identification and characterization of small molecule …...ii identification and characterization...

90
Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3'-Phosphatase by Nathalie Moatti A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Molecular Genetics University of Toronto © Copyright by Nathalie Moatti 2012

Upload: others

Post on 30-Sep-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3'-Phosphatase

by

Nathalie Moatti

A thesis submitted in conformity with the requirements for the degree of Master of Science

Department of Molecular Genetics University of Toronto

© Copyright by Nathalie Moatti 2012

Page 2: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

ii

Identification and Characterization of Small Molecule Inhibitors of

Polynucleotide Kinase 3’-Phosphatase

Nathalie Moatti

Master of Science

Department of Molecular Genetics

University of Toronto

2012

Abstract

DNA lesions arise constantly in cells and are repaired by a variety of DNA repair pathways.

Polynucleotide kinase 3’-phosphatase (PNKP) aids repair by phosphorylating 5’-hydroxyl DNA

termini and dephosphorylating 3’-phosphate DNA termini for the completion of repair by DNA

ligases. This activity is critical in vivo because DNA breaks do not usually possess ligatable

termini.

PNKP knockdown sensitizes cells to several DNA damaging agents, including the topoisomerase

I (TOP1) inhibitor camptothecin - analogs of which are being developed into chemotherapeutic

drugs - because the resolution of stalled TOP1-DNA complexes requires processing by PNKP.

We hypothesize that small molecule inhibitors of PNKP could bolster the effects of radio- and

chemotherapies on cancer cells.

I have identified eight compounds that effectively inhibit human PNKP and, with reduced

potency, T4 PNK in vitro. These compounds act by reversibly inhibiting the substrate-enzyme

interaction but they do not appear to sensitize U2OS cells to camptothecin.

Page 3: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

iii

Acknowledgments

I would like to thank my supervisor, Dr. Daniel Durocher, for his guidance, encouragement, and

support during my time in the lab – without a doubt it helped me get through the long periods of

troubleshooting. I would also like to thank the members of the Durocher lab, not only for their

constant willingness to help, but also simply for being great people to work with. For many

reasons, I am often reminded of how grateful I am to have joined this lab. I am indebted to

Thomas Sun, Fred Vizeacoumar, and Alessandro Datti of the SMART robotics facility for all of

their help with my screen. I also owe many thanks to my supervisory committee members, Drs.

Frank Sicheri and Peter Roy, as well as our collaborators in Dr. Mark Glover’s lab at the

University of Alberta and Dr. David Uehling at the Ontario Institute for Cancer Research. Lastly,

thanks to my family and friends for their support and great efforts to understand what I’ve been

working on these past two years.

Page 4: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

iv

Table of Contents

Acknowledgments.......................................................................................................................... iii

Table of Contents ........................................................................................................................... iv

List of Tables ................................................................................................................................. vi

List of Figures ............................................................................................................................... vii

List of Appendices ......................................................................................................................... ix

1 Introduction .................................................................................................................................1

1.1 DNA repair is critical for maintaining genomic stability and preventing tumorigenesis ....1

1.2 Targeted cancer therapies, like poly-ADP ribose polymerase inhibitors, kill tumor

cells with fewer side effects than chemotherapy .................................................................2

1.3 Polynucleotide kinase 3’-phosphatase processes certain types of damaged DNA

termini to allow for the completion of DNA repair by ligation ...........................................4

1.4 The importance of PNKP for DNA repair during development ..........................................7

1.5 Inhibitors of PNKP’s phosphatase activity would be useful tools for research and

clinical applications .............................................................................................................8

2 Results .......................................................................................................................................10

2.1 Development of screen conditions, Version 1 ...................................................................10

2.2 Development of screen conditions, Version 2 ...................................................................17

2.3 Homogeneous time-resolved FRET-based screen .............................................................24

2.4 Putative inhibitors of hPNKP.............................................................................................26

2.5 Validation and IC50 measurement of putative hPNKP inhibitors by HTRF ......................27

2.6 Optimization of gel-based phosphatase assay ....................................................................29

2.7 Measurement of IC50 values by gel-based phosphatase assay ...........................................30

2.8 Structure activity relationship analysis ..............................................................................31

2.9 Optimization of fluorescence polarization-based substrate-binding assay ........................37

2.10 Substrate-binding assay confirms that compounds prevent a 3'-phosphorylated

oligonucleotide from binding to hPNKP ...........................................................................38

2.11 Inhibitors of hPNKP display different degrees of reversibility .........................................39

2.12 Inhibitors of hPNKP can act on T4 PNK with reduced potency .......................................41

2.13 Inhibitors of hPNKP do not show a clear effect in U2OS cells co-treated with

camptothecin ......................................................................................................................44

Page 5: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

v

3 Discussion and future directions ...............................................................................................51

4 Materials and Methods ..............................................................................................................57

4.1 Transformation of E. coli. ..................................................................................................57

4.2 SDS-PAGE. .......................................................................................................................57

4.3 Quantitation of protein .......................................................................................................57

4.4 Cloning of human PNKP-D171A ......................................................................................58

4.5 Purification of human PNKP and PNKP-D171A ..............................................................58

4.6 Purification of T4 DNA Ligase..........................................................................................60

4.7 Preparation of 3'-phosphorylated double-stranded DNA substrates. .................................63

4.8 Measurement of PNKP-dependent ligation by homogeneous time-resolved

fluorescence HTRF) in a 384-well format, Version 1. ......................................................63

4.9 Measurement of PNKP-dependent ligation by HTRF in a 384-well format, Version 2. ..64

4.10 Measurement of PNKP-independent ligation by HTRF in a 384-well format, Versions

1 and 2. ...............................................................................................................................64

4.11 Screening............................................................................................................................65

4.12 Calculation of z’ factor, signal-to-noise (S/N), signal-to-background (S/B), and

percent error. ......................................................................................................................66

4.13 Preparation of polyacrylamide sequencing gels. ...............................................................67

4.14 Phosphatase gel assay. .......................................................................................................67

4.15 Fluorescence polarization (FP) substrate binding assay. ...................................................68

4.16 Measurement of the Kd of hPNKP-D171A in the FP substrate binding assay. .................68

4.17 Reversibility assay. ............................................................................................................69

4.18 Tissue culture. ....................................................................................................................69

4.19 ATP Lite cell viability assay. .............................................................................................70

Tables .............................................................................................................................................71

References Cited ............................................................................................................................70

Appendices .....................................................................................................................................75

Page 6: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

vi

List of Tables

Table 1a Buffers ............................................................................................................................ 71

Table 2a Sequences of oligonucleotide substrates (5' to 3') ........................................................ 73

Table 1 Original hit compounds ................................................................................................... 74

Table 2 Analog compounds .......................................................................................................... 76

Page 7: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

vii

List of Figures

Figure 1.3.1 PNKP is critical for the repair of DNA breaks with 3’-phosphate and/or 5’-hydroxyl

termini ............................................................................................................................................. 7

Figure 2.1.1 Optimization of HTRF screen conditions ................................................................ 13

Figure 2.1.2 Optimization of the order of addition, stability of reagents at room temperature, and

hPNKP concentration for HTRF screen conditions ...................................................................... 15

Figure 2.1.3 Screening the Prestwick Chemical Library confirmed that the optimized conditions

were amenable to high-throughput screening ............................................................................... 17

Figure 2.2.1 Further optimization of the HTRF assay was undertaken to find non-saturating

conditions with high statistical validity ........................................................................................ 20

Figure 2.2.2 The T4 Ligase and hPNKP concentrations were optimized to prevent the ligation

reaction from causing a kinetic lag ............................................................................................... 22

Figure 2.2.3 Optimization of DNA concentration, ligation product length, and acceptor

fluorophore concentration to maximize signal-to-background ..................................................... 24

Figure 2.3.1 Screening of the OICR HTS chemical collection under primary and secondary

screen conditions to identify putative hPNKP inhibitors .............................................................. 26

Figure 2.5.1 Several putative inhibitors were validated as inhibitors of hPNKP with a range of

potencies, while others could not be reconfirmed ........................................................................ 28

Figure 2.6.1 A gel-based assay was developed to directly measure the dephosphorylation

activity of PNKP ........................................................................................................................... 30

Page 8: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

viii

Figure 2.7.1 Dose response experiments were performed for each of the five original hits using

the gel assay, and IC50 values were measured .............................................................................. 31

Figure 2.8.1 Structure activity relationship analyses of the heterocyclic ring and the fragment

common to OICR7 and OICR9..................................................................................................... 33

Figure 2.8.2 Structure activity relationship analyses using analogs of OICR79 ......................... 35

Figure 2.8.3 Structure activity relationship analysis of OICR79 uncovered two modifications

that did not abolish inhibition ....................................................................................................... 37

Figure 2.10.1 Fluorescence polarization-based direct binding assays were carried out to

determine if the compounds could compete with a 3’-phosphorylated oligonucleotide substrate

for binding to hPNKP-D171A ...................................................................................................... 39

Figure 2.11.1 Reversibility assays demonstrated that most of the compounds were at least

slightly reversible .......................................................................................................................... 41

Figure 2.12.1 Gel-based assays conducted with T4 PNK demonstrate that some, but not all, of

the hPNKP inhibitors are capable of inhibiting this distant ortholog ........................................... 43

Figure 2.13.1 The viability of U2OS cells co-treated with 5 nM camptothecin (CPT) was not

significantly impaired relative to their DMSO-treated counterparts ............................................ 45

Figure 4.6.1 hPNKP and T4 Ligase were purified from BL21 (DE3) CodonPlus E. coli cells .. 62

Figure 4.16.1 FP reactions were conducted with hPNKP for proof of concept and to establish

optimal reaction conditions ........................................................................................................... 69

Page 9: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

ix

List of Appendices

PNKP inhibitors ............................................................................................................................ 75

Analogs that did not effectively inhibit PNKP ............................................................................. 76

Page 10: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

1

1 Introduction

1.1 DNA repair is critical for maintaining genomic stability and preventing tumorigenesis

DNA lesions arise constantly as a consequence of cellular metabolism or in response to

genotoxic agents, requiring immediate attention so that the cell may divide safely (Branzei and

Foiani, 2008). In order to repair these lesions effectively, there are a variety of DNA repair

pathways available depending on the nature of the damage, which allow cells to continue

functioning normally (Branzei and Foiani, 2008). However, if these pathways fail to mend even

one DNA lesion, the cell and its lineage can incur mutations or larger-scale genomic aberrations

with potentially disastrous consequences including cell death, especially if that break is double-

stranded (Jackson, 2002).

Cancer is a disease that arises when a cell loses its ability to control proliferation, causing the cell

to divide incessantly and form one or more tumors (Croce, 2008). This unrestrained growth is

borne from genomic translocations and/or mutations that activate oncogenes and disable tumor

suppressor genes (Croce, 2008). The term “oncogene” signifies a gene whose protein product

originally played a role in the regulation of proliferation, differentiation, or apoptosis, but has

since become mutated in such a way that causes it to promote or allow constitutive growth,

which can lead to tumorigenesis (Croce, 2008). Conversely, tumor suppressor genes act to

stabilize the genome and limit proliferation when the cell is stressed; for example, when faced

with a genotoxic agent (Weinberg, 1991). Proteins involved in the DNA damage response

(DDR) fall into this latter category, as they act to maintain the integrity of the genome and

perform checkpoints for cell division, which arrest the cell cycle until genomic stability is

recovered.

Page 11: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

2

2

In cases of familial breast, ovarian, and prostate cancer, 5-20% are traced to heterozygotic

germline mutations in BRCA1 or BRCA2 (Venkitaraman, 2002), two tumor suppressor genes that

are critical for homologous recombination (HR) - the least mutagenic form of double-strand

break repair (DSBR). Although BRCA1/2+/-

cells are capable of efficiently and accurately

repairing double-strand breaks (DSBs), it has been proposed that tumorigenesis is initiated by a

loss-of-heterozygosity event, wherein the sole functional BRCA1/2 allele acquires a nonsense

mutation (Miki et al., 1994; Wooster et al., 1995). This complete loss of BRCA1/2 activity

creates genomic instability in the cell and acts as a “driver mutation” by promoting the

development of new mutations in other genes, some of which are likely to exacerbate the

problem (Shah, 2008). In fact, it has been suggested that the inactivation of DDR factors is a

critical step in the formation of all cancers, as the aberrant growth of cancer cells creates

excessive DNA replication stress, which would normally activate the DDR and trigger cell cycle

arrest or cell death (Jackson and Bartek, 2009).

1.2 Targeted cancer therapies, like poly-ADP ribose polymerase inhibitors, kill tumor cells with fewer side effects than chemotherapy

When it comes to treating cancer, there are two main types of drugs available – chemotherapies

and targeted therapies. Chemotherapies include DNA damaging agents like cisplatin, which acts

by creating DNA inter-strand cross-links and invokes a number of pathways – cell cycle arrest

and p53-induced apoptosis, for instance – and results in preferential killing of rapidly diving

cells (Siddik, 2003). Consequently, these drugs effectively eliminate cancer cells but they also

harm the body’s naturally rapidly-diving cells, such as hair and gut cells, resulting in a slew of

unwelcome side effects. On the other hand, targeted therapies work on the basis of the specific

Page 12: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

3

genetic aberrations present in a given patient’s cancer cells so that they theoretically harm only

the abnormal cells and cause minimal side effects to healthy cells, thus creating a much larger

treatment window (Sawyers, 2004). One class of targeted therapies that has garnered massive

attention over the last few years is that of poly-ADP ribose polymerase (PARP) inhibitors.

PARP-1 is an enzyme with myriad roles in vivo, one of which is in the detection and repair of

single-strand breaks (SSBs), formed directly or as a product of base excision repair (BER;

Helleday, 2011; Satoh and Lindahl, 1992). When a base is damaged, the removal of it and a few

adjacent bases forms an unprotected SSB, which is a substrate for rapid recognition and transient

binding by PARP-1 (Strom et al., 2011). Upon binding to the SSB, PARP-1 is activated and

recruits XRCC1-Ligase III to initiate single-strand break repair (SSBR; El-Khamisy et al., 2003).

Importantly, PARP-1 is also hypothesized to mediate a distinct pathway of replication repair by

enabling the restart of stalled replication forks (Bryant et al., 2009). Inhibitors of PARP1 have

shown promise in the treatment of BRCA1/2 defective cancers because PARP-1 and BRCA1/2

exhibit a synthetic lethal relationship; that is, cells can survive the inactivation of PARP-1 or

BRCA1/2 alone, but they die when the two enzymes are lost concurrently (Bryant et al., 2005;

Farmer et al., 2005). As such, PARP inhibitors are effective in killing BRCA1/2 cancer cells,

while doing minimal harm to normal, BRCA1/2 positive cells (Kling, 2009). Thanks to the

promising results seen in tissue culture (Bryant et al., 2005; Farmer et al., 2005), the effects of

several PARP inhibitors, such as Olaparib (Astra Zeneca, UK), Veliparib (Abbott Laboratories,

USA) and Iniparib (Sanofi Aventis, France), on BRCA1/2 tumors are currently being investigated

in clinical trials. Furthermore, because the efficacy of PARP inhibitors in BRCA1/2 cancer cells

is thought to be due to the dual loss of PARP- and HR-mediated replication fork-restart

(Helleday, 2011), there is reason to believe that PARP inhibitors can be useful in treating other

Page 13: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

4

HR-deficient “BRCA-like” cancers (Anders et al., 2010) or as potentiators of chemo- and radio-

therapy (Zaremba and Curtin, 2007).

In 2008, a siRNA-based study conducted by Turner et al. identified genes whose silencing would

sensitize CAL51 metastatic mammary adenocarcinoma cells (Gioanni et al., 1990) to the potent

PARP inhibitor KU0058948 (Farmer et al., 2005). The CAL51 cells employed by the authors

were diploid, BRCA1/2-wild type (WT) and TP53-WT, which they reasoned would facilitate the

identification of new determinants of sensitivity to PARP inhibitors with minimal interference

from other somatic mutations (Turner et al., 2008). Their analysis revealed that, out of the six on-

target hits identified, loss of polynucleotide kinase 3’-phosphatase (PNKP) was the most

effective in sensitizing CAL51 and cervical carcinoma HeLa cells to PARP inhibition, with a

striking 63-fold increase in sensitivity to the PARP inhibitor (Turner et al., 2008). These results

strongly suggest that inhibitors of PNKP could also be promising drugs for the treatment of

BRCA1/2 or HR-deficient cancers as monotherapies, or as sensitizers to PARP inhibitors,

chemotherapy, and/or radiation therapy.

1.3 Polynucleotide kinase 3’-phosphatase processes certain types of damaged DNA termini to allow for the completion of DNA repair by ligation

Mammalian PNKP was first identified in the 1980s and subsequently characterized in the late

1990s (Habraken and Verly, 1983; Karimi-Busheri et al., 1999; Pheiffer and Zimmerman, 1982).

