characterization of structure-function...

150
1 CHARACTERIZATION OF STRUCTURE-FUNCTION CORRELATIONS OF NOVEL JAK2 SMALL MOLECULE INHIBITORS AND THEIR MECHANISMS OF ACTION By ANURIMA MAJUMDER A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2011

Upload: others

Post on 03-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

1

CHARACTERIZATION OF STRUCTURE-FUNCTION CORRELATIONS OF NOVEL JAK2 SMALL MOLECULE INHIBITORS AND THEIR MECHANISMS OF ACTION

By

ANURIMA MAJUMDER

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2011

Page 2: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

2

© 2011 Anurima Majumder

Page 3: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

3

To my Mom, Dad and Arjun for their constant love and support

Page 4: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

4

ACKNOWLEDGMENTS

I would like to acknowledge all those people who have made this dissertation

possible and because of whom my graduate experience has been one that I will cherish

forever. First, I would like to thank my mentor, Dr. Peter P. Sayeski. I consider myself

fortunate to have had Peter as my advisor. His patience, support, encouragement and

constant guidance throughout my graduate studies has helped me overcome problems

and achieve my goals. He not only taught me research techniques, but also guided me

on how to improve my scientific writing and presentation skills. Peter has been more

than just a mentor for graduate studies by being readily available to help me out with

research problems, career options as well as any other questions that I have had.

Second, I would like to thank the members of my supervisory committee, Drs. Kathleen

Shiverick, Thomas Rowe, Brian Law and William Slayton, for their invaluable guidance,

insight and advice through the course of my graduate studies.

I would also like to thank a few people who have taught me or helped me with

various techniques over these past four years. Special thanks to Andrew Magis and

Lakshmanan Govindasamy for helping me with the molecular docking studies. I would

also like to thank Marjorie Chow and Kanchana Karrupiah. The two-dimensional gel

electrophoresis studies would not have been possible without them. In addition, I also

thank Doug Smith and Steve McClellan for their help with fluorescent microscopy and

flow cytometry, respectively.

I would next like to thank all past and present members of the Sayeski Lab:

Jacqueline Sayyah, Robert Blair, Nicholas Figueroa, Shigeharu Tsuda, Dr. Sung Park,

Annet Kirabo, Kavitha Gnanasambandan and Rebekah Baskin. They not only provided

an environment conducive to learning and research but, also made lab life a lot more

Page 5: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

5

enjoyable. I am going to miss all the fun stuff we did together both inside and outside

lab. Special thanks to my friends for their support and companionship during my stay in

Gainesville. Thank you for being there for me when times were stressful.

Lastly, I would like thank my family for their unending advice, encouragement, love

and support without which I would never have been able to complete my graduate

studies. I am deeply indebted to my parents for their constant wisdom and

encouragement. I have come so far in life only because of them and the values that

they have instilled in me. I would also like to thank my little sister, Anusmita, for

reducing my stress by providing comic relief. I am also grateful to my in-laws for their

support and encouragement. Finally, thanks to my husband, Arjun, for bearing with me

and my mood swings and keeping me sane during times of stress. His love,

understanding and support have been invaluable.

Page 6: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

6

TABLE OF CONTENTS page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES ............................................................................................................ 9

LIST OF FIGURES ........................................................................................................ 10

LIST OF ABBREVIATIONS ........................................................................................... 12

ABSTRACT ................................................................................................................... 13

CHAPTER

1 INTRODUCTION .................................................................................................... 16

Tyrosine Kinases .................................................................................................... 16 Jak2 Tyrosine Kinase.............................................................................................. 17

History .............................................................................................................. 17 Structure ........................................................................................................... 18

Jak/STAT Signaling Paradigm .......................................................................... 20 Jak2 and Cancer ..................................................................................................... 21

Jak2 in Solid tumors ......................................................................................... 21 Jak2 in Hematological Malignancies ................................................................ 22

Jak2 chromosomal translocations and hematological malignancies .......... 23 Jak2 point mutations and hematological malignancies .............................. 23

Myeloproliferative Neoplasms ................................................................................. 24 Jak2 Mutations in Myeloproliferative Neoplasms .............................................. 25

Jak2 Inhibitors for the Treatment of Myeloproliferative Neoplasms .................. 27 Rationale for Studies............................................................................................... 34

2 A46, A BENZOTHIOPHENE DERIVED COMPOUND, SUPPRESSES JAK2-MEDIATED PATHOLOGIC CELL GROWTH .......................................................... 42

Materials and Methods............................................................................................ 43 Drug.................................................................................................................. 43

Cell Culture ....................................................................................................... 44 Cell Proliferation Assay .................................................................................... 44

Enzyme-linked Immunosorbent Assay ............................................................. 44 Cell Cycle Assay .............................................................................................. 44

Apoptosis Assay ............................................................................................... 45 Western Blotting ............................................................................................... 45

Colony Formation Assay .................................................................................. 45 Clonogenic Assay ............................................................................................. 46

Statistical Analysis ............................................................................................ 46 Results .................................................................................................................... 46

Page 7: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

7

A46 Inhibits Jak2-V617F-dependent Cell Proliferation ..................................... 46 A46 Inhibits the Jak/STAT Signaling Pathway .................................................. 48

A46 Induces G1/S Cell Cycle Arrest in HEL Cells ............................................ 49 A46 Induces Apoptosis in HEL Cells ................................................................ 50

A46 Suppresses Cytokine-independent Pathologic Cell Growth of Jak2-V617F Positive Bone Marrow Cells, ex vivo .................................................. 52

Discussion .............................................................................................................. 53

3 STRUCTURE-FUNCTION CORRELATION OF G6, A NOVEL SMALL MOLECULE INHIBITOR OF JAK2: INDISPENSABILITY OF THE STILBENOID CORE ..................................................................................................................... 64

Experimental Procedures ........................................................................................ 65 Drugs ................................................................................................................ 65

Cell Culture ....................................................................................................... 66 Cell Proliferation Assay .................................................................................... 66

Enzyme-linked Immunosorbent Assay ............................................................. 66 Cell Lysis and Immunoprecipitation .................................................................. 66

Western Blotting ............................................................................................... 67 Apoptosis Assay ............................................................................................... 67

Real Time PCR Analysis .................................................................................. 68 Patient Sample ................................................................................................. 68

Colony Forming Unit-Erythroid Colony Formation Assay ................................. 68 Computational Docking .................................................................................... 69

Statistical Analysis ............................................................................................ 70 Results .................................................................................................................... 70

A Stilbenoid Core Is Essential for Time- and Dose-dependent Inhibition of Jak2-V617F-dependent Cell Growth ............................................................. 70

Indispensability of the Stilbenoid core in Decreasing Phosphorylation of STAT3 and STAT5 ........................................................................................ 72

Induction of Apoptosis in HEL Cells by G6 and its Derivatives ......................... 73 Stilbenoid Core Bearing Derivatives of G6 Suppress Pathologic Cell Growth

of Patient-derived Bone Marrow Cells, ex vivo .............................................. 74 Computational Docking of G6 and its Derivatives into the ATP Binding

Pocket of the Jak2 Kinase Domain ............................................................... 75 Discussion .............................................................................................................. 77

4 CELL DEATH INDUCED BY THE JAK2 INHIBITOR, G6, CORRELATES WITH CLEAVAGE OF VIMENTIN FILAMENTS ............................................................... 90

Experimental Procedures ........................................................................................ 91 Drugs ................................................................................................................ 91

Reagents .......................................................................................................... 92 Cell Culture ....................................................................................................... 92

2-D Differential In Gel Electrophoresis (2-D DIGE) .......................................... 92 Cell Lysis .......................................................................................................... 94

Western Blotting ............................................................................................... 94

Page 8: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

8

Immunofluorescence ........................................................................................ 95 Cell Proliferation Assay .................................................................................... 95

In vivo Animal Model ........................................................................................ 95 Bone Marrow Immunohistochemistry ............................................................... 96

Statistical Analysis ............................................................................................ 96 Results .................................................................................................................... 96

G6 Treatment Induces Time- and Dose-Dependent Degradation of Vimentin . 96 G6 Treatment Induces Marked Reorganization of Vimentin Intermediate

Filaments within Cells.................................................................................... 98 G6-induced Cleavage of Vimentin is Jak2-mediated ........................................ 99

G6-induced Cleavage of Vimentin is Independent of de novo Protein Synthesis and Caspase Activity, but Calpain-dependent .............................. 99

The Mobilization of Calcium is Essential and Sufficient for the Cleavage of the Intermediate Filament Protein Vimentin ................................................ 101

Cleavage of Vimentin is Sufficient to Reduce HEL Cell Viability .................... 102 G6 Treatment Decreases the Levels of Vimentin Protein, in vivo ................... 103

Discussion ............................................................................................................ 104

5 CONCLUSIONS AND PERSPECTIVES ............................................................... 116

Importance of Designing/Identifying a Jak2 Specific Inhibitor ............................... 117 Characterization of the Novel Jak2 Inhibitor, A46 ................................................. 119

Stilbenes and Benzothiophenes as Potential Scaffolds for the Design of Novel Jak2 Small Molecule Inhibitors .......................................................................... 120

Comparison of Two Novel Jak2 Inhibitors, G6 and A46........................................ 122 Potential of Jak2 Inhibitor Therapy for Myeloproliferative Neoplasms .................. 124

Characterization of the Link Between G6-induced Cell Death and Cleavage of Vimentin............................................................................................................. 129

LIST OF REFERENCES ............................................................................................. 134

BIOGRAPHICAL SKETCH .......................................................................................... 150

Page 9: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

9

LIST OF TABLES

Table page 1-1 Preclinical characterization of Jak2 inhibitors ..................................................... 39

1-2 Clinical characterization of Jak2 inhibitors .......................................................... 40

2-1 Chemical characteristics of A46 ......................................................................... 57

3-1 Percent growth inhibition of G6 and its derivatives ............................................. 80

5-1 GI50 of other Jak2 inhibitors in HEL cells .......................................................... 133

Page 10: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

10

LIST OF FIGURES

Figure page 1-1 Tyrosine kinase catalyzed phospho-transferase reaction ................................... 36

1-2 Schematic representation of the Jak2 protein showing the regions where the somatic mutations identified in myeloproliferative neoplasm (MPN) patients are commonly found ........................................................................................... 37

1-3 Schematic representation of the Jak/STAT signaling paradigm ......................... 38

2-1 A46 inhibits Jak2-V617F-dependent cell proliferation ......................................... 58

2-2 A46 inhibits the Jak/STAT signaling pathway ..................................................... 59

2-3 A46 arrests HEL cells in G1 phase of the cell cycle ........................................... 60

2-4 A46 induces apoptosis in HEL cells .................................................................... 61

2-5 A46-induced apoptosis is mediated by the down regulation/cleavage of Bcl-2 family proteins Bim, Bax and Bid ........................................................................ 62

2-6 A46 suppresses cytokine-independent pathologic cell growth of Jak2-V617F positive bone marrow cells, ex vivo .................................................................... 63

3-1 Time- and dose-dependent effect of G6 and its derivatives on the HEL cell proliferation and Jak2 phosphorylation ............................................................... 81

3-2 Dose-dependent effect of G6 and its derivatives on the proliferation of Ba/F3-EpoR-Jak2-V617F cells ...................................................................................... 82

3-3 Inhibition of phosphorylation of STAT3 and STAT5 by G6 and its derivatives .... 83

3-4 Induction of apoptosis in HEL cells by G6 and its derivatives ............................. 84

3-5 Treatment with G6 or its stilbenoid derivatives leads to HEL cell death via the intrinsic apoptotic pathway.................................................................................. 85

3-6 Suppression of Jak2-V617F mediated pathologic cell growth in patient-derived bone marrow cells by G6 and its derivatives, ex vivo............................. 86

3-7 Molecular surface representation of the ATP binding site of Jak2 ...................... 87

3-8 Molecular docking of G6 and its derivatives into the ATP binding pocket of Jak2 .................................................................................................................... 88

3-9 Docking of G6 and its derivatives into the ATP binding pocket of Jak2 .............. 89

Page 11: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

11

4-1 Identification of vimentin as a differentially expressed protein between vehicle treated and G6 treated HEL cells ..................................................................... 108

4-2 G6 treatment induces time- and dose-dependent degradation of vimentin ...... 109

4-3 G6 treatment induces marked reorganization of vimentin intermediate filaments within cells ......................................................................................... 110

4-4 G6-induced cleavage of vimentin is Jak2-mediated ......................................... 111

4-5 G6-induced cleavage of vimentin is independent of de novo protein synthesis and caspase activity, but calpain-dependent .................................................... 112

4-6 Mobilization of calcium is essential and sufficient for the cleavage of vimentin 113

4-7 Cleavage of vimentin is sufficient to reduce HEL cell viability........................... 114

4-8 G6 treatment decreases the levels of vimentin protein, in vivo ......................... 115

5-1 Surface representation of the structure of Jak2, indicating possible sites for inhibitor targeting .............................................................................................. 132

Page 12: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

12

LIST OF ABBREVIATIONS

Jak2 Janus Kinase 2

JH1 Jak Homology 1

JH2 Jak Homology 2

SH2 Scr Homology 2

FERM 4.1 Ezrin Radixin Moesin

GPCR G-protein Coupled Receptor

STAT Signal Transducers and Activators of Transcription

MPN Myeloproliferative Neoplasms

PV Polycythemia Vera

ET Essential Thrombocythemia

PMF Primary Myelofibrosis

HEL Human Erythroleukemia

DMSO Dimethyl Sulfoxide

ELISA Enzyme-linked Immunosorbent Assay

G1/S Gap1/Synthesis

DTT Dithiothreitol

SDS-PAGE Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis

FITC Fluorescein Isothiocyanate

IDPN Beta, beta’-iminodipropionitrile

Page 13: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

13

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

CHARACTERIZATION OF STRUCTURE-FUNCTION CORRELATIONS OF NOVEL JAK2 SMALL MOLECULE INHIBITORS AND THEIR MECHANISMS OF ACTION

By

Anurima Majumder

August 2011

Chair: Peter P. Sayeski Major: Medical Sciences-Physiology and Pharmacology

Jak2 is a cytoplasmic tyrosine kinase that is involved in signaling via a diverse

range of ligands, including cytokines and growth factors. Hyperkinetic Jak2 tyrosine

kinase has been linked to various neoplastic disorders, including solid tumors and

hematological malignancies. In 2005, a Jak2 gain-of function mutation, Jak2-V617F,

was identified in a majority of patients with myeloproliferative neoplasms (MPNs). This

discovery spurred a great deal of interest in identifying small molecule inhibitors that

target Jak2, as effective inhibitors may have significant therapeutic potential for Jak2-

mediated pathologies and may also serve as useful research tools to study Jak2-

mediated signaling in general. In this dissertation, we first characterize a novel

benzothiophene based small molecule Jak2 inhibitor, A46, which we have identified by

structure-based drug design. We show that A46 specifically inhibited the proliferation of

Jak2-V617F expressing cells in both a time- and dose-dependent manner. Cells

exposed to 10 µM of A46 for 48 hours or more were unable to recover after drug

removal. A46 also caused a corresponding decrease in the phosphorylation of Jak2

and STAT3 proteins within these cells. Cell growth inhibition correlated with an induction

of cell cycle arrest and promotion of apoptosis. Moreover, we report that this compound

Page 14: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

14

inhibited the pathologic growth of primary Jak2-V617F expressing bone marrow cells,

ex vivo. Collectively, our data demonstrate that the benzothiophene based compound,

A46, suppresses Jak2-mediated pathogenesis, thereby making it a potential candidate

drug against Jak2-mediated disorders.

Our lab has previously identified a novel Jak2 inhibitor called G6 using structure-

based drug design. G6 showed promising results in preclinical studies and inhibited

Jak2 tyrosine kinase-mediated pathologic cell growth in vitro, ex vivo and in vivo. Here,

we identified a structure-function correlation of this compound. Specifically, we showed

that the stilbenoid core in G6 is essential for its ability to i) inhibit Jak2-V617F-

dependent cell proliferation, ii) suppress phosphorylation of key signaling molecules

involved in the Jak/STAT pathway, such as Jak2, STAT3 and STAT5, iii) induce

apoptosis in HEL cells via the intrinsic apoptotic pathway, iv) inhibit pathologic growth of

patient-derived Jak2-V617F-positive bone marrow cells, ex vivo and v) form strong

docking interactions with the ATP-binding pocket of Jak2 kinase domain. As such, we

demonstrated that G6 has a stilbenoid core that is indispensable for maintaining its Jak2

inhibitory potential.

Having shown that G6 specifically inhibits Jak2 kinase activity and suppresses

Jak2-mediated cellular proliferation, we next wanted to elucidate the molecular and

biochemical mechanisms by which G6 inhibits Jak2-mediated cellular proliferation. For

this, we treated Jak2-V617F expressing human erythroleukemia (HEL) cells for 12

hours with either vehicle control or 25 µM of the drug and compared protein expression

profiles using two-dimensional gel electrophoresis. One differentially expressed protein

identified by electrospray mass spectroscopy was the intermediate filament protein,

Page 15: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

15

vimentin. It was present in DMSO treated cells, but absent in G6 treated cells. HEL cells

treated with G6 showed both time- and dose-dependent cleavage of vimentin as well as

a marked reorganization of vimentin intermediate filaments within intact cells. In a

mouse model of Jak2-V617F mediated human erythroleukemia, G6 also decreased the

levels of vimentin protein, in vivo. The G6-induced cleavage of vimentin was found to be

Jak2-dependent and calpain-mediated. Furthermore, we found that intracellular calcium

mobilization is essential and sufficient for the cleavage of vimentin. Finally, we show

that the cleavage of vimentin intermediate filaments, per se, is sufficient to reduce HEL

cell viability. Collectively, these results suggest that G6-induced inhibition of Jak2-

mediated pathogenic cell growth is concomitant with the disruption of intracellular

vimentin filaments. As such, this work describes a novel pathway for the targeting of

Jak2-mediated pathological cell growth.

Page 16: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

16

CHAPTER 1

INTRODUCTION

Tyrosine Kinases

Tyrosine kinases are enzymes that catalyze the transfer of the γ-phosphate of

ATP to tyrosine residues of protein substrates (Fig. 1-1). These enzymes are critical

components of important signaling pathways that regulate vital physiological processes

such as cell proliferation, survival, differentiation and apoptosis. Abnormal tyrosine

kinase activity has however been linked to cancer, immunodeficiency, artherosclerosis,

cardiovascular disease etc.

Tyrosine kinases can be divided into two broad subfamilies; receptor tyrosine

kinases (RTKs) and non-receptor tyrosine kinases (NRTKs). The RTKs usually possess

a ligand binding extracellular domain, a transmembrane domain and a catalytic

intracellular kinase domain. These membrane-spanning kinases, therefore, have

intrinsic kinase activity and can undergo autophosphorylation upon activation by ligand

binding. The RTK family includes the insulin receptor and several growth factor

receptors, such as epidermal growth factor receptor and platelet-derived growth factor

receptor [1]. The NRTKs are usually cytoplasmic and possess a kinase domain, but lack

receptor-like features such as extracellular and transmembrane domains [1]. However,

these tyrosine kinases have additional domains that enable them to interact with other

signaling molecules, such as 1) an N-terminal myristylation/palmitoylation domain for

anchoring the kinase to the plasma membrane, 2) a pleckstrin homology (PH) domain

for binding to lipids, 3) an SH2 domain to bind phospho-tyrosines on other proteins, and

Portion of this chapter has been reproduced from Jak2 Inhibitors for the treatment of Myeloproliferative Neoplasms. Drugs of the Future 2010; 35(8): 651-660 with permission from 2010 Prous Science, S. A. U. or its licensors.

Page 17: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

17

4) an SH3 domain to bind to proline-rich sequences on other proteins [2]. Unlike growth

factor receptors, cytokine receptors do not have an intrinsic kinase domain. However,

these receptors associate with non-receptor tyrosine kinases and can thereby induce

tyrosine phosphorylation of the receptors and a variety of cellular proteins [3]. The

NRTK family includes the Src, Abl, Csk and FAK kinase subfamilies among many

others. The Janus kinases form a distinct family of cytoplasmic tyrosine kinases that

have a critical role to play in diverse signaling pathways that are essential for the normal

physiology and function.

Jak2 Tyrosine Kinase

History

Jak2, a cytosolic protein of approximately 130 kDa in mass, is a member of the

Janus family of cytoplasmic tyrosine kinases. Other known members of this family are

Tyk2, Jak1 and Jak3. These kinases are ubiquitously expressed in a variety of different

cell types with the exception of Jak3, which is predominantly expressed in cells of

hematopoietic origin [4]. Tyk2 (Tyrosine kinase 2), the first member of the Janus

tyrosine kinase family to be cloned, was identified by screening a T-cell cDNA library

using the sequence of the c-fms tyrosine kinase domain via a low stringency

hybridization technique [5]. The kinase domains of Jak1 and Jak2 were cloned by

polymerase chain reaction (PCR) using degenerate oligonucleotides complementary to

highly conserved kinase domains of the Src family members [6]. These kinase domain

sequences were then used to clone the full length Jak1 and Jak2 proteins [7, 8]. Jak3

was also cloned using similar PCR-based techniques [9, 10]. Initially, this family was

named as ‘Just Another Kinase’. However, subsequent sequence homology studies

revealed that a unique feature of all the members of this family of tyrosine kinases is

Page 18: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

18

that they have two potential kinase domains at their C-terminus. Thus, this family was

renamed as Janus Kinase after Janus, the Roman God with two opposite faces.

Structure

The unique structure of the Jak kinases clearly distinguishes them from other

members of the protein tyrosine kinase family. Similar to other members of the Jak

family, Jak2 has seven highly conserved Jak homology (JH) domains, JH1 through JH7,

numbered from the C- to the N-terminus, respectively [11] (Fig. 1-2). A hallmark feature

of all Janus kinases is the presence of two domains (JH1 and JH2) in the carboxyl half

of the protein that both have extensive homology to the kinase domain of other protein

tyrosine kinases. However, only the JH1 domain has functional kinase activity whereas

the JH2, or pseudokinase domain, has an important role in limiting the activity of the

kinase domain. The JH1 domain is a highly conserved kinase domain which contains

the activation loop, the critical sites of primary phosphorylation (Tyr1007 and Tyr 1008),

and the ATP-binding region. Phosphorylation of Tyr1007 residue in the activation loop is

essential for maximal Jak2 kinase activity [12]. The JH2 domain, which is adjacent to

the JH1 domain, negatively regulates both the basal as well as the ligand induced levels

of phosphotransferase activity [13, 14] by physically interacting with the JH1 domain

[15]. Even though this domain is structurally homologous to the JH1 domain, it lacks

certain critical amino acid residues that are essential for functional kinase activity and

hence is not catalytically active [16].

The amino half of Jak2 consists of domains JH3 through JH7. The JH3-JH4 region

weakly resembles the canonical Src Homology 2 (SH2) domain [17] and is hence called

the SH2-like domain, but its function is yet to be elucidated [18]. The JH4-JH7 domains

are N-terminal to the putative SH2 domain and are collectively called the FERM (4.1,

Page 19: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

19

Ezrin, Radixin, Moesin) domain. This domain is primarily involved in the binding to and

crosstalk with other cellular proteins [19]. For example, regions within the JH6 and JH7

domains mediate interactions with cell surface receptors by associating with conserved

Box 1 structural motifs on the cytokine receptors [20, 21].

The three-dimensional crystal structure of the entire Jak2 molecule has not yet

been resolved. However, a portion of the Jak2 kinase domain has been crystallized

recently [22]. This crystal structure gives important insights into the precise atomic

architecture of the Jak2 tyrosine kinase domain. Structurally, the kinase domain is

highly conserved among all tyrosine kinases thereby making it challenging to

specifically target Jak2 over the other kinases. However, this paper reports that the

Jak2 kinase domain has some unique features that can help distinguish it from the

kinase domains of other tyrosine kinases and thus help in achieving specificity. Firstly,

Jak family members possess an extra loop structure between amino acids 1056 and

1078, called the Jak2 insertion loop, which is relatively mobile and solvent accessible

[22]. The serine at position 1056 is conserved between all members of the Jak family

and is solvent exposed, suggesting that this residue might have an important role to

play in phosphorylation-dependent regulatory role in Jak2 function [22]. Analysis of the

Jak2 kinase domain crystal structure shows that the swing of the N-terminal lobe

towards the C-terminal lobe markedly narrows the active site of Jak2, thereby making it

more constricted and closed when compared to the active site of other kinases [22].

Comparisons of the electrostatic surface potentials around the ATP-binding sites

suggest that Jak2 is more positively charged than Jak1, whereas Jak3 is less charged

overall [23]. These structural differences between Jak2 and other closely related

Page 20: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

20

kinases, though subtle, might prove useful in achieving selectively for Jak2 over other

kinases. Recent crystal structures of the Jak2 kinase domain have undoubtedly

improved our understanding of the structure, function and regulation of Jak2 kinase.

However, future crystal structure analysis of the entire Jak2 molecule will be critical in

further developing our knowledge about these kinases and in attaining selectivity for

them.

Jak/STAT Signaling Paradigm

Jak2 is an important downstream signaling molecule activated by a variety of

cytokines, growth factors and G-protein coupled receptor (GPCR) ligands. An overview

of the canonical Jak/STAT signaling pathway, as activated by cytokines and growth

factors, is shown in Fig. 1-3. Briefly, signaling is initiated by binding of a ligand to its

cognate cell surface receptor resulting in receptor dimerization. Receptor dimerization

brings the receptor-associated Jak proteins in close proximity to each other allowing

them to transphosphorylate one another on specific tyrosine residues; Tyr 1007/1008 in

the case of Jak2 [12]. An activated Jak can in turn phosphorylate specific tyrosine

residues on the cytoplasmic tails of the receptors, thereby creating docking sites for

SH2-domain containing proteins such as the Signal Transducers and Activators of

Transcription (STAT) proteins. Receptor-bound STAT monomers are then

phosphorylated by Jak2 on specific tyrosine residues. Phosphorylated STATs form

homo- or hetero-dimers which translocate to the nucleus where they bind to gene

promoter elements to modulate gene transcription. Thus, Jak/STAT signaling results in

a signal cascade from extracellular binding and activation of a cell surface receptor to

changes in gene transcription in the nucleus.

Page 21: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

21

The important role that Jak kinases play in mediating cytokine signaling was first

characterized using mutant cell lines that did not respond to interferons. Analysis of this

particular mutant cell line showed that it lacked the tyrosine kinase Tyk2 and expressing

this kinase exogenously restored the ability of this cell to respond to interferons [24].

Specific Jak kinases, either alone or in combination with other Jak kinases, is

preferentially activated depending on the type of receptor that is being stimulated. For

example, binding of erythropoietin (Epo) to its cognate receptor activates Jak2 [25],

whereas binding of interleukin-3 (IL-3), interleukin-5 (IL-5) or granulocyte macrophage-

colony stimulating factor (GM-CSF) to their associated receptors activate both Jak2 and

Jak1 [26-28]. Thus, it was shown that Jak2 can associate with diverse cytokine and

growth factor receptors. Subsequently, it was shown that Jak2 is a versatile signaling

molecule that can also be activated by G-protein coupled receptors, such as the AT1

receptor [29], and by non-conventional ligands, such as reactive oxygen species [30].

The Jak/STAT signaling pathway can modulate cell growth, survival, differentiation and

death and is therefore critical for maintaining normal physiology. Deregulation of this

signal transduction pathway has been implicated in various disease states. On one

hand, Jak2 knockout mice die embryonically at day 12.5 due to lack of definitive

erythropoiesis [31, 32], suggesting that this kinase has a non-redundant role to play in

the erythropoiesis process. On the other side, hyperactive Jak2 signaling promotes cell

proliferation and prevents apoptosis, thereby leading to various neoplastic disorders,

including solid cancers and hematological malignancies.

