dna oxidation and base excision repair in lung and …

93
DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND LIVER OF 4-(METHYLNITROSAMINO)-1-(3-PYRIDYL)-1- BUTANONE TREATED MICE by Neeraj Gupta A thesis submitted to the Department of Pharmacology and Toxicology In conformity with the requirements for the degree of Master of Science Queen’s University Kingston, Ontario, Canada (April, 2011) Copyright © Neeraj Gupta, 2011

Upload: others

Post on 09-Jun-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND

LIVER OF 4-(METHYLNITROSAMINO)-1-(3-PYRIDYL)-1-

BUTANONE TREATED MICE

by

Neeraj Gupta

A thesis submitted to the Department of Pharmacology and Toxicology

In conformity with the requirements for

the degree of Master of Science

Queen’s University

Kingston, Ontario, Canada

(April, 2011)

Copyright © Neeraj Gupta, 2011

Page 2: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

ii

Abstract

4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is a potent pulmonary

carcinogen found in unburned tobacco and tobacco smoke. To exert its carcinogenic effect, NNK

is metabolically activated to reactive intermediates that can damage DNA by alkylation or

pyridyloxobutylation. NNK also has the ability to induce DNA oxidation and alter DNA repair

activities that can result in deficient repair and potentially exacerbate carcinogenesis. Base

excision repair (BER) is a ubiquitous DNA repair system that mainly repairs oxidative DNA

damage. The goal of this study was to determine the effect of NNK on DNA oxidation status and

BER activity in A/J mouse lung and liver. Female mice were treated with 10 mol of NNK i.p.

and lung and liver were isolated 1, 2 and 24 hours post administration. DNA was isolated from

lung and liver, and the formation of 8-hydroxydeoxyguanosine (8-OHdG, a biomarker of DNA

oxidation) was assessed by high-performance liquid chromatography with electrochemical

detection. At 1, 2 and 24 hours in both murine lung and liver, there was no statistically

significant difference in 8-OHdG levels (n = 4, P > 0.05) between control and NNK-treated mice.

To assess BER, cell-free whole tissue nuclear protein extracts from liver and lung were prepared

and incubated with a plasmid substrate containing oxidative DNA damage. In vivo treatment

with NNK did not alter BER activity in lung or liver compared to control mice (n=3 or 4, P >

0.05). These experiments indicate that acute treatment with a tumourigenic dose of NNK does

not significantly stimulate oxidative DNA damage or significantly alter BER activity in murine

lung and liver.

Keywords: 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, oxidative DNA damage, base

excision repair

Page 3: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

iii

Co-Authorship

This research was conducted by the candidate, Neeraj Gupta, under the supervision of Dr.

Thomas E. Massey.

Page 4: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

iv

Acknowledgements

First and foremost, I would like to thank my supervisor, Dr. Thomas Massey, for his

guidance and support during this project. I would also like to thank current and former lab

members for their advice, assistance and friendship: Ms. Sandra Graham, Mr. Mason Curtis, Dr.

Pamela Brown, and Dr. Katherine Guindon. Additionally, I would like to thank the faculty,

administrative staff and students of the Department of Pharmacology and Toxicology for their

assistance and advice. I would like to thank my thesis committee members, Dr. Louise Winn and

Dr. James Reynolds, for giving this project additional direction and focus. I would also like to

thank Dr. Donald Maurice for his helpful advice on preparing plasmid.

This research would not be possible without the financial support from the Canadian

Institute of Health Research, the Department of Pharmacology and Toxicology, and the School of

Graduate Studies and Research.

Finally I would especially like to thank my family for their constant love, support and

encouragement during the entire course of this project.

Page 5: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

v

Table of Contents

Abstract ............................................................................................................................................ ii

Co-Authorship ................................................................................................................................ iii

Acknowledgements ......................................................................................................................... iv

Table of Contents ............................................................................................................................. v

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

List of Abbreviations .................................................................................................................... viii

Chapter 1 GENERAL INTRODUCTION ....................................................................................... 1

1.1 Statement of research problem ............................................................................................... 1

1.2 Multi-stage process of carcinogenesis ................................................................................... 1

1.2.1 Tumour initiation ............................................................................................................ 2

1.2.2 Tumour promotion .......................................................................................................... 3

1.2.3 Tumour progression ........................................................................................................ 4

1.3 Lung cancer and smoking ...................................................................................................... 4

1.4 Human lung cancer classification .......................................................................................... 7

1.4.1 Modelling lung cancer in mouse ..................................................................................... 8

1.5 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) ...................................................... 9

1.5.1 NNK and lung cancer .................................................................................................... 10

1.5.2 Metabolic pathways of NNK ........................................................................................ 14

1.5.3 Cytochrome P450 enzymes and bioactivation of NNK ................................................ 17

1.5.4 NNK and DNA adducts ................................................................................................ 17

1.6 Base excision repair and oxidative stress ............................................................................. 19

1.6.1 Genetic damage induced by ROS ................................................................................. 22

1.6.2 NNK and DNA oxidation ............................................................................................. 23

1.7 Base excision repair (BER) pathway ................................................................................... 25

1.7.1 Decision between short and long patch repair in BER ................................................. 29

1.7.2 BER and carcinogenicity .............................................................................................. 30

1.8 NNK and DNA repair systems ............................................................................................ 32

1.9 Research hypothesis and objectives ..................................................................................... 34

Chapter 2 MATERIALS AND METHODS .................................................................................. 35

2.1 Reagents ............................................................................................................................... 35

2.2 Animal Treatments............................................................................................................... 35

Page 6: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

vi

2.3 Isolation of DNA from lung and liver .................................................................................. 36

2.4 DNA digestion ..................................................................................................................... 37

2.5 Determination of 8-OHdG and 2’-dG levels by high-performance liquid chromatography

with electrochemical detection .................................................................................................. 38

2.6 Cell-free whole tissue nuclear protein extract preparation .................................................. 38

2.7 Preparation of 8-OHdG adducted plasmid ........................................................................... 40

2.8 In vitro base excision repair assay ....................................................................................... 41

2.9 Data analysis ........................................................................................................................ 44

Chapter 3 RESULTS...................................................................................................................... 45

3.1 Levels of 8-OHdG following in vivo treatment with NNK.................................................. 45

3.2 Characterization of 8-OHdG adducted plasmid DNA substrate for in vitro repair assay .... 48

3.3 Optimization of nuclear protein extract amount for in vitro repair assay ............................ 51

3.4 Optimization of incubation time for in vitro repair assay .................................................... 51

3.5 Effect of in vivo treatment of mice with NNK on BER in lung and liver ............................ 53

Chapter 4 GENERAL DISCUSSION ............................................................................................ 57

4.1 In vivo treatment with NNK does not induce oxidative DNA damage ................................ 57

4.2 In vivo treatment with NNK does not alter overall BER activity ........................................ 60

4.3 Conclusions .......................................................................................................................... 65

4.4 Future directions .................................................................................................................. 65

REFERENCES .............................................................................................................................. 68

Page 7: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

vii

List of Figures

Figure 1.1 Formation of tobacco specific nitrosamines (TSNA) from major (nicotine) and minor

(nornicotine, anabasine and anatabine) tobacco alkaloid precursors ..................................... 11

Figure 1.2 Pathways of NNK metabolism ..................................................................................... 15

Figure 1.3 Proposed mechanism of generation of ROS by uncoupling of CYP during NNK

metabolism ............................................................................................................................. 26

Figure 1.4 Mechanisms of BER pathway depicting short patch and long patch repair ................. 27

Figure 2.1 Chromatograms demonstrating resolution of 2’-dG and 8-OHdG, isolated from lung

DNA of mice following in vivo treatment with saline ........................................................... 39

Figure 2.2 Schematic of in vitro DNA repair synthesis assay on damaged plasmid DNA by

nuclear extracts isolated from whole tissue. .......................................................................... 42

Figure 2.3 UV-illuminated ethidium bromide stained agarose gel of linearized plasmid DNA and

phosphor image demonstrating the ability of cell-free nuclear protein extracts to catalyze in

vitro DNA repair synthesis .................................................................................................... 43

Figure 3.1 Effect of in vivo treatment with NNK on 8-OHdG levels in lung DNA ...................... 46

Figure 3.2 Effect of in vivo treatment with NNK on 8-OHdG levels in liver DNA ...................... 47

Figure 3.3 Characteristics of 8-OHdG adducted pBluescript SK+ plasmid by photo-oxidation of

methylene blue ....................................................................................................................... 49

Figure 3.4 Optimization of 8-OHdG adducted plasmid for in vitro BER assay ............................ 50

Figure 3.5 Optimization of protein amount for in vitro BER assay ............................................... 52

Figure 3.6 Optimization of incubation time for in vitro BER assay .............................................. 54

Figure 3.7 Effect of in vivo treatment with NNK on in vitro BER in mouse lung extracts towards

8-OHdG adducted plasmid .................................................................................................... 55

Figure 3.8 Effect of in vivo treatment with NNK on in vitro BER in mouse liver extracts towards

8-OHdG adducted plasmid .................................................................................................... 56

Page 8: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

viii

List of Abbreviations

A adenine

AC adenocarcinoma

ad libitum free feeding

ADP adenosine diphosphate

amol atto (10-18

) moles

AP apurinic/apyrimidinic site

BER base excision repair

bp basepair

BP benzo[ ]pyrene

C cytosine

oC degree(s) Celsius

Ci curie(s)

cm centimetre(s)

CYP cytochrome P450

dATP deoxyadenosine triphosphate

dCTP deoxycytidine triphosphate

DFO deferoxamine mesylate

2’-dG 2’-deoxyguanosine

dGTP deoxyguanosine triphosphate

DNA deoxyribonucleic acid

5’dRP 5’-deoxyribose-5-phosphate

dTTP deoxythymidine triphosphate

EDTA ethylenediaminetetraacetic acid

Page 9: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

ix

e.g. exempli gratia (for example)