Like its well-known T4 counterpart, mammalian PNKP contains a 5’-kinase domain and a 3’-

phosphatase domain - together termed the “catalytic domain” – which enable this enzyme to

phosphorylate the 5’- ends of DNA and de-phosphorylate the 3’-ends of DNA (Bernstein et al.,

2005). However, unlike T4 PNK, mammalian PNKP also has a forkhead associated (FHA)

domain, which is responsible for recruiting PNKP to DNA breaks (Bernstein et al., 2005).

Page 14: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

5

FHA domains are common among a number of DNA repair proteins and are responsible for a

range of protein-protein interactions owing to their affinity for phosphothreonine, but typically

not phosphoserine, residues (Mohammad and Yaffe, 2009). In the case of PNKP, the FHA

domain mediates interactions with X-ray repair cross complementing group 1 (XRCC1) and X-

ray repair cross complementing group 4 (XRCC4), which function as scaffolding proteins during

SSBR and the non-homologous end-joining (NHEJ) arm of DSBR, respectively, to recruit

several DNA repair factors to the site of the break, including PNKP (Lee et al., 2000;

Whitehouse et al., 2001). PNKP is also recruited by XRCC1 to DSBs in the alternative end-

joining pathway of DSBR (Audebert et al., 2004), but not HR (Karimi-Busheri et al., 2007).

The interaction between the PNKP FHA domain and XRCC1 is rather unique, as the FHA binds

to a vicinal phosphothreonine-phosphoserine motif in XRCC1 (XRCC1; Ali et al., 2009), which

is generated by casein kinase 2 (CK2) upon DNA damage (Loizou et al., 2004). Conversely, the

interaction between the FHA domain and CK2-phosphorylated XRCC4 appears to occur via a

specific phosphothreonine residue in XRCC4, with residues at positions +1 and -2 relative to the

pThr (pT+1 and pT-2, respectively) contributing to binding specificity (Chappell et al., 2002;

Koch et al., 2004). Intriguingly, for both XRCC1 and XRCC4, the FHA domain exhibits a lack

of selectivity for the pT+3 residue, which is unique because pT+3 specificity is a feature of all

other FHA domains studied to date (Bernstein et al., 2005; Pennell et al., 2010).

Additionally, it has been shown that the PNKP FHA binds to XRCC1 with a stoichiometry of 2:1

in a hierarchical and cooperative fashion: binding of the vicinal pSer/pThr motif by one PNKP

FHA facilitates the interaction of a second PNKP FHA with another CK2-phosphorylated

threonine nearby (Ali et al., 2009); presumably, this 2:1 stoichiometry aids in increasing the

processivity of the XRCC1-based multiprotein complex at damaged SSBs (Ali et al., 2009).

Page 15: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

6

Aside from DSBs mended by HR, the DNA end-processing functions of PNKP are critical for

the repair of most DNA breaks because all known DNA polymerases and ligases require 5’-

phosphate and 3’-hydroxyl termini as substrates (Doherty and Suh, 2000), but most SSBs and

DSBs are “dirty”, and thus require processing to produce conventional termini before repair can

be completed (Caldecott, 2008). Specifically, ionizing radiation, reactive oxygen species (ROS),

the process of BER, and Topoisomerase I (Top1) poisons such as camptothecin (CPT) frequently

create 5’-OH and/or 3’-phosphate groups (Figure 1.3.1; Caldecott, 2008). Accordingly, it has

been shown that the recruitment of PNKP to SSBs by XRCC1 is crucial for timely SSBR in cells

following oxidative damage (Breslin and Caldecott, 2009). Notably, the importance of PNKP in

SSBR was attributed to its 3’-phosphatase activity because overexpression of wild-type but not

phosphatase-dead PNKP was sufficient to restore rapid SSBR when the PNKP-XRCC1

interaction was abolished (Breslin and Caldecott, 2009).

Page 16: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

7

Figure 1.3.1 PNKP is critical for the repair of DNA breaks with 3’-phosphate and/or 5’-hydroxyl

termini. Upon detection of a damaged base (top), APE1 creates an apurinic/apyrimidinic (AP) site with

both types of damaged ends. Reactive oxygen species (ROS; middle), produced by exogenous sources or

as a natural consequence of ATP production, attack the sugars in DNA, which often leads to 3’-phosphate

ends as well as 5-‘hydroxyl ends to a lesser extent. The stalling of topoisomerase I (Top1; bottom), due to

poisons like camptothecin, leads to detection and processing by tyrosyl-DNA phosphodiesterase 1

(Tdp1), which generates one or both types of damaged termini via removal of Top1. Processing of these

breaks by PNKP creates conventional DNA termini, which allows the appropriate polymerases and

ligases to complete the repair process.

1.4 The importance of PNKP for DNA repair during development

During development, the rapid proliferation of stem and progenitor cells creates a great deal of

oxidative stress for the DNA replication machinery and this can lead to mutations and/or cell

death if replication errors are not dealt with appropriately (O'Driscoll and Jeggo, 2008). It has

been proposed that neuroprogenitor cells are particularly sensitive to DSBs, and an accumulation

of DNA damage can induce large-scale apoptosis in this population, resulting in defective

neuronal development; accurate DNA repair is therefore critical at this early stage (Ciccia and

Elledge, 2010). This hypothesis is thought to explain why a number of neurological disorders

have been attributed to mutations in DNA repair proteins; for example, defects in ataxia

telangiectasia mutated (ATM) are responsible for ataxia telangiectasia (Savitsky et al., 1995),

and ataxia oculomotor apraxia is due to a defect in Aprataxin (APTX; Moreira et al., 2001).

Page 17: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

8

In line with the findings of Breslin and Caldecott (2009), which outlined the importance of the

phosphatase activity of PNKP in maintaining rapid SSBR after oxidative stress, mutations in

PNKP have been identified as the origin of the autosomal recessive disorder MCSZ

(microcephaly with seizures; Shen et al., 2010). MCSZ is characterized by microcephaly, early-

onset seizures, developmental delays, and variable behavioural problems (Shen et al., 2010).

Furthermore, lymphocytes from MCSZ patients were found to be significantly impaired in their

abilities to repair hydrogen peroxide- and CPT-induced damage (Shen et al., 2010).

1.5 Inhibitors of PNKP’s phosphatase activity would be useful tools for research and clinical applications

The body of research surrounding PNKP points to its importance in effecting rapid and accurate

DNA repair in the SSBR and NHEJ pathways. Taking into consideration that NHEJ is the main

method of DSBR in vivo (Mao et al., 2008), it follows that inhibiting PNKP would severely

hinder most SSBR and DSBR, which could prove to be even more effective than PARP

inhibitors in eliminating HR-impaired tumor cells. Furthermore, evidence for the sensitization of

A549 lung cancer cells to gamma radiation and camptothecin by stable down-regulation of

PNKP (Rasouli-Nia et al., 2004) makes a strong case for the potential to increase the efficacy of

certain classic cancer therapies – such as radiation therapy and Top1 poisons – by combining

them with PNKP inhibitors (Bernstein et al., 2008). Toward this end, as well as to generate new

tools for the study of PNKP, my work has focused on the identification of PNKP inhibitors

through high-throughput screening, and characterization with follow-up assays.

In order to query a variety of chemical structures, I developed a high-throughput screen based on

homogeneous time-resolved fluorescence resonance energy transfer (HTR-FRET or HTRF)

technology, which measured PNKP-dependent ligation of 3’-phosphorylated DNA substrates.

Page 18: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

9

Importantly, I used 3’-phosphorylated DNA to search for inhibitors of PNKP’s 3’-phosphatase

activity as opposed to its 5’-kinase activity because 3’-phosphates are far more common than 5’-

hydroxyls at damaged DNA ends in vivo (Buchko and Weinfeld, 1993; Coquerelle et al., 1973;

Henner et al., 1982). Also, despite the fact that Ape1 and Aptx are capable of mending 3’-

phosphates, it has been demonstrated that their phosphatase activities pale in comparison to that

of PNKP (Reynolds et al., 2009; Wiederhold et al., 2004) – specifically, 3’-phosphates are

removed by PNKP at a rate at least ten-fold greater than that of Aptx and nearly 1000-fold

greater than that of Ape1 (Takahashi et al., 2007; Wiederhold et al., 2004).

Primary and secondary HTRF-based screening of more than 142,000 compounds yielded nine

hits, five of which still exhibited inhibition of PNKP when obtained commercially and tested

with freshly dissolved stocks. I measured the relative potencies of these five compounds by

determining the concentration of compound at which dephosphorylation is inhibited by 50% -

termed an “IC50 value” - with HTRF- and gel-based assays. In addition to these five “hits”, I

acquired several analog compounds from commercial sources and tested their potency in order to

perform structure-activity relationship analyses. I further characterized the compounds and active

analogs by assessing their degree of reversibility and abilities to inhibit the distant ortholog T4

PNK. Finally, I assessed the ability of the inhibitors to sensitize U2OS cells to CPT, but they did

not appear to significantly reduce the viability of CPT-treated cells compared to control cells.

The sum of this work reinforces that the five original hits identified, as well as the three active

analog compounds, are true inhibitors of human PNKP and may prove useful in the development

of biologically active PNKP inhibitors for research or clinical purposes, but further study will be

required to determine their effectiveness in cells.

Page 19: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

10

2 Results

2.1 Development of screen conditions, Version 1

In an effort to develop a high-throughput assay for measuring the phosphatase activity of PNKP,

several techniques were investigated in our lab by Marella Canny before she found that

homogenous time-resolved fluorescence resonance energy transfer (HTR-FRET or HTRF)

provided high signal-to-background ratios and low variability. HTRF is a FRET-based

technology that employs a Europium cryptate as a donor fluorophore capable of emitting

fluorescence up to 100 times longer than more common donor fluorophores like Cy3 (Bazin et

al., 2002). Because of this long-lived donor emission, the delay between donor excitation and

measurement of acceptor fluorescence is much longer than it would be in a standard FRET

measurement, and this is beneficial because it allows any short-lived background fluorescence to

dissipate before the measurement is taken, which makes for stronger signal-to-background ratios

(Bazin et al., 2002).

The use of cross-linked allophycocyanin (symbolized “XL-665”) as the acceptor fluorophore is

the second feature that makes HTRF unique because the Europium cryptate/XL-665 FRET pair

has an exceptionally large Forster distance of 9 nm, which means that they exhibit a much higher

efficiency of energy transfer (Bazin et al., 2002). The Europium cryptate is conjugated to an anti-

2,4-dinitrophenol (DNP) antibody (“DNPK”), and XL-665 is conjugated to a streptavidin

molecule (“SAXL”) so that the two can be targeted to molecules of interest that are tagged with

DNP or biotin, respectively. In order to measure the phosphatase activity of PNKP, Marella

designed complimentary DNA substrates wherein one double-stranded DNA (dsDNA) has a 5’-

DNP tag, the other dsDNA has a 3’-biotin tag, and they each have a 3’-phosphate end (Table 2).

Page 20: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

11

Marella’s early experiments indicated that the dephosphorylation of these DNA substrates could

be measured by HTRF with reactions that contained 44 nM Homo sapiens PNKP (hPNKP) and

74 nM T4 Ligase in HTRF buffer (Table 1) with 0.1 mg/ml BSA. Under these conditions, the

HTRFphos-1 and HTRFphos-2 substrates (Table 2) were incubated at 20 nM. SAXL and DNPK

were diluted 100-fold in PBS (Table 1) and added separately at the end of the

dephosphorylation-ligation reaction. Once hPNKP dephosphorylated the DNA substrates, T4

Ligase was then able to ligate them due to their complimentary 5’ overhangs. The ligation of the

two DNA strands would bring the donor and acceptor fluorophores sufficiently close together to

produce a HTRF signal. The “background” levels of HTRF were then measured by performing

the assay with samples that lack enzyme and should therefore not contain ligated product, which

was then used to normalize HTRF signal between experiments (i.e. “signal/background”).

Though Marella’s studies showed proof of concept, a number of parameters required testing and

optimization in order to adapt this assay for high-throughput compound screening. Because the

compounds to be tested are dissolved in DMSO, it was necessary to determine if the

concentration of DMSO had an effect on the assay. I accomplished this by carrying out the

aforementioned HTRF reactions with 1%, 2.5%, 5%, or no DMSO. The results of this

experiment indicated that the inclusion of up to 5% DMSO in the reaction did not significantly

affect HTRF signal (Figure 2.1.1.a).

Another parameter that required optimization was the concentration of potassium fluoride (KF)

in the DNPK solution: according to the manufacturer (Cisbio Bioassays, USA), reactions

containing Europium cryptate require 100 – 400 mM KF to protect the cryptate from quenchers

that may be present in the reaction at the time of HTRF measurement. Also, the manufacturer

states that KF is only required at the time of HTRF measurement; therefore, I introduced the KF

Page 21: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

12

with the DNPK solution (Table 1) because that was the last component to be added to the

reaction before HTRF was read.

I carried out HTRF reactions in the presence of 100, 200, 300, or 400 mM KF. I observed that,

although variability remained the same across KF concentrations of 200 mM or greater, there

was an apparent decrease in signal with decreasing KF concentration (Figure 2.1.1.b). Notably,

HTRF signal was very poor at 100 mM KF. On this basis, I concluded that the final

concentration of KF in HTRF reactions should be 400 mM.

The next condition that I optimized was the duration of the dephosphorylation-ligation reaction.

To achieve this, I performed HTRF reactions as previously described and reactions were allowed

to proceed for 0.5, 1, 1.5, 2, 2.5, or 3 h at room temperature. To ensure that the incubation time

with fluorophores was consistent between time-points, I started the reactions every 30 min, and

then SAXL and DNPK solutions were added to all wells after 3 h. The results of the time course

with 44 nM hPNKP and 74 nM T4 Ligase showed high amounts of signal at each time point

(Figure 2.1.1.c) and I determined that a 1.5 h incubation time would provide the greatest signal

window within a reasonable timeframe.

Another important optimization step was that of hPNKP and DNA concentrations. Although the

aforementioned optimizations were performed under previously established conditions, it was

necessary to verify that the enzyme and substrate concentrations were optimal in my hands.

hPNKP was diluted in PNKP dilution buffer (Table 1) and reactions were carried out as

described with 88, 44, 22, 8.8 or 0.88 nM hPNKP (Figure 2.1.1.d). Because maximizing signal

was considered paramount, I concluded that 44 nM of hPNKP (50 ng) was still an appropriate

concentration to work with. I then performed HTRF reactions at 44 nM hPNKP with final

Page 22: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

13

HTRFphos-1 and HTRFphos-2 (Table 2) DNA concentrations of 0, 5, 10, 20, 35, 50, 200, or 250

nM (Figure 2.1.1.e). From this experiment, I found that maximum signal was achieved at 35-50

nM DNA, so I selected 40 nM of DNA substrates as the optimal DNA concentration for this

assay.

Figure 2.1.1 Optimization of HTRF screen conditions. (a) HTRF assays were carried out with different

final concentrations of DMSO to verify that DMSO concentration would not affect signal. (b) HTRF

assays were carried out at 0 - 400 mM KF to determine which concentration produced maximum signal.

(c) A time course for the HTRF reaction was carried out to select an incubation time that maximized

signal. (d) hPNKP was titrated in HTRF reactions to determine which concentration produced maximum

signal. (e) DNA was titrated with 42 nM SAXL and 2 nM DNPK fluorophores in HTRF reactions to

determine the optimal concentration of DNA for use with the aforementioned concentration of

fluorophores. Error bars for (a) and (d) represent standard error of the mean (SEM) for at least two

independent experiments. Error bars for (b), (c), and (e) represent the SEM for two technical replicates.

Until this point, I had prepared SAXL and DNPK solutions by separately diluting each reagent

100-fold in PBS (Table 1), and these were added sequentially at the end of the reaction; however,

to optimize this assay for a high-throughput screen it was necessary to determine if the order of

fluorophore addition could be modified to minimize the number of addition steps. To this end, I

Page 23: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

14

carried out HTRF reactions wherein SAXL and DNPK were diluted and added separately as

before, diluted and added together, or diluted and added together with the DNA substrates

(Figure 2.1.2.a). The results of this experiment suggested that SAXL and DNPK could be

diluted and added together at the end of the reaction, albeit with a small loss of signal. I also

subsequently carried out reactions in which SAXL was either added at the end of the reaction

with DNPK or included in the reaction with the DNA substrates, and this experiment

demonstrated that the latter order of addition led to a marked increase in signal (Figure 2.1.2.b),

so I adopted this order of addition for the screen.

Due to the nature of high-throughput screening, it was also important to verify that the HTRF

reagents were stable for long periods of time at room temperature as each screen run can take

several hours. To test this, I prepared dilutions of SAXL and DNPK in PBS (Table 1) every 2 h,

up to 6 h. HTRF reactions were initiated 4.5 h after the first SAXL and DNPK dilutions were

made and incubated for 1.5 h. I ended the reactions at the same time by adding the SAXL and

DNPK solutions that had been prepared at different times (i.e. 2 h, 4 h, 6 h ago or freshly made)

to the wells. The data suggested that the HTRF fluorophores were indeed stable at room

temperature for up to 6 h after being diluted to their working concentrations (Figure 2.1.2.c).