Jak2 and Cancer

Jak2 in Solid tumors

Page 22: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

22

A deregulated Jak/STAT signaling pathway has been linked to several solid

tumors, including breast, prostate, ovarian, head and neck cancers [33-38]. Jak2 can

cause constitutive tyrosine phosphorylation of ErbB2, an oncogene over expressed in

human breast cancer, leading to tumor progression and metastasis [39]. It is also known

that overexpression of BRCA1 leads to a constitutively active Jak2-STAT3 signaling in

human prostate cancer cells [40]. Epidermal growth factor stimulation constitutively

activates STAT3, thereby leading to head and neck carcinogenesis by activation of anti-

apoptotic mechanism [37]. Additionally, it has been demonstrated that hyperactivation of

the Jak/STAT3 signal transduction pathway contributes directly to ovarian cancer

growth and invasiveness [38].

Jak2 in Hematological Malignancies

Apart from its role in promoting growth and metastasis of solid tumors, a

constitutively active Jak kinase signaling has also been implicated in various

hematological malignancies. Hematopoiesis, the process of generating blood cells from

a self-renewing population of multipotent hematopoietic stem cells, is regulated by a

group of soluble factors called cytokines. The binding of cytokines to their cognate

receptors initiates signal transduction pathways that govern survival, proliferation,

differentiation, and apoptosis of the hematopoietic cells. A deregulation of these

signaling pathways can lead to the development of different hematological disorders,

such as leukemias and myeloproliferative neoplasms. Cytokine receptors lack an

intergral cytoplasmic kinase domain and hence signal via association with cytoplasmic

tyrosine kinases, such as the Jak kinase family members [3, 41]. Jak2 is widely involved

in cytokine signaling. Cytokines, such as erythropoietin, thrombopoietin, growth

Page 23: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

23

hormone, prolactin and GM-CSF, signal exclusively through Jak2 [42, 43]. Hence, it is

unsurprising that hyper activation of Jak2 kinase activity can cause overproduction of

blood cells, thereby leading to the development of neoplastic hematological

malignancies.

Jak2 chromosomal translocations and hematological malignancies

Several studies have linked specific Jak2 chromosomal translocations to human

neoplastic growth. For example, an abnormal fusion protein that has the helix-loop-helix

dimerization domain of TEL, an ETS family transcription factor, fused to the catalytic

domain of Jak2 was detected in a patient with early B-precursor acute lymphobastic

leukemia and another diagnosed with atypical chronic myeloid leukemia [44, 45]. The

phenotypes of these two patients were diverse because of the fact that the TEL-Jak2

fusion proteins had resulted from two distinct translocation events; a t(9:12)(p24;p13) in

the former case and an a t(9:15:12)(p24;q15;p13) in the latter. However, in both cases,

the translocation event resulted in constitutive activation of the Jak2 kinase domain and

its downstream signaling pathways [46]. Recent studies have also reported the

presence of a BCR-Jak2 fusion protein arising from a t(9:22)(p24;q11.2) translocation in

patients with typical and atypical chronic myelogenous leukemia [47-49]. In other

instances, a t(8:9) translocation event results in a PCM1-Jak2 fusion protein that

contains the coiled coil domains of PCM1 and the tyrosine kinase domain of Jak2 [50-

52]. This fusion protein has been implicated in several hematological disorders including

acute erythroid leukemia, atypical chronic myeloid leukemia, T-cell lymphoma, and

myeloproliferative neoplasms.

Jak2 point mutations and hematological malignancies

Page 24: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

24

In addition to chromosomal translocations, point mutations in the Jak2 allele also

cause constitutive Jak2 activation and subsequent activation of its downstream

signaling pathways. Substitution and deletion mutations in Jak2 have been detected in

many patients with different hematological disorders. For example, Jak2 amino acids

682-686 were found to be deleted (Jak2-∆IREED) in a patient with Down syndrome and

B-cell precursor acute lymphoblastic leukemia [53]. In another instance, a novel Jak2-

T875N mutation was identified in an actue megakaryoblastic leukemic cell line using a

combination of mass spectrometry and growth inhibition assays [54]. Jak2-K607N [55]

and Jak2-L611S [56] are other Jak2 point mutations that have been identified in patients

with acute myeloid leukemia and acute lymphoblastic leukemia, respectively. Overall,

these studies demonstrate that the Jak2 locus is susceptible to diverse genetic

altrerations that hyper activate the catalytic activity of Jak2 kinase, thereby leading to

the development of hematological malignancies.

Myeloproliferative Neoplasms

Myeloproliferative Neoplasms (MPNs) are a group of heterogeneous diseases

arising from a transformed hematopoietic stem cell and characterized by the presence

of excessive numbers of one or more terminally differentiated blood cells of the myeloid

lineage such as erythrocytes, thrombocytes or white blood cells. These differentiated

cells spread and accumulate in the bone marrow as well as in the peripheral blood and

other sites of extramedullary hematopoiesis due to excess proliferation and/or

decreased apoptosis of hematopoietic progenitors. MPNs have a high propensity to

progress to malignant leukemias. Excess production of red blood cells is a hallmark of

polycythemia vera (PV) and too many platelets is a classical characteristic of essential

Page 25: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

25

thrombocythemia (ET) whereas primary myelofibrosis (PMF) is characterized by bone

marrow fibrosis and impaired hematopoietic stem cell function.

Jak2 Mutations in Myeloproliferative Neoplasms

The year 2005 was a landmark year for the field of MPNs as five different groups,

working independently, identified a somatic Jak2 Val 617 to Phe substitution mutation

(Jak2-V617F) in a large number of patients with myeloproliferative neoplasms (MPNs)

[57-61]. It is interesting to note that each group used a different approach to arrive at the

result. The group led by William Vainchenker [57] began by culturing erythroid cells from

PV patients in the absence of exogenous cytokines. The cells were then treated with

different small molecule inhibitors in an effort to identify the factors and signaling

pathways required for PV erythroid cell proliferation. It was observed that Jak2 inhibition

can suppress PV erythroid proliferation and that siRNA mediated silencing of Jak2 can

also inhibit erythroid cell proliferation. This led them to sequence JAK2 in PV patient

samples and hence the presence of the Jak2-V617F mutation was discovered [57].

Robert Kralovics and his group [59] [62] followed up on the important discovery that loss

of heterozygosity at the 9p24 chromosomal locus as a result of uniparental disomy

(UPD) is present in a large number of PV patients [62]. Genes in this region of UPD

were sequenced in MPN patient samples and the Jak2-V617F allele was detected [59].

Two other groups used candidate gene approaches to identify the Jak2-V617F

mutation. Anthony Green and coworkers [58] were sequencing genes involved in

signaling pathways known to be implicated in oncogenic transformations when they

discovered this mutant allele in MPN. On the other hand, Gary Gilliland’s group [60]

detected the presence of the Jak2-V617F mutation while sequencing conserved

domains of all tyrosine kinases on a large set of MPN patients who had submitted

Page 26: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

26

samples via an internet-based collection protocol. Joe Zhao’s group [61] performed a

large scale DNA sequencing analysis of candidate tyrosine kinases and phosphatases

and thereby identified the presence of the gain-of-function Jak2 mutation in a majority of

PV patients.

These five groundbreaking studies, as well as other subsequent studies, reported

the presence of the Jak2-V617F mutation in over 90% of patients with PV and about

50% of patients with ET and PMF. A guanine to thymine mutation acquired in the

hematopoietic stem cells results in a substitution of valine to phenylalanine at codon

617 in the exon 14 of the JH2 pseudokinase domain of Jak2. It is believed that this

mutation at position 617 in the autoinhibitory pseudokinase domain of Jak2 allows the

kinase to evade inhibition and leads to a constitutively active Jak-STAT signaling

pathway [62, 63] and induces cytokine independent growth of cells [57, 60]. The co-

expression of Jak2-V617F and an ectopic erythropoietin receptor (EpoR) in the IL-3-

dependent hematopoietic cell line Ba/F3 transforms these cells to cytokine independent

growth [57, 60, [63]. Subsequently, it has also been shown that expression of this

mutation in both murine bone marrow transplant models [64, 65] as well as transgenic

models [66-71] is sufficient for the development of MPN-like phenotypes in recipient

mice. These reports strongly suggest that the Jak2-V617F mutation plays a critical role

in the pathogenesis of myeloproliferative neoplasms.

Although the Jak2-V617F mutation on exon 14 is the predominant disease-

associated allele in myeloproliferative neoplasms, several other Jak2 exon 14

mutations, such as C616Y [72] and D620E [73], have been identified in V617F-

negative, MPN patients. Moreover, about sixteen different Jak2 exon 12 mutations have

Page 27: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

27

been detected to date in Jak2-V617F-negative PV patients [74]. These exon 12

mutations are usually insertions, deletions or substitutions in the Jak2 sequence

spanning amino acids 538-543 (Fig. 1-2). This region is highly conserved and links the

SH2-like and the pseudokinase domains of Jak2. A recent study suggests that this

linker region acts as a switch in relaying signals from receptor binding to Jak2 kinase

activation by flexing the pseudokinase domain hinge [75]. Finally, a subset of patients

with Jak2-V617F-negative ET and PMF were found to carry MPLW515L/K mutations in

the thrombopoietin receptor, which also leads to the deregulation of the Jak/STAT

signaling pathway via activation of wild type Jak2 protein [76, 77]. Collectively, these

studies clearly demonstrate that hyperkinetic Jak2 has a crucial role to play in the

pathogenesis of MPNs as well as in a number of hematological malignancies.

Therefore, the clinical development of Jak2 inhibitors is of potential value for patients

with these hematological disorders and is currently an area of active research.

Jak2 Inhibitors for the Treatment of Myeloproliferative Neoplasms

Currently available treatment options for MPN patients are limited. For PV/ET

patients, the available therapies aim to reduce myeloproliferation and

thrombotic/hemorrhagic complications associated with it. As such, a PV/ET patient is

mostly treated with phlebotomy/plateletpheresis or myelosuppressive agents such as

hydroxyurea [78-80]. On the other hand, in PMF patients, treatment is directed towards

management of symptoms such as anemia and splenomegaly. Therefore, agents that

can improve cytopenia (erythropoietin, androgens) or induce myelosuppression (such

as hydroxyurea, thalidomide) are routinely used for treating these individuals [80, 81]. In

certain cases, more invasive techniques, such as splenectomy or radiation, are used to

treat severe splenomegaly in MPN patients. Unfortunately, these treatment strategies

Page 28: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

28

are more palliative and not curative in nature. The only therapy that has been shown to

be curative in patients with myeloproliferative neoplasms is stem cell transplantation.

Although this approach is potentially curative, risks associated with it, such as graft-

versus-host disease and nonrelapse mortality, severely limit its application for therapy

[81]. Given the critical role that constitutive Jak2 tyrosine kinase activity plays in the

pathophysiology of myeloproliferative neoplasms, it became an attractive molecule for

therapeutic targeting and prompted researchers to start the search for potent and

selective Jak2 small molecule inhibitors. Interestingly, the first widely available Jak2

inhibitor dates back to 1995 when Meydan et al. identified AG490 via a high throughput

screen of potential tyrosine kinase inhibitors [82]. However, it was subsequently

reported that although AG490 was a potent Jak2 inhibitor, it had poor specificity [83].

The discovery of the Jak2-V617F mutation in 2005 and its identification in a vast

majority of patients with MPNs provided a new thrust to the search for identification of

small molecule inhibitors that specifically target Jak2. Given the fact that the Jak2-

V617F mutation leads to sustained kinase signaling, similar to the effect of the BCR-

ABL fusion protein in chronic myelogenous leukemia (CML), it was expected that a Jak2

inhibitor would be as effective in MPN therapy as imatinib is in CML [84]. Moreover, in

2006, the resolution of a crystal structure encoding a portion of the Jak2 kinase domain

provided a valuable tool for designing inhibitors that could be highly specific for Jak2

[22].

Several pharmaceutical companies have developed Jak2 inhibitors that are

currently under study for the treatment of MPNs. The clinical trials conducted thus far

have focused on patients with PMF because it is the most serious condition among the

Page 29: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

29

different MPNs. PMF patients are characterized by fibrous scar tissue in their bone

marrow, impaired hematopoiesis and high levels of pro-inflammatory cytokines and

growth factors. These symptoms significantly reduce the quality of life of these patients

and as mentioned above, effective therapies are limited for these patients. The results

from four clinical trials in these patients are publicly available; the inhibitors tested were

INCB018424, CEP-701, TG101348, and XL019 (Fig. 4).

INCB018424 is a potent Jak2 inhibitor which has an in-vitro IC50 of 2.8 nM [85]. In

primary cultures, this inhibitor has an ability to suppress erythroid progenitor colony

formation from Jak2-V617F positive PV patients with an IC50 of 67 nM vs. healthy

donors (IC50 > 400 nM). In a mouse model of Jak2-V617F mediated MPN, treatment

with 180 mg/kg/day of INCB018424 significantly decreased splenomegaly, reduced

levels of some circulating cytokines, preferentially eliminated neoplastic cells, and

increased animal survival without myelosuppressive or immunosuppressive effects. In

clinical phase I/II trials, this drug was generally well tolerated; the maximum tolerated

dose (MTD) was 25 mg PO BID [86]. INCB018424 treatment resulted in reduced

splenomegaly in over 93% of the patients irrespective of Jak2 mutational status [87].

Treatment with this drug inhibited production of pro-inflammatory cytokines in all PMF

patients [88]. There was also significant improvement in quality of life and weight of the

patients with treatment [86]. However, the Jak2-V617F allele burden showed only a

modest decrease of 13% in the marrow and 9% in the peripheral blood [89], suggesting

that this drug might be inhibiting downstream Jak/STAT signaling in cells with

hyperkinetic Jak2 rather than altering the mutant allele burden. There were also no

reports of improvement or normalization of either the diseased bone marrow or the

Page 30: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

30

spleen tissues in any of the treated patients. No significant changes in fibrosis score,

bone marrow cellularity or circulating CD34+ cells were observed [90]. Finally,

thrombocytopenia and myelo-suppression were the most common side effects of drug

treatment [86]. Phase III clinical studies for this drug have recently been initiated

(www.clinicaltrials.gov).

CEP-701 (also called lestaurtinib), an indolocarbazole alkaloid, is an orally

available tyrosine kinase inhibitor that has anti-Jak2 tyrosine kinase activity. This drug is

a known inhibitor of Fms-like tyrosine kinase 3 (FLT3) and is currently being evaluated

in clinical trials for treating acute myelogenous leukemia patients with FLT3 mutations

[91, 92]. Lestaurtinib inhibits Jak2 in vitro, ex vivo, and in vivo [93]. It potently inhibits

Jak2 kinase activity in vitro with an IC50 of 1 nM and suppresses ex vivo erythroid colony

formation from primary CD34+ cells isolated from MPN patients at a concentration of

100 nM. CEP-701 treatment is also able to reduce phosphorylation of STAT3, STAT5

and other downstream signaling molecules of the Jak/STAT signaling pathway in vitro.

With respect to in vivo studies, this drug was able to inhibit the proliferation of Jak2-

V617F bearing HEL 92.1.7 cells xenografted into nude mice at a dose of 30 mg/kg BID.

The results of a phase II clinical study of CEP-701, with twenty two Jak2-V617F positive

PMF patients, were recently published [94]. These patients were treated with an oral

dose of 80 mg twice a day. 27% of the patients (6 out of 22) showed clinical

improvement with this treatment (i.e. the lowest level of response as per the

International Working Group for Myelofibrosis Research and Treatment criteria). The

responding patients included three who only showed a reduction in spleen size, two

who achieved transfusion independency, but only one who had a reduction in spleen

Page 31: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

31

size in conjunction with improvement in cytopenia [94]. Median time for response was

about three months and the duration of the response was fourteen months or higher.

There was an observed decrease in the levels of phosphorylated STAT3 from baseline

levels in the responding patients [94]. Most critically, no improvements were seen either

in the marrow fibrosis or in the Jak2-V617F allele burden in any of the treated patients.

Finally, 36% of the treated patients (8 out of 22) experienced toxic effects; principally,

myelo-suppression (anemia and thrombocytopenia) and gastrointestinal problems such

as diarrhea and nausea [94].

TG101348 is a potent and selective inhibitor of Jak2. It is an ATP-competitive

inhibitor that shows a high selectivity for Jak2 over other Jak family members [65, 95].

Among all the Jak2 inhibitors that are currently in clinical trials, TG101348 seems to be

the most selective for Jak2. Previous studies with this compound have shown that it

inhibits Jak2 kinase activity with an IC50 of 3 nM in vitro [65, 95]. Furthermore, it was

found to suppress erythroid colony formation from primary progenitor cells with an IC50

of about 300-600 nM [96]. The in vivo therapeutic efficacy of TG101348 in a murine

model of Jak2-V617F mediated PV was also evaluated in a study which found that mice

treated with 120 mg/kg BID showed a decrease in Jak2V617F mutant allele burden,

hematocrit values, leukocyte counts and spleen sizes [65]. TG101348 was also

evaluated in a phase I/II dose escalation study with twenty eight myelofibrosis patients

[97, 98]. The maximum tolerable dose was found to be 680 mg/day, taken once daily.

64% of the patients showed a decrease in spleen size of more than 50%. Fourteen

patients, who initially had leukocytosis, experienced improvement in their WBC counts

post treatment. Mutant allele burden was found to decrease in 32% of the Jak2-V617F

Page 32: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

32

positive patients enrolled in the study. The most frequent toxicities observed were

nausea/vomiting (64%) and diarrhea (50%) [97]. No changes in levels of plasma

cytokines were detected in patients treated with this drug [98]. Thrombocytopenia,

neutropenia and anemia were the other side effects that correlated with drug treatment

[97].

XL019 inhibits Jak2 kinase activity with an IC50 of 2 nM. In EPO stimulated primary

erythroid ex vivo cultures, STAT5 phosphorylation was inhibited with an IC50 of 64 nM

[99]. In in vivo studies using the HEL cell xenograft model, treatment with this inhibitor

reduced tumor growth in a dose-dependent manner; 60% inhibition at a dosage of 200

mg/kg BID and 70% inhibition at a dosage of 300 mg/kg BID relative to vehicle control

treated animals [99]. The effect of XL019 administration on human subjects was tested

in a phase I/II clinical study in patients with PMF post-PV or post-ET [100]. To date, 21

patients have been studied over multiple doses ranging from 25 mg to 300 mg, using

different schedules of administration (either once daily or three times weekly). Adverse

neurotoxicity was observed in patients receiving doses >100 mg (65). However, the

drug was well tolerated at the lower doses of 25 to 50 mg daily or 25 mg three times

weekly. A greater than 50% reduction in spleen size was achieved in 42% of the

patients. Reduction in leukocytosis and circulating blast cells and improvement of

anemia, pruritis, and fatigue were also observed [100]. The drug associated toxicities

were mostly related to adverse neurological effects such as peripheral neuropathy,

formication, balance disorder and confusion [99]. Adverse hematological side effects

were however not seen in treated patients. Although XL019 showed some beneficial

effects, it has been withdrawn from clinical trials due to the neurotoxicity concerns.

Page 33: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

33

A summary of the preclinical data pertaining to INCB018424, CEP-701,

TG101348, and XL019 is shown in Table 1-1 and a summary of the clinical trial

regimens for the same four drugs is displayed in Table 1-2. Data from these initial

reports about the clinical trials of Jak2 inhibitors show that the good preclinical

efficiencies demonstrated by these compounds in murine models of myeloproliferative

neoplasms did not translate to clinical trials in human patient populations. However,

there are also a number of other Jak2 inhibitors that are currently being evaluated in

early clinical trials, such as SB1518 and CYT387 (www.clinicaltrials.gov).

SB1518 is a potent, selective, orally active, ATP competitive Jak2 inhibitor that

inhibits Jak2-V617F activity with an IC50 of 19 nM (66). This compound was found to

inhibit the proliferation of Ba/F3 cells that had been transfected with erythropoietin

receptor (EpoR) and mutant Jak2-V617F with an IC50 of 81 nM [101]. In a mouse model

of MPD, established by injection of Ba/F3-JAK2V617F cells into nude mice, SB1518 was

found to produce therapeutic effects such as normalization of elevated white blood

cell count and reduction of GFP-labeled BaF3 cells in the peripheral blood, resolution of

hepatosplenomegaly, reduction of phospho-STAT5 in diseased organs, prolonged

survival and alleviation of terminal-stage anemia and thrombocytopenia [101].

CYT387, an aminopyrimidine derivative, inhibits Jak1/Jak2 with an IC50 of 11 and

18 nM respectively [102]. It inhibits the growth of Ba/F3-JAK2V617F and human

erythroleukemia (HEL) cells (IC50 ~1500 nM). The drug selectively suppressed the in

vitro growth of erythroid colonies harboring JAK2-V617F from polycythemia vera (PV)

patients, an effect that was attenuated by exogenous erythropoietin [102]. In a murine

MPN model, CYT387 normalized white cell counts, hematocrit, spleen size, and

Page 34: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

34

restored normal levels of inflammatory cytokines. Furthermore, treatment with this drug

was able to reduce the Jak2-V617F allelic burden, but not eliminate the mutant cells

[103]. With a number of other putative Jak2 inhibitors currently in various stages of

preclinical studies, it is hopeful that the list of Jak inhibitors in clinical trials will grow

significantly in the years to come and a potent and specific small molecule inhibitor that

is able to cure Jak2-mediated pathologies will soon be identified.

Rationale for Studies

Jak2, a cytoplasmic tyrosine kinase, has a critical role to play in hematopoiesis

and activating mutations in this kinase are associated with a number of hematological

disorders. Recently, several somatic point mutations, such as the Jak2-V617F, that

constitutively activate the Jak/STAT signaling pathway have been identified in the

majority of patients with myeloproliferative neoplasms (MPNs). This discovery made

Jak2 an attractive molecule for therapeutic targeting in MPNs and has been the driving

force behind the development of small molecule inhibitors that specifically target Jak2.

Since then, many different Jak2 inhibitors have been identified using a variety of

screening techniques and have been characterized preclinically as well as clinically.

However, as we have discussed in the previous section, initial reports from the clinical

trials of Jak2 inhibitors have not lived up to the expectations. These inhibitors were able

to alleviate symptoms in patients, but failed to actually reverse the progression of the

disease. On the basis of these reports from early clinical trials of Jak2 inhibitors, we can

conclude that there is still an unmet clinical need for Jak2 inhibitors that can

successfully cure patients with myeloproliferative neoplasms.

The goal of the study presented in the following chapters is to identify and

characterize the structure-function correlation of novel Jak2 inhibitors. Here, we first

Page 35: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

35

present a study in which we identify and characterize a novel benzothiophene-based

Jak2 inhibitor, called A46. Using structure-based virtual screening, our group recently

identified a novel small molecule inhibitor of Jak2 named G6 [104]. We showed that G6

has a specific inhibitory effect on Jak2 kinase activity as measured by in vitro enzyme

assays and an immunoassay ELISA [104]. We also demonstrated that this compound

exhibits exceptional therapeutic efficieny, in vivo [105]. Here, we perform structure-

function analysis of this novel Jak2 inhibitor, G6. Additionally, we also attempt to

elucidate the molecular and biochemical mechanisms by which G6 exerts its anti-Jak2

activity. The overall aim of the study presented here is to better understand the

mechanism of action of novel Jak2 inhbitors – both from a structural as well as a

functional point of view.

Page 36: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

36

Figure 1-1. Tyrosine kinase catalyzed phospho-transferase reaction. Tyrosine kinases catalyze the transfer of the γ-phosphate of ATP to tyrosine residues of protein substrate.

Protein Substrate Protein Substrate

ATP ADP

Tyrosine Kinase

Page 37: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

37

Figure 1-2. Schematic representation of the Jak2 protein showing the regions where

the somatic mutations identified in myeloproliferative neoplasm (MPN) patients are commonly found. Jak2 has seven highly conserved Jak homology or JH domains, JH1-JH7. The Jak2-V617F point mutation, the predominant disease-associated allele in MPNs, is on exon 14 of the JH2 or pseudokinase domain of Jak2. Other exon 14 mutations have also been identified in MPN patients who lack the V617F mutation. Additionally, about sixteen different exon 12 mutations have been detected in Jak2-V617F-negative PV patients. These mutations cluster in a region spanning from amino acids 538-543.

Page 38: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

38

Figure 1-3. Schematic representation of the Jak/STAT signaling paradigm. (a) Binding of the ligand to its cognate cell-surface receptor causes receptor dimerization. (b) Receptor dimerization brings the receptor associated Jak molecules in close proximity, such that they can trasphosphorylate one another on specific tyrosine residues (c) Active Jaks can now phosphorylate tyrosine residues on the cytoplasmic tail of the receptor. (d) STAT proteins bind to these phosphorylated tyrosine residues on the receptor and are also phosphorylated by receptor associated active Jaks. (e) Phosphorylated STAT proteins form dimers which translocate to the nucleus. (f) STAT dimers bind to the promoter region of specific target genes and modulate gene transcription, producing biological effects such as cell proliferation, differentiation, survival and growth.

Page 39: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

39

Table 1-1. Preclinical characterization of Jak2 inhibitors. The Jak2 IC50 was derived from cell-free assays using recombinant Jak2 protein. The HEL cell IC50 was determined using in vitro cultures of the Jak2-V617F expressing HEL cells. The ex vivo IC50 values were measured in primary cultures isolated from MPN patients. The in vivo drug dosage (mg/kg) is shown along with the animal model that was employed for that specific study.

Drug Jak2 IC50

(nM) HEL cell IC50

(nM) Ex Vivo IC50

(nM)

In Vivo Dosage

(mg/kg) (Mouse model used)

INCB018424

2.8

186

67

180 daily (Ba/F3-Jak2V617F

xenograft)

CEP-701 1 30-100 100 30 twice daily (HEL xenograft)

TG101348 3 300 300-600 120 twice daily

(Jak2V617F induced PV-bone marrow

transplant)

XL019 2 60-200 64 200-300 twice daily (HEL xenograft)

Page 40: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

40

Table 1-2. Clinical characterization of Jak2 inhibitors. Shown is a list of the four drugs tested in clinical trials and an overview of the treatment regimen. PMF – Primary myelofibrosis, post-PV/ET MF – post polycythemia vera (PV)/essential thrombocythemia (ET) myelofibrosis.

Drug Company Phase of

Development

Clinical Trial Dose (mg)

Route of Administration

Patient Population

Studied

Number of Patients

Clinical/Molecular Responses

Observed Toxicities

INCB018424 Incyte Phase I / II Phase III

25 twice daily

Oral PMF and post-PV/ET

MF

>100 Decrease in splenomegaly irrespective of

Jak2 mutational status, improved quality of life and weight, reduce

levels of inflammatory

cytokines, modest decrease in Jak2

V617F allele burden, no change

in bone marrow fibrosis

Thrombocytopenia, myelosuppression

CEP-701

Cephalon

Phase II

80 twice

daily

Oral

Jak2V617F- positive PMF

22

Reduced spleen

size, Improvement in cytopenia, two patients achieved

transfusion independency,

decrease in phospho-STAT3,

no improvement in bone marrow fibrosis, no significant

improvement in allele burden

Myelosuppression

(Anemia and thrombocytopenia),

gastrointestinal problems (diarrhea

and nausea)

Page 41: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

41

Table 1-2. Continued

Drug Company Phase of

Development

Clinical Trial Dose (mg)

Route of Administration

Patient Population

Studied

Number of Patients

Clinical/Molecular Responses

Observed Toxicities

TG101348 TargeGen Phase I / II 680 once daily

Oral PMF and post-PV/ET

MF

28 More than 50% decrease in spleen size, reduction in Jak2-V617F allele burden, marked

reduction in leukocytosis,

improvement in constitutional symptoms like

pruritus, no change in levels of plasma

cytokines

Nausea / vomiting, diarrhea,

thrombocytopenia, neutropenia,

anemia

XL019

Exelixis

Phase I / II

(Discontinued)

25-50

once daily

Oral

Primary or post-PV/ET

MF

21

Decrease in

splenomegaly, reduction in

leukocytosis and circulating blast

cells, improvement of anemia, pruritus

and fatigue

Neurotoxicities

such as peripheral neuropathy, formication,

balance disorder and confusion

Page 42: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

42

CHAPTER 2 A46, A BENZOTHIOPHENE DERIVED COMPOUND, SUPPRESSES JAK2-

MEDIATED PATHOLOGIC CELL GROWTH

Jak2, a member of the Janus family of cytoplasmic tyrosine kinases, is

ubiquitously expressed and is a key mediator of signal transduction and gene

transcription. A variety of cytokines, growth factors and GPCR ligands can activate

Jak2, trigger the canonical Jak/STAT signaling cascade and cause physiological effects

such as regulation of cell survival, development, growth, and proliferation. The critical

role of Jak2 in embryonic development has been demonstrated by gene deletion studies

in which Jak2 knock-out mice were found to be embryonic lethal due to absence of

definitive erythropoiesis that led to profound anemia [31, 32]. Aberrant Jak2 activity has,

however, been associated with several different pathological conditions. Constitutive

activation of the Jak-STAT pathway promotes aberrant cell proliferation and can lead to

the development of hematological malignancies and myeloproliferative neoplasms

(MPNs) [44, 45, 48]. MPNs include three pathogenetically related disorders;

polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis

(PMF). These disorders arise from a transformed hematopoietic stem cell and are

characterized by increased production of blood cells of the myeloid origin.