EtBr ethidium bromide

Fe iron

Fen1 flap endonuclease I

g gram(s)

g relative centrifugal force

G guanine

GC-MS gas chromatography-mass spectrometry

HEPES N-2-hydroxyethylpiperazine-N-2-ethanesulfonic acid

H2O2 hydrogen peroxide

HPB 4-hydroxy-1-(3-pyridyl)-1-butanone

HPLC-ECD high-performance liquid chromatography with

electrochemical detection

hr hour(s)

i.e. id est (that is)

in vitro outside the living body

in vivo inside the living body

i.p. intraperitoneal

iso-NNAC 4-(methylnitrosamino)-4-(3-pyridyl)butyric acid

iso-NNAL 4-(methylnitrosamino)-4-(3-pyridyl)-1-butanol

kb kilobasepair

kg kilogram(s)

K-ras Kirsten-ras oncogene

Lig III DNA ligase III

A microampere(s)

Page 10: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

x

Ci microcurie(s)

g microgram(s)

L microlitre(s)

M micromolar

mol micromole(s)

M molar

min minute(s)

mg milligram(s)

7-mG 7-methylguanine

MGMT O6-methylguanine-DNA methyltransferase

mL millilitre(s)

mm millimetre(s)

mM millimolar

mRNA messenger ribonucleic acid

MUTYH adenine-DNA glycosylase

mV millivolt(s)

nA nanoampere(s)

NAB N'-nitrosoanabasine

NAT N'-nitrosoanatabine

NER nucleotide excision repair

ng nanogram(s)

nm nanometre(s)

NMWL nominal molecular weight limit

NNA 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanal

NNAL 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol

Page 11: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

xi

NNK 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone

NNN N'-nitrosonornicotine

NO2- nitrite

NO3- nitrate

NSCLC non-small cell lung cancer

1O2 singlet oxygen

O2- superoxide radical anion

OGG1 8-oxoguanine glycosylase 1

OH hydroxyl radicals

8-OHdG 8-hydroxydeoxyguanosine

O6-mG O

6-methylguanine

O4-mT O

4-methylthymine

OPB 4-oxo-4-(3-pyridyl)butanal

OPBA 4-oxo-4-(3-pyridyl)butyric acid

32P phosphorous-32

P < probability less than

P > probability greater than

p16 p16ink4a

tumour suppressor gene

PCNA proliferating cell nuclear antigen

POB pyridyloxobutyl

Pol DNA polymerase beta

RF-C replication factor C

RGS regulator of G-protein signaling

RNA ribonucleic acid

ROS reactive oxygen species

Page 12: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

xii

SCC squamous cell cancer

SCLC small cell lung cancer

SD standard deviation

SDS sodium dodecyl sulfate

SEM standard error of the mean

T thymine

TCDD 2,3,7,8-tetrachlorodibenzo-p-dioxin

TSNA tobacco specific nitrosamines

U enzymatic unit(s)

UV ultraviolet

v/v volume per volume

v/v/v volume per volume per volume

W watt(s)

WHO World Health Organization

x times

XRCC1 X-ray repair cross-complementing protein

2xYT yeast extract tryptone

~ approximately

> greater than

< less than

plus or minus

Page 13: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

1

Chapter 1

GENERAL INTRODUCTION

1.1 Statement of research problem

Lung cancer is currently the leading cause of cancer death worldwide. The widespread

use of cigarettes and tobacco products was responsible for the rise in lung cancer incidence

during the 20th century. A potent pulmonary carcinogen that most likely plays a major role in

tobacco-induced lung cancer is 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). NNK is

found in cigarettes and tobacco smoke and a substantial amount of experimental data has shown

NNK to induce lung tumours in a variety of laboratory animals. Since cigarettes are widely

available and consumed by so many, understanding the mechanism of action of major tobacco-

specific carcinogens like NNK, is critical for developing novel preventative or treatment therapies

for tobacco-induced lung cancer. The current goal of the research is to investigate the mechanism

of action of NNK and in particular; 1) to assess the extent of DNA oxidation in murine lung and

liver after in vivo treatment with NNK; 2) to determine if in vivo treatment with NNK can alter

the activity of base excision repair (BER), a major DNA repair system.

1.2 Multi-stage process of carcinogenesis

Cancer can be defined as the loss of regulatory mechanisms that manage homeostasis and

normal growth of cells. The transformation of a healthy cell into a cancerous one is dependent on

the accumulation of a number of genetic and epigenetic changes. Abnormalities in the genome

and epigenome (changes in gene expression caused by mechanisms that do not involve the DNA

sequence) can alter the way a cell functions in its microenvironment and can result in cancer.

These changes in cell function, dubbed the hallmarks of cancer, include upregulation of growth

signals and receptors, insensitivity to growth inhibition, evasion of apoptosis, limitless replication

Page 14: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

2

potential, sustained angiogenesis, tissue invasion and metastasis (Hanahan and Weinberg, 2000).

The development of cancer typically follows a multistage process and can be described by a

model involving three stages; initiation, promotion and progression.

1.2.1 Tumour initiation

The initiation stage of carcinogenesis involves the transformation of a healthy cell into a

preneoplastic cell. During initiation, the molecular structure of DNA is irreversibly altered by an

initiating agent (either chemical, physical or biological in nature) in a dose dependent manner

(Pitot et al., 1981). The resulting products are initiated cells that are difficult to detect due to

similar histology and phenotype to normal cells. Initiating agents and their metabolites introduce

non-lethal damage to the genome and/or epigenome by covalently binding to DNA, inducing

strand breaks, distorting the helix structure or modifying components such as nitrogenous bases

or sugar-phosphate backbone (Pitot et al., 1981). It is well established that the DNA binding

capacity of a carcinogen is correlated to its carcinogenic potency. Initiated cells require one

round of DNA replication for the damage to be permanent (Bertram, 2000). An initiated cell will

continue to exist and replicate unless the DNA damage is repaired or the cell is eliminated.

Abnormal alterations in DNA can be attributed to endogenous factors or exogenous

agents such as UV light, ionizing radiation, and chemical carcinogens. Endogenous damage to

DNA can arise from reactive molecular byproducts from normal cellular processes. Metabolism

of oxygen during aerobic respiration is a major source of reactive oxygen species that can oxidize

DNA and cause genetic mutations (Klaunig and Kamendulis, 2004). DNA also undergoes

frequent spontaneous damage due to the inherent instability of the DNA helix structure. For

example, in a given cell, depurination occurs 104 times a day and is caused by the breakage of the

N-glycosidic bond connecting nitrogenous base to deoxyribose sugar (Bertram, 2000). Apurinic

Page 15: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

3

sites in DNA can result in random base insertion, potentially giving rise to mutations during

replication or transcription.

Chemicals are of major importance in causing damage to DNA during the initiation stage

of carcinogenesis. It is estimated that over 80% of all cancer deaths in industrialized nations are

attributed to exogenous chemical factors found in diet, tobacco, alcohol, and occupational hazards

(Luch, 2005). Most chemical carcinogens are not themselves directly carcinogenic. A few

carcinogens like ethylene oxide, bis(chloromethyl) ether and nitrogen mustard gas are considered

“direct carcinogens” as they can directly interact with biological macromolecules without

metabolic activation (Luch, 2005). Most chemical carcinogens require in vivo metabolic

activation to form reactive electrophilic intermediates that covalently bind to nucleophilic

macromolecules including DNA, RNA and proteins (Heidelberger, 1977; Luch, 2005; Miller and

Miller, 1981). These chemicals include aromatic and heterocyclic amines, aminoazo dyes,

polyaromatic hydrocarbons, N-nitrosamines and halogenated olefins (Luch, 2005).

1.2.2 Tumour promotion

The promotion stage of carcinogenesis involves the selective clonal expansion of the

initiated cell by long term exposure to promoting agents. This stage is considered to be reversible

and dose-dependent (Klaunig and Kamendulis, 2004). Compared to initiation, direct changes to

DNA structure or genetic code are not observed during promotion (Pitot et al., 1981). Promoting

agents (including hormones, drugs, metabolites or other chemicals) bind to surface, cytoplasmic

or nuclear receptors of the initiated cell, induce changes in transcription and alter gene expression

(Pitot et al., 1981). For example, chemical compounds such as benzo[a]pyrene (BP) and 2,3,7,8-

tetrachlorodibenzo-p-dioxin (TCDD) can act as tumour promoters through arylhydrocarbon

receptor-mediated signal transduction. Both chemicals can bind arylhydrocarbon receptors in the

Page 16: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

4

cytoplasm, dimerize and translocate to the nucleus where the complex bind DNA response

elements and regulate expression of several key proteins. Genes that promote growth and

differentiation are upregulated while genes that promote apoptosis are downregulated (Luch,

2005). BP and TCDD can also alter expression patterns of cell cycle proteins such as cyclins via

arylhydrocarbon receptors. Taken together, transcriptional changes in factors that control growth,

differentiation, apoptosis and cell cycle ultimately result in the clonal expansion of the initiated

cells.

1.2.3 Tumour progression

Progression is the final stage of carcinogenesis and occurs when the preneoplastic cells

are converted into cancer cells. The progression stage is irreversible and cells undergo karyotypic

change characterized by aneuploidy and chromosomal instability that can be observed by light

microscopy (Pitot et al., 1981). A loss of genetic integrity results in increased growth rate,

increased invasiveness, and changes in biochemical and morphological characteristics that drives

formation of the primary tumour.