Lastly, I repeated the titration of hPNKP to ensure that the concentration of enzyme was

optimized for the screening conditions, and I determined that a concentration of 8.8 nM hPNKP

per reaction (10 ng) was the most suitable (Figure 2.1.2.d).

Page 24: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

15

Figure 2.1.2 Optimization of the order of addition, stability of reagents at room temperature, and

hPNKP concentration for HTRF screen conditions. (a,b) Different methods of preparing HTRF reagents

were tested to determine which order of addition produced maximum signal. (c) HTRF reagents were

tested after being left at room temperature to verify that the reagents would be stable for the duration of a

screen run. (d) hPNKP was again titrated to check that the concentration was appropriate with the newly

optimized HTRF conditions. Error bars represent the SEM for two technical replicates.

To verify that this assay was amenable to high-throughput screening, I used the optimized assay

to screen the Prestwick Chemical Library (Prestwick Chemical, France), which consists of 1200

FDA-approved drugs (Figure 2.1.3.a). According to the manufacturer, this library contains

compounds with a high degree of chemical and pharmacological diversity, as well as

bioavailability. From this screen, I identified seven inhibitory compounds (Figure 2.1.3.b).

However, further testing under hPNKP-independent HTRF conditions showed that these

compounds were inhibiting both PNKP-dependent and –independent ligation reactions (Figure

2.1.3.c).

Page 25: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

16

Importantly, the z’ factor was quantified for this screen as a measure of the its statistical validity,

because it represents a screen’s capacity for identifying hits by taking into account the assay’s

signal window as well as dynamic range (Zhang et al., 1999). Specifically, a ratio of 0.5 is the

minimum value considered acceptable for high-throughput screening because it signifies clear

separation of positive and negative controls plus or minus three standard deviations (Zhang et al.,

1999).

Based on this pilot screen’s z’ factor of 0.73 ± 0.02, this trial demonstrated that the assay was

compatible with high-throughput screening. Therefore, despite the fact that this screen did not

identify any putative hPNKP inhibitors, it did prove that the assay produces statistically valid

data and can reliably identify primary hit compounds.

Page 26: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

17

Figure 2.1.3 Screening the Prestwick Chemical Library confirmed that the optimized conditions were

amenable to high-throughput screening. (a) Control-based analyses were performed to identify the

compounds that reduced HTRF signal by greater than three standard deviations from the mean positive

control (light blue line) for each plate. The mean negative controls for each plate are represented by the

dark blue line. FA = fluorescence emitted at 665 nm by SA-XL (acceptor); FD = fluorescence emitted at

620 nm by DNPK (donor); HTRF = (FA/FD) × 10,000 (b) Seven compounds were found to inhibit the

HTRF reactions, representing a 0.58% hit rate. (c) A secondary HTRF assay measuring hPNKP-

independent ligation was used to conclude whether or not the reduction in signal observed in the screen

was specific to hPNKP inhibition. Because all of the compounds caused inhibition in this secondary

screen, they were not considered to be inhibitors of hPNKP.

2.2 Development of screen conditions, Version 2

I developed the first version of the screen in such a way as to maximize the signal window with

little regard for the kinetics of the reaction. While these conditions led to a high degree of

statistical validity, they were not ideal for an inhibitor screen because assaying a reaction at

equilibrium can mask the effects of weak inhibitors, which have the potential to be developed

into more potent inhibitors. In the case of such inhibitors, measuring the assay in the linear

portion is key because a weakly inhibited reaction will exhibit a slower initial velocity, but it will

Page 27: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

18

ultimately reach a level of signal similar to that of an uninhibited reaction at saturation

(Copeland, 2005). Consequently, I undertook further optimizations to find conditions that were

not saturating but still maintained a high degree of statistical validity.

First, I noted that, although a time course was performed early in the optimization process, the

chosen incubation time of 1.5 h resulted in a highly saturated reaction; therefore, the incubation

time required adjustment to alleviate the reaction’s saturation. It was also important to ensure

that the reactions were being efficiently stopped at the desired time-point.

I tested two approaches for stopping the reactions: addition of EDTA, which is a chelator of

magnesium – the divalent metal necessary for hPNKP and T4 Ligase activity - or addition of the

classic phosphatase inhibitor sodium orthovanadate (Na3VO4, Gordon, 1991). First, I determined

the concentrations of EDTA and Na3VO4 at which the reaction is inhibited by 50%, i.e. their IC50

values, by pre-incubating the reaction mix with EDTA at 0.1 – 50 mM or Na3VO4 at 100 nM –

20 mM (Figure 2.2.1.a-b). I found the IC50 of EDTA to be 7.6 mM (Figure 2.2.1.a), while that

of Na3VO4 was estimated to be about the same (Figure 2.2.1.b). I then carried out time courses

wherein reactions were stopped with either 50 mM EDTA, 10 mM Na3VO4, or 20 mM Na3VO4

(Figure 2.2.1.c). Notably, it appeared that, when reactions were stopped with Na3VO4, the HTRF

ratios (Fl645×10,000/Fl620) were within their typical ranges but the fluorescence intensity in each

channel was about 4-7 times lower than what was typically observed. This suggested that

Na3VO4 was quenching fluorescence, which was further supported by the observation that

fluorescence intensities were 2-fold lower for the wells containing 20 mM Na3VO4 than the wells

containing 10 mM Na3VO4 (data not shown). Furthermore, when reactions were stopped with

Na3VO4, the HTRF ratios at t0 were 2 – 3 times higher than what was typically observed; this

was not the case for reactions stopped with EDTA, so I concluded that addition of 50 mM EDTA

Page 28: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

19

was better suited for stopping the dephosphorylation and ligation reactions. Furthermore, to

minimize the number of additions during the screen, EDTA was included in the DNPK solution

(Table 1) without negatively impacting signal.

The presence of BSA in the screen was another condition that I reconsidered. I had added BSA

to the reactions as a carrier for the enzymes of interest, but its high concentration in the screen

could make it capable of binding compounds and sequestering them from the enzymes of

interest, which could produce false negatives and reduce sensitivity. To test if this could be

occurring, I assessed the effects of three putative hPNKP inhibitors by HTRF in the presence and

absence of BSA at 0.1 mg/ml (Figure 2.2.1.d). I found that the inclusion of BSA reduced the

inhibitory effects by 20-25% for two of the three compounds that were tested, with little effect

on the uninhibited reaction (Figure 2.2.1.d). Consequently, I excluded BSA from future HTRF

reactions.

In attempting to find an incubation time that would fall in the linear portion of the reaction curve,

I observed that the reaction proceeded too quickly at 8.8 nM hPNKP, so the concentration of

hPNKP had to be reduced to generate a longer linear portion (Figure 2.2.1.e). However, the

reaction curve under the current conditions also appeared to be biphasic, which suggested that

the concentration of T4 Ligase may not be saturated, as a reporter enzyme should be (Figure

2.2.1.e). Consequently, I optimized both T4 Ligase and hPNKP concentrations once again.

Page 29: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

20

Figure 2.2.1 Further optimization of the HTRF assay was undertaken to find non-saturating conditions

with high statistical validity. (a) HTRF reactions were pre-incubated with different concentrations of

EDTA to determine the IC50 value. (b) Using the same technique, the IC50 value of sodium orthovanadate

(Na3VO4) was estimated. (c) A time course was carried out, with reactions stopped with 10 mM or 20

mM Na3VO4, or 50 mM EDTA, to observe which method of halting the reaction would be most effective.

(d) Different putative hPNKP inhibitors identified from an earlier screen were tested in reactions with and

without BSA to evaluate whether or not BSA could mask the inhibitory effects of compounds. (e) New

time courses performed with more early time-points indicated that there was a lag occurring in the first 15

minutes of the reaction. Error bars for (a) and (b) represent the SEM for at least two independent

experiments. Error bars for (d) and (e) represent the SEM from two technical replicates.

I re-optimized the T4 Ligase concentration by performing HTRF reactions with 40, 75, 150, 300,

400, 600, 800, or 1000 nM T4 Ligase (Figure 2.2.2.a). The experiment indicated that the

previous T4 Ligase concentration of 74 nM was far from saturating, as hypothesized. To

Page 30: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

21

guarantee that T4 Ligase was sufficiently saturating, I selected 800 nM T4 Ligase as the best

concentration for the remainder of the screen.

Next, I titrated hPNKP in reactions at 0.1, 0.3, 1, 3, and 10 nM (Figure 2.2.2.b). Although the

linearity of the curve within the first 15 minutes had improved, the excessive drop in activity

between 3 nM and 1 nM was curious and suggestive of the protein being unstable at

concentrations below 3 nM in PNKP dilution buffer (Table 2). This observation led me to repeat

the experiment with the addition of 0.02% Tween-20 to PNKP dilution buffer (Table 1), which

could increase the protein’s stability at lower concentrations and also reduce its potential to stick

to the polypropylene tube. Remarkably, the inclusion of Tween-20 in the dilution buffer led to an

increase in the initial velocities across all hPNKP concentrations and appreciable levels of

activity for reactions with less than 3 nM hPNKP. I subsequently repeated the experiment with

the liquid handling robotics platform to ensure that the best hPNKP concentration and incubation

time would be chosen for the screen (Figure 2.2.2.c). Based on these experiments, I determined

that 1 nM hPNKP and 20 minutes for the dephosphorylation-ligation reaction were suitable

parameters for the remainder of the screen.

Page 31: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

22

Figure 2.2.2 The T4 Ligase and hPNKP concentrations were optimized to prevent the ligation reaction

from causing a kinetic lag. (a) The T4 Ligase concentration was titrated to find a concentration at which

the ligation reaction would no longer be limiting. (b) hPNKP was diluted in PNKP dilution buffer (Table

1) and titrated into HTRF reactions with 800 nM T4 Ligase. (c) hPNKP was diluted in PNKP dilution

buffer with 0.02% Tween-20 and titrated into HTRF reactions with 800 nM T4 Ligase to increase the

stability of hPNKP at low concentrations. Error bars in (a) represent the SEM from two independent

experiments.

The change of hPNKP and T4 Ligase concentrations meant that the concentration of HTRFphos-

1 and HTRFphos-2 substrates (Table 2) should once again be adjusted. I also tested different

concentrations of the SAXL acceptor fluorophore to determine if signal could be increased

(Figure 2.2.3.a-b). I carried out the experiment with 1 nM (Figure 2.2.3.a) or 3 nM hPNKP

(Figure 2.2.3.b) and a 20 minute reaction time. The results suggested that 120 nM DNA

substrates and 84 nM SAXL fluorophore produced the best signal without using an excessive

amount of the expensive fluorophore at the hPNKP concentrations tested.

Finally, another approach was taken to improve upon HTRF signal: shortening the DNA

substrates. According to the FRET equation - percent energy transfer = 1 / [1 + (r/R0)6, where r is

the distance between acceptor and donor fluorophores, and R0 is the Förster distance, at which

Page 32: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

23

50% of the energy is transferred from donor to acceptor – with a Förster distance of 90 Å, only

28% of the energy is transferred from the Europium cryptate in DNPK to the XL-665

fluorophore in SAXL when the ligated DNA is 31 nucleotides (nt) – or about 105 Å - long.

However, by shortening the ligated DNA product to 25 nt (85 Å), the efficiency of energy

transfer more than doubles to 59%, which in theory would increase the signal over background. I

tested the newly optimized conditions side-by-side with the 31 nt and 25 nt substrates, and, as

hypothesized, the shorter substrates more than doubled the signal over background (Figure

2.2.3.c). I carried out a final time course at 1, 2, and 4 nM hPNKP using the newly optimized

conditions and shorter DNA substrates, and 2 nM hPNKP with a 20 minute incubation time was

found to provide sufficient signal without completely saturating the reaction (Figure 2.2.3.d).

Unfortunately, these conditions were not totally amenable to high-throughput screening because

once applied to a test run of ten plates, there was a substantial increase in variability, which

greatly reduced statistical validity such that the z’-factors were hovering around 0.5. In an effort

to reduce variability, 240 ml polypropylene reservoirs were utilized as the reagent source instead

of 2 ml 96-well plates. However, the use of reservoirs as source plates demanded a greater

consumption of reagents, which was excessive in the case of the SAXL fluorophore. To

minimize the costs associated with this increased consumption, I adjusted the concentration of

SAXL once more. I carried out reactions with 42, 60, or 84 nM SAXL, and 2 nM or no hPNKP

(Figure 2.2.3.e). I selected 60 nM SAXL as the best concentration going forward, not only

because it would consume less reagent, but also because it gave better signal-to-background

ratios (Figure 2.2.3.e). The combination of using reservoirs as source plates and reducing SAXL

from 84 nM to 60 nM greatly benefited the statistical validity of the screen, with z’ factors

increasing from ~ 0.5 to a more acceptable level of greater than 0.65.

Page 33: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

24

Figure 2.2.3 Optimization of DNA concentration, ligation product length, and acceptor fluorophore

concentration to maximize signal-to-background. (a, b) The concentration of DNA (31 nt product) was

titrated in HTRF reactions at 1 nM hPNKP (a) or 3 nM hPNKP (b). (c) HTRF assays were conducted

with DNA substrates that would create 31 or 25 nt ligation products to verify that the shorter product

would produce greater HTRF signal. (d) A time course was carried out at different hPNKP concentrations

using the HTRFphos-3 and HTRFphos-4 substrates, which generate 25 nt ligation products; inset shows a

close-up on the first 15 minutes of the reaction. (e) Lower concentrations of SAXL were tested under the

optimized conditions. Error bars in (c) and (e) represent the SEM from two technical replicates.

2.3 Homogeneous time-resolved FRET-based screen

To find putative inhibitors of hPNKP, the HTRF assay was used to probe the small molecule

library assembled by the Ontario Institute for Cancer Research (OICR; Toronto, Canada) with

ten, twenty, or thirty plates screened per day. I included positive and negative controls in each

plate so that each plate could be analyzed independently. In order to maintain statistical validity,

I repeated plates with z’-factors below 0.50. Compounds that caused a reduction in signal by

more than three standard deviations were designated as “primary hits”, and these were selected

for secondary screening. At the end of the “first half” of the screen (plates 1-250, Version 1 of

Page 34: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

25

the HTRF assay), 574 primary hits were uncovered from the 80,000 compounds that were

assayed, giving a hit rate of 0.72% (Figure 2.3.1.a). From the “second half” of the screen (plates

251-445, Version 2 of the HTRF assay), 357 hits were identified from the 62,400 compounds

tested, giving a hit rate of 0.57% (Figure 2.3.1.b).

Because the primary HTRF-based screen uses phosphatase-dependent ligation of DNA substrates

as the readout for hPNKP activity, the primary screen is inherently going to identify inhibitors of

both ligation and dephosphorylation. There is also the possibility that some apparent inhibitors

are results of non-specific effects, e.g. fluorescence quenching or DNA intercalation.

Consequently, I employed a secondary HTRF-based screen to eliminate the compounds that

reduced the HTRF signal for reasons other than hPNKP inhibition. To do so, the protocol for the

secondary screen was identical to that of the primary screen, except that hPNKP was eliminated

from the reactions and the DNA substrates contained ligatable 3’-hydroxyl groups (HTRFOH-1

and -2, Version 1; or HTRFOH-3 and -4, Version 2; Table 2). With this method, only

compounds that caused hPNKP-independent inhibition will inhibit reactions in both the primary

and the secondary screens; therefore putative hPNKP inhibitors were hits in the primary screen

but showed little to no inhibition in the secondary screen (Figure 2.3.1.c-d). Using this

approach, I found a total of nine putative inhibitors of hPNKP (Table 3), which equals an overall

hit rate of 0.0063%.

Page 35: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

26

Figure 2.3.1 Screening of the OICR HTS chemical collection under primary and secondary screen

conditions to identify putative hPNKP inhibitors. (a) 80,000 compounds were tested under saturating

(Version 1) screen conditions. The mean fold-change in activity is represented by the red line, and the

blue lines represent plus or minus three standard deviations away from the mean. (b) More than 62,000

compounds were tested under non-saturating (Version 2) screen conditions. (c) After testing the primary

hits in the secondary screen (Version 1) for inhibition of hPNKP-independent ligation, four hits were

identified as putative hPNKP inhibitors, circled in red. (d) After testing the primary hits in the secondary

screen (Version 2) for inhibition of hPNKP-independent ligation, five hits were identified as putative

hPNKP inhibitors, circled in red.

2.4 Putative inhibitors of hPNKP

Interestingly, with input from Dr. David Uehling of the OICR, we noticed some common

structural motifs among the putative hPNKP inhibitors. One feature was the presence of pyrrole,

pyrazine and/or thiophene rings, which were found in all nine hits. Another feature, which was

found in the first four hits of the screen (OICR1-4, Table 3), was the presence of a carboxylic

acid group at the end of an aliphatic chain attached to a five- or six-membered ring. The final

apparent feature was the presence of an amide group within the structures of the five compounds

Page 36: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

27

identified from the “second half” of the screen (OICR5-9, Table 3). Intriguingly, compounds

OICR7 and OICR9 have a common fragment, which contains two of the three aforementioned

features - a pyrrole ring, and a carboxylic acid or amide group attached to a benzene ring

(OICR79, Table 4). The importance of these structural motifs will be revisited later with

structure-activity relationship (SAR) analysis.