A somatic Jak2 mutation (Jak2-V617F) was identified in patients with

myeloproliferative neoplasms including 90% of polycythemia vera (PV) patients and

about 50% of patients with essential thrombocythemia (ET) and primary myelofibrosis

(PMF) [57-61]. A valine to phenylalanine substitution at codon 617 (V617F) in the

pseudokinase domain of Jak2 in hematopoietic stem cells results in a constitutively

active Jak-STAT signaling pathway that induces cytokine hypersensitivity and growth

factor independent growth of these cells. The presence of this mutation has also been

Page 43: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

43

detected in other hematological malignancies such as acute myeloid leukemia, chronic

myelomonocytic leukemia and chronic neutrophilic leukemia [106]. The co-expression of

Jak2-V617F and an ectopic erythropoietin receptor (EpoR) in the IL-3-dependent

hematopoietic cell line Ba/F3, transforms these cells to cytokine independent growth

[63]. Subsequently, it has also been shown that expression of this mutation in both

murine bone marrow transplant models [64, 107] as well as transgenic models [66, 67,

70, 71] is sufficient for the development of MPN-like phenotypes in recipient mice.

These reports strongly suggest that the Jak2-V617F mutation plays a causative role in

the pathogenesis of myeloproliferative neoplasms.

Unfortunately, as of present, there are no effective treatment options available for

these patients. Specifically, current treatment options are directed towards management

of symptoms and are palliative rather than being curative. The presence of the Jak2-

V617F mutation in a majority of MPN patients suggests that identification of Jak2

specific inhibitors is an important step towards developing an effective targeted therapy

for MPNs. Our laboratory has been actively involved in the identification of Jak2

inhibitors and has previously reported three small molecules with anti-Jak2 activity [104,

105, 108, 109]. Here, we used structure-based drug design to identify a benzothiophene

based structure, 1-benzothiophen-2-yl-(4-dimethylaminophenyl)methanol (termed as

A46), and show that this compound suppresses Jak2-mediated pathological cell growth

in vitro and ex vivo.

Materials and Methods

Drug

The small molecule A46 was obtained from the National Cancer

Institute/Developmental Therapeutics Program (NCI/DTP). The drug was solubilized in

Page 44: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

44

dimethyl sulfoxide (DMSO) at a concentration of 10 mM. The drug was stored at -20⁰C

in small aliquots to avoid repeated freeze thaw cycles.

Cell Culture

HEL and Raji cells were purchased from the American Type Culture Collection

(ATCC) and CMK cells from the German Collection of Microorganisms and Cell

Cultures DSMZ. SET-2 cells were a kind gift from Dr. Gary Reuther at the Moffitt Cancer

Center and Research Institute, Tampa, FL. These cells were cultured in RPMI 1640

(Mediatech) supplemented with 10% fetal bovine serum (FBS), penicillin, streptomycin

and L-glutamine at 37⁰C and 5% CO2.

Cell Proliferation Assay

HEL, Raji, CMK and SET-2 cells were plated in 96-well plates at a concentration of

approximately 5 × 104 cells per well. The cells were then treated with either 0.25%

DMSO or A46 for the indicated periods of time or concentrations. Cell viability was

assessed by trypan blue exclusion staining or an MTS assay as indicated.

Enzyme-linked Immunosorbent Assay

HEL cells, treated with either 0.25% DMSO or increasing doses of A46 for 48 hrs,

were lysed and analyzed by Enzyme-linked Immunosorbent Assay (ELISA) for detection

of phospho-Jak2 and phospho-STAT5 protein levels. For this, Jak2 [pY1007/pY1008]

and STAT5b [pY699] ELISA kits (Invitrogen) were used according to the manufacturer’s

protocol.

Cell Cycle Assay

The CycleTESTTM PLUS DNA Reagent Kit (BD Biosciences) was used to analyze

the DNA obtained from HEL cells suspensions. The cells were first incubated with

Page 45: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

45

0.25% DMSO or increasing doses of A46 for 48 hrs and then analyzed using a

FACSCalibur flow cytometer (BD Biosciences) as per the manufacturer’s protocol.

Apoptosis Assay

Induction of apoptosis in HEL cells was determined with the FITC AnnexinV

Apoptosis Detection Kit (BD Pharmigen) as per the manufacturer’s protocol. The cells

were incubated with either 0.25% DMSO or increasing doses of A46 for 48 hrs, stained

with AnnexinV and Propidium Iodide (PI) and then analyzed using a FACSCalibur flow

cytometer (BD Biosciences) to determine the percentage of cells undergoing apoptosis.

Western Blotting

HEL cells were treated with the indicated concentrations of A46 for 24 hrs. The

cells (~ 1 x 107) were then lysed in 0.8 ml of ice-cold RIPA buffer and protein

concentration was determined using a Bradford assay (Bio-Rad). Protein samples (~50

μg) were separated by gel electrophoresis. The separated proteins were then

transferred to nitrocellulose membranes and blotted with the indicated antibodies. All

the antibodies used were purchased from Cell Signaling Technology except for Cyclin

D1 which was from Santa Cruz Biotechnology. Western blots were visualized using the

enhanced chemi-luminescence system (Perkin-Elmer).

Colony Formation Assay

Following an Institutional Review Board approved protocol, a residual bone

marrow aspirate was obtained from a de-identified Jak2-V617F positive female

diagnosed with Essential Thrombocythemia. The marrow-derived mononuclear cells

were first washed in Iscove’s Modified Dulbecco’s Medium (IMDM) and then cultured in

human methylcellulose complete medium lacking erythropoietin and thrombopoietin

(R&D Systems). Cells were plated in 35-mm petri dishes at a concentration of

Page 46: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

46

approximately 4 × 105 cells/ml in the absence or presence of increasing doses of A46.

Thrombopoietin (50 ng/ml) was added to the indicated samples. The dishes were

incubated at 37⁰C and 5% CO2 in a humidified atmosphere for 14 days following which

the number of colony forming units-megakaryocytes (CFU-Megs) were counted.

Clonogenic Assay

Bone marrow cells from Jak2-V617F transgenic mice [67] were harvested,

cultured ex vivo in Iscove’s Modified Dulbecco’s Medium (IMDM), and treated with 25

µM of A46 for the indicated periods of time. Post treatment, the drug was washed away

and the cells (approximately 3 x 104 cells/ 35 mm dish) were plated in MethoCult

medium lacking cytokines. Five days later, the number of granulocyte/macrophage and

erythroid colony forming units were counted and plotted as a function of treatment

group.

Statistical Analysis

Results are expressed as mean +/- SEM. For statistical evaluation of time-

dependent responses to A46, a two-way analysis of variance (ANOVA) was used. For

the analysis of inhibition of phosphorylation, induction of cell cycle arrest, induction of

apoptosis and suppression of pathologic cell growth and clonogenic potential ex vivo, a

Student’s t-test was employed. Data were assumed to be statistically significant when p

< 0.05.

Results

A46 Inhibits Jak2-V617F-dependent Cell Proliferation

Using an in silico structure based drug design approach, we screened a library of

small molecules from the NCI/DTP repository as described previously [109] and

identified a small molecule, termed A46, which bound the ATP-binding pocket of Jak2

Page 47: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

47

with a favorable energy score. Table 2-1 lists the chemical name, NSC number,

chemical structure, and molecular weight of this compound. We next wanted to test the

ability of this compound to suppress Jak2-V617-mediated cell proliferation, in vitro. For

this, we used the human erythroleukemia (HEL 92.1.7) cell line, which is homozygous

for the Jak2-V617F mutation [110] and has a transformed proliferative phenotype that is

driven by constitutively active Jak2-V617F signaling [111]. HEL cells were treated either

with DMSO or with 10 µM of A46 for increasing periods of time. Viable cell numbers for

each treatment were determined using trypan blue exclusion and hemocytometer.

When compared to vehicle treated cells, we found that A46 significantly reduced viable

cell numbers in a time-dependent manner (Fig. 2-1A).

We next wanted to determine if the suppressive effects of A46 treatment on HEL

cell growth were reversible. For this, HEL cells were first exposed to 10 µM of A46 for 0,

8, 12, 24, 48 and 72 hrs. At the end of each time point, the drug was washed away and

the cells were resuspended in fresh growth medium. They were then allowed to grow for

an additional 72 hrs in the absence of the inhibitor. Viable cell numbers were

determined at the end of the 72 hour recovery period. We found that ~19 hrs of initial

exposure to A46 prevented 50% of the cells from recovering from the treatment (Fig. 2-

1B). For cells that had been treated with A46 for 48 hrs or more, fewer than 20% were

able to recover after removal of the drug (Fig. 2-1B), suggesting that after a finite time of

initial treatment, the effect of the drug on HEL cell growth is largely irreversible.

To determine the specificity of A46 for Jak2-V617F-dependent cell growth, the

drug was applied to four different cell lines and cell viability was assessed. The cell

lines were (i) HEL (erythroleukemia) cells that are homozygous for the activating Jak2-

Page 48: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

48

V617F mutation, (ii) Raji (Burkitt lymphoma) cells that have a translocation between the

c-Myc gene on chromosome 8 and the heavy chain locus on chromosome 14, (iii) SET-

2 (essential thrombocythemia) cells that are heterozygous for the Jak2-V617F mutation,

and (iv) CMK (acute megakaryocytic leukemia) cells that harbor a Jak3-A572V

mutation. Each of these cell lines was treated with increasing doses of A46. After 72 hrs

of treatment, we found that A46 markedly inhibited the growth of the Jak2-V617F

expressing HEL (Fig. 2-1C & D) and SET-2 (Fig. 2-1C) cells in a dose dependent

manner with a GI50 of ~ 400 nM for both cell types. The viability of Raji cells (Fig. 2-1C &

D) was, however, not affected by A46 treatment (GI50 > 25,000 nM). On the other hand,

the Jak3-A572V expressing CMK cells had intermediate susceptibility to the drug (Fig.

2-1D) with a GI50 of ~2,500 nM, suggesting that A46 is selective for Jak2-V617F

expressing cell lines over Jak3 and c-Myc mutated cell lines. Collectively, the data in

Fig. 2-1 demonstrate that the novel small molecule inhibitor A46 selectively inhibits

Jak2-V617F dependent in vitro cell growth, and this inhibitory effect of the drug on HEL

cell growth is largely irreversible after a determinate period of initial drug exposure.

A46 Inhibits the Jak/STAT Signaling Pathway

Phosphorylation of Jak2 on tyrosine residues 1007/1008 is concomitant with

increased Jak2 kinase activity [12]. Activated Jak2 can in turn phosphorylate and

activate downstream signaling targets such as STAT5. Hence, we next wanted to see if

the A46 mediated reduction in HEL cell numbers directly correlated with the

suppression of the Jak/STAT signaling pathway. This study is important in order to

eliminate the possibility that A46 exerts its cell growth inhibitory effect via mechanisms

that are independent of the Jak/STAT signaling pathway. For this, we treated HEL cells

with increasing concentrations of A46 for 48 hours and then measured the levels of

Page 49: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

49

phospho-Jak2 and phospho-STAT5 proteins in these cells using an ELISA-based

assay. It is important to note that the phospho-protein levels were measured 48 h after

drug exposure rather than the 72 h used in Fig. 2-1 because far fewer viable cells exist

at the longer time point. We observed that A46 significantly reduced levels of both

phospho-Jak2 (Fig. 2-2A) and phospho-STAT5 (Fig. 2-2B) proteins in a dose-

dependent manner. These reductions in levels of phosphorylated proteins of the

Jak/STAT signaling pathway correlated well with the A46-mediated reductions in HEL

cell numbers. Thus, the data in Fig. 2-2 indicate that A46 inhibits HEL cell growth by

directly down regulating the phosphorylation of key signaling molecules of the Jak/STAT

pathway; namely, Jak2 and STAT5.

A46 Induces G1/S Cell Cycle Arrest in HEL Cells

To determine the mechanism of A46-mediated suppression of HEL cell growth, we

first analyzed the cell cycle distribution of cells as a function of drug treatment. Here, we

treated HEL cells with increasing doses of A46 for 48 hours and then performed cell

cycle analysis via flow cytometry. Again, analysis was performed 48 h after drug

exposure rather than the 72 h used in Fig. 2-1 because far fewer viable cells exist at the

longer time point. We found that exposure to A46 increased the percentage of cells in

G1 phase and correspondingly decreased the percentage of cells in S phase, and this

effect was dose-dependent (Fig. 2-3A).

Cell cycle progression is driven and regulated by the cyclic expression of

cyclin/CDK complexes [112]. Cyclin D1 levels play a critical role in promoting the

progression of the cell cycle from G1 to S phase [113] and is also known to be a

downstream target of STAT5 transcriptional regulation [114]. Therefore, in order to

confirm the induction of G1 phase arrest in these cells, we performed anti-cyclin D1

Page 50: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

50

western blot analysis on HEL cells that had been treated with increasing doses of A46

for 24 hours. We performed our western blot analysis 24 hrs after drug exposure rather

the 48 hrs used in Figs. 2-3A because we wanted to ensure there were sufficient

numbers of viable cells and hence, sufficient levels of cellular protein to analyze. We

found that A46 treatment decreased the levels of cyclin D1 protein in a dose-dependent

manner (Fig. 2-3B, upper panel), thereby confirming the A46-induced arrest of HEL

cells in the G1 phase of the cell cycle. Analysis of the same samples with an anti β-actin

antibody was used as a loading control (Fig. 2-3B, bottom panel). Overall, the data in

Fig. 2-3 demonstrate that A46 treatment induces a G1/S cell cycle arrest in HEL cells

via the down regulation of cyclin D1.

A46 Induces Apoptosis in HEL Cells

The Jak/STAT signaling pathway is known to have direct effects on cell survival

and apoptosis (28). Having shown that A46 inhibited HEL cell growth (Fig 2-1) and

induced cell cycle arrest (Fig. 2-3), we next wanted to determine if this drug causes

apoptotic cell death in HEL cells. For this, we used Annexin V/Propidium Iodide (PI)

double staining and flow cytometry. The treatment conditions were the same as in Fig.

2-3A. The data in Fig. 2-4A shows representative apoptosis assay staining profiles from

one experiment while Fig 2-4B is a quantitative graph of two independent experiments

showing the percentage of cells in early apoptosis (Annexin V positive/PI negative)

plotted as a function of treatment condition. These data show that A46 induces

apoptosis in HEL cells in a dose-dependent manner.

Cleavage of procaspases converts these proteins into their functionally active

forms. Active caspases can then act on and cleave their substrates, such as the DNA

repair enzyme PARP, thereby affecting the integrity of the cell and triggering apoptosis

Page 51: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

51

[115]. Cleavage of procaspases and PARP are therefore considered as hallmarks of

apoptotic induction. In order to confirm the induction of apoptosis by A46, we analyzed

the effect of A46 treatment on the cleavage of procaspase 3 and its downstream

substrate, PARP. HEL cells were treated with increasing doses of A46 for 24 hours and

procaspase-3 and PARP protein levels were measured via western blot analysis (Fig. 2-

5A). The samples were also blotted with an anti β-actin antibody to demonstrate equal

protein loading across all lanes (Fig. 2-5A). We found that A46 induced a dose-

dependent cleavage of PARP and a decrease in procaspase 3 expression, thereby

corroborating the data from the Annexin V/PI apoptosis assay (Fig. 2-4A & 2-4B).

Having confirmed the ability of A46 to induce apoptosis in HEL cells, we next

wanted to determine the mechanism by which this drug causes apoptotic cell death.

Apoptosis is regulated by members of the Bcl-2 family, many of which are direct

downstream gene targets of the Jak/STAT signaling pathway (28). Therefore, we next

monitored the expression of Bcl-2 family members in HEL cells exposed to different

doses of A46 for 24 hours via western blot analysis. We observed an A46-induced

dose-dependent decrease in expression BimEL, a pro-apoptotic member of the Bcl-2

family (Fig. 2-5B). Although Bim is a pro-apoptotic protein, its disappearance/down

regulation has been shown to be associated with the generation of cleaved pro-

apoptotic forms that positively regulate and amplify apoptotic signaling [116, 117].

Another pro-apoptotic Bcl-2 family protein, Bax, was also down regulated dose-

dependently in response to A46 treatment, although to a much lesser extent (Fig. 2-5B).

Cleavage of Bax in response to apoptotic cues is not uncommon and is known to

enhance its cell death function [118, 119]. Additionally, we found that A46 decreased

Page 52: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

52

the levels of the inactive precursor form of Bid (Fig. 2-5B), which is yet another pro-

apoptotic protein that is activated upon cleavage [120]. Levels of anti-apoptotic Bcl-2

protein did not change with A46 treatment (Fig. 2-5B). Lastly, the samples were blotted

with an anti β-actin antibody to demonstrate equal protein loading across all lanes (Fig.

2-5B). In summary, the data in Figs. 2-4 and 2-5 indicate that A46 induces apoptotic cell

death in HEL cells via the down regulation/cleavage of Bcl-2 family proteins Bim, Bax

and Bid. Overall, from the data in Figs. 2-3, 2-4, and 2-5, we can conclude that A46

inhibits HEL cell proliferation by arresting the cells at G1/S transition and inducing

apoptosis.

A46 Suppresses Cytokine-independent Pathologic Cell Growth of Jak2-V617F Positive Bone Marrow Cells, ex vivo

We next wanted to determine the ability of A46 to inhibit the pathologic cell growth

of patient-derived Jak2-V617F positive bone marrow cells, ex vivo. For this,

mononuclear cells isolated from the bone-marrow of a female essential

thrombocythemia (ET) patient, who was Jak2-V617F positive, were cultured in medium

lacking thrombopoietin (TPO) and in the absence or presence of increasing doses of

A46. Hematopoietic progenitor cells isolated from a normal individual will be unable to

grow in the absence of cytokine. However, the V617F positive progenitor cells from the

patient will be able to grow under such conditions because the Jak2-V617F mutation

confers thrombopoietin-independent growth. Additionally, bone marrow cells from MPN

patients are known to be hypersensitive to cytokine stimulation [121]. Hence, the ET

patient derived bone marrow cells were also cultured in the presence or absence of the

exogenously added thrombopoietin. The results obtained show that treatment of the

mutant bone marrow cells with A46 significantly suppressed pathologic cell growth in a

Page 53: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

53

dose-dependent manner (Fig. 2-6A). We also observed that exposure of these patient

derived marrow cells to exogenous thrombopoietin significantly increased the number of

colonies formed (Fig. 2-6A), suggesting that these mutant cells from the marrow of an

ET patient are indeed hypersensitive to cytokine.

We also carried out a clonogenic assay to determine if A46 could inhibit the

cytokine-independent colony forming ability of bone marrow cells obtained from Jak2-

V617F transgenic mice [67]. Jak2-V617F positive bone marrow cells were harvested

from these transgenic animals, cultured ex vivo, and then exposed to 25 µM of A46 for

the indicated periods of time. At the end of this initial treatment period, the drug was

washed away and the cells were plated in semi-solid MethoCult medium lacking

cytokines. Five days later, the number of granulocyte-macrophage (GM) and erythroid

(E) colony-forming units in each treatment group were counted and plotted as a function

of time of initial exposure to the drug. We found that A46 significantly reduced the

number of CFU-GMs and CFU-Es in a time-dependent manner (Fig. 2-6B), suggesting

that this drug can suppress the cytokine-independent colony forming ability of Jak2-

V617F expressing murine bone marrow cells. As such, data in Fig. 2-6 demonstrate that

A46 inhibits Jak2-V617F-dependent pathologic cell growth of patient derived bone

marrow cells and also suppresses the clonogenic growth potential of Jak2-V617F-

positive bone marrow cells from MPN mice, ex vivo.

Discussion

Aberrant Jak2 kinase activity has been linked to human disorders including

several hematological malignancies and the MPNs [44, 45, 48]. A Jak2 gain-of-function

mutation (Jak2-V617F) has been detected in over 90% of patients with PV and about

50% of patients with ET and PMF [57-61]. Jak2-V617F-negative MPN patients often

Page 54: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

54

carry other activating mutations either in Jak2 [72-74] or in upstream signaling

molecules, such as the thrombopoietin receptor, MPL [76, 77]. The critical role of Jak2

in the pathogenesis of these disorders coupled with the fact that there are no curative

treatment options currently available for MPN patients, has generated considerable

amount of interest in developing Jak2 inhibitors as potential therapeutics for MPNs.

In this study, we characterized a novel benzothiophene inhibitor of Jak2 that was

identified by in silico structure based drug screening. We show that this compound

inhibits Jak2-V617F mediated cell proliferation both time and dose dependently (Fig.

1A, C & D) and this inhibitory effect of the drug on HEL cell growth is largely irreversible

after ~48 hrs of initial exposure (Fig. 1B). An important parameter used in evaluating the

effectiveness and safety of a drug is its specificity since more selective drugs can be

safely administered at higher doses without causing severe side-effects. In the past,

Jak2 inhibitors have been reported to have non-specific off target effects. AG490, one of

the first Jak2 inhibitors identified, was a potent Jak2 inhibitor, but had poor specificity

[82, 83, 122]. Our data here show that the Jak2-V617F positive cell lines, HEL and

SET2, are significantly more sensitive to inhibition by A46 (GI50 ~300 nM, Fig. 1C) when

compared to Jak3-dependent CMK cells (GI50 ~2,500 nM, Fig. 1D) or c-myc-dependent

Raji cells (GI50 > 25,000 nM, Fig. 1C & D), thereby suggesting that A46 selectively

inhibits the proliferation of Jak2-V617F-dependent cell lines while having little to no

effect on other cells lines whose proliferation is driven by Jak2-independent

mechanisms.

We have also shown that the A46 induced cell growth inhibition correlates with

direct suppression of the Jak/STAT signaling (Fig. 2A & B) which eliminates the

Page 55: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

55

possibility that A46 might be exerting its cell growth inhibitory effect via mechanisms

that are independent of the Jak/STAT signaling pathway. Our results indicate that the

mechanism by which A46 suppresses Jak2-V617F-mediated HEL cell proliferation is via

the induction of both G1/S cell cycle arrest (Fig. 3) and apoptosis (Fig. 4). Deregulation

of the Jak/STAT signaling promotes cell proliferation and blocks apoptosis resulting in

disease pathogenesis. Hence, a better understanding of the mechanism of Jak2-

inhibition induced cell death may lead to the development of more effective therapeutic

strategies for treating MPN patients, including combination therapies of Jak2 inhibitor

and apoptotic modulator mimetics. Here, we show that A46 induces apoptotic cell death

in HEL cells via the down regulation/cleavage of Bcl-2 family proteins Bim, Bax and Bid.

There are conflicting reports on the effect of apoptosis induction on the expression of

the proapoptotic protein Bim; some studies suggest that Bim is up regulated during

apoptosis [123, 124] whereas others suggest that Bim is cleaved during apoptosis

generating active and proapoptotic cleaved fragments (30, 31).

Jak2 small molecule inhibitors currently represent a diverse number of chemical

structures including pyrazines, pyrimidines, azaindoles, aminoindazoles, deazapurines,

stilbenes, benzoxazoles, and quinoxalines [125]. Our work here is significant in that it is

the first report indicating that benzothiophene based compounds, such as A46, possess

Jak2 inhibitory potential. Benzothiophenes are heterocyclic structures known to have

pro- and anti-estrogenic [126], anti-lipoxygenase [127] and anti-fungal properties [128].

Not surprisingly, several approved pharmaceuticals including raloxifene, zileuton, and

sertaconazole have benzothiophene based chemical structures. Molecules with a

benzothiophene core have also been reported to inhibit tubulin, cysteine and serine

Page 56: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

56

proteases (such as cathepsins K and L and thrombin), herpes simplex virus type I

replication and opioid receptor analgesics [129]. These heterocyclic structures are

relatively stable and their reactive site allows for subsequent derivatization, suggesting

that A46 is quite amenable to future lead optimization. As such, benzothiophenes may

be a new class of scaffolds for Jak2 small molecule inhibitors. In summary, our data

collectively show that the novel benzothiophene small molecule inhibitor of Jak2, A46,

inhibits Jak2-V617F-mediated pathological cell growth in vitro and ex vivo. As such, this

compound may perhaps serve as a lead therapeutic agent for the treatment of Jak2-

V617F mediated pathogenesis.

Page 57: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

57

Table 2-1. Chemical characteristics of A46. A46 is shown here along with its chemical name, NSC #, chemical structure, and molecular weight.

Compound Name

Chemical Name NSC # Structure Molecular

Weight

A46 1-benzothiophen-2-yl-(4-

dimethylaminophenyl)methanol 40282

283.3874

Page 58: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

58

Figure 2-1. A46 inhibits Jak2-V617F-dependent cell proliferation. A) HEL cells were treated with either DMSO or 10 μM A46 for 0, 24, 48 or 72 hours. Viable cell numbers for each treatment were determined by trypan blue exclusion staining using a hemocytometer. Each sample was measured in triplicate. Shown is one of two sets of representative results. *p < 0.05 with respect to DMSO. B) HEL cells were treated with 10 μM A46 for 0, 8, 12, 24, 48 and 72 hours. At the end of each treatment, the cells were washed, placed in fresh medium and cultured in the absence of the inhibitor for an additional 72 hours. The number of viable cells in each sample was then determined. Each sample was measured in triplicate. Shown are the means ± S.E. from two independent experiments. HEL (C & D), SET-2 (C), Raji (C & D) and CMK (D) were treated with increasing doses of A46 for 72 hours. The percent viable cells from each condition were determined either by trypan blue exclusion staining (Fig. 1C) or via an automated cell viability assay using the MTS reagent and a plate reader (Fig. 1D). Each sample was measured in triplicate.

Page 59: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

59

Figure 2-2. A46 inhibits the Jak/STAT signaling pathway. HEL cells were treated with 0, 0.09, 0.3, 0.9, 3, 9 and 30 μM of A46 for 48 hours. The cell lysates were then analyzed by ELISA for the measurement of phospho-Jak2 [pY1007/pY1008] (A) and phospho-STAT5 [pY699] (B) protein levels. Each condition was run in triplicate. Shown are the means ± S.D. of two independent experiments.

Page 60: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

60

Figure 2-3. A46 arrests HEL cells in G1 phase of the cell cycle. HEL cells were treated with 0, 0.1, 0.3, 1, 3, 10 and 30 μM of A46. A) After 48 hrs of treatment, cell cycle distribution of treated cells was determined as a function of drug treatment using flow cytometry. Shown are the means ± S.E. of two independent experiments. B) Following 24 hrs of treatment with the indicated concentrations of A46, cells were lysed and analyzed by immunoblotting with an anti-cyclin D1 (top) or an anti-β-actin antibody (bottom). Shown is one of two representative results.