1.3 Lung cancer and smoking

Globally lung cancer is the leading cause of cancer death among men and second leading

cause of cancer death among women, accounting for 1.5 million new cases and 1.3 million deaths

annually (World Health Organization, 2010). In Canada, lung cancer is the leading cause of

cancer-related death and the second most diagnosed cancer among men and women (Canadian

Cancer Society, 2010). Lung cancer was diagnosed in 24, 000 Canadians and claimed over 20,

000 lives in 2010 (Canadian Cancer Society, 2010). Once diagnosed, the 5-year prognosis of

lung cancer is poor with only 10 to 15% of patients surviving (Sekido et al., 1998; Wood et al.,

2004).

Page 17: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

5

An extensive amount of epidemiologic data collected in the past 50 years has clearly

established the link between cigarette smoking and lung cancer. It is estimated that smoking and

exposure to tobacco smoke cause 85 to 90% of lung cancer cases in men and 75 to 80% of lung

cancer cases in women (Hecht, 1999; Subramanian and Govindan, 2007). Cigarette smoke is

chemically complex and currently contains over 4000 chemicals, 50 that are known carcinogens

including polycyclic aromatic hydrocarbons, aromatic amines and tobacco specific nitrosamines

(Health Canada, 2010). The particulate phase of cigarette smoke contains over 3500 compounds

including carcinogens (Hecht, 1999). Unburned tobacco contains at least 16 known carcinogens

(Hecht, 2003) and smokeless tobacco products that are rising in popularity and are perceived as a

safer alternative to cigarettes, contain 30 known carcinogens (Boffetta et al., 2008). It should be

noted that cigarettes and tobacco smoke can also increase the risk for other serious health

problems including heart disease, stroke and chronic lung disease. They have also been linked to

cancer of the larynx, oesophagus, mouth, bladder, cervix, pancreas, and kidneys (World Health

Organization, 2010).

Since the mid 1950s there has been a decline in smoking prevalence in Canada and other

developed countries like the US and UK (Giovino, 2002). In 1985, 35% of Canadians aged 15

and older were considered current smokers, but by 2008 the rate dropped to 15% (Health Canada,

2010). The decline in smoking prevalence in Canada is due to a combination of factors including

tax increases, advertising restrictions, prominent health warnings, smoking restrictions in public

places and educational campaigns over the past 60 years (Cunningham, 1996).

Even though smoking rates have declined, in recent years the drop has started to level off

suggesting that a proportion of the population (approximately 4.9 million Canadians) will

continue to smoke cigarettes despite increased efforts to curb the behaviour (Health Canada,

2010). These individuals are at an increased risk of developing lung cancer compared to former

Page 18: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

6

smokers and never-smokers (Sekido et al., 1998). In addition, individuals who are never-smokers

but are exposed to tobacco smoke from direct smoking exhalation (mainstream smoke) or an idle

burning cigarette (sidestream smoke) have an increase risk of lung cancer (Hecht, 1999). Never-

smokers are also exposed to carcinogens and particulate matter from cigarette smoke that have

been trapped in hair, skin, and fabrics (Health Canada, 2010). Former smokers are also at risk of

lung cancer as smoking cessation reduces the risk, but cancer risk will never return to levels of

never-smokers (Sekido et al., 1998). Although the decline in smoking rates in developed

countries like Canada are encouraging, the global statistics offer a stark reality of tobacco use.

The World Health Organization (WHO) has termed the global use of cigarettes an

epidemic. There are currently more than 1 billion smokers with nearly 80% of them living in low-

and middle-income countries (World Health Organization, 2010). Global tobacco consumption is

currently increasing 3.4% each year. Cigarettes are widely available and accessible with 5.5

trillion cigarettes being produced and consumed annually (World Health Organization, 2010).

More than 5 million people die per year from tobacco use and by 2030 this number is expected to

rise to 8 million people (World Health Organization, 2010). Hence, tobacco use is the leading

cause of preventable premature death. Large epidemics of lung cancer are expected in China and

India where approximately 40% of all smokers reside (Hecht, 1999). In China it is estimated that

several million cases of lung cancer will arise by 2050 based on current smoking prevalence and

population growth (Liu et al., 1998).

Although smoking and exposure to tobacco smoke are responsible for the majority of

lung cancer cases, genetic variation likely plays a role in pulmonary carcinogenesis. Only 20% of

smokers actually develop lung cancer, suggesting that some individuals may have genetic

susceptibility towards carcinogenesis (Wei and Spitz, 1997). A likely gene candidate that may

contribute to familial lung cancer and lung cancer susceptibility is RGS17 found on chromosome

Page 19: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

7

6q23-25 (You et al., 2009). The RGS17 protein is a member of the regulator of G-protein

signaling (RGS) family that can indirectly enhance or inhibit the activities of downstream

proteins (e.g. adenylate cyclase) and secondary messengers (e.g. cyclic AMP) (You et al., 2009).

Lung tumours from cancer patients have been shown to accumulate high levels of RGS17 mRNA

transcripts compared to normal lung tissue, suggesting a potential role in carcinogenesis (You et

al., 2009). Other factors such as genetic variation in an individual’s ability to metabolize

carcinogens into genotoxic intermediates, detoxify those intermediates, and repair damaged DNA

can also determine susceptibility to carcinogenesis. Polymorphisms in genes encoding proteins

that participate in such reactions are associated with lung cancer risk (Schwartz, 2004).

1.4 Human lung cancer classification

The lung is comprised of over 40 unique cell types that perform a variety of functions

such as gas exchange at the alveoli, blood-barrier at the alveolar junction, immunity against

pathogens and removal of particulate matter from the respiratory tract. In 98% of lung cancer

cases the epithelial cells lining the bronchiolar airway of the lung are transformed into cancerous

cells called carcinomas (Travis et al., 2004). Lung cancer has heterogeneous tumour biology

compared to other common cancers such as breast, colon and prostate (Borczuk et al., 2003). It is

histologically categorized into two major classes; small cell lung cancer (SCLC) and non-small

cell lung cancer (NSCLC). Other rare tumour types include sarcomatoid carcinoma, salivary

gland tumours and carcinoid tumours (Travis et al., 2004).

SCLC represent 15 to 25% of all lung cancer cases with tumours centrally located in

larger airways such as the primary or secondary bronchi (Sekido et al., 1998). Compared to

normal cells of the bronchi, SCLC cells are small and have scant cytoplasm, ill defined cell

borders, finely granular chromatin, and inconspicuous or absent nucleoli (Jackman and Johnson,

Page 20: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

8

2005). SCLC is an aggressive form of cancer that has rapid replication rate and undergoes early

metastasis (Wood et al., 2004). Although most individuals’ SCLC initially respond to

chemotherapy and radiation therapy, prognosis is poor with 3% survival over 3 years (Chute et

al., 1999). SCLC occurs in individuals with smoking history and has a strong association with

tobacco use (Hanrahan and Glisson, 2010).

Non-small cell lung cancer (NSCLC) consist of 75 to 85% of lung cancer cases and are

further divided into major subtypes; squamous cell carcinoma (SCC) (30% of all lung cancer

cases), adenocarcinoma (AC) (40%) and large cell carcinoma (9%) (Sekido et al., 1998).

Development of SCC and AC are commonly associated with smoking. SCC originate in the

central bronchus while AC are found in peripheral lung, in distal airways and alveoli of the lung

(Corvalan and Wistuba, 2010).

1.4.1 Modelling lung cancer in mouse

Lung cancers in both mice and humans share morphologic, histogenic and molecular

features. Some strains of mice are particularly more sensitive to chemical induction of lung

cancer from tobacco specific carcinogens or cigarette smoke (Meuwissen and Berns, 2005). The

A/J and SWR mice are the most sensitive mouse strains, while others range from intermediate

sensitivity (O20, BALB/c) to more resistant (CBA, C3H) and highly resistant (C57BL/6 and

DBA) (Meuwissen and Berns, 2005). Several pulmonary adenoma susceptibility loci have been

mapped to oncogenes and tumour suppressor genes and can be attributed to sensitivity between

strains of mice. For example, polymorphisms in intron 2 of the K-ras oncogene are found in most

susceptible strains of mice and are thought to induce major increases in K-ras expression as well

as alter sensitivity to respiratory carcinogens (You et al., 1993; Chen et al., 1994). The K-ras

protein is a GTPase that activates and recruits other proteins in signal transduction pathways to

Page 21: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

9

facilitate growth of cells. Polymorphisms also occur at higher frequency in sensitive mice at a

susceptibility locus on chromosome 4 that encodes the tumour suppressor gene p16ink4a

(p16)

(Zhang et al., 2002; Herzog et al., 1999). The p16 protein is critical in regulating progression in

the cell cycle and in particular G1/S cell cycle transition. Mutations in K-ras and p16 gene have

also been well documented in human SCLC and NSCLC, suggesting development of lung

cancers between mice and humans could occur in a similar fashion (Sekido 1998).

The development of AC typically involves a series of morphologically distinct

preneoplastic changes (Horio et al., 1996). They include the sequential stages of hyperplasia,

adenoma, carcinoma in situ and finally metastatic carcinoma in both mice and humans (Stoner,

1998; Malkinson, 1998). In mouse models, the induction of lung tumours with chemical

carcinogens typically results in adenomas and ACs. One way in which mice and humans differ is

that malignant AC rarely metastasize in mouse models, as mice usually die from respiratory

distress caused by carcinoma masses in lung that obstructs gas exchange (Malkinson, 1998). The

cause of death in human AC patients is usually related to consequences of metastasis (Malkinson,

1998).