2.5 Validation and IC50 measurement of putative hPNKP inhibitors by HTRF

I acquired each putative inhibitor anew as a powder from commercial sources. I dissolved the

compounds in DMSO and assayed them at a range of concentrations in order to establish IC50

values, provided that the compounds still inhibited the reaction. Unfortunately, OICR4 was no

longer available, so it could not be subjected to any follow-up experiments. Furthermore,

compounds OICR1, OICR5, and OICR8 did not reproduce inhibition when tested from freshly

dissolved powders (Figure 2.5.1.a); we posit that the apparent inhibitory effects of these

compounds in the screen were due to breakdown products or impurities present in the library

source plates.

The remaining five compounds did exhibit inhibition of the HTRF reaction after being freshly

dissolved in DMSO, so I measured the IC50 values of these five compounds in order to determine

their relative potencies (Figure 2.5.1.b). I observed a wide range of potencies with these

compounds, with OICR2 having an IC50 value of ~ 0.5 µM, and OICR3 having an IC50 value of

~ 40 µM. Although OICR2 and OICR3 may appear superficially similar, their Tanimoto

coefficient – which describes the similarity of two molecules, and has a minimum of 0 and a

maximum of 1 (Tanimoto, 1957) – is 0.40, which indicates that they are chemically dissimilar,

and this explains the difference in their IC50 values.

Page 37: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

28

Figure 2.5.1 Several putative inhibitors were validated as inhibitors of hPNKP with a range of potencies,

while others could not be reconfirmed. (a) Compounds OICR1, OICR5, and OICR8 did not reconfirm

when tested from freshly dissolved powders. (b) The remaining five hits – OICR2, OICR3, OICR6,

OICR7 and OICR9 – did reconfirm with IC50 values from ~ 0.5 µM (strongest inhibitor, OICR2) to ~ 40

µM (weakest inhibitor, OICR3). Error bars represent the SEM from at least two independent experiments.

Page 38: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

29

2.6 Optimization of gel-based phosphatase assay

I developed a gel-based assay based on the methods of Freschauf et al. (2009) so that the

putative inhibitors could be validated by directly measuring phosphatase activity, thus abolishing

the need for T4 Ligase and HTRF fluorophores as reporters. Classically, the 3’-phosphorylation

status of DNA that has been 5’-labeled with 32

P would be followed by electrophoresis on a

polyacrylamide sequencing gel. The presence of the 3’-phosphate imparts a greater negative

charge to the phosphorylated DNA, which causes it to migrate faster on the gel (Figure 2.6.1.a).

However, thanks to the modern availability of fluorescent labels, these experiments were carried

out with Cy5-labeled DNA in lieu of radioactively-labeled DNA (GELphos, Table 2).

To maintain consistency and be able to directly compare the results from the gel-based

experiments with those from the HTRF-based experiments, I carried out all of the phosphatase

gel assays in HTRF buffer (Table 1) at 2 nM hPNKP and 120 nM GELphos DNA substrate

(Table 2). However, to ensure that the reactions were not saturated, I conducted a time course to

select an optimal reaction time. I quantified the percentage of de-phosphorylated DNA at each

time-point using ImageQuant 5.0 (Molecular Dynamics, USA) and plotted a reaction curve

(Figure 2.6.1.b). Based on this data, I selected 15 minutes as a suitable reaction time; although

this time point does not fall exactly in the linear portion of the reaction, it does appear that ~ 50%

of the DNA has been de-phosphorylated and the reaction is still progressing, albeit at a slower

rate.

Page 39: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

30

Figure 2.6.1 A gel-based assay was developed to directly measure the dephosphorylation activity of

PNKP. (a) The activity of PNKP on the 3’-phosphorylated DNA substrate leads to a band shift because

the loss of the negative charge imparted by the 3’-phosphate causes the DNA to migrate more slowly on

the gel. (b) A time course was performed with 2 nM hPNKP and 120 nM DNA to select a reaction time

that would allow for the dephosphorylation of about 50% of the DNA.

2.7 Measurement of IC50 values by gel-based phosphatase assay

Using the above reaction conditions, each putative inhibitor was assayed at a range of

concentrations in order to establish IC50 values from this direct assay. Remarkably, the IC50

values generated by the gel assay were significantly lower than those from HTRF experiments,

by a factor of about 10-20 (Figure 2.7.1). Since the hPNKP and DNA substrate concentrations

were maintained between the HTRF and gel experiments, I hypothesized that this disparity may

be due to nonspecific - albeit benign - binding of the compounds to the T4 Ligase. Such

nonspecific binding would titrate the compound away from hPNKP and raise the apparent IC50

value. Additionally, the DNA sequences of the HTRFphos and GELphos substrates (Table 2) are

not identical, so this may contribute to the differences in IC50 values. Alternatively, it is possible

that the gel assay is simply more sensitive than the HTRF assay.

Page 40: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

31

Figure 2.7.1 Dose response experiments were performed for each of the five original hits using the gel

assay, and IC50 values were measured. (a-e) The phosphatase activity of hPNKP was measured directly by

gel assay, which eliminated the use of a reporter enzyme and thus confirmed that the compounds are

impeding hPNKP from processing 3' phosphate DNA termini. Error bars represent the SEM from at least

two independent experiments.

2.8 Structure activity relationship analysis

The three structural motifs common to the putative hPNKP inhibitors identified by the screen –

amide groups, carboxylic acid groups, and five-membered heterocyclic rings - were of great

interest to us, so I utilized structure-activity relationship (SAR) analysis to assess their roles in

inhibiting hPNKP. Toward this end, I obtained a number of analog compounds in which these

features were individually modified or moved relative to the original structure (Table 4).

Furthermore, I acquired a number of analogs of OICR79 in order to investigate whether this

minimal fragment could be made more potent (Table 4).

Page 41: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

32

I employed HTRF experiments as well as subsequent gel-based experiments to assess how the

modifications affected the compounds’ potency. Based on analogs whose pyrrole rings were

removed (OICR9A) or converted to –N(CH3)2 (OICR9B), I determined that the pyrrole ring was

necessary for PNKP inhibition by OICR9 derivatives (Figure 2.8.1.a). The lack of inhibition by

analog OICR7A further reinforced the importance of the pyrrole ring, since changing its position

relative to the rest of the structure also abolished inhibitory activity (Figure 2.8.1.b).

OICR79 - the common fragment from OICR7 and OICR9 - was also tested by HTRF (Figure

2.8.1.c). We surmised that this structure could be responsible for the inhibition of hPNKP by

OICR7 and OICR9, and if this was the case, OICR79 should prove to be at least as inhibitory as

the original hits. The IC50 value of OICR79 was found to be on the same scale as that of OICR7

(compare Figure 2.8.1.c and Figure 2.5.1.b), which suggests that this portion of OICR7 may be

entirely responsible for hPNKP inhibition. However, it should be noted that the IC50 values from

the gel assay are not as consistent, with OICR79 appearing to be ~ 7 times less potent than the

parent compounds. Owing to this discrepancy, the importance of the rest of the OICR7 and

OICR9 structures should not be discounted entirely and their roles should be further studied by

testing new analogs in which they are modified.

Since the common fragment portion of OICR9 contains an amide group on the benzene ring

rather than a carboxylic acid group, we would have liked to test an amide version of this

fragment as well, but such a compound is not currently available.

Page 42: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

33

Figure 2.8.1 Structure activity relationship analyses of the heterocyclic ring and the fragment common to

OICR7 and OICR9. (a) Analogs of OICR9 may appear to inhibit hPNKP at concentrations > 100 µM, but

closer inspection of the fluorescence emitted in the acceptor and donor emission channels (below; “SAXL

em.” and “DNPK em.” respectively) suggest that this is due to fluorescence quenching by high

concentrations of compound. (b) HTRF assays demonstrated that shifting the position of the heterocyclic

ring in OICR7 also abolished activity. (c) OICR79, the fragment common to OICR7 and OICR9,

displayed inhibition of hPNKP of a magnitude similar to the parent compounds (left). This inhibition was

confirmed by the phosphatase gel assay (right), although the IC50 value is about seven times greater than

those of the parent molecules. Error bars represent the SEM from at least two independent experiments.

Functional groups in red represent groups that differ between the parent and analog compound.

Page 43: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

34

To learn more about the OICR79 fragment, I examined several analogs (Table 4) for their ability

to inhibit hPNKP. The addition of a methyl group at C6 of the benzene ring in OICR791 and

OICR793 (Table 4) caused a reduction in potency to the point that they only produced

observable inhibition at 500 µM in HTRF reactions (Figure 2.8.2.a-b); perhaps the presence of a

methyl group near the pyrrole ring imposes steric hindrance, which impedes the pyrrole ring

from binding effectively to hPNKP.

Another analog that virtually lost its activity was OICR792, which has a C=C(NH2)2 group

attached to C2 of the pyrrole ring (Figure 2.8.2.c; Table 4). The consistent loss of potency

brought on by the addition of functional groups on or near the pyrrole ring of the parent

compound further strengthens the argument for its importance. Interestingly, analog OICR795,

which had the pyrrole and carboxylic acid groups in ortho rather than meta positions on the

benzene ring (Table 4), was less potent but not completely inactive with an IC50 of ~ 200 µM

(Figure 2.8.2.d). This inhibition, although considerably weaker than that of the parent

compound, was curious because it appeared to be greater than that of OICR791 and OICR793,

which have methyl groups added to the same position. I hypothesized that this discrepancy arose

because the carboxyl group bound better to hPNKP in an ortho rather than meta position, and the

inclusion of an ortho methyl group with the meta carboxyl group threw a proverbial wrench in

the works, making binding very difficult for the carboxyl group. The validation of this

hypothesis would be aided by a structure of hPNKP complexed with inhibitor.

Page 44: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

35

Figure 2.8.2 Structure activity relationship analyses using analogs of OICR79. (a, b) HTRF dose

response assays demonstrated that the addition of a methyl group ortho to the pyrrole ring was

detrimental to inhibition, with the first ortho position (a, OICR791) impeding inhibition the most. (c)

HTRF dose response assays revealed that adding a –C=C(NH2)2 group to C2 of the pyrrole ring hindered

inhibition, too. (d) Shifting the two functional groups to ortho instead of meta positions also reduced

inhibition almost 100-fold by HTRF. Error bars represent the SEM from at least two independent

experiments.

One analog that displayed a reduced but still appreciable level of inhibition with an IC50 value of

~ 30 µM was OICR796 (Figure 2.8.3.a; Table 4); not only did this compound have a –CHOCH3

group rather than a -COOH group on C3 of the benzene ring, but it also had a –C=NCH3 group

attached to C2 of the pyrrole ring. We chose to test this analog because it had been identified as a

hit in a primary screen for inhibitors of the protein phosphatase Scp1, which is a member of the

Page 45: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

36

HAD superfamily of phosphatases like hPNKP (Kamenski et al., 2004). However, because there

are two modifications in this compound relative to OICR79, it cannot be determined what role

each of them plays in the reduced inhibition.

Finally, in contrast to the aforementioned analogs that eliminated or reduced potency, OICR794

led to a significant increase in activity via the addition of another carboxyl group to the second

meta position (Figure 2.8.3.b). I posit that the symmetrical carboxyl group enhances the binding

of the compound to hPNKP, perhaps making additional contacts with the protein, and this

reinforces the importance of the carboxyl group, which is common among the primary hits.

Page 46: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

37

Figure 2.8.3 Structure activity relationship analysis of OICR79 uncovered two modifications that did not

abolish inhibition. (a) With a C=NCH3 group added to C2 of the pyrrole ring and CHOCH3 replacing the

COOH group, OICR796 exhibited relatively weak activity by HTRF (left) but appreciable activity

according to the gel assay (right). (b) With an additional COOH group on the second meta position of the

benzene ring, OICR794 demonstrated about two times stronger inhibition than OICR79 by both HTRF

and gel assays. Error bars represent the SEM from at least two independent experiments.

2.9 Optimization of fluorescence polarization-based substrate-binding assay

Fluorescence polarization (FP) is a widely used technique for measuring the binding of a

fluorescently-labeled ligand to a protein because FP is proportional to the tumbling properties of

the fluorophore (Pagano et al., 2011). When a small fluorescently labeled ligand is free in

Page 47: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

38

solution, its fluorophore tumbles quickly and therefore does not have a stable orientation; a

consequence of this rapid movement is that the fluorophore has poor capability for polarizing

fluorescent light upon excitation (Pagano et al., 2011). Conversely, when the ligand is bound to a

protein, it becomes stabilized in space because the much larger protein tumbles very slowly, and

this causes a significant increase in FP upon excitation of the fluorophore (Pagano et al., 2011).

Based on the methods of Coquelle et al. (2011), I utilized this technique to measure the binding

of a FAM-labeled 3’-phosphorylated penta-nucleotide substrate to hPNKP. For this assay, I

employed the phosphatase-dead D171A mutant of hPNKP so that it would bind the substrate but

not act on it.

To determine the optimal concentration of the FPphos substrate (Table 2) for this assay, I

measured the fluorescence intensity of substrate in FP buffer (Table 1) with a Beacon 2000

variable temperature fluorescence polarization system (Invitrogen, USA) at 510 nm. I selected a

concentration of 12.5 nM as a suitable working concentration for the FP assay as it gave a

fluorescence intensity of ~1000, which was low enough to prevent saturation of the detectors

(data not shown). I then titrated hPNKP-D171A in FP reactions, and analysis of this data

determined that the Kd of hPNKP under these conditions was 41.6 nM (see Methods, Figure

4.15.1). Based on these results, I chose 50 nM hPNKP-D171A as the concentration of protein to

use in the assay, since it bound ~50% of the substrate at equilibrium.

2.10 Substrate-binding assay confirms that compounds prevent a 3'-phosphorylated oligonucleotide from binding to hPNKP

Using the conditions outlined above, I measured an IC50 value for each of the original hits as

well as OICR79, OICR794 and OICR796 - the three analogs that exhibited measurable levels of

inhibition by HTRF. Remarkably, all of the compounds tested were capable of preventing the

Page 48: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

39

substrate from binding to hPNKP-D171A, with IC50 values either similar to or stronger than

those measured by HTRF (Figure 2.10.1a-h).

Figure 2.10.1 Fluorescence polarization-based direct binding assays were carried out to determine if the

compounds could compete with a 3’-phosphorylated oligonucleotide substrate for binding to hPNKP-

D171A. (a-h) A range of compound concentrations were incubated with hPNKP-D171A and substrate

was added to the reaction and allowed to come to equilibrium prior to measurement of FP. The typical

sigmoidal dose-response curves for each compound indicated that these compounds were displacing DNA

from the phosphatase active site, although the mechanism by which this is achieved cannot be concluded.

Error bars represent the SEM from at least two independent experiments.

2.11 Inhibitors of hPNKP display different degrees of reversibility

The extent of reversibility of an inhibitor can provide insight into the mechanism by which the

inhibitor works; for example, irreversible inhibitors generally act by covalently modifying the

target enzyme so that it is permanently incapable of acting on its substrate (Copeland, 2005).

Page 49: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

40

Conversely, inhibitors can be slowly or rapidly reversible; in such cases, the compound interacts

with the enzyme through non-covalent bonds such as hydrophobic interactions and ionic bonds

(Copeland, 2005).

To assess the reversibility of the hPNKP inhibitors, I performed an assay in which hPNKP was

incubated at 100-fold over the reaction concentration (i.e. at 200 nM) with ten times the IC50 of a

given compound (e.g. 76 µM OICR7) – which is enough to inhibit the enzyme by 90% - and

then diluted in reaction buffer 100-fold – leaving a concentration of compound at which hPNKP

would be inhibited by 9% (Copeland, 2005). I followed the reactions with a time course and

established the compound’s reversibility based on how quickly the enzyme recovered its activity

upon dilution (Figure 2.11.1). From the reaction curves, I determined that compounds OICR2

and OICR3 are slowly reversible, while the other compounds are very slowly reversible. In fact,

OICR6, OICR7 and OICR79 appear to hinder the recovery of hPNKP activity so much so that

they could be considered virtually irreversible.

Page 50: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

41

Figure 2.11.1 Reversibility assays demonstrated that most of the compounds were at least slightly

reversible. (a) OICR2 and OICR3 were slowly reversible because the recovery of hPNKP activity was

slow and gradual over the time course. (b) OICR9 was somewhat reversible because a small amount of

hPNKP activity was recovered after 22 min. OICR6, OICR7 were virtually irreversible. (c) Analogs

OICR794 and OICR796 were also very slowly reversible, with hPNKP recovering to similar levels as

with OICR9. Analog OICR79 was virtually irreversible, with an effect similar to that of OICR7.