Page 61: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

61

Figure 2-4. A46 induces apoptosis in HEL cells. HEL cells were treated with 0, 0.1, 0.3,

1, 3, 10 and 30 μM of A46 for 48 hours, stained with annexin V-FITC and propidium iodide and then analyzed by flow cytometry to determine the level of apoptosis in the treated cells. A) Shown are representative flow cytometry profiles from one of two independent experiments. B) Quantification of the percentage of cells in early apoptosis as a function of drug treatment. Shown are the means ± S.E. of two independent experiments.

Page 62: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

62

Figure 2-5. A46-induced apoptosis is mediated by the down regulation/cleavage of Bcl-2 family proteins Bim, Bax and Bid. HEL cells were treated with 0, 0.1, 0.3, 1, 3, 10 and 30 μM of A46 for 24 hours. The whole cell lysates were then analyzed by western blotting with a series of antibodies as indicated: A) anti-poly(ADP-ribose) polymerase (PARP), anti-caspase 3 and anti-β-actin, B) anti-Bim, -Bax, -Bid, -Bcl-2 and -β-actin. Shown are the results from one of two independent experiments.

Page 63: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

63

Figure 2-6. A46 suppresses cytokine-independent pathologic cell growth of Jak2-

V617F positive bone marrow cells, ex vivo. A) Patient-derived bone marrow mononuclear cells were cultured in semisolid medium in the presence of increasing doses of A46, with or without thrombopoietin (TPO). At the end of 14 days, the numbers of megakaryocyte colony-forming units (CFU-Megs) were counted and plotted as a function of treatment condition. Each condition was measured in duplicate. *p < 0.05 with respect to 0 μM A46 (-TPO), #p < 0.05 with respect to 0 μM A46 (+TPO), and §p < 0.05 (+ TPO) vs. (-TPO). B) Jak2-V617F-positive bone marrow cells from transgenic mice were treated with 25 μM A46 for 0, 12 and 24 hours, ex vivo. At the end of each treatment, the drug was washed away and the cells were cultured in drug-free semi-solid medium lacking cytokines for another 5 days. The numbers of granulocyte-macrophage (GM) and erythroid (E) colony-forming units in each sample were counted and plotted as a function of treatment condition. Each sample was measured in duplicate. Shown is one of two representative experiments. *p < 0.05 vs. 0 hr for CFU-GM, #p < 0.05 vs. 0 hr for CFU-E.

Page 64: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

64

CHAPTER 3 STRUCTURE-FUNCTION CORRELATION OF G6, A NOVEL SMALL MOLECULE

INHIBITOR OF JAK2: INDISPENSABILITY OF THE STILBENOID CORE

Jak2 plays a critical role in animal development, as mice that are devoid of a

functional Jak2 allele die during embryonic development due to a lack of hematopoiesis

[31, 32]. Deregulation of the Jak/STAT signaling pathway promotes cell growth and

prevents apoptosis in a variety of solid tumors and hematological malignancies such as

acute lymphoid leukemia and chronic myeloid leukemia [45, 48, 52, 130]. Additionally, a

somatic Jak2 mutation (Jak2-V617F) is found in a high number of myeloproliferative

neoplasm (MPN) patients including 90% of polycythemia vera (PV) patients and

approximately 50% of patients with essential thrombocythemia (ET) and primary

myelofibrosis (PM) [57-61]. MPNs are a group of heterogeneous diseases arising from a

transformed hematopoietic stem cell and characterized by excessive numbers of one or

more terminally differentiated blood cells of the myeloid lineage such as erythrocytes,

thrombocytes or white blood cells. A guanine to thymine mutation in hematopoietic stem

cells results in a substitution of valine to phenylalanine at codon 617 in exon 14 of the

JH2 pseudokinase domain of Jak2. It is believed that this mutation allows the kinase to

evade negative feedback inhibition thereby leading to a constitutively active Jak/STAT

signaling pathway characterized by growth factor independent cell growth [57, 60].

Given the critical role that Jak2 plays in the pathophysiology of MPNs,

identification of specific Jak2 inhibitors has become an important step towards the

development of an effective targeted therapy for these disorders. Using structure-based

This research was originally published in the Journal of Biological Chemistry. Structure-function correlation of G6, a novel small molecule inhibitor of Jak2: indispensability of the stilbenoid core. J Biol Chem. 2010; 285(41): 31399-407. © The American Society for Biochemistry and Molecular Biology.

Page 65: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

65

virtual screening, our group recently identified a novel small molecule inhibitor of Jak2

named G6 [104]. We showed that G6 has a specific inhibitory effect on Jak2 kinase

activity as measured by in vitro enzyme assays and an immunoassay ELISA [104].

Examination of the chemical structure of G6 revealed the presence of a central

stilbenoid core. Stilbenoids are a group of naturally occurring compounds having a wide

range of biological activities. For example, resveratrol, piceatannol, 3,4,5,4′-

tetramethoxystilbene and 3,5,4′-trimethoxy-trans-stilbene, are all stilbenoids and are

known to have cytotoxic, anti-proliferative, pro-apoptotic, anti-angiogenic or tumour

suppressive effects [131-135].

We hypothesized that the central stilbenoid core in the G6 structure has a critical

role to play in mediating its Jak2 inhibitory potential. To test this, five derivative

compounds of G6, namely D21, D23, D25, D28 and D30, having structural similarity to

the original lead compound, were procured from the National Cancer Institute. Two of

these compounds, D28 and D30, have a stilbenoid core present in their chemical

structure similar to G6 whereas the three other compounds, D21, D23 and D25, lack the

stilbenoid core. We report here that the core stilbenoid structure present in G6 is

essential for maintaining its ability to inhibit Jak2 kinase activity.

Experimental Procedures

Drugs

G6 and its five structurally related derivative compounds (D21, D23, D25, D28 and

D30) were obtained from the National Cancer Institute/Developmental Therapeutics

Program (NCI/DTP) which maintains a repository of approximately 140,000 compounds.

Each compound was solublized in dimethyl sulfoxide at a concentration of 10 mM and

stored at -20⁰C.

Page 66: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

66

Cell Culture

Human Erythroleukemia (HEL) cells were purchased from the American Type

Culture Collection. Ba/F3-EpoR-Jak2-V617F cells were created as described before

[136]. Both cell lines were cultured in RPMI 1640 (Mediatech) supplemented with 10%

fetal bovine serum (FBS), penicillin, streptomycin and L-glutamine at 37⁰C and 5% CO2.

Cell Proliferation Assay

HEL cells or Ba/F3-EpoR-Jak2-V617F cells were plated in 96-well plates and

treated with either 0.25% DMSO or varying concentrations of G6 and its derivatives for

the indicated periods of time. Cell viability was assessed either by trypan blue exclusion

staining or by [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-

sulfophenyl)- 2H-tetrazolium, inner salt] (MTS) (Promega) as per the manufacturer’s

protocol.

Enzyme-linked Immunosorbent Assay

HEL cells were treated with either 0.25% DMSO or 25 μM of the inhibitor

compounds for 48 hrs and the cell lysates were analyzed by ELISA for detection of

phoshpho-Jak2, phospho-STAT3 and phospho-STAT5. Jak2 [pY1007/pY1008], STAT3

[pY705] and STAT5b [pY699] ELISA kits were purchased from Invitrogen and used

according to the manufacturer’s protocol.

Cell Lysis and Immunoprecipitation

HEL cells were treated with the different drugs for the indicated periods of time.

Cells (~ 107) were then lysed in 0.8 ml of ice-cold RIPA buffer and protein concentration

was determined using a Bradford assay (Bio-Rad). Cell lysates (~2500 μg) were then

immunoprecipitated by incubating with 2 μg of the appropriate antibody and 20 μl of

Protein A/G beads (Santa Cruz Biotechnology) for 4 hrs at 4⁰C with constant shaking.

Page 67: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

67

The protein complexes were washed thrice with IP wash buffer (25 mM Tris pH 7.5, 150

mM NaCl, and 0.1% Triton X-100) and then resuspended in SDS sample buffer.

Immunoprecipitated proteins were separated by SDS-PAGE and then transferred onto

nitrocellulose membranes. The anti-STAT3 and anti-STAT5 antibodies used for

immunoprecipitation were from Santa Cruz Biotechnology. For whole cell protein

lysates, ~50 μg of soluble protein was separated via SDS-PAGE and then transferred to

nitrocellulose membranes for analysis by western blotting.

Western Blotting

Nitrocellulose membranes were first blocked with 5% milk/TBST solution and then

probed with the different primary antibodies. The immuno-reactive bands were then

visualized using the enhanced chemi-luminescence system (Western Lightning Ultra,

Perkin-Elmer). The following antibodies were used at the indicated dilutions: phospho-

STAT3 (Santa Cruz Biotechnology and Cell Signaling at 1:500 each), STAT3 (Santa

Cruz Biotechnology, 1:1000), phospho-STAT5 (Cell Signaling, 1:500), STAT5 (Santa

Cruz Biotechnology, 1:1000), and STAT1 (Santa Cruz Biotechnology, 1:1000). The

following were all obtained from Cell Signaling and used at a 1:500 dilution; poly (ADP-

ribose) polymerase (PARP), Bcl-2, Bax, Bim, and Bid.

Apoptosis Assay

Induction of apoptosis in HEL cells was determined with the FITC AnnexinV

Apoptosis detection kit (BD Pharmigen) per the manufacturer’s protocol. The cells were

incubated with 0.25% DMSO or 25 μM of inhibitors for 48 hrs and then analyzed using a

FACSCalibur flow cytometer (BD Biosciences).

Page 68: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

68

Real Time PCR Analysis

The mRNA levels of Bcl-xL and GAPDH were measured by quantitative real-time

PCR analysis. Total RNA was extracted from HEL cells, treated with 25 μM of the

different drugs for 8 and 24 hours, using the RNeasy Mini Kit (Qiagen) as per

manufacturer’s protocol. 2 μg of each RNA sample was reverse transcribed into cDNA

in a final reaction volume of 20 μl using the High Capacity cDNA Reverse Transcription

Kit (Applied Biosystems). TaqMan Gene Expression Assays (Applied Biosystems)

Hs02758991_g1 (GAPDH) and Hs00236329_m1 (Bcl-xL) were used to detect the levels

of expression of these genes. GAPDH gene expression was used as an internal loading

control. The real-time polymerase chain reaction (PCR) was then performed with

TaqMan universal PCR Master Mix (Applied Biosystems) in a final reaction volume of

20 μl in a StepOne Real-Time PCR System as per manufacturer’s protocol.

Patient Sample

Bone marrow aspirates consisting of mononuclear cells were obtained from a de-

identified Jak2-V617F positive female diagnosed with polycythemia vera (WHO criteria)

at the UF & Shands Teaching Hospital as per an Institutional Review Board approved

protocol.

Colony Forming Unit-Erythroid Colony Formation Assay

Marrow-derived mononuclear cells were washed in IMDM and cultured in human

methylcellulose complete medium without Epo (R&D Systems) at a concentration of

approximately 4 × 105 cells/ml in the presence or absence of G6 or its structurally

related derivatives. 0.9 mL of the cultures was placed in 35-mm petri dishes and

incubated at 37⁰C and 5% CO2 in a humidified atmosphere for 14 days following which

the number of erythroid colony forming units (CFU-E) were counted.

Page 69: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

69

Computational Docking

The molecular docking program DOCK6 [137, 138] was used to study the

interactions of Jak2 with ATP, the ATP analog ACP (adenosine-5′-[β, γ-methylene]

triphosphate), G6, and the structurally related derivative compounds. The template used

was the crystal structure of Jak2 kinase domain in complex with the Jak2 inhibitor 5B3

(PDB ID: 3E64) [139]. The coordinates for ATP, ACP and each of the small molecules

were generated and energy minimized using PRODRG [140]. Subsequently, CHIMERA

[141] was used to add atomic charges to these small molecules. After removing the 5B3

molecule, the coordinates of the Jak2 kinase domain were saved in PDB format. To

prepare the protein for docking, hydrogen atoms and partial electrostatic charges were

first added to the molecule. A molecular surface of the Jak2 kinase domain was then

generated using the DMS tool in CHIMERA. The program SPHGEN was used to

generate spheres around the active site for identification of the target pocket on the

protein for small molecule docking. The grid file, necessary for rapid grid-based energy

score evaluation, was then generated around the identified docking site using the GRID

module in DOCK6. Each compound was docked in 1000 different orientations using

flexible dock and a net energy GRID score was generated for each, based on the van

der Waals and electrostatic interactions between the compound and the residues in the

binding pocket. The favorable binding orientations were selected on the basis of the

energy GRID scores generated for each orientation, wherein a favorable binding

orientation would have a more negative score. Finally, the docking results were

analyzed visually using COOT [142] and CHIMERA. Surface electrostatic potentials

were calculated using APBS [143] and PYMOL [144] was used to generate the figures.

Page 70: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

70

Statistical Analysis

For statistical evaluation of time-dependent responses to the different inhibitor

compounds, a two-way analysis of variance was used. For analysis of inhibition of

phosphorylation, induction of apoptosis, modulation of gene expression and

suppression of pathologic cell growth ex vivo, a Student’s t-test was employed. Data

were assumed to be statistically significant when p < 0.05.

Results

A Stilbenoid Core Is Essential for Time- and Dose-dependent Inhibition of Jak2-V617F-dependent Cell Growth

The human erythroleukemia (HEL 92.1.7) cell line is homozygous for the Jak2-

V617F mutation and this gain-of-function mutation is responsible for its transformed

phenotype [110, 145]. Proliferation of HEL cells is mediated by the constitutively active

Jak2-V617F signaling which promotes a G1/S phase transition, thereby leading to

increased cellular proliferation [111]. G6 and its five structurally related derivatives were

therefore first analyzed for their ability to inhibit the Jak2-V617F dependent proliferation

of HEL cells. Viable cell numbers were determined by trypan blue exclusion and

hemocytometer after 72 hrs. Each sample was measured in triplicate. Inhibition by G6

was arbitrarily set at 100% and the percent inhibition for all the other compounds

relative to G6 was defined as 1.00 - (Δ drug / Δ vehicle control). Table 3-1 summarizes

the percent growth inhibition for each of the six compounds. We found that the stilbene-

containing derivatives (D28 and D30) had high growth inhibition potentials whereas

those compounds lacking the stilbenoid core (D21, D23 and D25) had low growth

inhibition potentials.

Page 71: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

71

To determine the ability of each of these compounds to inhibit Jak2-V617F

mediated HEL cell proliferation, cells were treated either for varying periods of time or

with increasing concentrations of G6 or its derivatives. Viable cell numbers for each

treatment were determined. When compared to vehicle treated cells, we found that G6

and its stilbenoid derivatives (D28 and D30) significantly reduced viable cell numbers in

a time-dependent manner whereas the non-stilbenoid derivatives (D21, D23 and D25)

did not (Fig. 3-1A). We also found that G6 and the two stilbenoid derivatives (D28 and

D30) markedly inhibited the growth of HEL cells in a dose-dependent manner (Fig. 3-

1B) whereas the three non-stilbenoid derivatives (D21, D23, and D25) showed no

growth inhibition of HEL cells (Fig. 3-1C).

Phosphorylation of Jak2 at tyrosine residues 1007/1008 is concomitant with higher

kinase activity and increased cellular proliferation [61]. Therefore, we next wanted to

determine whether the presence of the stilbenoid core is critical for reduction of

phospho-Jak2 levels within treated cells. Phospho-Jak2 levels were measured 48 hrs

after drug exposure rather the 72 hrs used in Figs. 3-1A – C as far fewer viable cells

exist at the longer time point. Exposure of HEL cells to stilbenoid core bearing

compounds (G6, D28, and D30) significantly decreased the levels of phospho-Jak2

(Fig. 3-1D) when compared to those derivatives that lack the stilbenoid core (D21, D23,

and D25).

We next wanted to determine if the ability of G6 to inhibit Jak2-V617F mediated

cell proliferation was valid for other cell lines with constitutively active Jak2. For this, we

studied the ability of these compounds to inhibit Ba/F3 cells stably expressing Jak2-

V617F. The introduction of the Jak2-V617F cDNA into these cells via retroviral

Page 72: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

72

transduction confers cytokine independent growth that is entirely Jak2-V617F

dependent [136]. Cells were treated with various doses of the different drugs and viable

cell numbers were assessed after 72 hrs using an MTS assay. G6 and its stilbenoid

derivatives (D28 and D30) showed significant inhibition of cell growth in a dose

dependent manner (Fig. 3-2A) when compared to the non-stilbenoid derivatives (D21,

D23 and D25) (Fig. 3-2B). Collectively, these data demonstrate that the stilbenoid core

is essential for maintaining the ability of G6 to inhibit Jak2-V617F dependent cell growth

and the reduced cell growth correlates with significantly reduced levels of phospho-

Jak2.

Indispensability of the Stilbenoid core in Decreasing Phosphorylation of STAT3 and STAT5

In the canonical Jak/STAT signalling pathway, an active Jak2 phosphorylates

STAT proteins such as STAT3 and STAT5, which ultimately translocate to the nucleus

and modulate gene transcription [60]. Hence, we wanted to determine if the stilbenoid

core is critical for the inhibition of STAT phosphorylation. HEL cells were treated for 48

hrs with the different compounds and phospho-STAT levels were determined. G6 and

its stilbenoid derivatives (D28 and D30) significantly decreased phospho-STAT3 levels

as measured by ELISA (Fig. 3-3A) and western blot analysis (Fig. 3-3B). Similarly, only

the stilbenoid containing compounds (G6, D28 and D30) significantly decreased

phospho-STAT5 levels as measured by ELISA (Fig. 3-3C) and western blot analysis

(Fig. 3-3D). As such, these data confirm the ability of G6 and its stilbenioid derivatives

to downregulate the phosphorylation of key signaling molecules involved in Jak2-

dependent pathological cell growth; namely, STAT3, and STAT5.

Page 73: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

73

Induction of Apoptosis in HEL Cells by G6 and its Derivatives

Deregulation of the Jak/STAT signaling pathway is known to promote cell

proliferation and prevent apoptosis in several different cancers [146]. Given that G6 and

its stilbenoid core containing derivatives inhibited Jak2-V617F dependent cell

proliferation and Jak/STAT activation, we next wanted to determine whether these

drugs induce apoptotic death. HEL cells treated with G6 and the stilbeniod derivatives

(D28 and D30) exhibited a significant increase in the percentage of cells in early

apoptosis when compared to the DMSO or the non-stilbenoid (D21, D23, and D25)

treated cells (Fig. 3-4A). Fig 3-4B is a quantitative graph of four independent

experiments showing the amount of apoptosis plotted as a function of treatment

condition. We observed that the percentage of cells in early apoptosis increased from

7.45% in the DMSO treated control to 27.8% in G6 treated, 31.3% in D28 treated and

34.2% in D30 treated HEL cells, whereas it remained almost unchanged for the non-

stilbenoid treated cells (Fig. 3-4B).

Jak2/STAT signaling is known to positively regulate cell growth by directly

increasing expression of the anti-apoptotic marker, Bcl-xL, via STAT-binding elements

present in its promoter region [114, 147]. To determine if the presence of the stilbenoid

core correlates with reduced levels of Bcl-xL, we measured Bcl-xL mRNA levels in cells

treated with the different compounds. The stilbenoids (G6, D28 and D30), significantly

decreased Bcl-xL expression in HEL cells when compared to DMSO or the non-

stilbenoids (D21, D23 and D25) both at 8 hrs (Fig. 3-5A) and 24 hrs (Fig. 3-5B) of

treatment. As such, these data indicate that the presence of the stilbenoid core does in

fact correlate with markedly reduced levels of the proliferative marker, Bcl-xL.

Page 74: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

74

The intrinsic apoptotic pathway is regulated by members of the Bcl-2 family [148].

Therefore, we next monitored the expression of Bcl-2 family members in HEL cells

treated with 25 μM of the different compounds for 24 hours. As expected, the stilbenoid

derivates (G6, D28 and D30) induced greater cleavage of PARP, a marker of apoptosis,

when compared to the non-stilbenoids (D21, D23 and D25) (Fig. 3-5C). We also

observed a stilbenoid dependent increase in the protein levels of Bim, a pro-apoptotic

member of the Bcl-2 family (Fig. 3-5D).

The pro-apoptotic protein, Bid, is present in the cytosol as an inactive precursor.

Upon proteolytic cleavage, its active form is generated, which translocates to the

mitochondria and induces mitochondrial damage and destabilization [120]. We found

that the stilbenoid compounds decreased the levels of the inactive precursor form of

Bid, a hallmark of Bid cleavage and subsequent apoptosis. We found that the protein

levels of Bax and Bcl-2 did not change with any treatment (Fig. 3-5D). Lastly, the

samples were blotted with an anti-STAT1 antibody to demonstrate equal protein loading

across all lanes (Fig. 3-5D). Overall, the data in Figs. 3-4 and 3-5 indicate that the

stilbenoid core of G6 is critical for its ability to induce apoptotic cell death in HEL cells

via the intrinsic apoptotic pathway by downregulation of anti-apoptotic Bcl-xL, up-

regulation of pro-apoptotic Bim, and cleavage of Bid.

Stilbenoid Core Bearing Derivatives of G6 Suppress Pathologic Cell Growth of Patient-derived Bone Marrow Cells, ex vivo

We next wanted to determine whether the stilbene core is also essential for its

ability to inhibit pathologic cell growth of patient-derived bone marrow cells, ex vivo. For

this, we used a colony formation assay which measures the number of erythroid colony

forming units (CFU-E) that are produced when marrow derived stem cells are cultured,

Page 75: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

75

ex vivo. Stem cells isolated from a normal individual will be unable to grow in the

absence of exogenously added cytokine. However, the Jak2-V617F mutation confers

cytokine independent, cell growth. Here, mononuclear cells isolated from the bone-

marrow of a Jak2-V617F positive female polycythemia vera patient, were cultured in

medium lacking erythropoietin. DMSO or 5 μM of inhibitor was added as indicated. Our

previous work has shown that 5 μM of G6 inhibits ~50% of the cytokine independent

growth of Jak2-V617F expressing, marrow derived stem cells [104]. The results here

show that treatment of the primary cultures with either G6 or the stilbenoids (D28 and

D30) significantly blocked cytokine-independent pathologic cell growth when compared

to the DMSO and non-stilbenoid (D21, D23, and D25) treated samples (Fig. 3-6). As

such, data in Fig. 3-6 demonstrate that the stilbenoid core of G6 is essential for

reducing Jak2-dependent pathologic cell growth of human bone marrow mononuclear

cells cultured ex vivo.

Computational Docking of G6 and its Derivatives into the ATP Binding Pocket of the Jak2 Kinase Domain

Using the known structure of the Jak2 kinase domain [139], ATP, the ATP analog

ACP, G6, and each of its five structurally related derivatives, were docked into the ATP

binding pocket. The goal was to analyze the potential interactions of these compounds

with amino acids in this binding region. The ATP binding pocket of Jak2 and the

important residues clustered in this pocket have been described previously [16, 22].

Generation of the electrostatic surface potential for the Jak2 kinase domain showed the

predicted presence of acidic and basic patch residues clustered at the ATP binding

pocket (Fig. 3-7). We found that both ATP and ACP docked extremely well into this

pocket (Fig. 3-8A) with GRID scores of -76.81 kcal/mole and -62.37 kcal/mole,

Page 76: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

76

respectively. Furthermore, the docked molecules showed excellent correlation with the

known crystal structures of ACP and ATP complexed with kinase domains of other

proteins [149-152].

We found that the stilbenoids had higher binding affinities when compared to the

non-stilbenoids as indicated by their more negative energy scores. G6, D28, D30, D21,

D23 and D25 had energy scores of -75.46, -63.24, -72.26, -27.69, -38.90 and -48.91

kcal/mole, respectively. The docking orientations of G6, D28 and D30 into the pocket

(Fig. 3-8B) were very similar to that of ATP/ACP (Fig. 3-7) and distinctly different from

that of the non-stilbenoids (Fig. 3-8C). The docking conformation of each of the

stilbenoid inhibitors into the ATP binding pocket also correlated well (r.m.s.d < 2Å) with

the previously reported structure of another Jak2 inhibitor, 5B3, in complex with the

Jak2 kinase domain (Fig. 3-8D).

Examination of the most favorable docking orientations for G6 and each of its

derivatives showed the presence of hydrogen bonds (< 3.5Å) and van der Waals

interactions between the stilbenoid derivatives and the ATP binding pocket of Jak2 (Fig.

3-9). Specifically, the stilbenoid core containing derivatives exhibited van der Waals

interactions with many of the hydrophobic residues in the binding pocket such as V863,

L855, L983, L932, Y931, A880 and V911. The stilbenoid containing derivatives also

formed hydrogen bond interactions with several important surrounding residues

including D994, R980, E930 and L932. The non-stilbenoid derivatives however, did not

show any such hydrogen bond interactions with these critical residues of the Jak2 ATP

binding pocket. Overall, these data demonstrate the importance of the stilbenoid core

Page 77: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

77

structure for stronger docking interactions of the drug with the ATP binding pocket of

Jak2.

Discussion

Somatic Jak2 mutations, such as the Jak2-V617F, which result in deregulated

Jak/STAT signaling, have been identified in numerous patients with myeloproliferative

neoplasms and are known to play a primary role in the pathogenesis of these diseases

[57-61]. Therefore, identification of novel compounds having inhibitory effects against

hyperkinetic Jak2 has become an attractive therapeutic strategy in MPN. Using

structure-based virtual screening, our group recently identified a novel Jak2 small

molecule inhibitor called G6 [104].

In this report, we describe a structure-function correlation of this inhibitor

compound. We demonstrate that it has a central stilbenoid core in its structure which is

indispensible for maintaining its ability to inhibit Jak2 kinase activity. G6 and its

stilbenoid core containing derivatives (D28 and D30) effectively inhibit Jak2-V617F

mediated HEL cell proliferation in a time- and dose-dependent manner.

Correspondingly, they are also capable of suppressing the phosphorylation of Jak2,

STAT3 and STAT5 proteins. Furthermore, these stilbenoids significantly induce

apoptosis in treated cells via the intrinsic pathway and possess the ability to block ex

vivo pathologic cell growth of bone marrow cells isolated from a Jak2-V617F positive

MPN patient.

Stilbenes are a group of compounds with a wide range of diverse biological

activities. Stilbenoids, such as resveratrol, piceatannol and diethylstilbestrol, are

reported to have anti-proliferative, anti-oxidative, anti-neovascularization and tumor-

suppressive effects [131-135]. Resveratrol has beneficial cardiovascular effects [153]

Page 78: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

78

whereas diethylstilbestrol is known to have estrogen activity [154]. Piceatannol, a

naturally occurring phenolic stilbenoid, is the only stilbenoid that is a known protein

tyrosine kinase inhibitor. It inhibits LMP2A, a viral tyrosine kinase implicated in diseases

associated with the Epstein-Barr virus as well as the non-receptor tyrosine kinases Syk

and Lck [155, 156]. Here, we report that G6, which is also a stilbene, has anti-Jak2

tyrosine kinase activity. More importantly, the trans stilbenoid core identified in all the

active compounds is now being subjected to bioisosteric replacements [157] that would

result in new Jak2 chemotypes with improved druglike properties.

Computational docking of G6 and its structurally related derivatives into the ATP

binding pocket of Jak2 revealed important interactions between the inhibitors and

specific residues within Jak2. For example, E930 and L932 are part of the important

hinge region of the Jak2 kinase domain and are known to be involved in adenine-

binding. We found that these residues were occupied by the stilbenoid containing

compounds, but not the non-stilbenoids. Another amino acid that interacted with the

docked stilbenoid inhibitors was D994, located in the highly conserved DFG motif. This

motif is part of the critical activation loop of the kinase and our group was the first to

identify the importance of this residue as it relates to Jak2 kinase inhibition [104]. We

show here that the stilbenoid core bearing inhibitors also had a hydrogen bond

interaction with R980, which is a part of the catalytic loop and is known to be one of the

residues involved in the coordination of magnesium ions [16, 22]. Thus, the ability of the

stilbenoid derivatives to simultaneously interact, in silico, with the hinge region, the DFG

motif, and the catalytic loop suggests that they may interfere with both ATP-binding

function and activation loop phosphorylation, thereby making them potent Jak2

Page 79: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

79

inhibitors. In summary, our data collectively show that the central stilbenoid core

structure is indispensable for maintaining anti-Jak2 kinase activity of the small molecule

inhibitor G6.