1.5 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)

N-nitrosamines (referred to nitrosamines in this thesis) are present in many common

consumer items including tobacco, cosmetics, beer and bacon (Bertram, 2000). Over 200

nitrosamines and their carcinogenic activity have been confirmed in laboratory animals (Hecht,

1999). Among consumer products, cigarettes and smokeless tobacco contain levels of

carcinogenic nitrosamines 100 to 1000 times greater than other products intended for

consumption (Hecht, 2003). A major class of nitrosamines found in cigarettes are tobacco

Page 22: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

10

specific nitrosamines (TSNA) (Figure 1.1). 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone is a

nicotine derived nitrosaminoketone (NNK) and considerable research indicates that it is likely to

be an important contributor to tobacco-induced cancers. Among TSNA characterized, NNK,

NNN and NAT are found in greatest quantities in cigarettes and NNK, NNN and NNAL are the

most carcinogenic TSNA (Hecht, 1998; Hecht, 2003). Typical cigarettes contain 1 to 2 g of

NNK for every gram of unburned tobacco (Hecht, 2002). Every gram of commonly used

smokeless tobacco contains 1 to 5 g of NNK and NNN (Boffetta et al., 2008).

TSNA are formed from nitrosation of different alkaloids during curing, aging,

fermentation and burning of tobacco (Spiegelhalder and Bartsch, 1996; Brunnemann et al., 1996).

Nicotine, the addictive component of cigarettes, is the major alkaloid found in tobacco

comprising 85 to 90% of total alkaloid content (Boffetta et al., 2008). NNK, NNAL and NNN

are specifically formed when nicotine undergoes a nitrosation reaction. The formation of TSNA

is dependent on nitrosating agents, such as nitrites (NO2-), that can originate from different

sources. Nitrate (NO3-), a common constituent in tobacco, can be reduced to nitrite which can

then specifically react with tobacco alkaloids forming TSNA (Spiegelhalder and Bartsch, 1996).

The formation of some TSNA has also been shown to occur in vivo in humans and experimental

animals (Osterdahl and Slorach, 1988; Carmella et al., 1997).

1.5.1 NNK and lung cancer

NNK is a potent systemic lung carcinogen found in unburned tobacco and tobacco smoke

which induces lung tumours in commonly used rodent models regardless of route of

administration. Tumours of the respiratory system have been observed in multiple studies using

NNK-treated mice, rats and hamsters. For example, female A/J mice treated with a total dose of

Page 23: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

11

Figure 1.1 Formation of tobacco specific nitrosamines (TSNA) from major (nicotine) and minor (nornicotine, anabasine and anatabine) tobacco

alkaloid precursors (modified from Hecht, 1998). Currently, 7 TSNA have been identified including 4-(methylnitrosamino)-1-(3-pyridyl)-1-

butanone (NNK), N'-nitrosonornicotine (NNN), N'-nitrosoanabasine (NAB), N'-nitrosoanatabine (NAT), 4-(methylnitrosamino)-1-(3-pyridyl)-1-

butanol (NNAL), 4-(methylnitrosamino)-4-(3-pyridyl)-1-butanol (iso-NNAL), and 4-(methylnitrosamino)-4-(3-pyridyl)butyric acid (iso-NNAC).

With the exception of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanal (NNA), all have been detected in tobacco. Note, NNA has only been

synthesized and detected in vitro, and therefore technically is not a TSNA (Hecht et al., 1978; Hecht, 1998).

Page 24: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

12

0.11 mmol of NNK i.p., induced an average of 36.7 lung tumours per mouse (Castonguay et al.,

1983). In contrast, a similar dose of NNN and NNAL induced only 1.6 and 26.3 lung tumours

per mouse respectively. Similarly, among strain A mice treated with different TSNA, NNK

induced more lung adenomas per mouse than either NNN or NNA (Hecht et al., 1978). F344

rats treated subcutaneously with a total dose of 3.4 mmol NNK over 20 weeks developed tumours

in the nasal cavity, liver and lungs, while the same dose of NNN only gave rise to tumours in the

nasal cavity (Hecht et al., 1980). Additionally, Syrian golden hamsters treated with low dose of

NNK (5 mol) induced respiratory tract tumours in 6 of 20 animals treated (Hecht et al., 1983).

The accumulation of animal data indicate that NNK is one of the most potent pulmonary

carcinogens among TSNA identified.

NNK has organoselectivity for lung tissue in experimental animals, mainly inducing

adenomas and ACs regardless of route of administration (i.e. topically, intraperitoneally,

intravenously, subcutaneously, intravesically (into the urinary bladder), or administered through

drinking water) (Hoffmann et al., 2001; LaVoie et al., 1987; Prokopczyk et al., 1991; Lijinsky et

al., 1991). In addition to being a potent pulmonary carcinogen, treatment with NNK results in

cancers of other tissues and organs. Tumours of the nasal cavity, oral cavity and liver occur in a

variety of rodents treated with NNK (Hecht and Hoffmann, 1988; Hecht, 2003). Furthermore,

NNK and NNAL are the only known pancreatic carcinogens in unburned tobacco and tobacco

smoke. Male F344 rats treated with NNK or NNAL in drinking water significantly induced ACs

of acinar or ductal origin in the exocrine pancreas (Rivenson et al., 1988). Pancreatic lesions are

spontaneous in control animals and rarely observed (Hecht, 2003). NNK could also be

responsible for tobacco-induced cervix cancer. Levels of NNK and its metabolites were elevated

in the cervix of women who smoke (Prokopczyk et al., 1997). Cervical cells and subcellular

Page 25: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

13

fractions of the cervix have the capacity to bioactivate NNK and could generate reactive

intermediates that damage DNA (Prokopczyk et al., 2001).

NNK’s capacity to specifically induce lung adenoma and AC may be important in

tobacco-induced lung cancers in humans. The tumours induced by NNK in rodent species are

morphologically and topographically similar to human bronchiolar and bronchioalveolar AC

(Wynder and Hoffmann, 1994). Among smokers, the incidence of AC has increased over the past

5 decades, surpassing SCC as the most common form of lung cancer. The analysis of lung cancer

cases from the late 1960s to the late 1980s has shown an increase in the prevalence and rate of

AC cases compared to SCC cases (Devesa et al., 1991; Auerbach and Garfinkel, 1991). Changes

in cigarette design and smoking behaviour have likely contributed to the shift in AC. The

introduction of filter tips to cigarettes traps the particulate phase of tobacco smoke but also

impedes the delivery of nicotine into the body. To compensate for decreased levels of nicotine,

cigarette users tend to inhale more deeply, and smoke more intensely to increase pulmonary

absorption of nicotine. This practice also disperses tobacco smoke carcinogens farther into the

respiratory tract and exposes the distal bronchi and bronchioalveolar junction to relatively high

levels of smoke carcinogens, including polyaromatic hydrocarbons, volatile aldehydes and

TSNA. Reduction of nicotine levels in low tar cigarettes has also resulted in similar changes in

smoking behaviour. Furthermore, the chemical components of cigarettes have been altered and

may have contributed to increased AC. Between 1959 and 1997, tobacco blends have

incorporated more air cured tobacco, resulting in cigarettes and tobacco products that have higher

nitrate and TSNA content, including NNK, while lowering the amount of polyaromatic

hydrocarbons (Hoffmann et al., 2001). More intense smoking, deeper inhalation and increased

TSNA content in cigarettes are considered to be major contributors in increasing lung AC among

smokers (Wynder and Hoffmann, 1994).

Page 26: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

14

1.5.2 Metabolic pathways of NNK

The main NNK metabolism pathways are illustrated in Figure 1.2. To exert

carcinogenicity, NNK requires metabolic activation that generates reactive metabolites that can

adduct DNA. Bioactivation or metabolic activation refers to the process where an inactive

compound is converted into active metabolites. NNK undergoes major reactions including

carbonyl reduction, pyridine oxidation, and -hydroxylation. Denitrosation and ADP adduct

formation of NNK have only been observed in vitro using rat liver microsomes (Castonguay et

al., 1991; Peterson et al., 1994). The carbonyl reduction of NNK, catalyzed by 11 -

hydroxysteroid dehydrogenase, cytosolic carbonyl reductase and aldo-keto reductase, result in the

major metabolite NNAL (Maser, 2004). In general, the reduction of NNK to NNAL occurs in

many tissues including lung and liver. Upon NNK administration, NNAL is rapidly formed and

is the predominate metabolite in blood (Hecht, 1998). NNAL is not a final detoxified product,

since it can undergo bioactivation and has carcinogenic activity (Rivenson et al., 1988;

Castonguay et al., 1983). The reconversion of NNAL to NNK via oxidation does not occur as

extensively as reduction, however it may be of interest as it could play a role in NNAL

carcinogenicity.

The -hydroxylation reactions of NNK and NNAL are of major interest as they result in

reactive intermediates that can covalently bind DNA, RNA and proteins. Like most carcinogens,

NNK and its metabolites require metabolic activation to exert carcinogenic effect. The -

hydroxylation occurs on the methyl or methylene carbons that are adjacent to the N-nitroso group.

Hydroxylation of the methyl carbon of NNK forms 4-(hydroxymethylnitrosamino)-1-(3-pyridyl)-

1-butanone (1), that decomposes to 4-oxo-4-(3-pyridyl)-1-butanediazohydroxide (2) and

formaldehyde. Compound (2) is highly reactive and can form bulky pyridyloxobutyl (POB)

adducts on DNA, RNA and protein. Compound (2) can also react with water forming 4-hydroxy-

Page 27: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

15

Figure 1.2 Pathways of NNK metabolism. Metabolites I and II are detoxified products produced by pyridine oxidation. Metabolites III to VII are

endpoints of -hydroxylation and are generated by reactive metabolites that pyridyloxobutylate , methylate and pyridylhydroxybutylate

DNA (modified from Smith et al., 2003).

Page 28: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

16

1-(3-pyridyl)-1-butanone (keto alcohol or HPB) (III). HPB can be quantified to determine the

extent of hydroxylation at NNK’s methyl carbon.