2.12 Inhibitors of hPNKP can act on T4 PNK with reduced potency

Although mammalian and T4 PNK(P) are each capable of performing 3’-phosphatase and 5’-

kinase activities, they exhibit a number of key differences; most notably, the presence of an

FHA domain in mammalian PNKP but not T4 PNK (Galburt et al., 2002). It is this FHA domain

that allows mammalian PNKP to be intimately involved in various forms of DNA repair because

the domain binds to phosphorylated XRCC1 and XRCC4, two integral components of the BER

and NHEJ pathways, respectively (Koch et al., 2004; Whitehouse et al., 2001). Additionally, T4

PNK behaves as a tetramer in vivo, whereas mammalian PNKP acts as a monomer (Galburt et

al., 2002). Furthermore, T4 PNK accepts RNA, DNA, and oligonucleotides as substrates, with a

preference for extended 5’ ends (Galburt et al., 2002); mammalian PNK, on the other hand,

Page 51: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

42

accepts only DNA that is at least eight nucleotides long, with a preference for recessed DNA

ends (Bernstein et al., 2005).

In the interest of determining how specific the inhibitors were towards hPNKP, if at all, IC50

measurements were performed using T4 PNK in the gel-based assay (Figure 2.12.1).

Remarkably, after factoring in the two-fold increase in T4 PNK concentration over that of

hPNKP in the same assay, the IC50 values for T4 PNK are up to 50-fold higher than those for

hPNKP, thus revealing a substantial degree of specificity for the human protein over its T4

ortholog. It should be noted that the comparisons had to be made between gel-based rather than

HTRF-based experiments because the IC50 values were quite high, and the high concentrations of

compound had a tendency to interfere with the HTRF measurements.

Overall, despite the considerable reduction in potency relative to hPNKP, most of the

compounds were capable of effectively inhibiting T4 PNK, with only three compounds

displaying poor inhibition.

Page 52: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

43

Figure 2.12.1 Gel-based assays conducted with T4 PNK demonstrate that some, but not all, of the

hPNKP inhibitors are capable of inhibiting this distant ortholog. (a-h) Gel-based assays were carried out

with 5 nM T4 PNK and 120 nM 3’-phosphorylated GELphos DNA (Table 2); most of the compounds -

OICR2 (a), OICR9 (e), OICR79 (f), OICR794 (g) and OICR796 (h) - exhibit sigmoidal dose-response

curves, indicating that they are functional inhibitors of T4 PNK. Compounds OICR3 (b), OICR6 (c), and

OICR7 (d) appear to inhibit T4 PNK at very high concentrations but their IC50values could only be

approximated because they inhibit so weakly. Error bars represent the SEM from at least two independent

experiments.

Page 53: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

44

2.13 Inhibitors of hPNKP do not show a clear effect in U2OS cells co-treated with camptothecin

It has been demonstrated that knockdown or inhibition of hPNKP can potentiate the effects of

certain DNA damaging agents on cells; in particular, the requirement of PNKP for the repair of

damage induced by Topoisomerase I stalling causes hypersensitivity to CPT in hPNKP-impaired

cells (Freschauf et al., 2010; Rasouli-Nia et al., 2004). In order to explore whether or not the

compounds under investigation could induce the same sensitivity, I assessed cell viability in

U2OS cells treated with DMSO or 0.1 – 100 µM compound of interest, and co-treated with

DMSO or CPT (Figure 2.13.1.a-g). I used 5 nM CPT to sensitize the cells because it was found

to kill about 50% of the cells after a 96 h treatment at the seeding density used.

Page 54: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

45

Figure 2.13.1 The viability of U2OS cells co-treated with 5 nM camptothecin (CPT) was not significantly

impaired relative to their DMSO-treated counterparts. (a) The indicated number of cells were treated with

a series of CPT concentrations for 96 h prior to the quantification of cell viability with the ATP Lite assay

(Perkin Elmer, USA) (b-i) Cells were treated for 96 h with a range of concentrations of each hPNKP

inhibitor plus DMSO or 5 nM CPT. Error bars represent the SEM from at least two independent

experiments.

Unfortunately, it does not appear that any of the compounds were capable of significantly

impairing the growth of CPT-treated U2OS cells relative to the control DMSO-treated cells

under these conditions. Some compounds did slightly impair cell survival at high doses, such as

OICR3 and OICR794 (Figure 2.13.1.b, f), but these effects were not vastly different between

DMSO and CPT co-treated cells.

There are a number of potential explanations for why the compounds did not sensitize the cells to

CPT as desired – for example, they cannot cross the cell membrane, they break down or become

Page 55: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

46

modified in the cell, they preferentially target other enzymes in the cell – but to elucidate why

the compounds did not act as potentiators of camptothecin will require additional experiments

examining each hypothesis. Furthermore, and perhaps most importantly, the assay may not have

been designed optimally for the detection of PNKP inhibition-dependent CPT sensitization, as it

has been shown that cells in S-phase may not require the activity of PNKP to recover from CPT-

mediated damage (Rasouli-Nia et al., 2004). realize

Another limitation of the experimental set up that we later recognized was that the ATP Lite

assay I used to measure cell viability cannot differentiate between live cells and senescent cells.

By overlooking DNA damage-induced senescence (Di Leonardo et al., 1994), it is possible that

an impact of the inhibitors on viability was missed.

Page 56: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

51

3 Discussion and future directions

The search for inhibitors of phosphatases has been challenging due to the nature of the enzymes’

structure. A common issue with inhibitors is a lack of selectivity, which is a consequence of

phosphatases’ highly conserved active sites (Barr, 2010). The second major concern regarding

phosphatase inhibitors is their low level of cell permeability, which is also a consequence of the

enzymes’ active sites: the highly polar active sites favor inhibitors with charged anionic

phosphate mimics that do not readily cross the cell membrane (Barr, 2010). However, in spite of

these common difficulties, the frequent finding that one or more phosphatases are involved in a

given disease has fueled countless searches for inhibitors of this class of enzymes. In the case of

PNKP, our search for an inhibitory compound was driven by the discovery that knockdown of

PNKP sensitizes cells to PARP inhibitors (Turner et al., 2008), Topoisomerase I inhibitors

(Rasouli-Nia et al., 2004), and ionizing radiation (Rasouli-Nia et al., 2004), which has

implications for the treatment of many types of cancer. This work was also encouraged by the

identification of selective inhibitors towards proteins in the same haloacid dehalogenase (HAD)

family of phosphatases as PNKP, such as Scp1 (Zhang et al., 2011).

The work described here has identified five unique compounds and several analog compounds

that based on the direct gel-based phosphatase assay (Figure 1.2.7.1), exhibit a range of

potencies, some of which are in the low nanomolar range. It is worth noting that the discovery of

only five inhibitors from a screen of more than 140,000 compounds amounts to a hit rate of

0.0035%, which is considered relatively low, as other high-throughput screens typically produce

hit rates that are greater by one to two orders of magnitude (Copeland, 2005). This may be due to

an unknown structural bias in the library, or it is possible that the structure of hPNKP supports

Page 57: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

52

only a limited range of moieties. The likelihood of the former could be assessed by screening

additional chemical libraries to see what sorts of inhibitors they contain, if any.

Despite the low hit rate, the inhibitors identified by this large screen are very informative as they

belie a penchant for hPNKP to bind to heterocyclic rings, amides and carboxylic acid (COOH)

groups – at least two of which were present in each of the compounds and analogs tested (Tables

3, 4). Furthermore, the examination of analogs of OICR9 that lacked the heterocyclic ring or

retained the nitrogen heteroatom but not the full ring structure - OICR9A and OICR9B,

respectively - demonstrated the importance of this motif in effecting inhibition. Conversely, the

importance of the carboxylic acid moiety could be contested, as OICR796 contains a CHOCH3

group in its stead and still displays a moderate level of potency. However, because there are also

modifications made to the heterocyclic ring in OICR796, it is not possible to say for certain what

effect the change from –COOH to –CHOCH3 had on the potency of the compound; to do so, it

would be necessary to test an analog where only a single change was made. Bearing that in mind

though, it raises the possibility that the presence of a moiety with trigonal planar geometry is

more important than a COOH group specifically; to determine if this is the case, there are many

more motifs that could be explored, with the potential for some to increase potency and/or

selectivity. Moreover, the presence of an uncharged group in lieu of the COOH group would be

beneficial because it could lead to an increase in the compound’s membrane permeability.

With respect to the compounds' mechanisms of action, the FP assay confirmed that they do

prevent the DNA from binding to hPNKP, but whether that occurs via competition for the active

site, binding to an allosteric site, or simply denaturing the protein could not be ascertained from

this approach. However, the latter possibility was largely eliminated as a consequence of the

reversibility assays, which demonstrated that the compounds are generally reversible, with some

Page 58: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

53

dissociating and relieving hPNKP inhibition more quickly than others. Other biochemical

experiments – for example, using circular dichroism (CD) spectroscopy to monitor changes in

PNKP secondary structure in the presence of substrate and/or inhibitor (Chen et al., 1974) could

provide additional evidence towards a mechanism of action. For example, if the addition of

inhibitor led to an increase in random structure, it would be suggestive of a noncompetitive

mechanism, whereby the binding of compound outside the active site causes a change in enzyme

structure that allosterically inhibits the enzyme.

Further insight into how the compounds work can be gleaned from the dose response

experiments with T4 PNK (Figure 2.12.1). Interestingly, across all dose response experiments

with hPNKP, regardless of the value of the IC50, the trend in relative potencies was maintained;

for example, OICR2 was always the most potent and OICR3, the least. However, this trend was

not completely upheld with T4 PNK; the most apparent difference being that, while OICR9 is

still relatively potent with an IC50 value of 5.74 µM, OICR7 with the common fragment does not

appear to inhibit T4 PNK. This contrast is curious since the compounds each contain nearly

identical active portions (based on the activity seen with OICR79), but this discrepancy is likely

due to structural differences in the vicinity of the common fragment's binding site on each

protein - that is, the binding site on T4 PNK may accommodate OICR9 but not OICR7, thus

preventing the latter from effecting inhibition of this ortholog. In fact, the finding that OICR79

can inhibit T4 PNK with an approximate ten-fold reduction in potency relative to OICR9

strongly implies that the dissimilar portions of OICR7 and OICR9 play an important role in

dictating the compounds' interactions with T4 PNK. These differences in activity towards

hPNKP and T4 PNK have implications for the inhibitors’ mechanisms of action because they

point to a structural difference between the two enzymes as being a component of the compound

Page 59: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

54

binding site. This hypothesis is bolstered by preliminary data in which the compounds exhibit a

similar, albeit slightly reduced degree of potency (~ 1.5-2 fold) towards murine PNKP, which is

considerably more similar in sequence and structure to hPNKP than is T4 PNK. A

straightforward method for deciphering the compounds’ mode of action is to crystallize them

with PNKP, preferably with the human homolog for which they are most effectual.

The work presented here collectively demonstrates the discovery of eight compounds capable of

inhibiting hPNKP in vitro with at least moderate potency (i.e. micromolar IC50 values);

unfortunately however, none of the compounds showed an ability to sensitize U2OS cells to

camptothecin in tissue culture. Generally, uncharged polar compounds have the highest degree of

cell permeability since they are best equipped to cross the amphipathic lipid bilayer that

constitutes a cell’s membrane. Because carboxylic acid groups have a net negative charge, their

presence can make compounds less permeable; therefore, it is possible that the hPNKP inhibitors

are not entering the cell efficiently and this would prevent them from being present at high

enough concentrations to have a significant effect on hPNKP. This hypothesis could be tested by

assaying analogs that contain a methoxy group in place of the carboxyl group, provided that such

modifications do not eliminate the compounds’ activity in vitro. There are also well-established

cell-based assays that could be used to investigate the compounds’ membrane permeability, such

as the Caco-2 cell permeability assay, which could also be used to assess the metabolism of the

compounds in these cells (van Breemen and Li, 2005).

Beyond the question of the compounds' membrane permeability, it is not certain that the viability

experiments were designed appropriately to assess the role of PNKP inhibition in surviving CPT-

induced DNA damage: using clonogenic assays to measure the viability of A549 lung cancer

cells, Rasouli-Nia et al. (2004) have demonstrated that the cells exhibit a biphasic response to

Page 60: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

55

CPT, with PNKP knockdown only sensitizing cells at doses greater than 1 µM. They postulated

that the drop in viability at lower doses was a consequence of the increased sensitivity of S phase

cells to CPT and that the inability of PNKP knockdown to sensitize these cells indicated that

PNKP was not critical for recovery from CPT during S phase. These findings have important

implications for my assessment of PNKP inhibitors in cells because they suggest that the choice

of CPT treatment conditions determines whether or not PNKP is needed for recovery and thus if

sensitization can take place. The conditions I employed for the cell viability assays - 5 nM CPT

for 72 h with no drug-free recovery time - are very different from those at which Rasouli-Nia et

al. (2004) demonstrated sensitization by PNKP knockdown - ≥ 1 μM CPT for 2 h and 2-3 weeks

recovery; therefore, I hypothesize that treating the cells for several days with a low dose of CPT

likely facilitated cell death in S-phase and caused PNKP activity to be inconsequential. This

hypothesis is bolstered by recently published work from Ray Chaudhuri et al. (2012), which

revealed that low doses of CPT are not associated with DSB formation, but rather with PARP-

dependent replication fork stalling and reversal. Overall, this suggests that the inability of the

PNKP inhibitors to sensitize cells to CPT may simply be a consequence of experimental design.

In order to accurately determine whether or not the compounds are effective in cells, it will be

key to repeat these experiments under conditions that trigger CPT-induced death in non-S phase

cells – that is, at higher doses of CPT with a shorter treatment time and increased recovery time.

Furthermore, the effects of PNKP knockdown on CPT sensitization published by Rasouli-Nia et

al. were shown by clonogenic assays, which quantify viability by measuring colony formation

following drug treatments. As a consequence, clonogenic assays account for the effects of both

apoptosis and senescence on viability, unlike the ATP Lite assay, which measures only

apoptosis. Because it is quite possible that PNKP inhibition decreases viability by promoting

senescence, it would be preferable to re-assess CPT sensitization by clonogenic assay.

Page 61: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

56

Additionally, it would be worthwhile to repeat the experiments using the A549 lung carcinoma

cell line, in which Rasouli-Nia et al. demonstrated the CPT-sensitizing effects of hPNKP

knockdown, as well as using ionizing radiation as the source of DNA damage.

In the event that the compounds continue to fail to produce CPT sensitization, it will be

necessary to further investigate their membrane permeability as well as whether or not they are

affecting their intended target, using the alkaline comet assay, for example, which measures the

accumulation of SSBs in cells (Pfuhler and Wolf, 1996). If the compounds are in fact inhibiting

hPNKP, and thus hindering the repair of SSBs, DNA breaks might accumulate in mono-treated

cells and should persist even longer in CPT-sensitized cells.

Furthermore, the selectivity of the inhibitor-hPNKP interaction, and therefore the likelihood of

off-target effects occurring in cells, could be assessed by testing the ability of the compounds to

inhibit other phosphatases within the aspartate-based HAD family of protein tyrosine

phosphatases (PTPs), as well as phosphatases that operate via other mechanisms; for example, a

cysteine-based PTP or a protein serine/threonine phosphatase (PSP). It would also be prudent to

examine whether or not the compounds have an effect on other enzymes that perform similar

processing functions or have similar domains, such as Aprataxin (APTX) and APTX- and

PNKP-like factor (APLF).

Overall, the work that I have outlined herein has identified several compounds that inhibit

hPNKP, and these effects appear to be due to more than simple denaturation based on dose-

response, reversibility, and species-specificity experiments. Although further studies would be

required to determine the actual mechanism by which inhibition is achieved and the degree of

selectivity these compounds show for PNKP, the search for effective hPNKP inhibitors has

certainly moved forward. Alas, these inhibitors failed to demonstrate an ability to kill CPT-

Page 62: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

57

sensitized cells, which is not particularly useful for the development of potential cancer

therapies, but there are many tools in medicinal chemistry that can be utilized to improve cell

permeability or reduce the potential for off-target effects. A collaboration that we have

undertaken with Dr. Mark Glover’s group at the University of Alberta may help answer many of

these questions, as they attempt to obtain and solve the structure of crystals of our compounds

bound to murine, and hopefully human, PNKP.

Page 63: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

57

4 Materials and Methods

4.1 Transformation of E. coli.

XL1-Blue E. coli cells (Stratagene, USA) were made ultra-competent according to the Inoue

method (Inoue et al., 1990). BL21 (DE3) CodonPlus E. coli cells were made chemically

competent as described by Sambrook and Russell (2001). Cells were incubated in a microfuge

tube with plasmid DNA for 15 min on ice, heat-shocked for 30 sec at 42 °C, then moved

immediately back to ice where they were incubated for 10 min. One ml of Luria Broth (LB)

media (Table 1) was added to each tube and cells were grown for 1 h at 37 °C with shaking.