Page 80: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

80

Table 3-1. Percent growth inhibition of G6 and its derivatives. G6 and its derivatives are shown along with their NSC #, chemical structure, molecular weight and % growth inhibition. Inhibition by G6 was arbitrarily set at 100% and the inhibition potential of the other compounds relative to G6 was evaluated using the relation 1.00 - (Δ drug / Δ vehicle control).

Compound Name

NSC #

Structure

MWa

% Growth Inhibition

G6

33994

438.652

100.00

D21 10618

227.3054

-5.00

D23 47911

241.3322 -1.80

D25 13109

379.5844 -0.61

D28 27647

339.4766 124.16

D30 600567

384.5606 108.32

Page 81: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

81

Figure 3-1. Time- and dose-dependent effect of G6 and its derivatives on the HEL cell

proliferation and Jak2 phosphorylation. (A) HEL cells were treated with either 0.25% DMSO or with 25 μM of G6 or its derivatives for 0, 24, 48 or 72 h. The number of viable cells in each sample was measured by trypan blue exclusion. Each sample was measured in triplicate. D23 vs. G6 (p = 1.22 × 10-10). (B) & (C) HEL cells were treated for 72 hours either with DMSO or with 0.01, 0.03, 0.1, 0.3, 1, 3, 10 and 30 μM of the indicated compounds. The viable cell numbers for each treatment were determined in triplicate. Shown is one of two representative results. (D) HEL cells were treated with 25 μM of the different drugs for 48 h. Cells were then analyzed by ELISA for the detection of phospho-Jak2 (pY1007/pY1008). Each experiment was run in triplicate. Shown is one of two representative results. *p<0.05 with respect to DMSO, #p<0.05 with respect to non-stilbenoids.

Page 82: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

82

Figure 3-2. Dose-dependent effect of G6 and its derivatives on the proliferation of

Ba/F3-EpoR-Jak2-V617F cells. (A) & (B) Ba/F3-EpoR-Jak2-V617F cells were treated for 72 hours either with DMSO or with 0.01, 0.03, 0.1, 0.3, 1, 3, 10 and 30 μM of the indicated compounds. The viable cell numbers for each treatment were determined in triplicate using an MTS assay. Shown is one of two representative results.

Page 83: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

83

Figure 3-3. Inhibition of phosphorylation of STAT3 and STAT5 by G6 and its

derivatives. HEL cells were treated with 25 μM of the different drugs for 48 h. Cells were then analyzed for the detection of phospho-STAT3 and phospho-STAT5 by both ELISA (A) & (C) and western blot analysis (B) & (D). Stilbenoid containing compounds (G6, D28 and D30) effectively inhibited phosphorylation of STAT3 (A) & (B), and phosphorylation of STAT5b (C) & (D). Shown is one of two representative results for each. *p<0.05 with respect to DMSO, #p<0.05 with respect to non-stilbenoids.

Page 84: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

84

Figure 3-4. Induction of apoptosis in HEL cells by G6 and its derivatives. HEL cells were treated with 25 μM of the different drugs for 48 hrs and then stained with Annexin V-FITC and propidium iodide followed by flow cytometric analysis. (A) Shown are representative flow cytometry profiles from one of four independent results. (B) Quantification of the number of cells in early apoptosis (i.e. annexin V positive and propidium iodide negative). Data shown are the mean +/- SD from four independent experiments. *p<0.05 with respect to DMSO, #p<0.05 with respect to non-stilbenoids.

Page 85: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

85

Figure 3-5. Treatment with G6 or its stilbenoid derivatives leads to HEL cell death via the intrinsic apoptotic pathway. HEL cells were treated with 25 μM of the different drugs for either 8 hours (A) or 24 hours (B). Bcl-xL mRNA levels were normalized to those of GAPDH and plotted as the fold change over DMSO control. Each sample was run in duplicate. Shown is a one of two representative results. *p <0.05 with respect to DMSO. (C) Cells were treated with either DMSO or 25 μM of the different drugs for 24 hrs. The whole cell lysates were then analyzed by western blotting with an anti-PARP antibody. Shown is one of three representative results. (D) Cells were treated with either DMSO or 25 μM of the different drugs for 24 hrs. Whole cell lysates were then serially analyzed by western blot analysis for the indicated proteins. Shown is one of three representative results.

Page 86: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

86

Figure 3-6. Suppression of Jak2-V617F mediated pathologic cell growth in patient-derived bone marrow cells by G6 and its derivatives, ex vivo. Marrow-derived mononuclear cells were cultured in semisolid media in the presence or absence of 5 μM G6 and its structurally related derivatives. At the end of 14 days of culture, the numbers of CFU-E were counted. Each condition was measured in duplicate. *p<0.05 with respect to DMSO, #p<0.05 with respect to non-stilbenoids.

Page 87: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

87

Figure 3-7. Molecular surface representation of the ATP binding site of Jak2. Molecular surface representation of Jak2 in complex with ATP and its analog ACP, shown here as stick models. ATP is colored yellow and ACP is green.

Page 88: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

88

Figure 3-8. Molecular docking of G6 and its derivatives into the ATP binding pocket of Jak2. Molecular docking of G6 and its derivatives into the ATP binding pocket of Jak2. ATP, the ATP analog ACP, G6, and each of its structurally related derivatives were docked into the ATP binding pocket of the known crystal structure of Jak2 kinase domain (PDB ID: 3E64). (A) Coiled representation of the structure of Jak2 with ATP and ACP docked into the ATP binding pocket. The nucleotide binding loop is blue, the hinge region is cyan, the catalytic loop is magenta, and the activation loop is red. Residues within this pocket that are critical for interactions with ATP and other docked drugs have been represented as spheres. Phosphorylation of Y1007 within the activation loop is necessary for the activation of kinase activity of Jak2. ATP (yellow) and ACP (green) had strong interactions with the pocket as indicated by their highly negative GRID scores. (B) G6 (pink), D28 (grey) and D30 (yellow) docked at the ATP-binding pocket of Jak2. (C) D21 (orange), D23 (magenta) and D25 (green) docked at the ATP-binding pocket of Jak2. (D) Comparison of the docking of ATP (yellow), ATP analog ACP (green) and G6 (pink) with the crystal structure of a Jak2 inhibitor (5B3) (blue) in complex with Jak2 kinase domain showed good correlation (r.m.s.d. < 2Å) between the structures.

Page 89: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

89

Figure 3-9. Docking of G6 and its derivatives into the ATP binding pocket of Jak2. G6 (pink) and its stilbenoid derivative D28 (grey) and D30 (yellow) showed hydrogen bond interactions (< 3.5Å) with several critical residues (E930, L932, R980 and D994) that make up the ATP binding pocket of Jak2. However, the nonstilbenoid derivatives D21 (magenta), D23 (orange) and D25 (yellow) show no such hydrogen bond interactions.

Page 90: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

90

CHAPTER 4 CELL DEATH INDUCED BY THE JAK2 INHIBITOR, G6, CORRELATES WITH

CLEAVAGE OF VIMENTIN FILAMENTS

Jak2 is a member of the Janus family of cytoplasmic tyrosine kinases. Other

members of this family include Jak1, Jak3 and Tyk2. Jak2 is activated by a variety of

cytokines, growth factors and GPCR ligands, resulting in signaling cascades that

regulate cell growth, proliferation and death. Upon binding of the ligand to its specific

receptor, the receptor-associated Jak proteins are activated via a phosphorylation

event. An activated Jak can in turn phosphorylate and activate the Signal Transducers

and Activators of Transcription (STAT) family of transcription factors. Phosphorylated

STATs dimerize and translocate to the nucleus where they modulate gene transcription.

Thus, the Jak/STAT pathway results in a signal cascade from binding and activation at

the plasma membrane to changes in gene transcription in the nucleus.

Hyperkinetic Jak2 promotes aberrant cell growth and prevents apoptosis. Hence,

constitutively active Jak/STAT signaling pathway has been implicated in a variety of

neoplastic disorders. Jak2 can become constitutively active by several different gene

alterations including specific chromosomal translocations and point mutations. Jak2

chromosomal translocations such as TEL-Jak2, REL-Jak2, BCR-Jak2 and PCM1-Jak2

lead to the development of a variety of leukemias, lymphomas and myelomas [44, 45,

130, 158-161]. Additionally, an activating Jak2 point mutation (Jak2-V617F) has been

linked to the myeloproliferative neoplasms (MPN) such as polycythemia vera (PV),

essential thrombocythemia (ET) and primary myelofibrosis (PMF) [57-61]. This valine to

phenylalanine substitution mutation present in codon 617 of the autoinhibitory

pseudokinase domain of Jak2 allows the kinase to evade negative regulation thereby

making it constitutively active. MPN patients bear this mutation in their marrow derived

Page 91: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

91

stem cells and are characterized by the overproduction of terminally differentiated blood

cells of the myeloid lineage such as red cells or platelets. Current therapies for MPN

patients include phlebotomy and hydroxyurea. While these treatments alleviate some

disease symptomologies, they are not curative in any way. Therefore, there is an

unmet clinical need for Jak2 inhibitors.

Using structure-based virtual screening, our group recently identified a novel

stilbenoid small molecule inhibitor of Jak2 named G6 [104]. We subsequently showed

that G6 specifically inhibits Jak2 mediated human pathologic cell growth in vitro, ex vivo,

and in vivo [105, 162]. We also demonstrated that G6 inhibits Jak2 mediated cell

proliferation via the suppression of key signaling molecules of the Jak/STAT pathway;

the consequence of this inhibition is G1/S cell cycle arrest and apoptosis [105, 162].

Here, we sought to elucidate the molecular and biochemical mechanisms by which

G6 inhibits Jak2-mediated cellular proliferation. For this, we compared protein

expression profiles between vehicle treated and G6 treated cells using two-dimensional

gel electrophoresis. The intermediate filament protein, vimentin, was one protein that

was differentially expressed between the two conditions. We therefore hypothesized

that the mechanism by which G6 inhibits Jak2-dependent cell proliferation involves

modification of this protein. In this study, our data supports this hypothesis as we show

that G6-induced inhibition of Jak2-mediated pathogenic cell growth correlates with the

specific cleavage and cellular reorganization of vimentin.

Experimental Procedures

Drugs

G6, obtained from the National Cancer Institute/Developmental Therapeutics

Program (NCI/DTP). The drug was solublized in dimethyl sulfoxide (DMSO) at a

Page 92: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

92

concentration of 10 mM. G6 was stored at -20⁰C in small aliquots to avoid repeated

freeze thaw cycles.

Reagents

AG490, Jak Inhibitor I, PD98059 and PP2 were purchased from Calbiochem.

Cycloheximide was purchased from Fisher Scientific. Caspase Inhibitor I (Z-VAD

(OMe)-FMK), Calpain Inhibitor V (Mu-Val-HPh-CH2F, Mu = morpholinoureidyl; HPh =

homophenylalanyl), Verapamil, BAPTA-AM, A23187 and 3’,3’-iminodipropionitrile

(IDPN) were purchased from Calbiochem.

Cell Culture

Human Erythroleukemia (HEL) cells were purchased from the American Type

Culture Collection and maintained in RPMI 1640 (Mediatech) supplemented with 10%

fetal bovine serum (FBS), penicillin, streptomycin and L-glutamine at 37⁰C and 5% CO2.

2-D Differential In Gel Electrophoresis (2-D DIGE)

HEL cells were treated either with vehicle control DMSO or 25 µM G6 for 12 hours.

The cell pellets were resuspended in ice cold buffer containing 0.3% SDS, 20 mM Tris

pH 8.0, 100 mM DTT, 10 µl protease inhibitor cocktail III (Calbiochem), 5 mM MgCl2 and

250 units of benzonase. The cell suspension was homogenized by sonication. Protein

was precipitated with 9 volumes of ice cold 10% trichloroacetic acid (TCA) in acetone

overnight at 4°C. Precipitated proteins were then dissolved in solubilization buffer (7 M

urea, 2 M thiourea, 4% CHAPS, 0.2% SDS and 20 mM Tris, pH 8.0). After

centrifugation at 43,000 rpm for 30 min, solubilized protein in the supernatant was

quantified using the EZQ Protein Assay Kit (Invitrogen). Proteins (100 µg per sample)

were minimally labeled with CyDye (GE Healthcare) as per the manufacturer’s protocol.

An internal standard, which is loaded on every gel, was created by mixing equal

Page 93: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

93

amounts of protein from all samples. Proteins from the DMSO treated samples were

labeled with Cy3 (green) and the G6 treated samples were labeled with Cy5 (red). The

internal standard was labeled with Cy2 (blue).

100 µg of Cy2 labeled internal standard, 100 µg of Cy3 labeled sample, 100 µg of

Cy5 labeled sample were mixed with 200 ug unlabeled internal standard. The mixture

was used to rehydrate a 24 cm pH 3 to 11 nl IPG strip (GE Healthcare) overnight in a

rehydration buffer (solubilization buffer with 100 nM DTT containing Orange G as

tracking dye) in the dark at room temperature. Three independent replicates of each

sample were run on three strips. IEF was carried out in IPGphor3 unit (GE Healthcare)

as per manufacturer’s recommendation. Temperature was maintained at 19°C

throughout focusing.

After completion of IEF, strips were first reduced in 15 ml of 50 mM Tris-HCl pH

6.8, 6 M Urea, 30% (v/v) glycerol, 2% (w/v) SDS, 100 mM DTT for 20 min in the dark at

room temperature, then alkylated in 15 ml of 50 mM Tris-HCl pH 6.8, 6 M Urea, 30%

(v/v) glycerol, 2% SDS, 2.5% idoacetamide for 20 min. After equilibration, strips were

transferred and mounted on an 8% - 16% precast Tris Glycine polyacrylamide gel

(Jule). Electrophoresis was carried out initially at 12ºC at 10 mA/gel for one hour and

then at constant current overnight at 12 mA/gel and a limit of 150 V until dye front

reached the bottom of the plate.

Gels were then scanned with Typhoon 9400 Variable Mode Imager (GE

Healthcare). The excitation/emission wavelengths for Cy2, Cy3 and Cy5 were 488/520,

532/580 and 633/670 nm, respectively. For each gel, images for the internal standard

as well as the control and experimental conditions were acquired. The digital images

Page 94: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

94

were then analyzed with DeCyder 2D version 7.0 (GE Healthcare). Information from

replicate gels was analyzed with BVA Module (Biological Variation Analysis). Spots

were selected by setting the fold difference threshold to 1.6-fold. Statistical significance

was estimated using Student’s t-test. Protein identification using electrospray mass

spectroscopy was done at the Scripps Research Institute.

Cell Lysis

Cells (~ 107) were washed with two volumes of ice-cold PBS and then lysed in 0.8

ml of ice-cold RIPA buffer (20 mM Tris pH 7.5, 10% glycerol, 1% Triton X-100, 1%

deoxycholic acid, 0.1% SDS, 2.5 mM EDTA, 50 mM NaF, 10 mM Na4P2O7, 4 mM

benzamidine, and 10 µg/mL aprotinin). Protein concentrations in the whole cell lysates

were determined using a Bradford assay (Bio-Rad). Cell lysates were then resuspended

in SDS sample buffer. Whole cell lysates (~ 30 μg) were separated by SDS-PAGE and

then transferred onto nitrocellulose membranes for analysis by western blotting.

Western Blotting

Nitrocellulose membranes were first blocked with 5% milk/TBST solution at room

temperature and then probed first with the different indicated primary antibodies

overnight at 4°C followed by the respective secondary antibodies (1:4000, GE

Healthcare). The immuno-reactive bands were then visualized using the enhanced

chemi-luminescence system (Western Lightning Ultra, Perkin-Elmer). The following

antibodies were used at the indicated dilutions: vimentin (Abcam and BD Biosciences,

each at 1:500), STAT1 (Santa Cruz Biotechnology, 1:1000), and β-actin (Cell Signaling,

1:500).

Page 95: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

95

Immunofluorescence

HEL cells were cultured in RPMI in 100 mm dishes and treated with 25 μM G6 for

the indicated periods of time. Following treatment, the cells were centrifuged, washed

and resuspended in 1X PBS. Cells were then plated onto poly-L-lysine coated 8-

chamber slides (Santa Cruz Biotechnology) and fixed at -20°C in a mixture of 50%

methanol and 50% acetone for 10 minutes. The fixed cells were then permeabilized with

0.2% Triton X-100 and blocked with 5% goat serum for 30 minutes at room

temperature. The samples were incubated overnight at 4°C with a primary antibody of

mouse anti-vimentin (BD Biosciences, 1:100) and washed 4X with PBS the following

morning. The samples were then incubated with a FITC-conjugated anti-mouse

secondary antibody (Santa Cruz Biotechnology) for one hour at room temperature. The

cells were again washed with PBS, mounted with UltraCruz DAPI containing mounting

media (Santa Cruz Biotechnology) and sealed with a cover slip. These cells were

imaged using a 100X objective on an inverted fluorescence microscope (Olympus).

Cell Proliferation Assay

HEL cells were plated in 96-well plates and treated with either 0.25% DMSO, 30

μM G6 or 2% IDPN for the indicated periods of time. Cell viability was then assessed for

each sample by trypan blue exclusion staining and hemocytometer.

In vivo Animal Model

The xenograft model of Jak2-V617F expressing HEL cells in NOD/SCID mice has

been described previously [105]. Briefly, 3 months old NOD/SCID mice were

randomized into 5 groups (n=6 per group). One group consisted of naive animals that

did not receive any treatment. All other groups received a single tail vein injection of 2 x

106 Jak2-V617F-positive HEL cells. Three weeks after HEL cell injection, the mice

Page 96: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

96

developed symptoms of a fully penetrant bone marrow malignancy. The mice then

began receiving intraperitoneal injections of either vehicle control (DMSO) or G6 at

doses of 0.1, 1, and 10 mg/kg/day for the next 21 days. At the end of the three week

treatment period, all groups of mice were euthanized and bone marrow tissues were

fixed in 10% neutral-buffered formalin and embedded in paraffin.

Bone Marrow Immunohistochemistry

Paraffin embedded bone marrow sections from each treatment group were

analyzed by anti-vimentin immunohistochemistry. Antigen retrieval was carried out first

by microwaving at 95°C for 20 min in 1mM EDTA-NaOH solution, pH 8.0. The section

were then cooled, blocked with Protein Block (DAKO), and incubated with anti-vimentin

antibody (Abcam, 1:100) for 2 hours at room temperature. Antigen-antibody complexes

were detected using biotinylated secondary antibodies and streptavidin-peroxidase

substrate (DAKO). Stained sections were then analyzed via a standard light microscope

(Nikon) at 40X and 100X magnifications.

Statistical Analysis

For statistical evaluation of time-dependent response of HEL cell viability to G6

and IDPN, a two-way analysis of variance was used. For analysis of differential

expression of proteins in 2-D DIGE and G6-induced degradation of vimentin using

densitometry, a Student’s t-test was employed. Data were assumed to be statistically

significant when p < 0.05.

Results

G6 Treatment Induces Time- and Dose-Dependent Degradation of Vimentin

The human erythroleukemia (HEL 92.1.7) cell line is homozygous for the Jak2-

V617F mutation [110, 145]. The presence of this mutation induces constitutively active

Page 97: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

97

Jak/STAT signaling and promotes a G1/S phase transition thereby driving increased

cellular proliferation [111]. We previously demonstrated that the Jak2 inhibitor, G6,

inhibits Jak2-V617F-mediated HEL cell proliferation and induces apoptosis [105, 162].

However, the specific mechanisms by which G6 does this are not known.

To gain some insight into the mechanism by which G6 reduces cell viability, HEL

cells were treated for 12 hours with either vehicle control (0.25% DMSO) or 25 μM G6.

The protein expression profiles of these two treatment conditions were compared using

two-dimensional gel-electrophoresis (Fig. 4-1A, 4-1B, 4-1C). One spot in particular was

present in the DMSO treated cells, but significantly reduced in the G6 treated cells (Fig.

4-1D & 4-1E; marked by arrows). That spot was excised and identified using electro

spray mass spectrometry as vimentin. In a separate two-dimensional gel

electrophoresis study, we compared the protein expression profiles between HEL cells

that had been treated with either DMSO or 25 μM G6 for 24 hours. Even at this longer

time point, the spot representing vimentin was still significantly reduced in the G6

treated cells when compared to the DMSO treated cells (data not shown).

To confirm that vimentin protein levels were decreasing with G6 treatment, protein

samples from both conditions were subjected to western blot analysis with an anti-

vimentin antibody. Consistent with the mass spectroscopy data, treatment of HEL cells

with G6 resulted in the disappearance of full-length vimentin (Fig. 4-1F). Of note, we

also observed the appearance of low molecular weight fragments of vimentin in the G6

treated cells (Fig. 4-1F).

To determine whether this effect was time- and dose-dependent, HEL cells were

treated either with 25 μM of G6 for increasing lengths of time or with varying doses of

Page 98: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

98

G6 for 24 hours. Whole cell lysates were then separated on SDS-PAGE and examined

by immunoblotting with an anti-vimentin antibody. Fig. 4-2A is a representative blot

showing that full-length vimentin was cleaved into low molecular weight fragments with

G6 treatment as a function of time. The same samples were then reprobed with an anti-

β-actin antibody to confirm equal protein loading and also to demonstrate the specificity

of G6 for vimentin over other cytoskeletal proteins such as β-actin. Quantification of all

blots using densitometry confirmed the total loss of full-length vimentin protein in

response to G6 treatment over time (Fig. 4-2B). Similarly, Fig. 4-2C is a representative

blot showing a dose-dependent cleavage of full-length vimentin in response to G6 and

Fig. 4-2D is a quantification of all blots. Collectively, the data in Figs. 4-1 and 4-2

demonstrate the ability of G6 to induce specific cleavage of the intermediate filament

protein vimentin. Furthermore, this effect is both time- and dose-dependent.

G6 Treatment Induces Marked Reorganization of Vimentin Intermediate Filaments within Cells

We next wanted to study the effect of G6 treatment on structure and cellular

distribution of intracellular vimentin filaments. For this, HEL cells were treated with 25

μM G6 for 0, 24 and 36 hours and then vimentin protein expression was analyzed via

indirect immunofluorescence. For the 0 hr time point, we found that vimentin was

distributed uniformly over the cytoplasm (Fig. 4-3A, 4-3D, & 4-3G). However, for the 24

and 36 hr time points, vimentin had a punctate staining pattern in the perinuclear region

of the cell (Fig. 4-3B, 4-3E, 4-3H & 4-3C, 4-3F, 4-3I, respectively).

As such, the data in Fig. 4-3 indicate that G6 treatment induces cellular

redistribution and aggregation of vimentin intermediate filament within HEL cells.

Page 99: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

99

G6-induced Cleavage of Vimentin is Jak2-mediated

Having already demonstrated the ability of G6 to induce specific cleavage of

vimentin (Fig. 4-2), we next wanted to determine if this G6-induced cleavage was Jak2-

dependent. For this, HEL cells were treated for 24 hours with increasing concentrations

of three different Jak2 inhibitors; G6, AG490 and Jak Inhibitor I. As a control, HEL cells

were also treated with non-Jak2 inhibitors; namely, the MAPK inhibitor, PD98059 and

Src family kinase inhibitor, PP2. Whole cell lysates were separated by SDS-PAGE and

immunoblotted with an anti-vimentin antibody. We observed that the Jak2-specific

inhibitors induced cleavage of vimentin dose-dependently (Fig. 4-4A) whereas the non-

Jak2 inhibitors had no effect on full-length vimentin (Fig. 4-4B). The loading of total

protein was confirmed by immunoblotting the same cellular lysates with an anti-STAT1

antibody (Fig. 4-4A & 4B). Thus, from Fig. 4-4 we conclude that G6-induced vimentin

degradation is Jak2-mediated.

G6-induced Cleavage of Vimentin is Independent of de novo Protein Synthesis and Caspase Activity, but Calpain-dependent

Given that G6 induces specific cleavage of vimentin (Fig. 4-2), we next wanted to

determine whether this G6-induced vimentin cleavage is dependent on de novo protein

synthesis. To assess this, HEL cells were first pretreated for 4 hours with increasing

doses of cycloheximide (CHX), an inhibitor of protein biosynthesis, and then treated with

increasing concentrations of G6 for 24 hours. Cycloheximide inhibits protein synthesis

by interfering with the translation elongation process of protein biosynthesis [163].

Western blot analysis of the cell lysates from the different treatment groups showed that

exposure to increasing doses of G6 induced a dose dependent cleavage of vimentin in

HEL cells which was not be blocked by pretreatment with cycloheximide (Fig. 4-5A),

Page 100: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

100

indicating that this G6-induced cleavage process does not require de novo protein

synthesis.

Vimentin is cleaved in response to G6 treatment into low molecular weight

fragments of vimentin (Fig. 4-2) suggesting that this process is mediated by a

protease/proteolytic enzyme. Caspases are a class of intracellular cysteine proteases

with roles in cytokine maturation, inflammation and apoptosis [164]. We previously

showed that G6 induces caspase 3/7 activation in a time-dependent manner in HEL

cells [105]. It has also been reported that vimentin is a caspase substrate and can be

cleaved by some caspases in vitro [165]. Therefore, we wanted to determine if G6-

induced vimentin cleavage is caspase-mediated. For this, we first pretreated HEL cells

with the pan-caspase inhibitor, Caspase Inhibitor I (zVAD-fmk), for 4 hours before

treating them with 30 μM and 60 μM G6 for 24 hours. The effect of caspase inhibition on

G6-dependent vimentin cleavage was then studied by western blotting the cell lysates

with an anti-vimentin antibody. We found that inhibition of caspases by zVAD-fmk was

unable to prevent G6-induced cleavage of vimentin (Fig. 4-5B), but was able to

significantly reduce the G6-induced cleavage of PARP (Fig. 4-5B), a substrate known to

be cleaved by caspases [115], thereby indicating that G6-induced cleavage of vimentin

is caspase-independent.

Calpain, a calcium-dependent neutral cysteine protease [166], is yet another

protease that is known to cleave vimentin [167, 168]. Hence, to examine whether G6-

induced vimentin cleavage is calpain-mediated, we pretreated HEL cells with a calpain

inhibitor, Calpain Inhibitor V (Mu-Val-HPh-CH2F), for 4 hours before exposing them to

increasing doses of G6 for 24 hours. Immunoblotting analysis of the HEL cell lysates

Page 101: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

101

showed that calpain inhibition prevented G6-induced cleavage of vimentin in a dose-

dependent manner (Fig. 4-5C), demonstrating that the protease involved in the

cleavage of vimentin in response to G6 treatment is calpain. Overall, the data in Fig. 4-5

indicate that the G6-induced cleavage of intermediate filament protein vimentin is

independent of de novo protein synthesis and caspase activity, but dependent on

calpain protease activity.

The Mobilization of Calcium is Essential and Sufficient for the Cleavage of the Intermediate Filament Protein Vimentin

Given that calpain is a calcium-dependent cysteine protease, we next investigated

the role of calcium in the G6-induced vimentin cleavage process. Specifically, we first

examined the effect of inhibiting the flux of extracellular calcium into cells by pretreating

the cells with verapamil. Verapamil blocks Ca2+ channels, principally the L-type channel,

thereby interfering with the extracellular influx of calcium ions. HEL cells were

pretreated with 30 μM verapamil for 4 hours before exposure to 30 μM G6 for 24 hours.

Cell lysates were then immunoblotted with an anti-vimentin antibody. We found that

inhibition of extracellular calcium ion influx into the cell via blockage of L-type calcium

channels did not have any effect on G6-induced cleavage of vimentin (Fig. 4-6A).