Like NNK, NNAL also undergoes similar -hydroxylation reactions to form electrophilic

metabolites. The α-hydroxylation of the methyl carbon of NNAL forms an intermediate (3),

which decomposes to formaldehyde and the DNA pyridylhydroxybutylating species, 4-hydroxy-

4-(3-pyridyl)-1-butanediazohydroxide (4). Compound (4) can react with water to form 4-

hydroxy-1-(3-pyridyl)-1-butanol (Diol) (VI) or cyclizes to 2-(3-pyridyl)tetrahydrofuran (pyridyl-

THF) (VII).

Hydroxylation of the methylene carbon of NNK results in the formation of 4-hydroxy-4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone (5) that spontaneously decomposes to methane

diazohydroxide and 4-oxo-4-(3-pyridyl)butanal (keto aldehdye or OPB) (6). Methane

diazohydroxide is a methylating agent and interacts with DNA at several bases. OPB can be

trapped as a bisulfite adduct to quantify -methylene hydroxylation or undergo further oxidation

to form 4-oxo-4-(3-pyridyl)butyric acid (IV) (keto acid of OPBA). Hydroxylation at the

methylene carbon of NNAL (7), ultimately yields the methylating species, methane

diazohydroxide (8) and a hydroxy aldehyde which can cyclize to 5-hydroxy-2-(3-

pyridyl)tetrahydrofuran (lactol) and could be further oxidized to 4-hydroxy-4-(3-pyridyl)butyric

acid (hydroxy acid) (V).

The major pathway of detoxification of NNK and NNAL occurs via pyridine oxidation.

Pyridine N-oxidation of NNK and NNAL generates 4-(methylnitrosamino)-1-(3-pyridyl-N-

oxide)-1-butanone (NNK-N-oxide) (I) and 4-(methylnitrosamino)-1-(3-pyridyl-N-oxide)-1-

butanol (NNAL-N-oxide) (II) respectively. These N-oxide products are readily excreted through

the urine. Glucuronidation of NNAL is another important detoxification route and can occur at

the carbinol or the pyridine nitrogen of NNAL (Not shown in Figure 1.2). In addition O-

Page 29: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

17

glucuronidation of compound 2 has been observed in rats and is a potential detoxified product

(Murphy et al., 1995).

1.5.3 Cytochrome P450 enzymes and bioactivation of NNK

The metabolic activation of NNK is catalyzed mainly by cytochrome P450 (CYP)

enzymes. In particular, the CYP1, CYP2 and CYP3 families of enzyme are primarily responsible

for catalyzing oxidation of xenobiotics, generating polar metabolites that can either be excreted or

remain within the body for further biotransformation. The liver is the major site of xenobiotic

metabolism, containing an abundance of CYP enzymes that are easily accessible for study. The

respiratory tract also contains CYPs and can be a site of metabolism for therapeutics and

xenobiotics that are inhaled or in the systemic circulation. The pharmacological and toxicological

effects of agents depend on the lung’s ability to activate or deactivate these agents via

biotransformation and in fact, the carcinogenicity of NNK depends on metabolic activation in the

respiratory system. Although human lung tissue contains similar hepatic CYPs involved in

xenobiotic metabolism, low level expression of pulmonary CYPs make it difficult to fully

characterize their identity and absolute levels. The levels of pulmonary CYPs are not uniform

throughout the lung and may vary depending on cell type and region. Enzymes that likely play a

major role in NNK hydroxylation in human lung and liver include CYP1A1, 1A2, 2A6, 2A13 and

2B6 (Nademanee, 1992; Smith et al., 1995; Smith et al., 1996; Patten et al., 1996; Su et al., 2000;

Jalas et al., 2003; Jalas et al., 2005).

1.5.4 NNK and DNA adducts

The metabolic activation of NNK and NNAL results in reactive metabolites that can

methylate and pyridyloxobutylate DNA. The methane diazohydroxide metabolite generated from

NNK metabolism, methylates DNA at several bases producing 7-methylguanine (7-mG), O6-

Page 30: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

18

methylguanine (O6-mG), and O

4-methylthymine (O

4-mT) (Hecht, 1998). NNK-induced

pyridyloxobutyl (POB) adducts are bulky DNA adducts and are detected at the N7-, N

2- and O

6-

positions of guanine and O2- positions of cytosine and thymine (Jalas et al., 2005). The extent of

NNK-induced DNA methylation and POB adduction are dependent on a number of factors

including the site of hydroxylation (the methyl or methylene carbon of NNK), levels of

bioactivation and detoxification, and ability to repair DNA adducts. NNK’s ability to induce both

methyl and POB adducts may be important for carcinogenicity as most other nitrosamines just

induce one type of DNA damage. Treatment with either NNN or N-nitrosodimethylamine, agents

that can only pyridyloxobutylate or methylate DNA respectively, have lower pulmonary

carcinogenicity compared to NNK which induce both types of adducts (Hoffmann et al., 1984;

Hecht et al., 1986).

Methyl and POB DNA adducts have been detected in the lungs of human smokers. DNA

isolated from peripheral lung and tracheobronchial tissue from smokers had significantly higher

levels of POB adducts compared to non-smokers (Foiles et al., 1991; Holzle et al., 2007). The 7-

mG adduct was found at higher levels in smokers compared to non-smokers (Mustonen et al.,

1993). Similarly, O6-mG was also detected in the lungs of smokers (Wilson et al., 1989). It

should be noted that methyl and POB adducts detected in smokers could be due to reactive

metabolites generated from NNK, but also from other nitrosamines including NNN and N-

nitrosodimethylamine found in cigarettes and tobacco smoke (Hecht, 2003).

The O6-mG adduct may be of particular importance in pulmonary carcinogenesis in

mice. O6-mG is strongly correlated with tumour multiplicity in mice treated with NNK or

acetoxymethyl-methylnitrosamine, an analog of NNK that exclusively methylates DNA (Peterson

et al., 2001). In addition, O6-mG adducts that are persistent and escape DNA repair, induce

GA transition mutations in DNA. GGTGAT mutations have been found in codon 12 of the

Page 31: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

19

K-ras oncogene in a high proportion of lung AC induced by NNK in A/J mice, suggesting a

possible role of O6-mG in tumour induction (Belinsky et al., 1989). Persistent POB adducts can

also induce mutations including GA and GT in codon 12 of K-ras gene in lung tumours of

NNK-treated mice (Ronai et al., 1993). Mutations at codons 12, 13 and 61 of K-ras can result in

uncontrolled growth and potentially develop into cancers. K-ras mutations are detected in 20 to

30% of human lung ACs and 15 to 20% of all non-small cell lung cancers, with frequent

mutations at codon 12 (Sekido et al., 2003). Lung AC from smokers frequently had more K-ras

mutations compared to ACs from former smokers and never-smokers (Slebos et al., 1991; Westra

et al., 1993).

NNK has also been shown to induce epigenetic changes by altering methylation status of

the p16 tumour suppressor gene. The specific methylation of cytosine residues in CG

dinucleotides that are found near the promoter region of genes, can silence expression via

chromatin remodelling. For example, in vivo treatment with NNK in rats induced

hypermethylation in CG dinucleotides in the promoter region of the p16 gene in 94% of lung AC

(Belinsky et al., 1998). Methylation of the promoter region was associated with reduced p16

protein levels that could impair cell cycle checkpoints and potentially contribute to carcinogenesis

(Belinsky et al., 1998). In vivo treatment with NNK is likely altering the activity of DNA

methyltransferases that are responsible for methylating cytosine residues in CG dinucleotides

(Belinsky, 1998).

1.6 Base excision repair and oxidative stress

An organism’s genome is vulnerable to a wide array of DNA damaging agents of

endogenous and exogenous origin. DNA repair systems are required to maintain genomic

integrity by identifying and removing damage from the DNA helix structure to prevent

Page 32: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

20

mutagenicity and cytotoxicity. Several types of DNA repair have been identified including base

excision repair (BER), mismatch repair, nucleotide excision repair (NER) and direct reversal

repair. Each type of repair is responsible for fixing specific types of DNA damage.

The BER pathway recognizes and replaces a wide array of damaged bases from DNA. A

major function of BER is to remove nucleotides that have been oxidized by reactive oxygen

species (ROS). ROS is a collective term that can refer to a free radical oxygen species that have

an unpaired electron in its valence shell; it also refers to certain non-radicals that are either strong

oxidizing reagents or easily converted to free radicals (Wiseman and Halliwell, 1996). The major

ROS generated in biological systems include superoxide radical anions (O2-), hydroxyl radicals

( OH), peroxyl radicals (RO2 ), alkoxyl radicals (RO ), hydrogen peroxide (H2O2) and singlet

oxygen (1O2) (Wiseman and Halliwell, 1996). Production of ROS can arise from both

endogenous and exogenous sources. Different environmental agents including tobacco smoke

and air pollutants contain ROS and can generate ROS. Chemical carcinogens (such as

benzo[ ]pyrene, aflatoxin and benzene), ionizing radiation and UV light can also induce ROS

(Loft and Poulsen, 1996).

In biological systems, a major source of endogenous ROS is the complete reduction of

oxygen via the electron transport chain in mitochondria during cellular respiration.

Approximately 4 to 5% of molecular oxygen that undergoes one-electron reduction is converted

to superoxide radical anions (Klaunig and Kamendulis, 2004). Mitochondrial superoxide

dismutase, an antioxidant enzyme, can catalyze the conversion of excess superoxide radical

anions to hydrogen peroxide (Klaunig and Kamendulis, 2004) while catalase and glutathione

peroxidase convert hydrogen peroxide to water. Excess hydrogen peroxide can leak from the

Page 33: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

21

mitochondria and into the cytosol (Yu, 1994). In the presence of ferrous iron, hydrogen peroxide

can decompose to hydroxyl radicals via the Fenton reaction:

Fe2+

+ H2O2 Fe3+

+ OH + OH-

Hydroxyl radicals are also formed in a similar manner when reduced forms of other transition

metals such as copper come into contact with hydrogen peroxide. Iron and copper are

sequestered in proteins including ferritin, transferrin, caeruloplasmin and metallothionein.