Cells were pelleted by spinning for 20 sec at maximum speed in a 5415 D table-top centrifuge

(Eppendorf, Germany). The supernatant was discarded, and the cells were resuspended in 100 µl

of LB media (Table 1). For XL1-Blue cells, 10% of the transformation mix was plated on one

LB plate containing 0.1 mg/ml ampicillin (LB + AMP; Table 1), and the remaining 90% was

plated on a second LB + AMP plate. For BL21 cells, 100% of the transformation mix was plated

on a single LB + AMP plate. Plates were incubated upside down in a 37 °C incubator overnight.

4.2 SDS-PAGE.

6X SDS-PAGE sample buffer (Table 1) was added to samples to a final concentration of 1X.

Samples were boiled for 5 min and then run on 8% or 10% SDS-PAGE gels (Table 1) for 45 min

at 200 V. Proteins in the gel were visualized by staining with Coommassie Brilliant Blue solution

(Table 1) for 5 min and then de-stained overnight in de-stain solution (Table 1).

4.3 Quantitation of protein

Protein samples were diluted 10-fold in 6 M guanidinium-HCl. A Nanodrop 8000

spectrophotometer was blanked with 6 M guanidinium-HCl and the concentration of protein was

determined by measuring absorbance at 280 nm.

Page 64: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

58

4.4 Cloning of human PNKP-D171A

The Quickchange PCR method was used to generate the phosphatase-dead hPNKP D171A

construct from the pProExHTA-hPNKP plasmid (M. Canny, A. Koch). Seventy-five ng of

plasmid DNA was amplified with 250 ng each of forward (5’-

GGTGAAACCCCAGGGCAAGGTGGCTGGCTTTGCGCTGGACGGGACGCTCATCACCA

CACG-3’) and reverse (5’-

CGTGTGGTGATGAGCGTCCCGTCCAGCGCAAAGCCAGCCACCTTGCCCTGGGGTTTC

ACC-3’) primers. The PCR reaction also contained 100 μM of each dNTP, 2.5 units of Pfu DNA

polymerase (Agilent Technologies, USA), and 1X Pfu buffer [20 mM Tris-HCl, pH 8.8, 2 mM

MgSO4, 10 mM (NH4)2SO4, 10 mM KCl, 0.1% (v/v) Triton X-100, 0.1 mg/ml BSA] for 19

cycles [95 °C for 30 sec, 55 °C for 1 min, 68 °C for 22 min] and a final extension step of 68 °C

for 30 min. The PCR product was incubated with 20 units of DpnI restriction endonuclease (New

England BioLabs, USA) at 37 °C for 3 h. Six individual colonies from freshly transformed XL1-

blue E. coli were picked and grown overnight at 37 °C in 2-ml LB + AMP cultures (Table 1).

The DNA was purified from these cultures using Miniprep Spin Columns (Qiagen, Inc.)

according to the manufacturer’s instructions. The Miniprep columns gave yields of 115-200 ng

plasmid DNA/µl, and plasmids were sequenced by ACGT DNA Technologies Corporation

(Toronto, Ontario) to verify that the mutation was in place and the reading frame was intact.

4.5 Purification of human PNKP and PNKP-D171A

Competent BL21 (DE3) CodonPlus E. coli were transformed with 200 ng pProExHTA-hPNKP

plasmid (M. Canny, A. Koch) or pProExHTA-hPNKP-D171A, plated on LB + AMP (Table 1),

and grown overnight at 37 °C. Freshly transformed E.coli BL21 Codon Plus cells were scraped

into 10 ml of LB media (Table 1) and used to inoculate 2 × 1L of LB + AMP media (Table 1).

When the cultures reached an OD600 of 0.8, hPNKP expression was induced at 16 °C by adding

Page 65: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

59

isopropyl β-D-1-thiogalactopyranoside (IPTG; Sigma, USA) to a final concentration of 100 μM.

Cultures were grown overnight at 16 °C and cells were harvested by spinning in a cold JLA8.1

rotor (Beckman Coulter, USA) for 10 min at 6000 × g. Cells were resuspended in 5 ml of cold

PNKP lysis buffer (Table 1) per gram of pellet. The resuspended cells were frozen at -20 °C for

overnight storage. To continue with the protein purification, cells were thawed in warm water

and lysed with a VibraCell sonicator (Sonics, USA) for seven cycles of 30 sec of bursts at 85%

power/30 sec on ice. Lysates were incubated with chicken egg lysozyme (Sigma, USA) at 0.2

mg/ml for 30 min at 4 °C while nutating and then spun at 18,000 rpm for 45 min in a JA25.5

rotor (Beckman Coulter, USA). The supernatants were filtered through a 60 ml syringe fitted

with a 0.8 µm filter (Thermo Scientific, USA) and then through a second 60 ml syringe fitted

with a 0.45 µm filter (Thermo Scientific, USA) into two 50 ml conical tubes. Fifteen-hundred µl

of agarose-Ni2+

-NTA bead slurry (Qiagen, Inc.) was added to a 15 ml conical tube and washed in

5 ml of lysis buffer (Table 1) by spinning at 500 × g for 2 min per wash for a total of three

washes. The washed beads were split evenly between the two 50 ml conical tubes of supernatant

and the tubes were nutated for 1.5 h at 4°C. During this time, a polypropylene 20 ml column

(Bio-Rad Laboratories, Inc.) was assembled at 4 °C and rinsed once with water and then once

with 70% ethanol. When the incubation was complete, 20 ml of bead-supernatant slurry was

transferred to the cold column and incubated for 5 min; the flow-through was then eluted into a

flask and these steps were repeated until all of the bead-supernatant slurry was loaded onto the

column. The column was then washed with 2 × 20 ml of cold PNKP lysis buffer (Table 1),

catching each flow-through in a new flask. Samples of uninduced and induced cells, supernatant

and pellet fractions, column flow-through and washes, and bead slurry from the column were

analyzed by SDS-PAGE in order to confirm the presence of hPNKP (Figure 4.6.1.a). Once the

presence of hPNKP was confirmed, 1 ml of cold PNKP lysis buffer (Table 1) and 100 μl of TEV

Page 66: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

60

protease (5 mg/ml; F. Sicheri) were added to the column. The column was tapped to mix and

incubated overnight at 4 °C to release hPNKP from the beads. Following the incubation, the

column contents were eluted into a 1.5 ml microfuge tube. Seven additional eluate fractions were

obtained by adding 1.5 ml cold PNKP lysis buffer (Table 1) to the column and eluting into

separate 1.5 ml microfuge tubes. Samples of beads before and after TEV incubation, and all

eluate fractions were analyzed by SDS-PAGE on a 10% SDS-PAGE gel (Table 1) in order to

confirm that hPNKP had been eluted (Figure 4.6.1.b). The eluate fractions containing hPNKP

(as determined from the gel) were combined and filtered through a 10 ml syringe fitted with a

0.22 µm filter (Millipore, USA) into a 15 ml conical tube. The concentration of protein in this

eluate was determined as described above and then transferred to 12-14 kDa MW cut-off dialysis

tubing (Spectrum Labs, USA). The protein was dialyzed overnight in PNKP dialysis buffer

(Table 1) at 4 °C. Following dialysis, the protein solution was filtered through a 10 ml syringe

fitted with a 0.22 µm filter (Millipore, USA). The concentration of hPNKP was determined as

described above and then divided into 100 μl aliquots in 1.5 ml microfuge tubes for long-term

storage at -20 °C.

4.6 Purification of T4 DNA Ligase.

Competent BL21 (DE3) CodonPlus cells were transformed with 120 ng of pT4 plasmid DNA

(courtesy of F. Sicheri), plated on LB + AMP (Table 1), and grown overnight at 37 °C. Freshly

transformed E. coli were scraped into 10 ml of LB media (Table 1) and transferred to a 15 ml

conical tube. Each of six beveled flasks containing 1 L LB + AMP (Table 1) media were

inoculated with 0.25 ml of cell suspension and grown at 37 °C until the cultures reached an

OD600 of 0.4, at which point the temperature of the incubator was decreased to 18 °C. The

cultures were grown until they reached an OD600 of 0.8, at which point T4 Ligase expression was

induced by adding IPTG (Sigma, USA) to a final concentration of 100 μM. The cultures were

Page 67: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

61

grown overnight at 18 °C and then harvested in cold JLA8.1 bottles by spinning in a JLA8.1

rotor (Beckman Coulter, USA) at 6000 rpm for 6 min at 4 °C. Cells were resuspended in 20 ml

of Ligase lysis buffer (Table 1) and transferred to cold 50 ml conical tubes, and stored overnight

at -20 °C. Cells were thawed the following day in warm water and divided into 4 tubes (~18 ml

per tube), which were then each brought up to 40 ml with cold Ligase lysis buffer (Table 1).

Sixty mg of chicken egg lysozyme (Sigma, USA), 400 µl of 8% sodium deoxycholate (Sigma,

USA), 400 µl of 100 mM PMSF (Sigma, USA) in isopropanol, and one large EDTA-free

complete protease inhibitor cocktail tablet (Roche, Canada) were added to each tube. Cells were

lysed with a VibraCell sonicator (Sonics, USA) for eight cycles of 20 sec of bursts at 85%

power/ 90 sec on ice. Lysates were centrifuged for 45 min at 18000 rpm at 4 °C in a cold JA25.5

rotor (Beckman Coulter, USA). The supernatant was filtered through a syringe fitted with a 0.8

µm filter (Thermo Scientific, USA) and then through a syringe fitted with a 0.45 µm filter

(Thermo Scientific, USA) into four 50 ml conical tubes. Three ml of agarose-Ni2+

-NTA bead

slurry (Qiagen, Inc.) was added to each conical tube and the tubes were nutated for 1 h at 4 °C.

Two polypropylene columns (Bio-Rad Laboratories, Inc.) were set up at 4 °C, washed once with

water and then once with 70% ethanol. The bead-supernatant slurry was loaded onto the columns

as described above for hPNKP, with each column containing ~3 ml of packed beads once fully

loaded. Each column was washed six times with 18 ml of cold Ligase lysis buffer (Table 1). T4

Ligase was retrieved from the column by loading 20 ml of Ligase elution buffer (Table 1) and

eluting; this step was repeated once for a total of two eluate fractions. Samples of uninduced and

induced cells, supernatant and pellet fractions, column flow-through and washes, and all eluates

were analyzed by SDS-PAGE in order to confirm the presence of T4 Ligase (Figure 4.6.1.c).

Page 68: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

62

Figure 4.6.1 hPNKP and T4 Ligase were purified from BL21 (DE3) CodonPlus E. coli cells. (a) The

presence of hPNKP protein in cells and lysates was confirmed prior to purification of the protein with

agarose-Ni2+

-NTA beads, TEV protease cleavage, and dialysis. (b) Final fractions were checked at the

completion of purification. Eluate fractions 1-6 were combined for dialysis. (c) Fractions were checked

for T4 Ligase prior to and following the application of lysates to agarose-Ni2+

-NTA beads.

Following elution, both eultion fractions were concentrated using an Amicon Ultra 30 kDa cut-

off centriprep column (Millipore, USA) to a volume of ~ 5 ml. To do so, 10 ml of T4 Ligase

elution buffer (Table 1) was first added to the column and spun through at 3,000 × g for 10 min

at 4 °C. The buffer was discarded and the protein was added to the column and similarly spun at

3,000 × g for 10 min at 4 °C. The flow-through was spun through the column repeatedly until it

reached a volume of ~ 5 ml, and then transferred to a cold 15 ml conical tube. The chamber of

the column was then rinsed with 1 ml of T4 Ligase elution buffer (Table 1) and this was added to

the conical tube as well. The concentration of protein in the conical tube was quantified as

described above. Next, two PD-10 desalting columns (GE Life Sciences, USA) were washed five

times with 4 ml of 2X T4 Ligase storage buffer (Table 1). Two and one-half ml of protein were

poured onto each column and eluted into a single cold 15 ml conical tube. The protein was then

Page 69: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

63

re-concentrated on an Amicon Ultra 30 kDa cut-off centriprep column (Millipore, USA) that had

been rinsed with 10 ml of 2X T4 Ligase storage buffer (Table 1). As before, the flow-through

was re-applied to the column, this time until it reached a volume of ~ 2.5 ml. The protein

concentration was quantified as above and an equal volume of sterile glycerol (Fischer Scientific,

USA) was added in order to store the purified protein in 50% glycerol at -20 °C.

4.7 Preparation of 3'-phosphorylated double-stranded DNA substrates.

Oligonucleotides were purchased from Integrated DNA Technologies (USA) or Eurofins MWG

Operon (USA). Complementary single-stranded oligonucleotides were diluted to 10-50 µM in 10

mM Tris-HCl, pH 7.5, 50 mM NaCl, and 1 mM EDTA. The tube was heated at 95 °C for 5 min

and then allowed to cool slowly at room temperature for at least 1 h.

4.8 Measurement of PNKP-dependent ligation by homogeneous time-resolved fluorescence HTRF) in a 384-well format, Version 1.

hPNKP was thawed on ice and diluted to 0.1 mg/ml in cold PNKP dilution buffer (Table 1).

HTRF buffer (Table 1) was prepared with 0.1 mg/ml BSA (Sigma, USA), 8.8 nM hPNKP and 74

nM T4 Ligase (HTRF-BLP buffer) in a 50 ml conical tube or a sterile glass bottle. HTRF-BP

buffer was prepared in an identical manner, except lacking T4 Ligase. Fifteen µl of HTRF-BLP

or HTRF-BP master mix was aliquoted into the requisite number of wells of an opaque black

polystyrene 384-well microplate (Corning, model # 3573). Five hundred nl of compound

dissolved in DMSO (Sigma, USA) was added to each reaction (or 500 nl of 100% DMSO for

controls) and incubated for 1 h at room temperature. To initiate reactions, 5 µl of SAXL-DNA

solution (Table 1) with 160 nM HTRFphos-1 (Table 2) and 160 nM HTRFphos-2 (Table 2) was

added to each well, giving final concentrations of 40 nM for each oligonucleotide. Unless the

experiment required a time course, the reaction was allowed to proceed for 1.5 h at room

temperature. To end reactions, 10 µl of DNPK solution (Table 1) was added to each well and

Page 70: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

64

incubated for 1 h at room temperature. SA-XL and DNPK fluorophores for HTRF were obtained

from Cisbio Bioassays, USA. HTRF signal was measured on a PheraStar microplate reader

(BMG Labtech, Germany). The donor fluorophore (DNPK) was excited at 337 nm and

fluorescence emission was measured simultaneously at 620 nm (Channel B, DNPK) and 665 nm

(Channel A, SAXL). HTRF ratios were calculated as (FI665×10,000)/FI620.

4.9 Measurement of PNKP-dependent ligation by HTRF in a 384-well format, Version 2.

hPNKP was thawed on ice and diluted to 4.9 μg/ml in cold PNKP dilution buffer (Table 1).

HTRF buffer (Table 1) was prepared with 2.1 nM hPNKP and 800 nM T4 Ligase (HTRF-LP

buffer) in a 50 ml conical tube or a sterile glass bottle. HTRF-P buffer was prepared in an

identical manner, except lacking T4 Ligase. Fifteen µl of HTRF-LP or HTRF-P buffer was

aliquoted into wells of an opaque black polystyrene 384-well microplate (Corning, model #

3573). Five hundred nl of compound dissolved in DMSO (Sigma, USA) was added to each

reaction (or 500 nl of 100% DMSO for controls) and incubated for 1 h at room temperature. To

initiate reactions, 5 µl of SAXL-DNA solution (Table 1) with 160 nM HTRFphos-3 (Table 2)

and 160 nM HTRFphos-4 (Table 2) was added to each well. Unless the experiment required a

time course, the reaction was allowed to proceed for 20 min at room temperature. To end

reactions, 10 µl of DNPK solution (Table 1) was added to each well and incubated for 1 h at

room temperature. HTRF signal was measured on a PheraStar microplate reader (BMG Labtech,

Germany) using the same settings as described above.

4.10 Measurement of PNKP-independent ligation by HTRF in a 384-well format, Versions 1 and 2.

Assays were carried out as described above except that hPNKP was left out of the reactions and

the oligonucleotides used in the SAXL + DNA solutions were either 160 nM HTRFOH-1 and

Page 71: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

65

160 nM HTRFOH-2 (Table 2) for Version 1, or 160 nM HTRFOH-3 and 160 nM HTRFOH-4

(Table 2) for Version 2.

4.11 Screening.

All mixes were prepared at the beginning of the day and aliquoted into source plates or reservoirs

[Polystyrene 96-well plates (#351177, Falcon, USA), polypropylene deep-well plates (P-DW-20-

C, Axygen, USA), or polypropylene reservoirs (RES-SW96-HP, Axygen, USA)]. If more than

one source plate was required for a given mix, or if a reservoir needed to be replenished during a

run, the remainder of the mix would be stored at 4 °C until needed.

For the first half of the screen (HTRF, Version 1), Biomek AP96 P20 and P250 tips (Beckman

Coulter, USA) were used. For the second half of the screen (HTRF, Version 2), FX-250-L-R

Maximum Recovery tips (Axygen, USA) and Biomek AP96 P20 tips (Beckman Coulter, USA)

were used. HTRF-(B)LP was dispensed with the 250 µl-capacity tips, while the SAXL + DNA

and DNPK solutions were dispensed with the 20 µl-capacity tips.

