (2011). High levels of CD34 +CD38 low/-CD123 + blasts are predictive of an

adverse outcome in acute myeloid leukemia: A groupe ouest-est des leucémies aiguës et

maladies du sang (GOELAMS) study. Haematologica.

https://doi.org/10.3324/haematol.2011.047894

Verhaak, R. G. W., Goudswaard, C. S., Van Putten, W., Bijl, M. A., Sanders, M. A., Hugens,

W., … Valk, P. J. M. (2005). Mutations in nucleophosmin (NPM1) in acute myeloid

leukemia (AML): Association with other gene abnormalities and previously established

gene expression signatures and their favorable prognostic significance. Blood.

https://doi.org/10.1182/blood-2005-05-2168

Von Tresckow, B., Morschhauser, F., Ribrag, V., Topp, M. S., Chien, C., Seetharam, S., …

Engert, A. (2015). An open-label, multicenter, phase I/II study of JNJ-40346527, a CSF-1R

inhibitor, in patients with relapsed or refractory Hodgkin lymphoma. Clinical Cancer

Research. https://doi.org/10.1158/1078-0432.CCR-14-1845

Walsh, N. C., Kenney, L. L., Jangalwe, S., Aryee, K.-E., Greiner, D. L., Brehm, M. A., & Shultz,

L. D. (2017). Humanized Mouse Models of Clinical Disease. Annual Review of Pathology.

https://doi.org/10.1146/annurev-pathol-052016-100332

Walter, M., Lucet, I. S., Patel, O., Broughton, S. E., Bamert, R., Williams, N. K., … Rossjohn, J.

(2007). The 2.7 Å Crystal Structure of the Autoinhibited Human c-Fms Kinase Domain.

Journal of Molecular Biology. https://doi.org/10.1016/j.jmb.2007.01.036

Wang, Q., Lu, Y., Li, R., Jiang, Y., Zheng, Y., Qian, J., … Yi, Q. (2018). Therapeutic effects of

CSF1R-blocking antibodies in multiple myeloma. Leukemia.

https://doi.org/10.1038/leu.2017.193

Wang, Y., Szretter, K. J., Vermi, W., Gilfillan, S., Rossini, C., Cella, M., … Colonna, M. (2012).

IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans

cells and microglia. Nature Immunology. https://doi.org/10.1038/ni.2360

Weber, G. F., Chousterman, B. G., He, S., Fenn, A. M., Nairz, M., Anzai, A., … Swirski, F. K.

(2015). Interleukin-3 amplifies acute inflammation and is a potential therapeutic target in

sepsis. Science, 347, 1260–1265. https://doi.org/10.1126/science.aaa4268

Wehrle-Haller, B. (2003). The role of Kit-ligand in melanocyte development and epidermal

homeostasis. In Pigment Cell Research. https://doi.org/10.1034/j.1600-0749.2003.00055.x

Weinberg, O. K., Seetharam, M., Ren, L., Seo, K., Ma, L., Merker, J. D., … Arber, D. A. (2009).

Clinical characterization of acute myeloid leukemia with myelodysplasia-related changes as

defined by the 2008 WHO classification system. Blood. https://doi.org/10.1182/blood-2008-

10-182782

Welch, J. S., Ley, T. J., Link, D. C., Miller, C. A., Larson, D. E., Koboldt, D. C., … Wilson, R.

K. (2012). The origin and evolution of mutations in acute myeloid leukemia. Cell.

https://doi.org/10.1016/j.cell.2012.06.023

Wiktor-Jedrzejczak, W., Bartocci, A., Ferrante, A. W., Ahmed-Ansari, A., Sell, K. W., Pollard,

J. W., & Stanley, E. R. (1990). Total absence of colony-stimulating factor 1 in the

macrophage-deficient osteopetrotic (op/op) mouse. Proceedings of the National Academy of

Sciences of the United States of America, 87, 4828–4832.

https://doi.org/10.1073/pnas.88.13.5937b

Yang, H., Tabe, Y., Saito, K., Jacamo, R., Ma, H., Ruvolo, V., … Konopleva, M. Y. (2018).

Page 201: Investigation of CSF1-CSF1R Signaling in AML-Bone Marrow ......mem) in particular led to significant increases in AML cell numbers and adhesion to stroma, compared to cells co-cultured

191

Mitochondrial Transfer Confers Microenvironment-Mediated Resistance to Oxphos

Inhibition in AML. Blood. https://doi.org/10.1182/BLOOD-2018-99-112933

Yang, Y. C., Ciarletta, A. B., Temple, P. A., Chung, M. P., Kovacic, S., Witek-Giannotti, J. A.

S., … Clark, S. C. (1986). Human IL-3 (multi-CSF): Identification by expression cloning of

a novel hematopoietic growth factor related to murine IL-3. Cell, 47, 3–10.

https://doi.org/10.1016/0092-8674(86)90360-0

Yarden, Y., Kuang, W. J., Yang-Feng, T., Coussens, L., Munemitsu, S., Dull, T. J., … Ullrich,

A. (1987). Human proto-oncogene c-kit: a new cell surface receptor tyrosine kinase for an

unidentified ligand. The EMBO Journal. https://doi.org/10.1002/j.1460-

2075.1987.tb02655.x

Yu, W., Chen, J., Xiong, Y., Pixley, F. J., Dai, X.-M., Yeung, Y.-G., & Stanley, E. R. (2008).

CSF-1 receptor structure/function in MacCsf1r-/- macrophages: regulation of proliferation,

differentiation, and morphology. Journal of Leukocyte Biology.

https://doi.org/10.1189/jlb.0308171

Zheng, X., Seshire, A., Rüster, B., Bug, G., Beissert, T., Puccetti, E., … Ruthardt, M. (2007).

Arsenic but not all-trans retinoic acid overcomes the aberrant stem cell capacity of

PML/RARα-positive leukemic stem cells. Haematologica.

https://doi.org/10.3324/haematol.10541