Oxidative stress has been shown to release transition metals from proteins and the released metals

can then participate in Fenton reactions to produce hydroxyl radicals (Halliwell and Aruoma,

1991).

Numerous enzymes that catalyze redox reactions can also contribute to the generation of

ROS. During CYP catalyzed metabolism, the generation of ROS can arise via different

processes. Redox cycling in the presence of molecular oxygen and uncoupling can result in the

generation of superoxide anion radical and hydrogen peroxide (Klaunig and Kamendulis, 2004).

Several CYPs have the ability to generate ROS during metabolism. CYP2E1 produces a

prolonged burst of ROS near the site of substrate oxidation during ethanol metabolism (Ekstrom

and Ingelman-Sundberg, 1989). Similarly, the metabolism of phenobarbital by CYP2B can

uncouple resulting in release of superoxide radical anion (Rice et al., 1994). Other enzymes can

also produce ROS when catalyzing reactions. Organelles derived from endoplasmic reticulum

called peroxisomes are responsible for breaking down fatty acid chains and contain high levels of

oxidase enzymes that generate hydrogen peroxide (Yu, 1994). The immune response can

generate excess ROS in a process called the respiratory burst. Activated macrophages and

hepatic Kupffer cells elicit rapid and transient increase in oxygen uptake to generate superoxide

anion radicals and hydrogen peroxide (Klaunig and Kamendulis, 2004). Release of ROS is

Page 34: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

22

important for a host’s defense and results in localized tissue inflammation and induction of

genotoxicity and cytotoxicity in foreign microbes (Yu, 1994).

1.6.1 Genetic damage induced by ROS

Excess production of ROS can shift cells into an oxidative state with deleterious

consequences to DNA. In a given cell, an estimated 105 oxidative DNA lesions are formed per

day (Fraga et al., 1990). A wide variety of modifications on pyrimidine and purine bases as well

as the sugar-phosphate backbone in DNA have been observed in the oxidative state. In addition,

ROS can directly and indirectly result in single or double stranded DNA breaks and abasic sites.

Although over 20 base lesions have been identified, the most abundant lesion detected is the

hydroxylation of C-8 in guanine residues known as 8-hydroxydeoxyguanosine (8-OHdG) (Cooke

et al., 2003). Oxidation of guanine can also result in the ring opened product 2,6-diamino-4-

hydroxy-5-formamidopyrimidine. Other abundant lesions of DNA oxidation include 8-

oxoadenine, 2-hydroxyadenine, 5-hydroxyctosine, cytosine glycol and thymine glycol (Loft and

Poulsen, 1996). The type of oxidative lesion generated depends on the type of ROS interacting

with DNA. Superoxide anion radical and hydrogen peroxide have low reactivity with DNA

directly (Wiseman and Halliwell, 1996). However, hydroxyl radicals are highly reactive and can

induce almost any type of oxidized DNA modification (Loft and Poulsen, 1996). Singlet oxygen

generated by UV light has high specificity for guanine residues resulting in 8-OHdG.

If left unrepaired, oxidative DNA damage can induce miscoding mutations in DNA. The

vast majority of oxidative lesions result in base substitutions such as transition or transversion

mutations. A transition mutation is a point mutation that changes purine nucleotides into the

opposite purine (A G) or pyrimidine nucleotides into the opposite pyrimidine (C T).

Conversely, a transversion mutation refers to substitution of a pyrimidine for a purine or vice

Page 35: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

23

versa. Base deletions and insertions are seldom observed as a result of oxidized DNA (Loft and

Poulsen, 1996).

Although ROS can modify all DNA bases, modifications of GC pairs are common in the

oxidized state. Hydroxyl radicals from ionizing radiation can induce potential GCCG or AT

base pair substitutions (Loft and Poulsen, 1996). Excess singlet oxygen can react with guanine

and induce GCAT base pair substitutions (McBride et al., 1992; Epe, 1991). ROS can also

interact with the deoxynucleotide pool resulting in oxidized nucleotides that can be

misincorporated in DNA during synthesis. For example, oxidized deoxyguanosine triphosphate

(dGTP) from the nucleotide pool can misincorporate opposite adenine (Cheng et al., 1992).

8-OHdG, a major oxidative lesion, is mutagenic in bacterial and mammalian cells that

can produce a GT transversion in oncogenes and tumour suppressor genes (Klaunig and

Kamendulis, 2004; Loft and Poulsen, 1996). 8-OHdG has been shown to induce mutations in

codon 12 of the K-ras oncogene in murine fibroblast cells (Jackson, 1994). Human fibroblasts

exposed to ROS generating systems induced GT and CA transversions at codon 249 of the

p53 tumour suppressor gene, an important mutation commonly seen in human tumours (Hussain

et al., 1994). Oxidative DNA damage has also been shown to induce transformation of normal

cells. Syrian hamster embryos treated with ROS generating systems developed 8-OHdG lesions

and underwent cellular transformation (Zhang et al., 2000). Antioxidants inhibited the formation

of 8-OHdG and cellular transformation in cells that were treated with ROS.

1.6.2 NNK and DNA oxidation

NNK has been shown to induce oxidative DNA damage in rodent species. F344 rats

treated with a single i.p. dose of NNK (20 mg/rat) had significantly higher 8-OHdG levels in lung

compared to controls (Chung and Xu, 1992). Treatment of female A/J mice with multiple doses

Page 36: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

24

of NNK (0.25 mg/mouse, 3 times weekly for 3 weeks by gavage) caused significantly elevated

levels of 8-OHdG in lung and liver DNA (Chung and Xu, 1992). Similarly, female A/J mice

treated with multiple doses of NNK via gavage (0.5 mg/mouse, 3 times per week for 3 weeks) or

a single dose of NNK i.p. (2 mg/mouse) had elevated levels of 8-OHdG in lung DNA (Rosa et al.,

1998). The use of antioxidant agents in conjunction with NNK, also suggests NNK can induce

oxidative DNA damage. Concomitant treatment with (–)-epigallocatechin-3-gallate, an

antioxidant found in green teas, significantly reduced the number of lung adenomas per mouse

and 8-OHdG levels in lungs of A/J mice chronically treated with NNK via gavage (Xu et al.,

1992). In the same study, (–)-epigallocatechin-3-gallate exerted little effect on levels of O6-mG

in NNK-treated mice, indicating a potential role of oxidation in carcinogenesis (Xu et al., 1992).

Oxidation of DNA can also result in other forms of genetic damage, such as DNA strand

breaks. Human lung fibroblasts incubated with NNK resulted in a dose dependent increase in

DNA strand breaks (Weitberg and Corvese, 1993). Co-treatment with NNK and

hypoxanthine/xanthine oxidase, enzymes that generate ROS, further increased the levels of strand

breaks compared to NNK alone. Conversely, concomitant treatment with NNK and oxygen

radical scavengers such as mannitol, catalase and superoxide dismutase protected DNA from

strand breaks, suggesting NNK can potentiate its effects through ROS (Weitberg and Corvese,

1993). Treatment with NNK can also induce micronuclei in rat fibroblast skin cells in a dose

dependent manner. Formation of micronuclei occurs when DNA fails to divide equally between

daughter cells during cell division due to DNA strand breaks by exogenous agents such as

chemicals or radiation. Superoxide dismutase, an antioxidant enzyme, reduced micronuclei

formation in rat skin fibroblasts treated with NNK, suggesting involvement of NNK mediated

ROS production in DNA strand breakage (Kim and Wells, 1996).

Page 37: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

25

The mechanism by which NNK induces oxidative DNA damage has not been fully

characterized. It is hypothesized that ROS can be generated during CYP metabolism of NNK.

During the first step of oxidation, iron(III) of the protoporphyrin-IX ring of a CYP creates a bond

with triplet oxygen. The iron(III)-oxygen complex is relatively stable; however superoxide anion

radicals (O2-) can be released, resulting in the breakdown or uncoupling of the oxidation

reactions catalyzed by CYP (Meunier et al., 2004). Additional superoxide anion radicals and

electrons can also be released during electron transfers in the CYP catalytic cycle. Microsomes

containing human CYP1A1, CYP1A2, CYP2B6 and CYP3A4 have been shown to produce

superoxide anion radicals during substrate metabolism (Puntarulo and Cederbaum, 1998). A

proposed mechanism of generation of ROS during CYP metabolism of NNK is outlined in Figure

1.3. This involves uncoupling during CYP metabolism of NNK, resulting in release of

superoxide anion radical. Although the mechanism has not been fully characterized, it appears

that NNK can generate ROS that can oxidize DNA.

1.7 Base excision repair (BER) pathway

Repair of oxidative damage to the genome by BER is carried out in three major steps:

recognition and excision of the damaged DNA bases, insertion of nucleotides and finally ligation

(Figure 1.4). The first step of BER is carried out by DNA glycosylases that recognize and

remove damaged or incorrect bases by hydrolyzing the N-glycosidic bond between base and

deoxyribose sugar (Christmann et al., 2003). In human cells, 11 different glycosylases have been

identified and characterized based on the types of modified bases removed (Christmann et al.,

2003). For example, 8-oxoguanine glycosylase 1 (OGG1) recognizes and releases 8-OHdG

paired with cytosine. Adenine-DNA glycosylase (MUTYH), identifies and removes 8-OHdG

lesions that have incorrectly mispaired with adenine during DNA replication in humans

Page 38: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

26

Figure 1.3 Proposed mechanism of generation of ROS by uncoupling of CYP during NNK

metabolism (modified from Yang and Smith, 1996). During -hydroxylation of NNK, the initial

oxidation step is believed to form an -nitrosamino radical and superoxide anion at the CYP

iron(III) protoporphyrin-IX ring (initial radical species not shown in figure). In some cases both

radical species recombine, in a process called oxygen rebound to form the alcohol intermediate

(1). In other cases uncoupling could occur and release superoxide anion from the CYP active site.