Therefore, we next studied the effect of chelating intracellular calcium on G6-mediated

vimentin cleavage. For this, we pretreated HEL cells with 10 μM BAPTA-AM for 2 hours

before treatment with 30 μM G6 for 24 hours. BAPTA-AM is membrane-permeable ester

form of the calcium chelator BAPTA. Once inside the cell, it is hydrolyzed by cytosolic

esterases into its active form and can chelate intracellular calcium. Results from the

western blot analysis showed that chelation of intracellular calcium protected vimentin

Page 102: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

102

from G6-induced cleavage (Fig. 4-6B), indicating that intracellular calcium has a critical

role to play in mediating the G6-induced cleavage of vimentin.

In the next experiment, we examined the effect of the calcium ionophore, A23187,

on vimentin protein levels within HEL cells. A23187 is a mobile ion-carrier that forms

stable complexes with divalent cations, such as calcium, and can hence be used for

increasing intracellular levels of calcium ions. Accordingly, HEL cells were treated with

10 μM A23187 for increasing periods of time and the cellular lysates were then western

blotted using an anti-vimentin antibody. We found that increasing intracellular calcium

levels via exposure to an ionophore is sufficient to induce cleavage of vimentin in HEL

cells (Fig. 4-6C), further confirming the essential role that calcium ions play in the

vimentin cleavage process. As such, data in Fig. 4-6 demonstrate that mobilization of

intracellular calcium ions is both essential and sufficient for the cleavage of the

intermediate filament protein, vimentin.

Cleavage of Vimentin is Sufficient to Reduce HEL Cell Viability

To determine how critical vimentin is to the viability of cells, we studied the effect

of vimentin cleavage on the survival of HEL cells. The drug 3’,3’-iminodipropiontrile

(IDPN) selectively disrupts vimentin intermediate filaments [169]. Therefore, we treated

HEL cells either with vehicle control DMSO, 30 μM G6 or 2% IDPN for 0, 6, 12, 24 or 48

hours. At each time point, the number of viable cells in each condition was determined

and cell lysates from those same conditions were immunoblotted with an anti-vimentin

antibody in order to correlate decreased cell numbers with increased vimentin cleavage.

We found that treatment with both G6 and IDPN time-dependently decreased viable cell

numbers (Fig. 4-7A) and this decrease in cell viability correlated with a corresponding

time-dependent cleavage of full-length vimentin in the G6 and IDPN treated cells (Fig.

Page 103: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

103

4-7B). Overall, the data in Fig. 4-7 demonstrate that the cleavage of vimentin

intermediate filaments is sufficient to reduce the viability of Jak2-V617F expressing HEL

cells.

G6 Treatment Decreases the Levels of Vimentin Protein, in vivo

Our data thus far indicate that treatment of HEL cells with G6 results in the

degradation and subsequent loss of vimentin protein, in vitro. To determine if this is

conserved in vivo, HEL cells were injected into the tail vein of NOD/SCID mice and

allowed to engraft into the bone marrow over the ensuing 21 days at which time the

mice began receiving daily intraperitoneal injections of either vehicle control (DMSO) or

G6 at doses of 0.1, 1, and 10 mg/kg/day for the next 21 days. At the end of the three

week treatment period, all groups of mice were euthanized and bone marrow was

analyzed for vimentin protein levels via anti-vimentin immunohistochemistry.

Representative stained sections from each treatment group are shown at 40X (Fig. 4-

8A) and 100X (Fig. 4-8B) magnification. We found that when a negative control IgG

antibody was used in place of the anti-vimentin primary antibody in the immuno-

histochemical procedure, only the hematoxylin counter stain was observed. Probing the

naïve bone marrow with the anti-vimentin antibody revealed strong staining in erythroid

cells, but not myeloid cells. HEL cell injection followed by DMSO treatment resulted in a

dramatic increase in the expression of vimentin protein when compared to naïve

animals. Treatment with 0.1 mg/kg/day of G6 did not produce any observable change in

the expression level of the protein when compared to DMSO treated mice. However,

treatment with 1 and 10 mg/kg/day of G6 clearly reduced the levels of vimentin protein

to those seen in the completely naïve animals. Hence, the data in Fig. 4-8 indicate G6

Page 104: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

104

treatment reduces HEL cell-induced vimentin expression in a dose dependent manner,

in vivo.

Discussion

Our group recently identified a novel stilbenoid Jak2 small molecule inhibitor

named G6 [104]. We subsequently showed that G6 specifically inhibits Jak2 mediated

human pathologic cell growth in vitro, ex vivo, and in vivo [105, 162]. We have also

demonstrated that G6 inhibits Jak2 mediated cell proliferation via the suppression of key

signaling molecules of the Jak/STAT pathway and with the induction of G1/S cell cycle

arrest and apoptosis [105, 162]. In this report, our objective was to determine the

mechanisms by which G6 exerts its inhibitory actions. To this end, we found that G6

treatment induced a time- and dose-dependent cleavage of the intermediate filament

protein, vimentin (Fig. 4-2). G6 treatment of HEL cells resulted in the movement of

vimentin from a uniform distribution in the cytoplasm to aggregates in the perinuclear

region of the cell (Fig. 4-3). The G6-mediated cleavage of vimentin is Jak2-dependent

(Fig. 4-4) and calpain-mediated (Fig. 4-5). The mobilization of intracellular calcium is

critical for G6-mediated vimentin cleavage (Fig. 4-6) and the cleavage of vimentin, per

se, is sufficient to reduce HEL cell viability (Fig. 4-7). Lastly, the ability of G6 to cleave

vimentin is conserved in vivo (Fig. 4-8). Taken together, these results describe a novel

mechanism by which G6 exerts its inhibitory actions.

Vimentin, a member of the Type III intermediate filament protein family, is a key

component of the cytoskeleton of the cell. It plays an important role in maintaining cell

shape and integrity as well as stabilizing cytoskeletal interactions such as adhesion,

migration and signaling. Apoptosis is associated with disruption of the cytoskeletal

network and caspase and/or calpain-induced cleavage of cytoskeletal proteins, such as

Page 105: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

105

vimentin, is known to occur in response to various inducers of apoptosis [170-174].

However, studies have reported that knockdown of vimentin does not induce significant

apoptosis per se, but cleavage or degradation of vimentin potentiates the therapeutic

effects of a drug [175, 176]. These studies also report that higher levels of vimentin

expression render cells more susceptible to drug-induced apoptosis. In agreement with

these previous reports, we found that Jak2-V617F expressing HEL cells have readily

detectable levels of vimentin protein and are very susceptible to drug inhibition and

subsequent loss of cell viability. However, in contrast to the earlier reports, we found

that the loss of vimentin, either by G6 or by IDPN treatment, was sufficient to

significantly decrease cell viability (Fig. 4-7). Possible explanations for these observed

differences include the fact that the earlier reports used siRNA to knockdown vimentin

mRNA levels whereas we used pharmacological inhibitors that resulted in decreased

vimentin protein levels. In addition, G6 also reduces Jak2 kinase activity [105, 162]

which presumably vimentin siRNA treatment does not. These differences

notwithstanding, our work here is significant in that we demonstrate that G6 treatment

results in vimentin cleavage and the subsequent loss of cell viability.

With respect to the signaling pathway that facilitates G6-mediated vimentin

cleavage, our data indicates that calcium plays a critical role in this process. For

example, the G6-induced cleavage of vimentin is mediated by the calcium-dependent

protease, calpain (Fig. 4-5C). Furthermore, we show that mobilization of intracellular

calcium is both essential and sufficient for cleavage of vimentin. Our data therefore

suggest that there is a close correlation between Jak2 kinase activity and the levels of

intracellular calcium ions. This is supported by previous work which indicates that

Page 106: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

106

erythropoietin (Epo)-induced activation of its cognate receptor, EpoR, inhibits calcium-

induced neurotransmitter release via the activation of Jak2 [177]. This report also shows

that this Epo-induced inhibition of calcium activity can be blocked by treatment with a

tyrosine kinase inhibitor, such as genistein, further confirming the importance of Jak2 in

mediating calcium-induced responses. Overall, these studies demonstrate that Jak2 can

modulate intracellular calcium levels. However, the exact protein target(s) that Jak2 may

phosphorylate in the calcium signaling pathway are not known.

We have previously reported that G6 treatment potently induces apoptosis in HEL

cells via the modulation of various Bcl-2 family proteins, such as Bcl-xL, Bim and Bid

[105, 162]. Previous studies have reported that elevation of intracellular calcium levels

can trigger apoptosis via the destabilization of the mitochondrial membrane and

subsequent activation of calcium-dependent calpain proteases [178-180]. Activation of

calpains can further lead to the cleavage and activation of apoptotic regulators of the

Bcl2 family, such as Bid [181]. Therefore, these studies suggest that calcium and

calcium-dependent proteases may have an important role to play in the G6-mediated

cell death/apoptosis process.

The Jak2-V617F mutation is found in a large percentage on MPN patients [57-61].

A previous study, which compared the mRNA expression profiles between healthy

individuals and MPN patients, reported vimentin to be one gene that was differentially

expressed in these two groups [182]. Specifically, MPN patients were found to have

elevated levels of vimentin mRNA when compared to non-diseased individuals. This

increase in vimentin gene expression correlated positively with the presence of the

Jak2-V617F mutation. In other words, significant over expression of vimentin was only

Page 107: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

107

observed in patients that were homozygous for the Jak2-V617F mutation. Thus, our

data here, which describes vimentin as one protein that is down regulated in response

to Jak2-V617F inhibition by G6, is noteworthy. Furthermore, our data which show that

the cleavage of vimentin is Jak2-dependent (Fig. 4-4), imply that pharmacological

inhibition of Jak2 is sufficient to induce cleavage of vimentin and subsequent loss of cell

viability (Fig. 4-7). When taken together with the MPN microarray data [182], our work

here suggests that there may be a link between hyper activation of Jak2 kinase and

over expression of vimentin. Furthermore, vimentin expression might be a potential

biomarker for the progression of Jak2-V617F mediated pathogenesis and for disease

regression via Jak2 inhibitory therapy. In summary, our data show that G6-induced

inhibition of Jak2-mediated cell growth correlates with the cleavage and subsequent

loss of intracellular vimentin filaments in vitro and in vivo. As such, this work describes a

novel mechanistic pathway for the targeting of Jak2-mediated pathological cell growth.

Page 108: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

108

Figure 4-1. Identification of vimentin as a differentially expressed protein between

vehicle treated and G6 treated HEL cells. HEL cells were treated with either 0.25% DMSO or with 25 μM of G6 for 12h. Proteins from the internal standard (A), DMSO treated (B) and G6 treated (C) were labeled with Cy2 (blue), Cy3 (green) and Cy5 (red), respectively. (D) An overlay of the three colored images is shown. One protein spot, indicated by the arrow, was differentially expressed between the vehicle treated and G6 treated samples. (E) Analysis of the images obtained from the 2-D DIGE using DeCyder 2D predicted this differentially expressed protein to be significantly downregulated in the G6 treated samples (p=0.01). The indicated protein was excised and identified using electro spray mass spectrometry as vimentin. (F) HEL cells were treated either with DMSO or 25 μM of G6 for 24 hr and analyzed by western blotting using an anti-vimentin antibody. The same samples were also blotted with an anti-STAT1 antibody to confirm equal loading across all lanes. Shown is one of three representative images.

Page 109: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

109

Figure 4-2. G6 treatment induces time- and dose-dependent degradation of vimentin.

HEL cells were treated either with 25 μM of G6 for varying lengths of time (A) or with increasing doses of G6 for 24 hours (C). Cell lysates were then separated on SDS-PAGE and immunoblottted with an anti-vimentin antibody. The same samples were then reprobed with an anti-β-actin antibody to confirm equal protein loading and also to demonstrate the specificity of G6 for vimentin over other cytoskeletal proteins such as β-actin. Shown is one of three independent results for each. Expression of full-length vimentin was quantified using densitometry and plotted as a function of either time (B) or dose (D) of G6 treatment. Data shown are the mean ± SE from three independent experiments. *p<0.05 with respect to 0 hr (B) or 0 μM (D).

Page 110: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

110

Figure 4-3. G6 treatment induces marked reorganization of vimentin intermediate filaments within cells. HEL cells, treated with 25 μM of G6 for 0, 24 or 36 h, were analyzed via indirect immunofluorescence for changes in the cellular distribution of vimentin in response to drug treatment. Vimentin was indirectly labeled with a FITC-conjugated secondary antibody (A, B, and C). The nuclei were counter stained with DAPI (D, E, and F). The images were then merged (G, H and I). Shown is one of two representative results.

Page 111: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

111

Figure 4-4. G6-induced cleavage of vimentin is Jak2-mediated. HEL cells were treated for 24 hrs with increasing concentrations of Jak2 specific inhibitors (G6, AG490 and Jak Inhibitor I) (A) or non-Jak2 inhibitors (MAPK inhibitor, PD98059 and c-Src inhibitor, PP2) (B). Whole cell lysates were separated by SDS-PAGE and immunoblotted with an anti-vimentin antibody. Loading of protein was confirmed using an anti-STAT1 antibody. Shown is one of three representative blots.

Page 112: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

112

Figure 4-5. G6-induced cleavage of vimentin is independent of de novo protein synthesis and caspase activity, but calpain-dependent. HEL cells were pretreated for 4 hours with either cycloheximide (CHX) (A), caspase inhibitor I (zVAD) (B) or calpain inhibitor V (C) and then treated with increasing doses of G6 for 24 hours. Whole cell lysates from the different treatment groups were then analyzed by western blotting with an anti-vimentin antibody. The same lysates were also probed with an anti-β-actin antibody to confirm equal protein loading across all lanes. Shown is one of three representative results for each.

Page 113: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

113

Figure 4-6. Mobilization of calcium is essential and sufficient for the cleavage of vimentin. HEL cells were first pretreated with either 30 μM verapamil for 4 hours (A) or 10 μM BAPTA-AM for 2 hours (B) and then treated with 30 μM G6 for 24 hours. Post treatment, the cells were lysed, proteins were separated by gel electrophoresis and then immunoblotted with an anti-vimentin antibody (upper panel) or an anti-β-actin antibody (lower panel) to confirm equal protein across all lanes. (C) HEL cells were treated with 10 μM A23187 for the indicated periods of time. Cellular lysates were then probed with an anti-vimentin antibody (top panel). An anti-β-actin antibody was used as a loading control (bottom panel). Shown is a representative blot from three independent experiments for each.

Page 114: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

114

Figure 4-7. Cleavage of vimentin is sufficient to reduce HEL cell viability. HEL cells

were exposed to either vehicle control (DMSO), 30 μM G6 or 2% IDPN for 0, 6, 12, 24 or 48 hours. (A) The numbers of viable cells at each time point were determined and plotted as a function of treatment condition. (B) Cell lysates from each treatment group were collected simultaneously and analysed by immunoblotting with either an anti-vimentin antibody or an anti-β-actin antibody. Shown is one of three representative results. *p<0.05 with respect to DMSO.

Page 115: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

115

Figure 4-8. G6 treatment decreases the levels of vimentin protein, in vivo. Anti-vimentin immuno-histochemistry was carried out on bone marrow sections from the indicated groups of animals. Shown are representative stained bone marrow sections from each treatment group at 40X (A) and 100X (B) magnifications.

Page 116: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

116

CHAPTER 5

CONCLUSIONS AND PERSPECTIVES

Jak2 was first discovered in 1992 by polymerase chain reaction (PCR)-based

techniques. Since then, this protein has been linked to many critical physiological

processes as well as several pathophysiological states. Specifically, hyperkinetic Jak2

tyrosine kinase has been implicated in diverse hematological disorders, including

myeloproliferative neoplasms and hence is an emerging therapeutic target for these

disorders. In this dissertation, we characterize the structure-function correlations of two

novel Jak2 small molecule inhibitors and their mechanisms of action. We have shown

here that A46, a novel benzothiophene based compound identified in our lab,

suppresses Jak2-mediated pathogenesis, thereby making it a potential candidate drug

against Jak2-mediated disorders. Our lab has previously identified yet another small

molecule inhibitor of Jak2, called G6, which shows remarkable therapeutic efficiency

against Jak2-mediated cell proliferation in vitro and in vivo [105]. Here, we conducted

structure-function studies to demonstrate that G6 has a stilbenoid core which is

indispensable for maintaining its Jak2 inhibitory potential, suggesting that naturally

occurring compounds, such as stilbenoids, might be a good scaffold for future

development of potent Jak2 small molecule inhibitors. In addition, we also investigate

the molecular and biochemical mechanisms by which G6 inhibits Jak2-mediated cell

proliferation. Specifically, we demonstrate that the intermediate filament protein vimentin

is cleaved in response to G6 treatment and show data which suggest that G6-induced

Portion of this chapter has been reproduced from Jak2 Inhibitors for the treatment of Myeloproliferative Neoplasms. Drugs of the Future 2010; 35(8): 651-660 with permission from 2010 Prous Science, S. A. U. or its licensors.

Page 117: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

117

inhibition of Jak2-mediated pathogenic cell growth correlates with the disruption of

intracellular vimentin filaments.

This work is significant for a number of reasons. Firstly, we identify and

characterize a novel small molecule inhibitor of Jak2, A46, which may perhaps serve as

a lead therapeutic agent for the treatment of Jak2-V617F mediated pathogenesis.

Secondly, we identify stilbenes and benzothiophenes as two new potential scaffolds for

designing potent Jak2 small molecule inhibitors. Lastly, this work describes a novel

mechanistic pathway for the targeting of Jak2-mediated pathological cell growth via

inducing cleavage of vimentin. As such, our data suggest that vimentin expression

might be a potential biomarker for the progression of Jak2-V617F mediated

pathogenesis and for disease regression via Jak2 inhibitory therapy. The implications

and perspectives of the studies presented in this dissertation are discussed in further

detail below.

Importance of Designing/Identifying a Jak2 Specific Inhibitor

More than half a century ago, Dameshek first described myeloproliferative

neoplasm (MPN) in an editorial [183]. He recognized that the different MPNs were all

characterized by proliferation of cells of the myeloid lineage, which he suggested might

result from a common ‘undiscovered stimulus’. The molecular pathogenesis of these

disorders remained elusive for about five decades after that till 2005, when five different

groups, each working independently, identified the presence of a Jak2-V617F mutation

in a majority of patients with MPNs [57-61]. These five groundbreaking studies, as well

as other subsequent studies, reported the presence of the Jak2-V617F mutation in over

90% of patients with PV and about 50% of patients with ET and PMF. In vivo studies

have subsequently demonstrated that expression of this mutation in murine models is

Page 118: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

118

sufficient for the development of fully penetrant MPN in these animals [64, 66-67, 70-71,

112]. These data strongly suggest that the Jak2-V617F mutation plays a critical role in

the pathogenesis of myeloproliferative neoplasms. There are about 22 cases of PV, 24

cases of ET and 1.46 cases of PMF per 100,000 people [184, 185]. Constitutively active

Jak/STAT signaling pathway has also been linked to various forms of neoplastic

disorders, including solid cancers and hematological malignancies, such as leukemias,

myelomas and lymphomas. Unfortunately, there are currently no effective treatments

available for such Jak2-mediated pathologies. Commonly used treatment strategies for

MPNs involve the use of phlebotomy or myelosuppressive agents such as hydroxyurea

and thalidomide. However, these treatments are only palliative in nature and provide

only temporary relief without actually curing the disease. Therefore, there is currently a

need to develop molecularly targeted therapies that will be able to cure Jak2-mediated

disorders. Identification of the hyperactivating Jak2 mutations associated with these

disorders has prompted the search for potent and selective Jak2 small molecule

inhibitors and has brought us a step closer to the development of an effective and

curative therapeutic strategy for such diseases.

Elucidating the role of Jak2 in normal physiology and development is challenging

since Jak2 knockout mice die embyonically around E12.5 [31-32]. Hence, one approach

to studying the role of Jak2 in later stages of development and in adults is to use a

potent and specific small molecule inhibitor of Jak2. Therefore, identification of a good

Jak2 inhibitor will not only be useful as a therapeutic drug for Jak2-mediated disorders,

but also be an important research tool for investigating the role of Jak2 in regulating

critical physiological processes. Yet another approach to understanding the importance

Page 119: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

119

of Jak2 in later stages of development and in adults is to induce tamoxifen-mediated

deletion of Jak2 at various stages of development and studying the effects of Jak2

deletion on tissue histology and survivability at each stage.

Characterization of the Novel Jak2 Inhibitor, A46

In the chapter 2 of this study, we show that the novel benzothiophene small

molecule inhibitor of Jak2, A46, inhibits Jak2-V617F-mediated pathological cell growth

in vitro and ex vivo. Specifically, we show that this compound inhibits Jak2-V617F

mediated cell proliferation both time and dose dependently with a GI50 of ~300 nM. The

GI50 of A46 is comparable to other Jak2 inhibitors that have moved into clinical trials,

such as TG101348 (Table 1-1) and lower than many other Jak2 inhibitors that have

been characterized preclinically (Table 6-1), suggesting that this compound can

potentially be developed into drug for use in treatment of MPN patients.

We have also demonstrated that A46 selectively inhibits the proliferation of Jak2-

V617F-dependent cell lines while having little to no effect on other cells lines whose

proliferation is driven by Jak2-independent mechanisms. Selectivity/specificity is a

desirable quality in a drug because it allows the drug to be administered in higher doses

without producing non-desirable side-effects. We have also shown that the A46 induced

cell growth inhibition correlates with direct suppression of the Jak/STAT signaling.

Additionally, our data indicate that the mechanism by which A46 suppresses Jak2-

V617F-mediated HEL cell proliferation is via the induction of both G1/S cell cycle arrest

and apoptosis. Moreover, we report that this compound inhibited the pathologic growth

of primary Jak2-V617F expressing bone marrow cells, ex vivo.

Although we have demonstrated that A46 inhibits Jak2-mediated pathological

growth in vitro and ex vivo, we next need to demonstrate the in vivo efficacy of this drug.

Page 120: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

120

For this, we can use a transgenic mouse model of Jak2-V617F driven myeloproliferative

neoplasia [67]. These trangenic mice develop fully penetrant MPN with symptoms such

as erythroid, megakaryocytic, and granulocytic hyperplasia in the bone marrow and

spleen, splenomegaly, reduced levels of plasma erythropoietin and thrombopoietin, and

increased levels of cytokine-independent hematopoietic progenitor cells in the

peripheral blood, spleen and bone marrow. We hypothesize that treating MPN mice with

A46 will result in reversal of the pathologic symptoms concommitant with suppression of

the hyperactive Jak/STAT signaling in these animals. In addition to the in vivo

characterization of A46, we also need to study the pharmacologic properties of this drug

including its half life and bioavailability, before it can be moved into clinical trials for

treatment of myeloproliferative neoplasms.

Stilbenes and Benzothiophenes as Potential Scaffolds for the Design of Novel Jak2 Small Molecule Inhibitors

Our group recently identified a novel stilbenoid small molecule inhibitor of Jak2

named G6 [104]. We subsequently showed that G6 specifically inhibits Jak2 mediated

human pathologic cell growth in vitro, ex vivo, and in vivo [105]. In chapter 3, we discuss

the structure activity relationship properties of this novel Jak2 inhibitor. G6 has a

stibenoid core in its chemical structure. We demonstrated that the core stilbenoid

structure present in G6 is indispensable for maintaining its ability to inhibit Jak2 kinase

activity. Specifically, we show that the stilbenoid core in G6 is essential for conserving

its ability to i) inhibit Jak2-V617F mediated cell growth, ii) suppress the phosphorylation

of key signaling molecules of the Jak/STAT signaling pathway, iii) induce apoptosis in

HEL cells, iv) suppress pathologic cell growth of Jak2-V617F expressing human bone

Page 121: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

121

marrow cells, ex vivo and v) form stronger docking interactions with the ATP-binding

pocket of Jak2 kinase domain.

Since the discovery of the Jak2-V617F mutation as the predominant mutation

present in a large proportion of MPN patients, there has been a drive to develop Jak2-

specific inhibitors as therapeutics for MPNs and other Jak2-mediated pathologies.

There have been several Jak2 inhibitors that have been identified and characterized

preclinically and clinically. However, a potent, specific and orally bioavailable Jak2

inhibitor that has the ability to cure Jak2-mediated disorders is yet to be identified.

AG490 was the first reported Jak2 inhibitor [82]. Later, it was shown that even though

this compound was a potent inhibitor of Jak2, it was highly non-specific and inhibited

several other kinases as well [83]. Consequently, it was used as a starting point for

designing more potent and/or selective Jak2 inhibitors. Specifically, AG490 was used as

a lead compound and several derivatives of it were identified. For example, the AG490

derivative WP1066 had better potency and specificity for Jak2 than AG490 [186, 187].

Similarly, LS104, which is structurally related to AG490, is a non-competitive inhibitor of

Jak2 and showed promising preclinical results and is now being evaluated in clinical

trials [188]. Recently, a variety of medicinal chemistry techniques such as library

screening, fragment-based drug discovery, scaffold morphing, molecular docking and

lead compound derivatization have been used to study structure-function correlations of

numerous Jak2 inhibitors. The objective of these studies is to identify core scaffolds and

functional groups that play crucial roles in potent Jak2 inhibition. Jak2 small molecule

inhibitors currently represent a diverse number of chemical structures including

pyrazines, pyrimidines, azaindoles, aminoindazoles, deazapurines, benzoxazoles, and

Page 122: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

122

quinoxalines [125]. Data presented in the chapters 2 and 3 of this dissertation

demonstrate that stilbenes and benzothiophenes may be a new class of scaffolds for

Jak2 small molecule inhibitors. Specifically, stilbenoids or stilbene derivatives are

naturally occurring compounds that are known to have a wide range of biological

activities including anti-proliferative, anti-oxidative, anti-neovascularization, tumor-

suppressive and anti-tyrosine kinase effects [131-135, 153, 154]. Moreover, the trans-

stilbenoid core identified in G6 and its active derivatives is amenable to bioisosteric

replacements that could potentially result in new Jak2 chemotypes with improved

druglike properties. Similarly, benzothiophenes, relatively stable heterocyclic structures,

have reactive sites that allow for subsequent derivatization, suggesting that A46 is also

quite amenable to future lead optimization.

Having demonstrated the importance of the stilbene core in maintaining G6-

mediated inhibition of Jak2, we next want to modify the positions and sizes of the

hydroxyl and amine functional groups on the core stilbene structure of G6 and study

how they impact the properties of the lead compound G6. The objective of such studies

is to see if derivatization of G6 can result in improvement of its potency, specificity or its

drug-like properties, such as bioavailability and solubility. Similar structure-activity

relationship studies also need to be done for the benzothiophene-based Jak2 inhibitor,

A46.

Comparison of Two Novel Jak2 Inhibitors, G6 and A46

In this work we have characterized a novel Jak2 inhibitor, A46. Specifically, we

show that this compound inhibits Jak2-V617F mediated HEL cell proliferation both time

and dose dependently with a GI50 of ~300 nM. On the other hand, G6, the other novel

Jak2 inhibitor, inhibits Jak2-V617F-mediated cell proliferation with a GI50 of ~4 μM [104,

Page 123: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

123

105]. A lower GI50 suggests that A46 is a more potent inhibitor of Jak2-mediated

pathological cell growth than G6, in vitro. However, we observed an interesting

characteristic of A46. In spite of its greater potency, this compound never kills all the

cells even at the highest dose (30 μM), in vitro. The inability of A46 to completely

eliminate viable cells was also observed in the ex vivo clonogenic assay that we

performed. In comparison, exposure of HEL cells to 10 μM G6 results in complete

elimination of viable cells after 72 hours, in vitro [105]. This study also demonstrates

that G6 has exceptional therapeutic efficacy, in vivo, in a mouse model of human

erythroleukemia. At a therapeutic dose of 1mg/kg/day, G6 is able to specifically remove

mutant cells from the bone marrow and cause significant improvement in both the bone

marrow and the spleen of the treated animals [105].

Like most other Jak2 inhibitors currently known, G6 targets and binds to the ATP-

binding pocket of Jak2. However, initial studies have shown that G6 is more efficacious

in an in vivo setting than all the other Jak2 inhibitors that have been developed so far.