The screen was carried out on a Biomek FX platform (Beckman Coulter, USA) using a Thermo

CRS Dimension 4 system (Thermo Fisher Scientific, USA). Plates were kept in a room

temperature incubator for each incubation period. Plates were staggered in order to ensure that

incubation times were kept constant - for example, for a 30-plate screening run, there was a 7

min delay between plates. For each plate, 15 µl of HTRF-(B)LP or HTRF-(B)P mix was

dispensed from the source plate or reservoir into the wells of an opaque black polystyrene 384-

well microplate (Corning, model # 3573). Then, 0.2 µl of 1 mM compounds were pinned into the

wells of the screen plate (columns 3-22) and 0.2 µl DMSO (Sigma, USA) was subsequently

pinned from a “control” polypropylene 384-well source plate into the screen plate (columns 1-2

and 23-24). The compounds and protein(s) were incubated together for 1 h in the room

Page 72: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

66

temperature incubator. Next, 5 µl of SAXL + DNA solution was dispensed into each well of the

screen plate and incubated in the room temperature incubator for the appropriate amount of time,

depending on the version of the HTRF assay being applied. Ten µl of DNPK solution (Table 1)

was then dispensed into each well of the screen plate and incubated for 1 h in the room

temperature incubator prior to the HTRF signal being measured on the Pherastar plate reader

(BMG Labtech, Germany). It should be noted that a 384-pin head was used for pinning

compounds and DMSO, but a 96-tip head was used with the tips for dispensing each reagent; as

such, screen reagents were aspirated and dispensed four times per plate to minimize quadrant

effects.

4.12 Calculation of z’ factor, signal-to-noise (S/N), signal-to-background (S/B), and percent error.

The z’ factor was calculated as a measure of the available signal window according to the

methods of Zhang et al. (1999), wherein µp and σp represent the mean HTRF signal and standard

deviation, respectively, for reactions containing PNKP and Ligase (positive control). µn and σn

represent the mean HTRF signal and standard deviation, respectively, in reactions containing

PNKP only (negative control).

z' factor = 1 - [(3σp + 3σn)/(µp - µn)]

S/N = (µp - µn)/√(σp2 + σn

2)

S/B = µp ÷ µn

% error = (σ × 100) ÷ µ

Page 73: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

67

4.13 Calculation of Tanimoto coefficient.

The SMILES codes for the compounds of interest were entered together into an online similarity

matrix calculator with default fingerprint settings of 1024 bit length and a search depth of 6

(Guha, 2012).

4.14 Preparation of polyacrylamide sequencing gels.

One large (41.9 cm × 33.3 cm) and one small (39.4 cm × 33.3 cm) siliconized optically clear gel

plate (Gel Company, USA) were aligned with 0.25 mm spacers (Gel Company, USA) between

them, and then clamped together with bulldog clips. The bottom of the gel was sealed with vinyl

tape (VWR, USA). 12% polyacrylamide gel mix (Table 1) was poured between the plates either

with a syringe fitted with an 18-gauge needle or directly from a glass beaker. A 20- or 42-well

0.25 mm comb (Gel Company, USA) was placed between the plates and the top of the gel was

clamped as well. The gel was allowed to polymerize overnight at room temperature.

4.15 Phosphatase gel assay.

hPNKP was thawed on ice and diluted to 4.9 μg/ml in cold PNKP dilution buffer (Table 1).

HTRF-P buffer was prepared in a microfuge tube with 2.1 nM hPNKP. For reactions with T4

PNK, the HTRF-P buffer was prepared in an identical manner except with 5.7 nM T4 PNK in the

place of hPNKP. HTRF-NEG buffer was prepared in an identical manner, except lacking

enzyme. Seventeen and one-half µl of HTRF-P or HTRF-NEG buffer was aliquoted into

microfuge tubes or a generic PCR plate (Sarstedt, Germany). Five hundred nl of compound

dissolved in DMSO (Sigma, USA) was added to each reaction (or 500 nl of 100% DMSO for

controls) and incubated for 1 h at room temperature. To initiate reactions, 2 µl of 1.2 µM

GELphos DNA (Table 2) was added to each well. Unless the experiment required a time course,

the reaction was allowed to proceed for 20 min at room temperature. Reactions were stopped by

adding 20 µl of 2X formamide loading buffer (90% formamide, 25 mM EDTA, 0.02%

Page 74: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

68

bromophenol blue, 0.02% xylene cyanol) and heating for 5 min at 95 °C. Four µl (0.2 pmol of

DNA) from each reaction was loaded into a 12% polyacrylamide sequencing gel, which had

been pre-run for 1 h at 40 W. Samples were run in 1X TBE (Table 1) at 40 W for 3 h or until the

bromophenol blue dye reached the bottom of the gel. DNA was visualized by scanning in the

Cy5 channel with a Typhoon 9400 variable mode imager (GE Healthcare, USA) and the

percentage of dephosphorylation was quantified with ImageQuant 5.2 software (Molecular

Dynamics, USA).

4.16 Fluorescence polarization (FP) substrate binding assay.

hPNKP-D171A was thawed on ice. Two-times FP-PNKP buffer was prepared in a microfuge

tube by diluting hPNKP-D171A to 50 nM in FP buffer (Table 1). FP-NEG buffer (2X) was

prepared in an identical manner, except lacking hPNKP. Twelve and one-half µl of 2X FP-PNKP

buffer was aliquoted into wells of an opaque black polystyrene 384-well microplate (Corning,

model # 3573) and was incubated with 0.5 µl compound dissolved in DMSO (or 0.5 µl 100%

DMSO for controls) for 1 h at room temperature. FPphos DNA (Table 2) was diluted to 25 nM

in FP buffer (Table 1) and 12.5 µl was added to all reactions and incubated for 15 min at room

temperature. Fluorescence polarization was measured with an Analyst HT (Molecular Devices,

USA) using the fluorescein filter (samples excited at 485 nm and emission measured at 530 nm).

4.17 Measurement of the Kd of hPNKP-D171A in the FP substrate binding assay.

The fluorescence polarization assay was carried out as above except with the final concentration

of hPNKP titrated from 5 nM to 10 µM, and the percentage of substrate bound was determined

by FP (Figure 2.9.1). GraphPad 5.0 (Prism, USA) software was used to analyze the data, and the

Kd for this reaction was found to be 41.6 nM (Figure 2.9.1).

Page 75: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

69

Figure 4.17.1 FP reactions were conducted with hPNKP for proof of concept and to establish optimal

reaction conditions. FP reactions were conducted with 5 nM – 10 µM hPNKP, and with a Kd of 41.6 nM,

50 nM was selected as the concentration of hPNKP at which to perform dose response experiments with

inhibitors.

4.18 Reversibility assay.

hPNKP was thawed on ice and diluted to 0.15 mg/ml (2.6 µM) in HTRF Buffer (Table 1). The

diluted hPNKP was incubated at 100 nM with 0.5 µl compound in HTRF buffer (Table 1) for 30

min at room temperature. The concentration of compound for this incubation was 10-times its

IC50 for 2 nM hPNKP. Five µl of hPNKP with compound was added to 495 µl of SAXL + DNA

solution (Table 1) containing 40 nM HTRFphos3 (Table 2) and 40 nM HTRFphos4 (Table 2),

with 800 nM T4 Ligase in HTRF buffer (Table 1) and quickly inverted a few times to mix.

Twenty µl of reaction mix was aliquoted into the appropriate number of wells of an opaque black

polystyrene 384-well microplate (Corning, model # 3573) and reactions were stopped at

appropriate time points with the addition of 10 µl of REV-SD solution (Table 1). Reactions were

incubated with REV-SD solution for 1 h at room temperature and HTRF signal was measured

with a PheraStar microplate reader (BMG Labtech, Germany) as before.

4.19 Tissue culture.

U2OS human osteosarcoma cells (Ponten and Saksela, 1967) were maintained in McCoy's

medium (Gibco, USA) supplemented with 5% fetal bovine serum (Gibco, USA), 10 IU penicillin

and 0.1 mg/ml streptomycin (Multicell Technologies, Inc., USA). Cells were maintained at 37

Page 76: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

70

°C and 5% CO2. Cells were split when they approached 80-90% confluency by treating with a

0.25 % trypsin-EDTA solution (Gibco, USA) and transferring to new flasks.

4.20 ATP Lite cell viability assay.

Cells were lifted from a non-confluent flask by trypsinizing for no more than five minutes, and

transferred to a sterile 15 ml conical tube. The concentration of cells was determined using a

hemocytometer and the appropriate amount of cells were diluted into 10 ml of warm media to

give 15,000 cells/ml. 100 μl of diluted cells (1500 cells) were seeded in the requisite number of

wells of an opaque, white, tissue culture-treated 96-well polystyrene Costar microplate (Corning,

model #3917) using a multichannel pipette and a sterile single-use polypropylene basin. Cells

were then incubated for 24 h at 37 °C/5% CO2 to allow them to attach to the plate. The following

day, hPNKP inhibitors and CPT were diluted to a 500X concentration in tissue culture-quality

DMSO (Sigma, USA) by serial dilution in a biological safety tissue culture cabinet. To prepare

media for treatments, 1 μl of compound (or DMSO for controls) was added for every 500 μl of

media and vortexed thoroughly. After the 24 h incubation, media was suctioned off and 100 µl of

media containing compounds and/or DMSO was added to the appropriate wells in duplicate.

Cells were returned to 37 °C/5% CO2for the 96 h drug treatment. To measure viability, the ATP

Lite cell viability kit (PerkinElmer, USA) was used as directed. ATP Lite Lysis buffer and ATP

Lite Substrate were thawed slowly until they reached room temperature. Fifty μl of Lysis buffer

was added to each well with a multichannel pipette and the plate was shaken at 700 rpm for 5

min. Then, 50 μl of Substrate was added to each well by multichannel pipette and the plate was

shaken again at 700 rpm for 5 min. Chemiluminescence was measured with an EnSpire 2300

multi-label plate reader (PerkinElmer, USA) after a 10 min delay to dark-adapt the plate.

Page 77: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

71

Tables

Table 1 Buffers

Coommassie dye

solution

0.2% Coommassie Brilliant Blue R-250, 40% methanol

De-stain solution 30% methanol, 5% glacial acetic acid

DNPK solution (3X) 5 ng DNPK, 0.02% Tween-20, 0.5X PBS (1.2 M KF, Version

1; and 150 mM EDTA pH 8.0, Version 2)

HTRF Buffer (1X) 50 mM Tris-HCl, pH 7.5, 10 mM MgCl2, 10 mM DTT, 1mM

ATP, 0.02% Tween-20

FP buffer (1X) 50 mM Tris-HCl, pH 7.5, 10 mM MgCl2, 10 mM DTT, 0.2%

Tween-20

Luria broth (LB) 0.5% yeast extract, 1% NaCl, 1% tryptone peptone

PBS (1X) 137 mM NaCl, 7.8 mM Na2HPO4, 2.7 mM KCl, 1.5 mM

KH2PO4

PNKP dialysis

buffer

50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 1 mM DTT, 20%

glycerol

PNKP dilution

buffer (1X)

50 mM Tris-HCl, pH 7.0, 100 mM KCl, 1 mM EDTA, 1mM

DTT

PNKP lysis buffer 50 mM Tris-HCl, pH 8.0, 500 mM NaCl, 200 μM PMSF,

0.04% β-mercaptoethanol, and two EDTA-free complete

protease inhibitor cocktail tablets (Roche) per 200 ml buffer

SAXL solution 50 ng SAXL, 1X PBS

SAXL-DNA

solution (4X)

50 ng SAXL, 4X DNA substrates, TE pH 7.0

REV-SD solution 71 ng SA-XL, 5 ng DNPK, 50 mM EDTA, 0.02% Tween; in

1X PBS

SDS-PAGE running

buffer (1X)

25 mM Tris, 200 μM glycine, 0.1% SDS

SDS-PAGE sample

buffer (1X)

83 mM Tris-HCl, pH 6.8, 1% SDS, 0.003% bromophenol

blue, 10% glycerol, 10 mM DTT

SDS-PAGE

separating gel

375 mM Tris-HCl, pH 8.8, 0.1% SDS, 0.05% ammonium

persulfate, 0.1% TEMED, 40% acrylamide:bisacrylamide

(29:1) to desired final concentration

SDS-PAGE stacking

gel

0.06 M Tris-HCl, pH 6.8, 0.1% SDS, 0.1% ammonium

persulfate, 0.1% TEMED, 40% acrylamide:bisacrylamide

(29:1) to desired final concentration

T4 Ligase buffer

(1X)

50 mM Tris-HCl, pH 7.5, 10 mM MgCl2, 10 mM DTT, 1 mM

ATP

T4 Ligase elution

buffer

50 mM sodium phosphate, pH 8.0, 300 mM NaCl, 300 mM

imidazole, 0.1% β-mercaptoethanol

T4 Ligase lysis

buffer

50 mM sodium phosphate, pH 8.0, 300 mM NaCl, 10 mM

imidazole, 0.1% β-mercaptoethanol

Page 78: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

72

Table 3 Buffers (continued)

T4 Ligase storage

buffer (1X)

10 mM Tris-HCl, pH 7.5, 50 mM KCl, 1 mM DTT, 50%

glycerol

T4 Ligase storage

buffer without

glycerol (2X)

20 mM Tris-HCl, pH 7.5, 100 mM KCl, 2 mM DTT

TBE buffer (1X) 89 mM Tris base, 89 mM boric acid, 10 mM EDTA

TE buffer 10 mM Tris-HCl, pH 7.0-8.0 as needed, 1 mM EDTA

Page 79: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

73

Table 2 Sequences of oligonucleotide substrates (5' to 3')

Bio14 ATTACCGTAATCAG-biotin 14 nt

Bio2 pATTACCGTAATCATG-biotin 15 nt

Cy5PNK Cy5-TAATCGAGCTCGAATTCACT 20 nt

DNP11OH DNP-GAATTCAGTCG 11 nt

DNP11phos DNP-GAATTCAGTCGp 11 nt

DNPOH DNP-AGTGAATTCGAGCTCG 16 nt

DNPphos DNP-AGTGAATTCGAGCTCGp 16 nt

FPphos 6-FAM-TCCTCp 5 nt

GELphos Cy5PNK + Oligo1phos annealed 20 nt dsDNA

HTRFOH-1 DNPOH + Oligo5 annealed 20 nt dsDNA

HTRFOH-2 Bio2 + Oligo2OH annealed 15 nt dsDNA

HTRFOH-3 DNP11OH + Oligo15 annealed 15 nt dsDNA

HTRFOH-4 Bio14 + Oligo10OH annealed 14 nt dsDNA

HTRFphos-1 DNPphos + Oligo5 annealed 20 ntdsDNA

HTRFphos-2 Bio2 + Oligo2phos annealed 15 nt dsDNA

HTRFphos-3 DNP11phos + Oligo15 annealed 15 nt dsDNA

HTRFphos-4 Bio14 + Oligo10phos annealed 14 nt dsDNA

Oligo1phos AGTGAATTCGAGCTCGp 16 nt

Oligo2OH pCATGATTACGG 11 nt

Oligo2phos CATGATTACGGp 11 nt

Oligo5 pTAATCGAGCTCGAATTCACT 20 nt

Oligo10OH pCTGATTACGG 10 nt

Oligo10phos pCTGATTACGGp 10 nt

Oligo15 pTAATCGACTGAATTC 15 nt

Page 80: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

74

Table 3 Original hit compounds

Compound Name Structure

OICR1

OICR2

OICR3

OICR4

OICR5

Page 81: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

75

Table 1 Original hit compounds (continued)

OICR6

OICR7

OICR8

OICR9

Page 82: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

76

Table 4 Analog compounds

Compound Name Structure

OICR7A

OICR9A

OICR9B

OICR79

OICR791

OICR792

Page 83: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

77

Table 2 Analog compounds (continued)

OICR793

OICR794

OICR795

OICR796

Table 4 Analog compounds Functional groups in red represent groups that have been modified relative

to the parent compound, OICR79, for the purpose of structure-activity relationship analysis.

Page 84: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

70

References Cited

Ali, A.A., Jukes, R.M., Pearl, L.H., and Oliver, A.W. (2009). Specific recognition of a multiply

phosphorylated motif in the DNA repair scaffold XRCC1 by the FHA domain of human PNK.

Nucleic Acids Res 37, 1701-1712.

Anders, C.K., Winer, E.P., Ford, J.M., Dent, R., Silver, D.P., Sledge, G.W., and Carey, L.A.

(2010). Poly(ADP-Ribose) polymerase inhibition: "targeted" therapy for triple-negative breast

cancer. Clin Cancer Res 16, 4702-4710.

Audebert, M., Salles, B., and Calsou, P. (2004). Involvement of poly(ADP-ribose) polymerase-1

and XRCC1/DNA ligase III in an alternative route for DNA double-strand breaks rejoining. J

Biol Chem 279, 55117-55126.