The resulting -nitrosamino radical (2) could fragment into nitric oxide (3) and an imine (4).

Nitric oxide could then be oxidized to nitrite (NO2-) while the imine could be hydrolyzed to an

aldehyde (5) and methylamine (6).

Page 39: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

27

Figure 1.4 Mechanisms of BER pathway depicting short patch and long patch repair (Christmann

et al., 2003). AP: apurinic/apyrimidinic; 5’dRP: 5’-deoxyribose-5-phosphate; Polβ, Polδ/ε:

DNA polymerases; Lig I, Lig III: DNA ligases; XRCC1: X-ray repair cross-complementing

protein 1; RF-C: replication factor C; Fen1: flap endonuclease 1; PCNA: proliferating cell

nuclear antigen.

Page 40: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

28

(Cooke et al., 2003). Glycosylases have been further divided into 2 major subtypes; type I and

type II. Type I glycosylase identify and remove modified bases leaving an apurinic or

apyrimidinic site (AP) in DNA (Christmann et al., 2003). Type II glycosylases recognize and

remove damaged bases, but can also cleave the DNA phosphodiester backbone by endogenous

3’-endonuclease activity, giving rise to a single strand break (Christmann et al., 2003). In the

case of type I glycosylases, the phosphodiester backbone of DNA is cleaved by a separate AP

endonuclease that can result in 5’-deoxyribose-5-phosphate (5’dRP) and 3’-hydroxyl DNA

termini (Fortini and Dogliotti, 2007).

After removal of the damaged base, depending on the type of BER, a single nucleotide or

series of nucleotides will be inserted. In short patch repair, DNA polymerase (Pol ) exerts

lyase activity and catalyzes the release of the hemiacetal form of 5’dRP formed at the DNA

terminus of the AP site (Christmann et al., 2003). The resulting gap is filled by a single

nucleotide by Pol . DNA ligase III (Lig III) participates in short patch repair and seals the DNA

backbone (Christmann et al., 2003). Both Lig III and Pol have been shown to bind with the

scaffolding protein X-ray repair cross-complementing protein 1 (XRCC1) for added stability and

function (Caldecott et al., 1994; Robertson et al., 2009). The net result of short patch repair is

replacement of the damaged base by a single nucleotide.

In long patch repair, Pol first inserts a single nucleotide at the AP site (Fortini and

Dogliotti, 2007). After Pol dissociation, further DNA synthesis is done by DNA polymerase

and , resulting in a longer repair patch (Christmann et al., 2003). Proliferating cell nuclear

antigen (PCNA) and replication factor C (RF-C) are recruited to stabilize DNA polymerase and

during nucleotide insertion (Stucki et al., 1998). The original 5’dRP flap structure is cleaved by

flap endonuclease I (FenI) (Klungland and Lindahl, 1997). PCNA forms a complex with FenI to

Page 41: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

29

stimulate its endonuclease activity. Finally, DNA ligase I interacts with PCNA and Pol to seal

the DNA backbone (Srivastava et al., 1998; Christmann et al., 2003). The net result of long patch

repair is replacement of the damaged base by a series of nucleotides, 2 to 15 nucleotides in

length.

Other accessory proteins may have a role during BER. The addition of purified p53

protein to cell free extracts or reconstituted BER pathway stimulates BER in vitro. Conversely,

immunodepletion of p53 from cell extracts and deficient p53 cell lines have decreased BER

activity (Zhou et al., 2001; Achanta and Huang, 2004). The p53 protein has been shown to

directly bind and stabilize AP endonuclease and OGG1 glycosylase to enhance BER activity

(Achanta and Huang, 2004). Furthermore, p53 has been shown to stabilize the interaction

between Pol and DNA termini at AP sites (Zhou et al., 2001).

1.7.1 Decision between short and long patch repair in BER

The decision to undergo short or long patch BER is not fully characterized. Several

hypotheses exist attempting to elucidate the switch between short and long patch repair. The

capacity to cleave the 5’dRP terminus by Pol plays an important role in determining which

pathway BER performs. Pol can cleave the 5’dRP terminus in hemiacetal form and completes

BER via the short patch pathway. Oxidized or reduced AP sites, 3’-unsaturated aldehydes or 3’-

phosphates are resistant to Pol lyase activity and result in further processing through the long

patch repair pathway (Nakamura et al., 2000; Robertson et al., 2009; Christmann et al., 2003).

The type of DNA termini produced at the AP site depend on the oxidative DNA lesion present as

well as the glycosylase used for repair (Fortini et al., 1999). Other factors such as relative

concentration of ATP and the stage of the cell cycle may also play a role in determining whether

long or short patch BER proceeds (Petermann et al., 2003; Fortini and Dogliotti, 2007).

Page 42: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

30

Although short and long patch BER most likely occur concurrently, the degree to which

each occurs has not been fully characterized. In vitro assays using cell free extracts suggest that

repair of 8-OHdG adducts is carried out predominantly by short patch repair (Dianov et al., 1998;

Fortini et al., 1999). However, in intact cells transfected with a plasmid containing an 8-OHdG

lesion, repair patches longer than one nucleotide were observed in 55 to 80% of plasmids,

suggesting that long patch repair of 8-OHdG also plays a role in intact cell systems (Sattler et al.,

2003).

1.7.2 BER and carcinogenicity

Deficient DNA repair can result in persistent DNA damage. If replication occurs before

the lesion is repaired, a permanent change in DNA may be introduced, which in turn could

potentially contribute to carcinogenesis. Knockout mice have been used to test the effects of

deficient BER on oxidative DNA damage and cancer susceptibility. Mice deficient in major

glycosylases had no drastic changes on phenotype and marginal effects on spontaneous

tumourigenesis. For example, OGG1 knockout mice developed a significantly higher number of

spontaneous lung adenocarcinoma (AC) and accumulated higher levels of 8-OHdG lesions in

liver compared to wild type mice (Sakumi et al., 2003). Mice deficient in MUTYH glycosylase

had increased incidence of spontaneous intestinal tumours (Sakamoto et al., 2007). Knockout

mice deficient in both OGG1 and MUTYH accumulated 8-OHdG lesions in lung and small

intestine, and had increased frequency of lung and ovarian tumours and lymphomas (Xie et al.,

2004; Russo et al., 2004). Although these studies have shown moderate effect on spontaneous

tumourigenesis, exposing knockout mice to pro-oxidants can exacerbate carcinogenesis.

Treatment with the pro-oxidant dimethylarsinic acid in OGG1-deficient mice resulted in a greater

number of lung tumours (Kinoshita et al., 2007). These types of experiments demonstrate that

BER can function as a protective mechanism during carcinogenesis.

Page 43: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

31

Human cancers have also been analyzed for mutations in major proteins of the BER

pathway. Low OGG1 activity is associated with higher risk of some cancers. Squamous cell

carcinoma (SCC) of the head and neck was significantly associated with low OGG1 activity in

peripheral blood mononuclear cells (Paz-Elizur et al., 2006). Furthermore, leukocytes and

peripheral blood mononuclear cells obtained from lung cancer patients had reduced capacity to

repair 8-OHdG lesions in DNA constructs compared to healthy controls (Paz-Elizur et al., 2003).

Smokers with low OGG1 activity also had higher risk of developing lung cancer (Paz-Elizur et

al., 2003). Low OGG1 activity in cancer patients could be due to polymorphisms, differences in

expression levels, protein stability, post-translational modifications, or presence or absence of

natural activators and inhibitors.

Deficiency in the MUTYH glycosylase has been linked to rare forms of familial

colorectal cancer in humans. A British family with a history of colorectal cancer had a germline

biallelic missense mutation in the MUTYH gene that resulted in 2 variants, both with a single

amino acid substitution (David et al., 2007). An in vitro study using the human MUTYH variants

in E. coli deficient in MutY (homologue of human MUTYH), found decreased repair of 8-OHdG

mispaired with adenine compared to wild type MUTYH (Chmiel et al., 2003). Tumours from the

family contained GT transversion mutations in the tumour suppressor gene adenomatous

polyposis coli, likely due to the accumulation of oxidative DNA lesions that could not be repaired

as a result of deficient BER (David et al., 2007). Thus far, this is the only BER germ line

mutation to be directly implicated in carcinogenesis (Paz-Elizur et al., 2008).

While deficient BER can result in elevated oxidative DNA damage and incidence of

cancer, conflicting evidence suggests that the mere presence of oxidative DNA damage may not

be necessary or sufficient to induce carcinogenesis in vivo (Cooke et al., 2003). Elevated

oxidative DNA damage is seen in several human pathological conditions (e.g. rheumatoid

Page 44: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

32

arthritis, systemic lupus, and neurodegenerative conditions) without increase in cancer

susceptibility (Cooke et al., 2003) and glycosylase knockout mice did not show increased

incidence of spontaneous carcinogenesis in several organ systems (Klungland et al., 1999;

Minowa et al., 2000; Paz-Elizur et al., 2008). Other DNA repair systems and redundant

glycosylases may be able to mitigate the effect of deficient BER. Mismatch repair and nucleotide

excision repair have the limited ability to repair 8-OHdG adducts which could act as a backup

repair mechanism in glycosylase knockout mice (Colussi et al., 2002; Reardon et al., 1997).