This raises an interesting question as to what makes G6 different from the other ATP-

site targeting Jak2 inhibitors. A possible reason for the higher efficacy of G6 might be its

unique stilbenoid chemical structure. The molecular docking studies presented in the

third chapter of this dissertation show that the stilbenoid core in G6 is critical for

maintaining its strong binding affinity for the ATP-binding pocket of Jak2. We show that

G6 has potential hydrogen bond interactions with several critical residues lining the

ATP-binding pocket, including D994, R980, E930 and L932. Potential hydrogen bond

interactions with D994 and R980 are unique to G6 and have not been identified for any

of the other Jak2 inhibitors currently available. Yet another possible reason for the

Page 124: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

124

better efficacy of G6 maybe the pharmacokinetic properties of G6. The pharmacokinetic

properties of G6 have not been characterized yet. However, it is possible that G6 might

have better bioavailability and/or ADME (Absorption, Distribution, Metabolism and

Excretions) characteristics when compared to the other inhibitors, which makes it more

efficacious than the others.

Potential of Jak2 Inhibitor Therapy for Myeloproliferative Neoplasms

The discovery of Jak2-activating mutations in the majority of MPN patients has

spurred the development of small molecule inhibitors that specifically target Jak2. To

date, a limited number of Jak2 inhibitors have been evaluated in clinical trials.

Interestingly, while these inhibitors exhibited moderate to good preclinical efficacy,

including murine models of human MPNs, those results have not translated well at the

level of clinical trials. In these trials, the primary clinical benefits observed have been

significant reduction in splenomegaly, patient weight gain and in some cases,

reductions in the Jak2 mutant allele burden and/or reduced levels of some inflammatory

cytokines. However, none of the inhibitors have yet been able to reverse the disease by

providing any improvement in the bone marrow fibrosis of the treated patients. The

reasons as to why these inhibitors have been disappointing in clinical trials are now an

area of investigative research. For example, one theory is that the genetics underlying

MPNs are more complex than first thought. For instance, it is unclear how a single point

mutation such as Jak2-V617F can lead to the development of three phenotypically

distinct disorders; namely, PV, ET, and PMF. Some hypothesize that there are

additional genetic and/or epigenetic events that contribute to the pathogenesis of these

disorders and in turn determine which of the three disorders a specific patient will

manifest [189].

Page 125: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

125

Recently, three independent groups showed that the presence of a genetic

haplotype leads to a predisposition for acquiring the Jak2-V617F mutation and

consequently developing MPN [190-192]. Results from recent epigenetic studies

suggest that hypermethylation of the Suppressors of Cytokine Signaling (SOCS) family

proteins, negative regulators of Jak/STAT signaling, may also have a role to play in the

pathogenesis of MPNs [193]. The presence of such genetic and epigenetic factors may

determine how a specific MPN manifests itself in a given individual and in turn, how that

individual responds to a specific treatment. This also raises the possibility that we might

have to consider the use of combination therapies for effective treatment of these

disorders instead of merely Jak2 monotherapy. Lastly, although this runs contrary to

current dogmatic drives as they relate to the development of Jak2 specific inhibitors,

perhaps inhibitors of the future need to be less specific for Jak2 and instead have some

off target properties as well. In other words, dirty might be better.

Another important factor to be considered when judging the limited success of the

Jak2 inhibitor clinical trials is the patient population being studied. These early clinical

trials have all been performed with myelofibrosis patients because it is the most severe

of the three classical MPNs. However, it must be kept in mind that a drug that does not

show significant improvement in these patients might still be efficacious if given to a

patient with a less severe MPN, such as PV, ET or even early stage PMF. Hence it will

be important to design more appropriate clinical trials that can address this important

issue before final conclusions are drawn regarding the efficacy of these drugs in

humans.

Page 126: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

126

Most of the efforts to identify Jak2 inhibitors have been focused on screening for

small molecules that target the ATP-binding pocket within the Jak2 kinase domain (Fig.

5-1A). However, since this pocket is highly conserved amongst all kinases, developing a

Jak2 inhibitor that does not inhibit other kinases is a challenge. The ATP-pocket

targeted inhibitors have another limitation as well. Specifically, they are unable to

distinguish between the wild-type and mutant Jak2 kinases. Hence, it is unsurprising

that these inhibitors, due to their potency against wild-type Jak2, cause

myelosuppression and anemia in treated patients. The occurrence of such adverse

side effects imposes an upper limit on the dosage of the drug that can be administered

to these patients.

Another dilemma is that the Jak2 activating mutations identified in MPN patients,

including V617F, are mostly present in the JH2 or pseudokinase domain of Jak2 and

not the kinase domain. Unfortunately, the crystal structure of Jak2 encoding both the

JH1 (kinase) and JH2 (pseudokinase) domains has not yet been resolved. As such, it is

currently not possible to design inhibitors that specifically target mutant forms of Jak2

over the wild-type protein. To complicate matters even further, one wonders whether an

inhibitor that is specifically designed against Jak2-V617F will be effective against the

dozens of other Jak2 mutations that are known to exist. That said, in the future, solving

the crystal structure of full length Jak2, or at least the JH1-JH2 domains, will be very

critical for the designing future Jak2 kinase inhibitors.

Attempts are also being made to design more specific Jak2 inhibitors by exploiting

the subtle differences between the kinase domain structures of Jak2 and other protein

kinases. Crystal structure analyses have shown that the ATP-binding site of the Jak

Page 127: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

127

kinases is much more constricted when compared to the other protein tyrosine kinases

[23]. It has also been shown that the electrostatic surface potential around the active-

site within Jak2 is more positively charged than other Jak family members, such as Jak3

(74). Moreover, several residues in the hinge region (such as M929, Y931, P934,

R938), the glycine loop (such as K857), the catalytic loop (such as I982), and the

activation loop (such as E1015) may potentially confer inhibitor selectivity to Jak2 over

other tyrosine kinases including other Jak family members (48). That said, only

completion of such studies will determine whether these structure-activity-relationships

do in fact exist.

Developing allosteric inhibitors to Jak2 might be an alternate approach for specific

Jak2 targeting. There are several other possible target sites on Jak2, other than the

ATP-binding site, against which allosteric inhibitors can be designed. The JH2 domain

of Jak2 has an autoinhibitory effect on the kinase domain [13, 14]. The mechanism for

the constitutive of Jak2-V617F involves the disruption of the inhibitory interactions

between the JH1 and JH2 domains at the JH1-JH2 interface and linker regions (Fig. 5-

1B; marked by a box) [194, 195]. Hence, this region is a possible target for allosteric

inhibition of Jak2. A recent study reported that the linker between the SH2-like domain

and the JH2 domain (Fig. 5-1C; boxed region) flexes the hinge between the JH1 and

JH2, thereby altering the accessibility of the kinase domain and leading to its activation

[75]. Incidentally, the exon 12 mutations identified in MPN patients are present in this

linker. Therefore, this linker region might also be a plausible site for allosteric targeting

of Jak2. The FERM domain negatively regulates the wild-type Jak2, but positively

regulates Jak2-V617F kinase activity [196]. Mutation/deletion of this domain also

Page 128: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

128

increases Jak2 kinase activity, further suggesting the role of the FERM domain in

regulating activity of Jak2 [197-199]. The FERM domain is also known to regulate Jak2

receptor association [20, 21]. Hence, the FERM domain might be another interesting

region to target for allosteric inhibition of Jak2. Overall, allosteric inhibitors that target

these alternative regulatory sites on Jak2 may provide more effective inhibitors that

specifically target mutant Jak2.

In summary, the causative nature of Jak2 somatic mutations in the pathogenesis

of MPNs was established in 2005. The first clinical trial testing a putative Jak2 small

molecule inhibitor for the treatment of MPNs initiated less than three years later. While

the initial data from these limited number of clinical trials have not lived up to the

expectations, it is evident that these inhibitors, even if not curative, can still have great

therapeutic benefit for MPN patients due to their ability to reduce some clinical

symptoms as well as improve the overall quality of life. However, one of the challenges

to implementing these Jak2 inhibitor therapies for MPN patients will be their prohibitive

costs. The symptoms of MPN patients are effectively managed by existing therapeutic

options, such as venesection/cytoreductive agent. Therefore, a shift towards the use of

more expensive Jak2 inhibitor therapy will be warranted only if these inhibitors show

some curative effects as opposed to just palliative ones. The issue of acquired

resistance can also become a potential problem with the use of potential Jak2 inhibitors

similar to that seen with other types of inhibitor based therapeutics, such as BCR-ABL

and FLT3 (FMS-like tyrosine kinase 3) inhibitors. In this time of rapid expectations, we

must be mindful that this area of research is still in its infancy. Current avenues of

investigation continue to expand our knowledge of Jak2 kinase and its potential

Page 129: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

129

inhibitors. With an increased knowledge of 1) the molecular pathogenesis of MPNs, 2)

the structural properties of wild type and mutant Jak2 proteins 3) the continued

identification of structure-activity-relationships between Jak2 and putative inhibitors and

4) the development of better animal models that more closely resemble the complex

nature of MPNs observed in humans, we are confident that one day, Jak2 inhibitors will

be a viable therapy for the treatment of myeloproliferative neoplasms.

Characterization of the Link Between G6-induced Cell Death and Cleavage of Vimentin

Our data in chapter 3 showed that the novel stilbenoid Jak2 inhibitor G6

specifically inhibits Jak2-mediated cell proliferation in a time and dose-dependent

manner. Therefore, we next wanted to understand the molecular and biochemical

mechanisms by which G6 inhibits Jak2-mediated cellular proliferation. For this, we

compared protein expression profiles between vehicle treated and G6 treated cells

using two-dimensional gel electrophoresis. We identified the intermediate filament

protein, vimentin, as one protein that was differentially expressed between the two

conditions. The data in chapter 4 show that G6-induced inhibition of Jak2-mediated

pathogenic cell growth correlates with the specific cleavage and cellular reorganization

of vimentin. Specifically, we demonstrate that G6 treatment induced a time- and dose-

dependent cleavage of the intermediate filament protein, vimentin. We also show that

G6 treatment of HEL cells resulted in the change in vimentin filament organization from

a uniform distribution in the cytoplasm to aggregates in the perinuclear region of the

cell. Our data provides evidence that this G6-mediated cleavage of vimentin is Jak2-

dependent and calpain-mediated. Additionally, we demonstrate that the mobilization of

intracellular calcium is critical for G6-mediated vimentin cleavage and the cleavage of

Page 130: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

130

vimentin, per se, is sufficient to reduce HEL cell viability. Lastly, our data also suggest

that the ability of G6 to induce cleavage of vimentin is conserved in vivo. Taken

together, these results describe a novel mechanism by which G6 exerts its inhibitory

effects.

Having established the critical role that calcium and the calcium-dependent

protease, calpain, plays in this G6-induced vimentin cleavage process, we next need to

determine the mechanism by which inhibition of Jak2 via G6 can induce calcium

mobilization and activation of calpain. For this, we need to study the effects of G6

treatment on the expression of important calcium-regulating genes, such as calmodulin

and calmodulin kinase. As of now, there is a missing link between the inhibition of Jak2

by the inhibitor G6 and the subsequent mobilization of calcium and activation of calpain,

which finally causes degradation of vimentin. Our data suggest that there is a close

correlation between Jak2 kinase activity and the levels of intracellular calcium ions.

However, the exact protein target(s) that Jak2 may phosphorylate in the calcium

signaling pathway are yet to be identified.

Having demonstrated the correlation between G6-induced cell death and cleavage

of vimentin, it is important to next determine if there is a direct causative relation

between them. For this, we need to first transfect HEL cells with a mutated vimentin

construct that is resistant to calpain cleavage and then treat them with G6. We expect

that the cells expressing this calpain-resistant vimentin protein will be protected from

cell death induced by the Jak2 inhibitor, G6. This experiment will help us in determining

if the Jak2 inhibitor, G6, induces cell death directly via the cleavage of vimentin

filaments. However, there are some technical difficulties in conducting this experiment.

Page 131: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

131

Calpain is a calcium-dependent protease that can recognize and cleave a wide variety

of substrates and a unique substrate identification site has not yet been characterized

for this enzyme. Hence, creating a calapin-resistant vimentin construct may be

technically challenging. Currently, due to this limitation, it was not possible to establish a

direct causative link between G6-induced cell death and cleavage of vimentin. Overall,

the data presented in this dissertation characterizes the structure-activity relationships

of novel Jak2 inhibitors and provide useful insight into the mechanisms by which these

inhibitors exert their anti-Jak2 effect.

Page 132: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

132

Figure 5-1. Surface representation of the structure of Jak2, indicating possible sites for

inhibitor targeting. (A) The ATP-binding pocket of Jak2 is marked by a box around it. This pocket has several loops/regions that are critical for the interaction of the Jak2 kinase domain with ATP. Rotating the structure shows the regions behind the ATP binding pocket (B & C). (B) The interface between the JH1 and JH2 domains, including the linker between these two domains, is indicated by the box. This interface is critical for regulating the autoinhibitory effect of the JH2 domain on the JH1/kinase domain. (C) The linker region between the FERM domain and the SH2-like domain is indicated by the box. This linker has a critical role to play in regulating the flexing of the hinge between the JH1 and JH2 domains, thereby effecting the interactions between these domains.

COLOR CODE: Pale green – FERM domain

Light pink – SH2-like domain

Light orange – JH2 (pseudokinase) domain

White – JH1 (kinase) domain

Cyan – Linker between FERM and SH2-like domains

Yellow – Linker between SH2-like and JH2 domains

Purple – Linker between JH2 and JH1 domains

Blue – Nucleotide loop

Green – Hinge region

Magenta – Catalytic loop

Red – Activation loop

Page 133: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

133

Table 5-1. GI50 of other Jak2 inhibitors in HEL cells. Other known Jak2 inhibitors in various stages of clinical and preclinical development are listed here along with their respective GI50 s in HEL cells.

Drug HEL cell IC50

(nM)

Reference

CEP-701 30-100 [93]

TG101209 152 [200]

INCB018424 186 [85]

TG101348 300 [65]

P1 510 [201]

CYT387 1804 [102]

WP1066 2000 [202]

G6 4000 [105]

Z3 15000 [109]

Page 134: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

134

LIST OF REFERENCES

[1] Hubbard SR, Till JH. Protein tyrosine kinase structure and function. Annu Rev Biochem. 2000;69:373-398.

[2] Pawson T. Protein modules and signalling networks. Nature. 1995;373:573-580.

[3] Kishimoto T, Taga T, Akira S. Cytokine signal transduction. Cell. 1994;76:253-262.

[4] Witthuhn BA, Silvennoinen O, Miura O, et al. Involvement of the Jak-3 Janus kinase in signalling by interleukins 2 and 4 in lymphoid and myeloid cells. Nature. 1994;370:153-157.

[5] Firmbach-Kraft I, Byers M, Shows T, Dalla-Favera R, Krolewski JJ. Tyk2, prototype of a novel class of non-receptor tyrosine kinase genes. Oncogene. 1990;5:1329-1336.

[6] Wilks AF. Two putative protein-tyrosine kinases identified by application of the polymerase chain reaction. Proc Natl Acad Sci U S A. 1989;86:1603-1607.

[7] Wilks AF. Cloning members of protein-tyrosine kinase family using polymerase chain reaction. Methods Enzymol. 1991;200:533-546.

[8] Harpur AG, Andres AC, Ziemiecki A, Aston RR, Wilks AF. JAK2, a third member of the JAK family of protein tyrosine kinases. Oncogene. 1992;7:1347-1353.

[9] Rane SG, Reddy EP. JAK3: a novel JAK kinase associated with terminal differentiation of hematopoietic cells. Oncogene. 1994;9:2415-2423.

[10] Takahashi T, Shirasawa T. Molecular cloning of rat JAK3, a novel member of the JAK family of protein tyrosine kinases. FEBS Lett. 1994;342:124-128.

[11] Giordanetto F, Kroemer RT. Prediction of the structure of human Janus kinase 2 (JAK2) comprising JAK homology domains 1 through 7. Protein Eng. 2002;15:727-737.

[12] Feng J, Witthuhn BA, Matsuda T, Kohlhuber F, Kerr IM, Ihle JN. Activation of Jak2 catalytic activity requires phosphorylation of Y1007 in the kinase activation loop. Mol Cell Biol. 1997;17:2497-2501.

[13] Saharinen P, Silvennoinen O. The pseudokinase domain is required for suppression of basal activity of Jak2 and Jak3 tyrosine kinases and for cytokine-inducible activation of signal transduction. J Biol Chem. 2002;277:47954-47963.

[14] Saharinen P, Takaluoma K, Silvennoinen O. Regulation of the Jak2 tyrosine kinase by its pseudokinase domain. Mol Cell Biol. 2000;20:3387-3395.

Page 135: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

135

[15] Saharinen P, Vihinen M, Silvennoinen O. Autoinhibition of Jak2 tyrosine kinase is dependent on specific regions in its pseudokinase domain. Mol Biol Cell. 2003;14:1448-1459.

[16] Lindauer K, Loerting T, Liedl KR, Kroemer RT. Prediction of the structure of human Janus kinase 2 (JAK2) comprising the two carboxy-terminal domains reveals a mechanism for autoregulation. Protein Eng. 2001;14:27-37.

[17] Bernards A. Predicted tyk2 protein contains two tandem protein kinase domains. Oncogene. 1991;6:1185-1187.

[18] Kampa D, Burnside J. Computational and functional analysis of the putative SH2 domain in Janus Kinases. Biochem Biophys Res Commun. 2000;278:175-182.

[19] Girault JA, Labesse G, Mornon JP, Callebaut I. Janus kinases and focal adhesion kinases play in the 4.1 band: a superfamily of band 4.1 domains important for cell structure and signal transduction. Mol Med. 1998;4:751-769.

[20] Tanner JW, Chen W, Young RL, Longmore GD, Shaw AS. The conserved box 1 motif of cytokine receptors is required for association with JAK kinases. J Biol Chem. 1995;270:6523-6530.

[21] Zhao Y, Wagner F, Frank SJ, Kraft AS. The amino-terminal portion of the JAK2 protein kinase is necessary for binding and phosphorylation of the granulocyte-macrophage colony-stimulating factor receptor beta c chain. J Biol Chem. 1995;270:13814-13818.

[22] Lucet IS, Fantino E, Styles M, et al. The structural basis of Janus kinase 2 inhibition by a potent and specific pan-Janus kinase inhibitor. Blood. 2006;107:176-183.

[23] Williams NK, Bamert RS, Patel O, et al. Dissecting specificity in the Janus kinases: the structures of JAK-specific inhibitors complexed to the JAK1 and JAK2 protein tyrosine kinase domains. J Mol Biol. 2009;387:219-232.

[24] Velazquez L, Fellous M, Stark GR, Pellegrini S. A protein tyrosine kinase in the interferon alpha/beta signaling pathway. Cell. 1992;70:313-322.

[25] Witthuhn BA, Quelle FW, Silvennoinen O, et al. JAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin. Cell. 1993;74:227-236.

[26] Silvennoinen O, Ihle JN, Schlessinger J, Levy DE. Interferon-induced nuclear signalling by Jak protein tyrosine kinases. Nature. 1993;366:583-585.

Page 136: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

136

[27] Quelle FW, Sato N, Witthuhn BA, et al. JAK2 associates with the beta c chain of the receptor for granulocyte-macrophage colony-stimulating factor, and its activation requires the membrane-proximal region. Mol Cell Biol. 1994;14:4335-4341.

[28] Lutticken C, Wegenka UM, Yuan J, et al. Association of transcription factor APRF and protein kinase Jak1 with the interleukin-6 signal transducer gp130. Science. 1994;263:89-92.

[29] Marrero MB, Schieffer B, Paxton WG, et al. Direct stimulation of Jak/STAT pathway by the angiotensin II AT1 receptor. Nature. 1995;375:247-250.

[30] Simon AR, Rai U, Fanburg BL, Cochran BH. Activation of the JAK-STAT pathway by reactive oxygen species. Am J Physiol. 1998;275:C1640-1652.

[31] Neubauer H, Cumano A, Muller M, Wu H, Huffstadt U, Pfeffer K. Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis. Cell. 1998;93:397-409.

[32] Parganas E, Wang D, Stravopodis D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93:385-395.

[33] Garcia R, Bowman TL, Niu G, et al. Constitutive activation of Stat3 by the Src and JAK tyrosine kinases participates in growth regulation of human breast carcinoma cells. Oncogene. 2001;20:2499-2513.

[34] Neilson LM, Zhu J, Xie J, et al. Coactivation of janus tyrosine kinase (Jak)1 positively modulates prolactin-Jak2 signaling in breast cancer: recruitment of ERK and signal transducer and activator of transcription (Stat)3 and enhancement of Akt and Stat5a/b pathways. Mol Endocrinol. 2007;21:2218-2232.

[35] Dagvadorj A, Collins S, Jomain JB, et al. Autocrine prolactin promotes prostate cancer cell growth via Janus kinase-2-signal transducer and activator of transcription-5a/b signaling pathway. Endocrinology. 2007;148:3089-3101.

[36] Dhir R, Ni Z, Lou W, DeMiguel F, Grandis JR, Gao AC. Stat3 activation in prostatic carcinomas. Prostate. 2002;51:241-246.

[37] Grandis JR, Drenning SD, Zeng Q, et al. Constitutive activation of Stat3 signaling abrogates apoptosis in squamous cell carcinogenesis in vivo. Proc Natl Acad Sci U S A. 2000;97:4227-4232.

[38] Silver DL, Naora H, Liu J, Cheng W, Montell DJ. Activated signal transducer and activator of transcription (STAT) 3: localization in focal adhesions and function in ovarian cancer cell motility. Cancer Res. 2004;64:3550-3558.

Page 137: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

137

[39] Yamauchi T, Yamauchi N, Ueki K, et al. Constitutive tyrosine phosphorylation of ErbB-2 via Jak2 by autocrine secretion of prolactin in human breast cancer. J Biol Chem. 2000;275:33937-33944.

[40] Gao B, Shen X, Kunos G, et al. Constitutive activation of JAK-STAT3 signaling by BRCA1 in human prostate cancer cells. FEBS Lett. 2001;488:179-184.

[41] Taniguchi T. Cytokine signaling through nonreceptor protein tyrosine kinases. Science. 1995;268:251-255.

[42] Ihle JN. The Janus protein tyrosine kinase family and its role in cytokine signaling. Adv Immunol. 1995;60:1-35.

[43] Rane SG, Reddy EP. Janus kinases: components of multiple signaling pathways. Oncogene. 2000;19:5662-5679.

[44] Lacronique V, Boureux A, Valle VD, et al. A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia. Science. 1997;278:1309-1312.

[45] Peeters P, Raynaud SD, Cools J, et al. Fusion of TEL, the ETS-variant gene 6 (ETV6), to the receptor-associated kinase JAK2 as a result of t(9;12) in a lymphoid and t(9;15;12) in a myeloid leukemia. Blood. 1997;90:2535-2540.

[46] Lacronique V, Boureux A, Monni R, et al. Transforming properties of chimeric TEL-JAK proteins in Ba/F3 cells. Blood. 2000;95:2076-2083.

[47] McWhirter JR, Galasso DL, Wang JY. A coiled-coil oligomerization domain of Bcr is essential for the transforming function of Bcr-Abl oncoproteins. Mol Cell Biol. 1993;13:7587-7595.

[48] Griesinger F, Hennig H, Hillmer F, et al. A BCR-JAK2 fusion gene as the result of a t(9;22)(p24;q11.2) translocation in a patient with a clinically typical chronic myeloid leukemia. Genes Chromosomes Cancer. 2005;44:329-333.

[49] Lane SW, Fairbairn DJ, McCarthy C, Nandini A, Perry-Keene J, Kennedy GA. Leukaemia cutis in atypical chronic myeloid leukaemia with a t(9;22) (p24;q11.2) leading to BCR-JAK2 fusion. Br J Haematol. 2008;142:503.

[50] Bousquet M, Quelen C, De Mas V, et al. The t(8;9)(p22;p24) translocation in atypical chronic myeloid leukaemia yields a new PCM1-JAK2 fusion gene. Oncogene. 2005;24:7248-7252.

[51] Murati A, Gelsi-Boyer V, Adelaide J, et al. PCM1-JAK2 fusion in myeloproliferative disorders and acute erythroid leukemia with t(8;9) translocation. Leukemia. 2005;19:1692-1696.

Page 138: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

138

[52] Reiter A, Walz C, Watmore A, et al. The t(8;9)(p22;p24) is a recurrent abnormality in chronic and acute leukemia that fuses PCM1 to JAK2. Cancer Res. 2005;65:2662-2667.

[53] Malinge S, Ben-Abdelali R, Settegrana C, et al. Novel activating JAK2 mutation in a patient with Down syndrome and B-cell precursor acute lymphoblastic leukemia. Blood. 2007;109:2202-2204.

[54] Mercher T, Wernig G, Moore SA, et al. JAK2T875N is a novel activating mutation that results in myeloproliferative disease with features of megakaryoblastic leukemia in a murine bone marrow transplantation model. Blood. 2006;108:2770-2779.

[55] Lee JW, Kim YG, Soung YH, et al. The JAK2 V617F mutation in de novo acute myelogenous leukemias. Oncogene. 2006;25:1434-1436.

[56] Kratz CP, Boll S, Kontny U, Schrappe M, Niemeyer CM, Stanulla M. Mutational screen reveals a novel JAK2 mutation, L611S, in a child with acute lymphoblastic leukemia. Leukemia. 2006;20:381-383.

[57] James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144-1148.

[58] Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054-1061.

[59] Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352:1779-1790.

[60] Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387-397.

[61] Zhao R, Xing S, Li Z, et al. Identification of an acquired JAK2 mutation in polycythemia vera. J Biol Chem. 2005;280:22788-22792.

[62] Kralovics R, Guan Y, Prchal JT. Acquired uniparental disomy of chromosome 9p is a frequent stem cell defect in polycythemia vera. Exp Hematol. 2002;30:229-236.

[63] Lu X, Levine R, Tong W, et al. Expression of a homodimeric type I cytokine receptor is required for JAK2V617F-mediated transformation. Proc Natl Acad Sci U S A. 2005;102:18962-18967.

[64] Lacout C, Pisani DF, Tulliez M, Gachelin FM, Vainchenker W, Villeval JL. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006;108:1652-1660.

Page 139: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

139

[65] Wernig G, Kharas MG, Okabe R, et al. Efficacy of TG101348, a selective JAK2 inhibitor, in treatment of a murine model of JAK2V617F-induced polycythemia vera. Cancer Cell. 2008;13:311-320.

[66] Marty C, Lacout C, Martin A, et al. Myeloproliferative neoplasm induced by constitutive expression of JAK2V617F in knock-in mice. Blood. 2010;116:783-787.

[67] Xing S, Wanting TH, Zhao W, et al. Transgenic expression of JAK2V617F causes myeloproliferative disorders in mice. Blood. 2008;111:5109-5117.

[68] Shide K, Shimoda HK, Kumano T, et al. Development of ET, primary myelofibrosis and PV in mice expressing JAK2 V617F. Leukemia. 2008;22:87-95.

[69] Tiedt R, Hao-Shen H, Sobas MA, et al. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood. 2008;111:3931-3940.

[70] Akada H, Yan D, Zou H, Fiering S, Hutchison RE, Mohi MG. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood. 2010;115:3589-3597.

[71] Li J, Spensberger D, Ahn JS, et al. JAK2 V617F impairs hematopoietic stem cell function in a conditional knock-in mouse model of JAK2 V617F-positive essential thrombocythemia. Blood. 2010;116:1528-1538.

[72] Zhang SJ, Li JY, Li WD, Song JH, Xu W, Qiu HX. The investigation of JAK2 mutation in Chinese myeloproliferative diseases-identification of a novel C616Y point mutation in a PV patient. Int J Lab Hematol. 2007;29:71-72.

[73] Schnittger S, Bacher U, Kern W, Schroder M, Haferlach T, Schoch C. Report on two novel nucleotide exchanges in the JAK2 pseudokinase domain: D620E and E627E. Leukemia. 2006;20:2195-2197.