Barr, A.J. (2010). Protein tyrosine phosphatases as drug targets: strategies and challenges of

inhibitor development. Future Med Chem 2, 1563-1576.

Bazin, H., Trinquet, E., and Mathis, G. (2002). Time resolved amplification of cryptate emission:

a versatile technology to trace biomolecular interactions. J Biotechnol 82, 233-250.

Bernstein, N.K., Karimi-Busheri, F., Rasouli-Nia, A., Mani, R., Dianov, G., Glover, J.N., and

Weinfeld, M. (2008). Polynucleotide kinase as a potential target for enhancing cytotoxicity by

ionizing radiation and topoisomerase I inhibitors. Anticancer Agents Med Chem 8, 358-367.

Bernstein, N.K., Williams, R.S., Rakovszky, M.L., Cui, D., Green, R., Karimi-Busheri, F., Mani,

R.S., Galicia, S., Koch, C.A., Cass, C.E., et al. (2005). The molecular architecture of the

mammalian DNA repair enzyme, polynucleotide kinase. Mol Cell 17, 657-670.

Branzei, D., and Foiani, M. (2008). Regulation of DNA repair throughout the cell cycle. Nat Rev

Mol Cell Biol 9, 297-308.

Breslin, C., and Caldecott, K.W. (2009). DNA 3'-phosphatase activity is critical for rapid global

rates of single-strand break repair following oxidative stress. Mol Cell Biol 29, 4653-4662.

Bryant, H.E., Petermann, E., Schultz, N., Jemth, A.S., Loseva, O., Issaeva, N., Johansson, F.,

Fernandez, S., McGlynn, P., and Helleday, T. (2009). PARP is activated at stalled forks to

mediate Mre11-dependent replication restart and recombination. Embo J 28, 2601-2615.

Bryant, H.E., Schultz, N., Thomas, H.D., Parker, K.M., Flower, D., Lopez, E., Kyle, S., Meuth,

M., Curtin, N.J., and Helleday, T. (2005). Specific killing of BRCA2-deficient tumours with

inhibitors of poly(ADP-ribose) polymerase. Nature 434, 913-917.

Buchko, G.W., and Weinfeld, M. (1993). Influence of nitrogen, oxygen, and nitroimidazole

radiosensitizers on DNA damage induced by ionizing radiation. Biochemistry 32, 2186-2193.

Caldecott, K.W. (2008). Single-strand break repair and genetic disease. Nat Rev Genet 9, 619-

631.

Page 85: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

71

Chappell, C., Hanakahi, L.A., Karimi-Busheri, F., Weinfeld, M., and West, S.C. (2002).

Involvement of human polynucleotide kinase in double-strand break repair by non-homologous

end joining. Embo J 21, 2827-2832.

Chen, Y.H., Yang, J.T., and Chau, K.H. (1974). Determination of the helix and beta form of

proteins in aqueous solution by circular dichroism. Biochemistry 13, 3350-3359.

Ciccia, A., and Elledge, S.J. (2010). The DNA damage response: making it safe to play with

knives. Mol Cell 40, 179-204.

Copeland, R.A. (2005). Evaluation of enzyme inhibitors in drug discovery. A guide for

medicinal chemists and pharmacologists. Methods Biochem Anal 46, 1-265.

Coquelle, N., Havali-Shahriari, Z., Bernstein, N., Green, R., and Glover, J.N. (2011). Structural

basis for the phosphatase activity of polynucleotide kinase/phosphatase on single- and double-

stranded DNA substrates. Proceedings of the National Academy of Sciences of the United States

of America 108, 21022-21027.

Coquerelle, T., Bopp, A., Kessler, B., and Hagen, U. (1973). Strand breaks and 5' end-groups in

DNA of irradiated thymocytes. Int J Radiat Biol Relat Stud Phys Chem Med 24, 397-404.

Croce, C.M. (2008). Oncogenes and cancer. N Engl J Med 358, 502-511.

Di Leonardo, A., Linke, S.P., Clarkin, K., and Wahl, G.M. (1994). DNA damage triggers a

prolonged p53-dependent G1 arrest and long-term induction of Cip1 in normal human

fibroblasts. Genes Dev 8, 2540-2551.

Doherty, A.J., and Suh, S.W. (2000). Structural and mechanistic conservation in DNA ligases.

Nucleic Acids Res 28, 4051-4058.

El-Khamisy, S.F., Masutani, M., Suzuki, H., and Caldecott, K.W. (2003). A requirement for

PARP-1 for the assembly or stability of XRCC1 nuclear foci at sites of oxidative DNA damage.

Nucleic Acids Res 31, 5526-5533.

Farmer, H., McCabe, N., Lord, C.J., Tutt, A.N., Johnson, D.A., Richardson, T.B., Santarosa, M.,

Dillon, K.J., Hickson, I., Knights, C., et al. (2005). Targeting the DNA repair defect in BRCA

mutant cells as a therapeutic strategy. Nature 434, 917-921.

Freschauf, G.K., Karimi-Busheri, F., Ulaczyk-Lesanko, A., Mereniuk, T.R., Ahrens, A., Koshy,

J.M., Rasouli-Nia, A., Pasarj, P., Holmes, C.F., Rininsland, F., et al. (2009). Identification of a

small molecule inhibitor of the human DNA repair enzyme polynucleotide kinase/phosphatase.

Cancer Res 69, 7739-7746.

Freschauf, G.K., Mani, R.S., Mereniuk, T.R., Fanta, M., Virgen, C.A., Dianov, G.L., Grassot,

J.M., Hall, D.G., and Weinfeld, M. (2010). Mechanism of action of an imidopiperidine inhibitor

of human polynucleotide kinase/phosphatase. J Biol Chem 285, 2351-2360.

Galburt, E.A., Pelletier, J., Wilson, G., and Stoddard, B.L. (2002). Structure of a tRNA repair

enzyme and molecular biology workhorse: T4 polynucleotide kinase. Structure 10, 1249-1260.

Gioanni, J., Le Francois, D., Zanghellini, E., Mazeau, C., Ettore, F., Lambert, J.C., Schneider,

M., and Dutrillaux, B. (1990). Establishment and characterisation of a new tumorigenic cell line

with a normal karyotype derived from a human breast adenocarcinoma. Br J Cancer 62, 8-13.

Page 86: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

72

Gordon, J.A. (1991). Use of vanadate as protein-phosphotyrosine phosphatase inhibitor. Methods

Enzymol 201, 477-482.

Guha, R. (n.d.) Similarity Matrix Calculator. In On the Web. Retrieved May 30, 2012, from

http://rguha.net/code/java/cdkws/sim.html.

Habraken, Y., and Verly, W.G. (1983). The DNA 3'-phosphatase and 5'-hydroxyl kinase of rat

liver chromatin. FEBS Lett 160, 46-50.

Helleday, T. (2011). The underlying mechanism for the PARP and BRCA synthetic lethality:

clearing up the misunderstandings. Mol Oncol 5, 387-393.

Henner, W.D., Grunberg, S.M., and Haseltine, W.A. (1982). Sites and structure of gamma

radiation-induced DNA strand breaks. J Biol Chem 257, 11750-11754.

Inoue, H., Nojima, H., and Okayama, H. (1990). High efficiency transformation of Escherichia

coli with plasmids. Gene 96, 23-28.

Jackson, S.P. (2002). Sensing and repairing DNA double-strand breaks. Carcinogenesis 23, 687-

696.

Jackson, S.P., and Bartek, J. (2009). The DNA-damage response in human biology and disease.

Nature 461, 1071-1078.

Kamenski, T., Heilmeier, S., Meinhart, A., and Cramer, P. (2004). Structure and mechanism of

RNA polymerase II CTD phosphatases. Mol Cell 15, 399-407.

Karimi-Busheri, F., Daly, G., Robins, P., Canas, B., Pappin, D.J., Sgouros, J., Miller, G.G.,

Fakhrai, H., Davis, E.M., Le Beau, M.M., et al. (1999). Molecular characterization of a human

DNA kinase. J Biol Chem 274, 24187-24194.

Karimi-Busheri, F., Rasouli-Nia, A., Allalunis-Turner, J., and Weinfeld, M. (2007). Human

polynucleotide kinase participates in repair of DNA double-strand breaks by nonhomologous end

joining but not homologous recombination. Cancer Res 67, 6619-6625.

Kling, J. (2009). PARP inhibitors blaze a trail in difficult-to-treat cancers. Nat Biotechnol 27,

784-786.

Koch, C.A., Agyei, R., Galicia, S., Metalnikov, P., O'Donnell, P., Starostine, A., Weinfeld, M.,

and Durocher, D. (2004). Xrcc4 physically links DNA end processing by polynucleotide kinase

to DNA ligation by DNA ligase IV. Embo J 23, 3874-3885.

Lee, K.J., Huang, J., Takeda, Y., and Dynan, W.S. (2000). DNA ligase IV and XRCC4 form a

stable mixed tetramer that functions synergistically with other repair factors in a cell-free end-

joining system. J Biol Chem 275, 34787-34796.

Loizou, J.I., El-Khamisy, S.F., Zlatanou, A., Moore, D.J., Chan, D.W., Qin, J., Sarno, S.,

Meggio, F., Pinna, L.A., and Caldecott, K.W. (2004). The protein kinase CK2 facilitates repair

of chromosomal DNA single-strand breaks. Cell 117, 17-28.

Mao, Z., Bozzella, M., Seluanov, A., and Gorbunova, V. (2008). DNA repair by nonhomologous

end joining and homologous recombination during cell cycle in human cells. Cell Cycle 7, 2902-

2906.

Page 87: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

73

Miki, Y., Swensen, J., Shattuck-Eidens, D., Futreal, P.A., Harshman, K., Tavtigian, S., Liu, Q.,

Cochran, C., Bennett, L.M., Ding, W., et al. (1994). A strong candidate for the breast and

ovarian cancer susceptibility gene BRCA1. Science 266, 66-71.

Mohammad, D.H., and Yaffe, M.B. (2009). 14-3-3 proteins, FHA domains and BRCT domains

in the DNA damage response. DNA Repair (Amst) 8, 1009-1017.

Moreira, M.C., Barbot, C., Tachi, N., Kozuka, N., Uchida, E., Gibson, T., Mendonca, P., Costa,

M., Barros, J., Yanagisawa, T., et al. (2001). The gene mutated in ataxia-ocular apraxia 1

encodes the new HIT/Zn-finger protein aprataxin. Nat Genet 29, 189-193.

O'Driscoll, M., and Jeggo, P.A. (2008). The role of the DNA damage response pathways in brain

development and microcephaly: insight from human disorders. DNA Repair (Amst) 7, 1039-

1050.

Pagano, J.M., Clingman, C.C., and Ryder, S.P. (2011). Quantitative approaches to monitor

protein-nucleic acid interactions using fluorescent probes. Rna 17, 14-20.

Pennell, S., Westcott, S., Ortiz-Lombardia, M., Patel, D., Li, J., Nott, T.J., Mohammed, D.,

Buxton, R.S., Yaffe, M.B., Verma, C., et al. (2010). Structural and functional analysis of

phosphothreonine-dependent FHA domain interactions. Structure 18, 1587-1595.

Pfuhler, S., and Wolf, H.U. (1996). Detection of DNA-crosslinking agents with the alkaline

comet assay. Environ Mol Mutagen 27, 196-201.

Pheiffer, B.H., and Zimmerman, S.B. (1982). 3'-Phosphatase activity of the DNA kinase from rat

liver. Biochem Biophys Res Commun 109, 1297-1302.

Ponten, J., and Saksela, E. (1967). Two established in vitro cell lines from human mesenchymal

tumours. Int J Cancer 2, 434-447.

Rasouli-Nia, A., Karimi-Busheri, F., and Weinfeld, M. (2004). Stable down-regulation of human

polynucleotide kinase enhances spontaneous mutation frequency and sensitizes cells to genotoxic

agents. Proc Natl Acad Sci U S A 101, 6905-6910.

Ray Chaudhuri, A., Hashimoto, Y., Herrador, R., Neelsen, K.J., Fachinetti, D., Bermejo, R.,

Cocito, A., Costanzo, V., and Lopes, M. (2012). Topoisomerase I poisoning results in PARP-

mediated replication fork reversal. Nat Struct Mol Biol 19, 417-423.

Reynolds, J.J., El-Khamisy, S.F., and Caldecott, K.W. (2009). Short-patch single-strand break

repair in ataxia oculomotor apraxia-1. Biochem Soc Trans 37, 577-581.

Sambrook, J., and Russell, D.W. (2001). Molecular cloning : a laboratory manual, 3rd edn (Cold

Spring Harbor, N.Y., Cold Spring Harbor Laboratory Press).

Satoh, M.S., and Lindahl, T. (1992). Role of poly(ADP-ribose) formation in DNA repair. Nature

356, 356-358.

Savitsky, K., Bar-Shira, A., Gilad, S., Rotman, G., Ziv, Y., Vanagaite, L., Tagle, D.A., Smith, S.,

Uziel, T., Sfez, S., et al. (1995). A single ataxia telangiectasia gene with a product similar to PI-3

kinase. Science 268, 1749-1753.

Sawyers, C. (2004). Targeted cancer therapy. Nature 432, 294-297.

Page 88: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

74

Shah, N.P. (2008). Bench to bedside: BRCA: from therapeutic target to therapeutic shield. Nat

Med 14, 495-496.

Shen, J., Gilmore, E.C., Marshall, C.A., Haddadin, M., Reynolds, J.J., Eyaid, W., Bodell, A.,

Barry, B., Gleason, D., Allen, K., et al. (2010). Mutations in PNKP cause microcephaly, seizures

and defects in DNA repair. Nat Genet 42, 245-249.

Siddik, Z.H. (2003). Cisplatin: mode of cytotoxic action and molecular basis of resistance.

Oncogene 22, 7265-7279.

Strom, C.E., Johansson, F., Uhlen, M., Szigyarto, C.A., Erixon, K., and Helleday, T. (2011).

Poly (ADP-ribose) polymerase (PARP) is not involved in base excision repair but PARP

inhibition traps a single-strand intermediate. Nucleic Acids Res 39, 3166-3175.

Takahashi, T., Tada, M., Igarashi, S., Koyama, A., Date, H., Yokoseki, A., Shiga, A., Yoshida,

Y., Tsuji, S., Nishizawa, M., et al. (2007). Aprataxin, causative gene product for EAOH/AOA1,

repairs DNA single-strand breaks with damaged 3'-phosphate and 3'-phosphoglycolate ends.

Nucleic Acids Res 35, 3797-3809.

Turner, N.C., Lord, C.J., Iorns, E., Brough, R., Swift, S., Elliott, R., Rayter, S., Tutt, A.N., and

Ashworth, A. (2008). A synthetic lethal siRNA screen identifying genes mediating sensitivity to

a PARP inhibitor. Embo J 27, 1368-1377.

van Breemen, R.B., and Li, Y. (2005). Caco-2 cell permeability assays to measure drug

absorption. Expert Opin Drug Metab Toxicol 1, 175-185.

Venkitaraman, A.R. (2002). Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell

108, 171-182.

Weinberg, R.A. (1991). Tumor suppressor genes. Science 254, 1138-1146.

Whitehouse, C.J., Taylor, R.M., Thistlethwaite, A., Zhang, H., Karimi-Busheri, F., Lasko, D.D.,

Weinfeld, M., and Caldecott, K.W. (2001). XRCC1 stimulates human polynucleotide kinase

activity at damaged DNA termini and accelerates DNA single-strand break repair. Cell 104, 107-

117.

Wiederhold, L., Leppard, J.B., Kedar, P., Karimi-Busheri, F., Rasouli-Nia, A., Weinfeld, M.,

Tomkinson, A.E., Izumi, T., Prasad, R., Wilson, S.H., et al. (2004). AP endonuclease-

independent DNA base excision repair in human cells. Mol Cell 15, 209-220.

Wooster, R., Bignell, G., Lancaster, J., Swift, S., Seal, S., Mangion, J., Collins, N., Gregory, S.,

Gumbs, C., and Micklem, G. (1995). Identification of the breast cancer susceptibility gene

BRCA2. Nature 378, 789-792.

Zaremba, T., and Curtin, N.J. (2007). PARP inhibitor development for systemic cancer targeting.

Anticancer Agents Med Chem 7, 515-523.

Zhang, J.H., Chung, T.D., and Oldenburg, K.R. (1999). A Simple Statistical Parameter for Use in

Evaluation and Validation of High Throughput Screening Assays. J Biomol Screen 4, 67-73.

Zhang, M., Cho, E.J., Burstein, G., Siegel, D., and Zhang, Y. (2011). Selective inactivation of a

human neuronal silencing phosphatase by a small molecule inhibitor. ACS Chem Biol 6, 511-

519.

Page 89: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

75

Appendices

I. PNKP inhibitors

Page 90: Identification and Characterization of Small Molecule …...ii Identification and Characterization of Small Molecule Inhibitors of Polynucleotide Kinase 3’-Phosphatase Nathalie Moatti

76

II. Analogs that did not effectively inhibit PNKP