[74] Scott LM, Tong W, Levine RL, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356:459-468.

[75] Zhao L, Dong H, Zhang CC, et al. A JAK2 interdomain linker relays Epo receptor engagement signals to kinase activation. J Biol Chem. 2009;284:26988-26998.

[76] Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3:e270.

[77] Pardanani AD, Levine RL, Lasho T, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood. 2006;108:3472-3476.

[78] Finazzi G, Barbui T. Evidence and expertise in the management of polycythemia vera and essential thrombocythemia. Leukemia. 2008;22:1494-1502.

Page 140: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

140

[79] Beer PA, Green AR. Pathogenesis and management of essential thrombocythemia. Hematology Am Soc Hematol Educ Program. 2009:621-628.

[80] Vannucchi AM, Guglielmelli P, Tefferi A. Advances in understanding and management of myeloproliferative neoplasms. CA Cancer J Clin. 2009;59:171-191.

[81] Rambaldi A. Therapy of myelofibrosis (excluding JAK2 inhibitors). Int J Hematol. 2010;91:180-188.

[82] Meydan N, Grunberger T, Dadi H, et al. Inhibition of acute lymphoblastic leukaemia by a Jak-2 inhibitor. Nature. 1996;379:645-648.

[83] Kleinberger-Doron N, Shelah N, Capone R, Gazit A, Levitzki A. Inhibition of Cdk2 activation by selected tyrphostins causes cell cycle arrest at late G1 and S phase. Exp Cell Res. 1998;241:340-351.

[84] Deininger MW, Druker BJ. Specific targeted therapy of chronic myelogenous leukemia with imatinib. Pharmacol Rev. 2003;55:401-423.

[85] Quintas-Cardama A, Vaddi K, Liu P, et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood. 2010;115:3109-3117.

[86] Rosen D, Herrington B, Bhargava P, Laucirica R, Verstovsek G. Correlation of tissue biopsy and fine needle aspiration cytology with positron emission tomography results. Patholog Res Int. 2011;2011:323051.

[87] Faderl S, Thomas DA, O'Brien S, et al. Augmented hyper-CVAD based on dose-intensified vincristine, dexamethasone, and asparaginase in adult acute lymphoblastic leukemia salvage therapy. Clin Lymphoma Myeloma Leuk. 2011;11:54-59.

[88] Manshouri T, Estrov Z, Quintas-Cardama A, et al. Bone Marrow Stroma-Secreted Cytokines Protect JAK2V617F-Mutated Cells from the Effects of a JAK2 Inhibitor. Cancer Res. 2011.

[89] Vigil CE, Wang SA, Cortes JE, et al. Dasatinib-Responsive Mast Cell Neoplasms As Initial Presentation of Chronic Myelogenous Leukemia in Blast Phase. J Clin Oncol. 2011.

[90] Bastidas N, Levine JP, Stile FL. The "Sweet Science" of Reducing Periorbital Lacerations in Mixed Martial Arts. Ann Plast Surg. 2011.

[91] Smith BD, Levis M, Beran M, et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood. 2004;103:3669-3676.

Page 141: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

141

[92] Knapper S, Burnett AK, Littlewood T, et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood. 2006;108:3262-3270.

[93] Hexner EO, Serdikoff C, Jan M, et al. Lestaurtinib (CEP701) is a JAK2 inhibitor that suppresses JAK2/STAT5 signaling and the proliferation of primary erythroid cells from patients with myeloproliferative disorders. Blood. 2008;111:5663-5671.

[94] Santos FP, Kantarjian HM, Jain N, et al. Phase 2 study of CEP-701, an orally available JAK2 inhibitor, in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. Blood. 2010;115:1131-1136.

[95] Geron I, Abrahamsson AE, Barroga CF, et al. Selective inhibition of JAK2-driven erythroid differentiation of polycythemia vera progenitors. Cancer Cell. 2008;13:321-330.

[96] Lasho TL, Tefferi A, Hood JD, Verstovsek S, Gilliland DG, Pardanani A. TG101348, a JAK2-selective antagonist, inhibits primary hematopoietic cells derived from myeloproliferative disorder patients with JAK2V617F, MPLW515K or JAK2 exon 12 mutations as well as mutation negative patients. Leukemia. 2008;22:1790-1792.

[97] Maures TJ, Su HW, Argetsinger LS, Grinstein S, Carter-Su C. Phosphorylation controls a dual-function polybasic nuclear localization sequence in the adapter protein SH2B1{beta} to regulate its cellular function and distribution. J Cell Sci. 2011;124:1542-1552.

[98] Cui TX, Lin G, LaPensee CR, et al. C/EBPbeta mediates growth hormone-regulated expression of multiple target genes. Mol Endocrinol. 2011;25:681-693.

[99] Zheng Y, Qin H, Frank SJ, et al. A CK2-dependent mechanism for activation of the JAK-STAT signaling pathway. Blood. 2011.

[100] Xu J, Zhang Y, Berry PA, et al. Growth hormone signaling in human T47D breast cancer cells: potential role for a growth hormone receptor-prolactin receptor complex. Mol Endocrinol. 2011;25:597-610.

[101] Wang X, Cowan JW, Gerhart M, et al. gamma-Secretase-mediated growth hormone receptor proteolysis: Mapping of the intramembranous cleavage site. Biochem Biophys Res Commun. 2011;408:432-436.

[102] Pardanani A, Lasho T, Smith G, Burns CJ, Fantino E, Tefferi A. CYT387, a selective JAK1/JAK2 inhibitor: in vitro assessment of kinase selectivity and preclinical studies using cell lines and primary cells from polycythemia vera patients. Leukemia. 2009;23:1441-1445.

Page 142: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

142

[103] Tyner JW, Bumm TG, Deininger J, et al. CYT387, a novel JAK2 inhibitor, induces hematologic responses and normalizes inflammatory cytokines in murine myeloproliferative neoplasms. Blood. 2010;115:5232-5240.

[104] Kiss R, Polgar T, Kirabo A, et al. Identification of a novel inhibitor of JAK2 tyrosine kinase by structure-based virtual screening. Bioorg Med Chem Lett. 2009;19:3598-3601.

[105] Kirabo A, Embury J, Kiss R, et al. The stilbenoid tyrosine kinase inhibitor, G6, suppresses Jak2-V617F-mediated human pathological cell growth in vitro and in vivo. J Biol Chem. 2011;286:4280-4291.

[106] Jelinek J, Oki Y, Gharibyan V, et al. JAK2 mutation 1849G>T is rare in acute leukemias but can be found in CMML, Philadelphia chromosome-negative CML, and megakaryocytic leukemia. Blood. 2005;106:3370-3373.

[107] Wernig G, Mercher T, Okabe R, Levine RL, Lee BH, Gilliland DG. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107:4274-4281.

[108] Sandberg EM, Ma X, He K, Frank SJ, Ostrov DA, Sayeski PP. Identification of 1,2,3,4,5,6-hexabromocyclohexane as a small molecule inhibitor of jak2 tyrosine kinase autophosphorylation [correction of autophophorylation]. J Med Chem. 2005;48:2526-2533.

[109] Sayyah J, Magis A, Ostrov DA, Allan RW, Braylan RC, Sayeski PP. Z3, a novel Jak2 tyrosine kinase small-molecule inhibitor that suppresses Jak2-mediated pathologic cell growth. Mol Cancer Ther. 2008;7:2308-2318.

[110] Quentmeier H, MacLeod RA, Zaborski M, Drexler HG. JAK2 V617F tyrosine kinase mutation in cell lines derived from myeloproliferative disorders. Leukemia. 2006;20:471-476.

[111] Walz C, Crowley BJ, Hudon HE, et al. Activated Jak2 with the V617F point mutation promotes G1/S phase transition. J Biol Chem. 2006;281:18177-18183.

[112] Ivanchuk SM, Rutka JT. The cell cycle: accelerators, brakes, and checkpoints. Neurosurgery. 2004;54:692-699; discussion 699-700.

[113] Stacey DW. Cyclin D1 serves as a cell cycle regulatory switch in actively proliferating cells. Curr Opin Cell Biol. 2003;15:158-163.

[114] Baker SJ, Rane SG, Reddy EP. Hematopoietic cytokine receptor signaling. Oncogene. 2007;26:6724-6737.

[115] Kumar S, Harvey NL. Role of multiple cellular proteases in the execution of programmed cell death. FEBS Lett. 1995;375:169-173.

Page 143: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

143

[116] Chen D, Zhou Q. Caspase cleavage of BimEL triggers a positive feedback amplification of apoptotic signaling. Proc Natl Acad Sci U S A. 2004;101:1235-1240.

[117] Gomez-Bougie P, Oliver L, Le Gouill S, Bataille R, Amiot M. Melphalan-induced apoptosis in multiple myeloma cells is associated with a cleavage of Mcl-1 and Bim and a decrease in the Mcl-1/Bim complex. Oncogene. 2005;24:8076-8079.

[118] Wood DE, Newcomb EW. Cleavage of Bax enhances its cell death function. Exp Cell Res. 2000;256:375-382.

[119] Gao G, Dou QP. N-terminal cleavage of bax by calpain generates a potent proapoptotic 18-kDa fragment that promotes bcl-2-independent cytochrome C release and apoptotic cell death. J Cell Biochem. 2000;80:53-72.

[120] Gross A, Yin XM, Wang K, et al. Caspase cleaved BID targets mitochondria and is required for cytochrome c release, while BCL-XL prevents this release but not tumor necrosis factor-R1/Fas death. J Biol Chem. 1999;274:1156-1163.

[121] Axelrad AA, Eskinazi D, Correa PN, Amato D. Hypersensitivity of circulating progenitor cells to megakaryocyte growth and development factor (PEG-rHu MGDF) in essential thrombocythemia. Blood. 2000;96:3310-3321.

[122] Gu Y, Zou Y, Aikawa R, et al. Growth hormone signalling and apoptosis in neonatal rat cardiomyocytes. Mol Cell Biochem. 2001;223:35-46.

[123] Will B, Siddiqi T, Jorda MA, et al. Apoptosis induced by JAK2 inhibition is mediated by Bim and enhanced by the BH3 mimetic ABT-737 in JAK2 mutant human erythroid cells. Blood. 2010;115:2901-2909.

[124] Rubert J, Qian Z, Andraos R, Guthy DA, Radimerski T. Bim and Mcl-1 exert key roles in regulating JAK2V617F cell survival. BMC Cancer. 2011;11:24.

[125] Baskin R, Majumder A, Sayeski PP. The recent medicinal chemistry development of Jak2 tyrosine kinase small molecule inhibitors. Curr Med Chem. 2010;17:4551-4558.

[126] Fuchs-Young R, Glasebrook AL, Short LL, et al. Raloxifene is a tissue-selective agonist/antagonist that functions through the estrogen receptor. Ann N Y Acad Sci. 1995;761:355-360.

[127] Carter GW, Young PR, Albert DH, et al. 5-lipoxygenase inhibitory activity of zileuton. J Pharmacol Exp Ther. 1991;256:929-937.

[128] Palacin C, Tarrago C, Ortiz JA. Sertaconazole: pharmacology of a gynecological antifungal agent. Int J Gynaecol Obstet. 2000;71 Suppl 1:S37-46.

Page 144: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

144

[129] Horton DA, Bourne GT, Smythe ML. The combinatorial synthesis of bicyclic privileged structures or privileged substructures. Chem Rev. 2003;103:893-930.

[130] Joos S, Granzow M, Holtgreve-Grez H, et al. Hodgkin's lymphoma cell lines are characterized by frequent aberrations on chromosomes 2p and 9p including REL and JAK2. Int J Cancer. 2003;103:489-495.

[131] Le Corre L, Chalabi N, Delort L, Bignon YJ, Bernard-Gallon DJ. Resveratrol and breast cancer chemoprevention: molecular mechanisms. Mol Nutr Food Res. 2005;49:462-471.

[132] Larrosa M, Tomas-Barberan FA, Espin JC. The grape and wine polyphenol piceatannol is a potent inducer of apoptosis in human SK-Mel-28 melanoma cells. Eur J Nutr. 2004;43:275-284.

[133] Kimura Y. New anticancer agents: in vitro and in vivo evaluation of the antitumor and antimetastatic actions of various compounds isolated from medicinal plants. In Vivo. 2005;19:37-60.

[134] Gosslau A, Chen M, Ho CT, Chen KY. A methoxy derivative of resveratrol analogue selectively induced activation of the mitochondrial apoptotic pathway in transformed fibroblasts. Br J Cancer. 2005;92:513-521.

[135] Belleri M, Ribatti D, Nicoli S, et al. Antiangiogenic and vascular-targeting activity of the microtubule-destabilizing trans-resveratrol derivative 3,5,4'-trimethoxystilbene. Mol Pharmacol. 2005;67:1451-1459.

[136] Pradhan A, Lambert QT, Griner LN, Reuther GW. Activation of JAK2-V617F by components of heterodimeric cytokine receptors. J Biol Chem. 2010;285:16651-16663.

[137] Kuntz ID, Blaney JM, Oatley SJ, Langridge R, Ferrin TE. A geometric approach to macromolecule-ligand interactions. J Mol Biol. 1982;161:269-288.

[138] Lang PT, Brozell SR, Mukherjee S, et al. DOCK 6: combining techniques to model RNA-small molecule complexes. RNA. 2009;15:1219-1230.

[139] Antonysamy S, Hirst G, Park F, et al. Fragment-based discovery of JAK-2 inhibitors. Bioorg Med Chem Lett. 2009;19:279-282.

[140] Schuttelkopf AW, van Aalten DM. PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr D Biol Crystallogr. 2004;60:1355-1363.

[141] Pettersen EF, Goddard TD, Huang CC, et al. UCSF Chimera--a visualization system for exploratory research and analysis. J Comput Chem. 2004;25:1605-1612.

Page 145: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

145

[142] Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta Crystallogr D Biol Crystallogr. 2004;60:2126-2132.

[143] Baker NA, Sept D, Joseph S, Holst MJ, McCammon JA. Electrostatics of nanosystems: application to microtubules and the ribosome. Proc Natl Acad Sci U S A. 2001;98:10037-10041.

[144] DeLano WL. Unraveling hot spots in binding interfaces: progress and challenges. Curr Opin Struct Biol. 2002;12:14-20.

[145] Martin P, Papayannopoulou T. HEL cells: a new human erythroleukemia cell line with spontaneous and induced globin expression. Science. 1982;216:1233-1235.

[146] Bromberg J. Stat proteins and oncogenesis. J Clin Invest. 2002;109:1139-1142.

[147] Socolovsky M, Fallon AE, Wang S, Brugnara C, Lodish HF. Fetal anemia and apoptosis of red cell progenitors in Stat5a-/-5b-/- mice: a direct role for Stat5 in Bcl-X(L) induction. Cell. 1999;98:181-191.

[148] Antonsson B, Martinou JC. The Bcl-2 protein family. Exp Cell Res. 2000;256:50-57.

[149] Biondi RM, Komander D, Thomas CC, et al. High resolution crystal structure of the human PDK1 catalytic domain defines the regulatory phosphopeptide docking site. EMBO J. 2002;21:4219-4228.

[150] Favelyukis S, Till JH, Hubbard SR, Miller WT. Structure and autoregulation of the insulin-like growth factor 1 receptor kinase. Nat Struct Biol. 2001;8:1058-1063.

[151] Lougheed JC, Chen RH, Mak P, Stout TJ. Crystal structures of the phosphorylated and unphosphorylated kinase domains of the Cdc42-associated tyrosine kinase ACK1. J Biol Chem. 2004;279:44039-44045.

[152] Till JH, Ablooglu AJ, Frankel M, Bishop SM, Kohanski RA, Hubbard SR. Crystallographic and solution studies of an activation loop mutant of the insulin receptor tyrosine kinase: insights into kinase mechanism. J Biol Chem. 2001;276:10049-10055.

[153] Pace-Asciak CR, Hahn S, Diamandis EP, Soleas G, Goldberg DM. The red wine phenolics trans-resveratrol and quercetin block human platelet aggregation and eicosanoid synthesis: implications for protection against coronary heart disease. Clin Chim Acta. 1995;235:207-219.

[154] Robinson TJ. Relationship of oestrogen and progesterone in oestrous behaviour of the ewe. Nature. 1954;173:878.

Page 146: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

146

[155] Geahlen RL, McLaughlin JL. Piceatannol (3,4,3',5'-tetrahydroxy-trans-stilbene) is a naturally occurring protein-tyrosine kinase inhibitor. Biochem Biophys Res Commun. 1989;165:241-245.

[156] Swanson-Mungerson M, Ikeda M, Lev L, Longnecker R, Portis T. Identification of latent membrane protein 2A (LMP2A) specific targets for treatment and eradication of Epstein-Barr virus (EBV)-associated diseases. J Antimicrob Chemother. 2003;52:152-154.

[157] Akwabi-Ameyaw A, Bass JY, Caldwell RD, et al. Conformationally constrained farnesoid X receptor (FXR) agonists: Naphthoic acid-based analogs of GW 4064. Bioorg Med Chem Lett. 2008;18:4339-4343.

[158] Barth TF, Melzner I, Wegener S, et al. [Biallelic mutation of SOCS-1 impairs JAK2 degradation and sustains phospho-JAK2 action in MedB-1 mediastinal lymphoma line]. Verh Dtsch Ges Pathol. 2005;89:234-244.

[159] Passamonti F, Rumi E, Pietra D, et al. Relation between JAK2 (V617F) mutation status, granulocyte activation, and constitutive mobilization of CD34+ cells into peripheral blood in myeloproliferative disorders. Blood. 2006;107:3676-3682.

[160] Mc Lornan DP, Percy MJ, Jones AV, Cross NC, Mc Mullin MF. Chronic neutrophilic leukemia with an associated V617F JAK2 tyrosine kinase mutation. Haematologica. 2005;90:1696-1697.

[161] Campbell PJ, Scott LM, Buck G, et al. Definition of subtypes of essential thrombocythaemia and relation to polycythaemia vera based on JAK2 V617F mutation status: a prospective study. Lancet. 2005;366:1945-1953.

[162] Majumder A, Govindasamy L, Magis A, et al. Structure-function correlation of G6, a novel small molecule inhibitor of Jak2: indispensability of the stilbenoid core. J Biol Chem. 2010;285:31399-31407.

[163] Schneider-Poetsch T, Ju J, Eyler DE, et al. Inhibition of eukaryotic translation elongation by cycloheximide and lactimidomycin. Nat Chem Biol. 2010;6:209-217.

[164] Talanian RV, Quinlan C, Trautz S, et al. Substrate specificities of caspase family proteases. J Biol Chem. 1997;272:9677-9682.

[165] Byun Y, Chen F, Chang R, Trivedi M, Green KJ, Cryns VL. Caspase cleavage of vimentin disrupts intermediate filaments and promotes apoptosis. Cell Death Differ. 2001;8:443-450.

[166] Chapman HA, Riese RJ, Shi GP. Emerging roles for cysteine proteases in human biology. Annu Rev Physiol. 1997;59:63-88.

Page 147: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

147

[167] Yoshida H, Murachi T, Tsukahara I. Degradation of actin and vimentin by calpain II, a Ca2+-dependent cysteine proteinase, in bovine lens. FEBS Lett. 1984;170:259-262.

[168] Traub P, Scherbarth A, Willingale-Theune J, Paulin-Levasseur M, Shoeman R. Differential sensitivity of vimentin and nuclear lamins from Ehrlich ascites tumor cells toward Ca2+ -activated neutral thiol proteinase. Eur J Cell Biol. 1988;46:478-490.

[169] Kumar N, Robidoux J, Daniel KW, Guzman G, Floering LM, Collins S. Requirement of vimentin filament assembly for beta3-adrenergic receptor activation of ERK MAP kinase and lipolysis. J Biol Chem. 2007;282:9244-9250.

[170] Schutte B, Nieland L, van Engeland M, Henfling ME, Meijer L, Ramaekers FC. The effect of the cyclin-dependent kinase inhibitor olomoucine on cell cycle kinetics. Exp Cell Res. 1997;236:4-15.

[171] Hashimoto M, Inoue S, Ogawa S, et al. Rapid fragmentation of vimentin in human skin fibroblasts exposed to tamoxifen: a possible involvement of caspase-3. Biochem Biophys Res Commun. 1998;247:401-406.

[172] Prasad SC, Thraves PJ, Kuettel MR, Srinivasarao GY, Dritschilo A, Soldatenkov VA. Apoptosis-associated proteolysis of vimentin in human prostate epithelial tumor cells. Biochem Biophys Res Commun. 1998;249:332-338.

[173] Morishima N. Changes in nuclear morphology during apoptosis correlate with vimentin cleavage by different caspases located either upstream or downstream of Bcl-2 action. Genes Cells. 1999;4:401-414.

[174] Liu X, Van Vleet T, Schnellmann RG. The role of calpain in oncotic cell death. Annu Rev Pharmacol Toxicol. 2004;44:349-370.

[175] Bargagna-Mohan P, Hamza A, Kim YE, et al. The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin. Chem Biol. 2007;14:623-634.

[176] Lahat G, Zhu QS, Huang KL, et al. Vimentin is a novel anti-cancer therapeutic target; insights from in vitro and in vivo mice xenograft studies. PLoS One. 2010;5:e10105.

[177] Kawakami M, Iwasaki S, Sato K, Takahashi M. Erythropoietin inhibits calcium-induced neurotransmitter release from clonal neuronal cells. Biochem Biophys Res Commun. 2000;279:293-297.

[178] Orrenius S, Zhivotovsky B, Nicotera P. Regulation of cell death: the calcium-apoptosis link. Nat Rev Mol Cell Biol. 2003;4:552-565.

Page 148: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

148

[179] Rizzuto R, Brini M, Murgia M, Pozzan T. Microdomains with high Ca2+ close to IP3-sensitive channels that are sensed by neighboring mitochondria. Science. 1993;262:744-747.

[180] Goll DE, Thompson VF, Li H, Wei W, Cong J. The calpain system. Physiol Rev. 2003;83:731-801.

[181] Mandic A, Viktorsson K, Strandberg L, et al. Calpain-mediated Bid cleavage and calpain-independent Bak modulation: two separate pathways in cisplatin-induced apoptosis. Mol Cell Biol. 2002;22:3003-3013.

[182] Kralovics R, Teo SS, Buser AS, et al. Altered gene expression in myeloproliferative disorders correlates with activation of signaling by the V617F mutation of Jak2. Blood. 2005;106:3374-3376.

[183] Kasper S, Breitenbuecher F, Hoehn Y, et al. The kinase inhibitor LS104 induces apoptosis, enhances cytotoxic effects of chemotherapeutic drugs and is targeting the receptor tyrosine kinase FLT3 in acute myeloid leukemia. Leuk Res. 2008;32:1698-1708.

[184] Flavin M, MacDonald M, Dolovich M, Coates G, O'Brodovich H. Aerosol delivery to the rabbit lung with an infant ventilator. Pediatr Pulmonol. 1986;2:35-39.

[185] Boros SJ, Campbell K. A comparison of the effects of high frequency--low tidal volume and low frequency--high tidal volume mechanical ventilation. J Pediatr. 1980;97:108-112.

[186] Iwamaru A, Szymanski S, Iwado E, et al. A novel inhibitor of the STAT3 pathway induces apoptosis in malignant glioma cells both in vitro and in vivo. Oncogene. 2007;26:2435-2444.

[187] Ferrajoli A, Faderl S, Van Q, et al. WP1066 disrupts Janus kinase-2 and induces caspase-dependent apoptosis in acute myelogenous leukemia cells. Cancer Res. 2007;67:11291-11299.

[188] Lipka DB, Hoffmann LS, Heidel F, et al. LS104, a non-ATP-competitive small-molecule inhibitor of JAK2, is potently inducing apoptosis in JAK2V617F-positive cells. Mol Cancer Ther. 2008;7:1176-1184.

[189] James C. The JAK2V617F mutation in polycythemia vera and other myeloproliferative disorders: one mutation for three diseases? Hematology Am Soc Hematol Educ Program. 2008:69-75.

[190] Kilpivaara O, Mukherjee S, Schram AM, et al. A germline JAK2 SNP is associated with predisposition to the development of JAK2(V617F)-positive myeloproliferative neoplasms. Nat Genet. 2009;41:455-459.

Page 149: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

149

[191] Olcaydu D, Harutyunyan A, Jager R, et al. A common JAK2 haplotype confers susceptibility to myeloproliferative neoplasms. Nat Genet. 2009;41:450-454.

[192] Jones AV, Chase A, Silver RT, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet. 2009;41:446-449.

[193] Vannucchi AM, Guglielmelli P, Rambaldi A, Bogani C, Barbui T. Epigenetic therapy in myeloproliferative neoplasms: evidence and perspectives. J Cell Mol Med. 2009;13:1437-1450.

[194] Dusa A, Mouton C, Pecquet C, Herman M, Constantinescu SN. JAK2 V617F constitutive activation requires JH2 residue F595: a pseudokinase domain target for specific inhibitors. PLoS One. 2010;5:e11157.

[195] Gnanasambandan K, Magis A, Sayeski PP. The constitutive activation of Jak2-V617F is mediated by a pi stacking mechanism involving phenylalanines 595 and 617. Biochemistry. 2010;49:9972-9984.

[196] Zhao L, Ma Y, Seemann J, Huang LJ. A regulating role of the JAK2 FERM domain in hyperactivation of JAK2(V617F). Biochem J. 2010;426:91-98.

[197] Funakoshi-Tago M, Pelletier S, Matsuda T, Parganas E, Ihle JN. Receptor specific downregulation of cytokine signaling by autophosphorylation in the FERM domain of Jak2. EMBO J. 2006;25:4763-4772.

[198] Wernig G, Gonneville JR, Crowley BJ, et al. The Jak2V617F oncogene associated with myeloproliferative diseases requires a functional FERM domain for transformation and for expression of the Myc and Pim proto-oncogenes. Blood. 2008;111:3751-3759.

[199] Robertson SA, Koleva RI, Argetsinger LS, et al. Regulation of Jak2 function by phosphorylation of Tyr317 and Tyr637 during cytokine signaling. Mol Cell Biol. 2009;29:3367-3378.

[200] Pardanani A, Hood J, Lasho T, et al. TG101209, a small molecule JAK2-selective kinase inhibitor potently inhibits myeloproliferative disorder-associated JAK2V617F and MPLW515L/K mutations. Leukemia. 2007;21:1658-1668.

[201] Mathur A, Mo JR, Kraus M, et al. An inhibitor of Janus kinase 2 prevents polycythemia in mice. Biochem Pharmacol. 2009;78:382-389.

[202] Verstovsek S, Manshouri T, Quintas-Cardama A, et al. WP1066, a novel JAK2 inhibitor, suppresses proliferation and induces apoptosis in erythroid human cells carrying the JAK2 V617F mutation. Clin Cancer Res. 2008;14:788-796.

Page 150: CHARACTERIZATION OF STRUCTURE-FUNCTION …ufdcimages.uflib.ufl.edu/UF/E0/04/32/10/00001/majumder_a.pdf · support and encouragement. Finally, thanks to my husband, Arjun, for bearing

150

BIOGRAPHICAL SKETCH

Anurima Majumder was born in 1984, in the small town of Durgapur, India. Even

as a child, Anurima was inspired by her role model, her father, to pursue a career in

academics and become a professor like him. She attended Carmel High School, where

she was first inspired to pursue a career in bioscience by her biology teacher, Mrs.

Geeta Menon. Anurima went on to earn her Bachelor of Engineering degree in

biotechnology from the Bengal College of Engineering and Technology, in August 2006.

In August 2007, she moved to Gainesville, Florida and joined the Interdisciplinary

Program in Biomedical Sciences at the University of Florida in pursuit of a Ph.D. She

began her graduate dissertation research under the guidance of Dr. Peter Sayeski in

May, 2008. Her research was primarily focused on studying structure-activity

relationships of novel Jak2 small molecule inhibitors. Anurima graduated in August 2011

and is now pursuing her postdoctoral studies at the University of Iowa.