Epidemiologic studies have also shown polymorphisms of major BER genes (OGG1 glycosylase,

AP endonuclease, XRCC1, etc.) are weakly associated with cancer and may only play a minor

role in carcinogenesis in humans (Hung et al., 2005a; Hung et al., 2005b; Vogel et al., 2004; Paz-

Elizur et al., 2008). Further studies need to be conducted to elucidate the specific contribution of

BER and oxidative DNA damage in different organ systems of experimental animals and humans,

particularly during oxidative and carcinogenic challenge.

1.8 NNK and DNA repair systems

Studies on the effects of NNK on DNA repair systems are limited. To date, only 2 types

of DNA repair have been shown to be affected by treatment with NNK; nucleotide excision

repair (NER) and direct reversal repair. The NER pathway recognizes and removes bulky

chemical adducts that can induce helix distortion in DNA. NER is also responsible for repairing

cyclobutane pyrimidine dimers and photoproducts, two major kinds of lesions generated from UV

light (de Laat et al., 1999). Using an in vitro NER assay, mice treated with 10 mol of NNK i.p.,

had reduced activity for NER of POB adducted plasmid in extracts prepared from lung tissue

(Brown and Massey, 2009). Interestingly, the same NNK treatment increased NER in liver

extracts, suggesting inter-organ differences in susceptibility of NNK tumourigenicity.

Page 45: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

33

Immunoblots showed an alteration in levels of key NER proteins, indicating that treatment with

NNK can also alter protein levels of NER.

Direct reversal repair is the direct removal of a specific DNA adduct by a specialized

class of proteins (Lindahl and Wood, 1999). For example, O6-methylguanine-DNA

methyltransferase (MGMT) irreversibly binds and removes alkyl groups from O6-alkylguanine.

In vivo treatment with NNK reduced MGMT activity in different lung cell types including type II

cells and non-ciliated bronchiolar epithelial cells (Belinsky et al., 1988). In addition, MGMT

activity was inhibited in cultured rat hepatocytes by OPB, a reactive intermediate generated by α-

hydroxylation of NNK (Liu et al., 1992). The decline of MGMT activity could be due to direct

adduct formation on the repair protein by reactive NNK metabolites (Mijal et al., 2004).

Treatment with NNK could also induce alkyl or POB adducts in DNA which can act as

competitive substrates during MGMT repair (Peterson et al., 2001). Some bulky POB adducts

like O6-[4-oxo-4-(3-pyridyl)-butyl]guanine can compete with O

6-alkylguanine for repair by

MGMT, resulting in reduced removal of O6-alkyl guanine by MGMT (Wang et al., 1997;

Peterson et al., 2001). These results demonstrate the possibility for NNK to alter the activity and

levels of protein of DNA repair pathways in lung and liver.

With respect to BER, in vivo treatment with NNK has been studied in female OGG1

knockout mice (Igarashi et al., 2009). This study found NNK-treated knockout mice had

significantly higher incidence and multiplicity of lung tumours compared to NNK-treated wild

type mice. Antioxidants inhibited enhanced lung carcinogenesis in NNK-treated knockouts and

NNK-treated wild type mice. These results suggest a potential role for an intact BER pathway in

protecting against NNK-induced pulmonary carcinogenesis.

Page 46: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

34

1.9 Research hypothesis and objectives

Lung cancer is the leading cause of cancer death worldwide. Although cigarettes are by

far the largest cause of lung cancer cases among men and women, the mechanism by which

cigarettes induce pulmonary carcinogenesis are not fully characterized. A potent pulmonary

carcinogen found at significant levels in tobacco products is the tobacco specific nitrosamine,

NNK. Treatment with NNK induced lung ACs in a variety of animal species. The metabolism of

NNK by CYP enzymes results in the formation of reactive intermediates that can methylate and

pyridyloxobutylate DNA and induce damage in major tumour suppressor genes and oncogenes.

In addition, NNK has the potential to induce oxidative DNA damage. DNA repair systems are

critical for protecting genomic integrity from chemical agents like NNK. Alterations in repair

activities can result in persistent DNA lesions that can potentially exacerbate carcinogenesis. The

overall objective of this research was to determine the ability of NNK to cause DNA oxidation in

an established mouse model of NNK lung tumourigenicity, as well as to assess potential effects

of NNK on BER activity in murine lung and liver. Treatment with NNK has been shown to alter

NER and MGMT repair systems and may have a similar effect on BER.

Hypothesis 1: In vivo treatment with NNK induces oxidative DNA damage in murine lung and

liver.

Objective 1: Determine the effect of in vivo treatment with NNK on levels of 8-OHdG in A/J

mouse lung and liver.

Hypothesis 2: In vivo treatment with NNK alters BER activity in murine lung and liver.

Objective 2: Determine the effect of in vivo treatment with NNK on overall BER activity in A/J

mouse lung and liver.

Page 47: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

35

Chapter 2

MATERIALS AND METHODS

2.1 Reagents

To evaluate oxidative DNA damage, chemicals and reagents were obtained as follows:

NNK from Toronto Research Chemicals (North York, ON); 8-OHdG, 2’-deoxyguanosine (2’-

dG), alkaline phosphatase, nuclease S1, nuclease P1 from Sigma (St. Louis MO); Ultrafree-MC

10,000 nominal molecular weight limit (NMWL) filter units from Millipore (Bedford, MA);

Wako DNA Extractor TIS kit from Wako Chemicals USA, Inc. (Richmond, VA). To assess BER

activity, chemicals and reagents were obtained as follows: proteinase K from Sigma (St. Louis,

Mo); Slide-A-Lyzer dialysis cassettes 3500 molecular weight cutoff from Thermo Scientific

(Rockford IL); EcoR1 from New England Biolabs (Beverley, MA); [32

P] dGTP from Perkin

Elmer (Waltham, MA). All other chemicals were reagent grade and were obtained from common

commercial suppliers.

2.2 Animal Treatments

Female A/J mice, aged 7 to 8 weeks (Jackson Laboratories, Bar Harbour, ME) were

housed with a 12 hour light/dark cycle and provided food and water ad libitum. Mice were

treated with a single tumourigenic dose of 10 mol NNK (~118 mg/kg NNK for mice that had an

average weight of ~17 g) in saline (0.1 mL, i.p.) or 0.9% saline control (0.1 mL, i.p.). This NNK

treatment protocol has been shown to induce 7 to 12 lung adenomas per A/J mouse 16 weeks later

(Hecht et al., 1989). To avoid the use of anesthetics that could interfere with biochemical

processes in lung or liver, mice were sacrificed by cervical dislocation 1, 2 or 24 hours following

NNK administration. The time points were selected to assess BER activity and levels of

oxidative DNA damage since NNK can induce substantial DNA damage in lung after the first

Page 48: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

36

few hours after NNK administration (Peterson and Hecht, 1991; Chung and Xu, 1992; Tjaelve,

1991). Lungs and livers were perfused with TE buffer (10 mM Tris, 1 mM EDTA, pH 7.9) and

excised. Organs were snap frozen in liquid nitrogen and stored at –80oC until nuclear extract

preparation or DNA isolation. For each time point, 20 mice were treated with NNK and another

20 mice with saline. To determine BER activity, 4 sets of lungs were pooled together to produce

1 lung nuclear protein extract while 1 liver was used to produce 1 liver nuclear protein extract.

To assess oxidative DNA damage, 1 set of lungs and 1 liver were required. A total of 120 mice

were required for conducting 4 independent experiments at 1, 2 and 24 hour time points.

2.3 Isolation of DNA from lung and liver

DNA was isolated from lung and liver tissue using a Wako DNA TIS Extractor kit,

according to the manufacturer’s recommendations with minor modifications: 0.1 mM

deferoxamine mesylate (DFO) was added to the lysis solution, enzyme reaction solution, and TE

buffer, while 0.3 mM DFO was added to the sodium iodide solution. DFO is an iron-chelating

agent that prevents Fenton reactions and subsequent production of hydroxyl radicals that could

artifactually oxidize DNA during sample preparation (Ravanat, 2005b). Briefly, lung and liver

tissue (~0.15 g and ~0.6 g respectively) was pulverized into powder using a mortar and pestle in

liquid nitrogen. Lysis solution was added to the tissue powder and homogenized to disrupt

cellular membranes. The whole tissue lysate was centrifuged (600 x g for 10 minutes at 4oC) and

RNAse, proteinase and an unspecified oxidation inhibitor were added and incubated at 37oC for 1

hour. The lysate was centrifuged (10,000 x g for 5 minutes at 4oC) and the supernatant kept.

DNA was precipitated with 300 L sodium iodide solution and washed with alcohol solutions.

The resulting DNA was dried and dissolved in TE buffer containing 0.1 mM DFO.

Concentration and purity of DNA were determined by spectrophotometry, using absorbance at

Page 49: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

37

260 nm and 260 nm/280 nm absorbance ratio respectively. For purity of DNA, absorbance ratios

between 1.8 and 2.0 were accepted. DNA was stored at –80oC until digestion steps.

2.4 DNA digestion

Thorough digestion of genomic DNA is necessary for accurate measurement of genomic

8-OHdG levels (Huang et al., 2001). Calf thymus DNA and pBluescript SK+ plasmid was used

to optimize DNA digestion to ensure accuracy in the measurement of 8-OHdG levels. Nuclease

P1 and S1 enzymes and incubation times were tested with calf thymus DNA, and selected based

on which combinations produced the lowest 8-OHdG levels (data not shown). Preliminary results

suggested that DNA digestion with nuclease P1 had the lowest levels of 8-OHdG. Incubating calf

thymus DNA with nuclease P1 and alkaline phosphatase for different incubation times (ranging

from total times of 15 minutes to 2 hours) did not seem to have an effect on lowering 8-OHdG

levels. The final digestion method was performed using the method described by Lan et al. with

minor modifications (Lan et al., 2000). Briefly, DNA was diluted with TE buffer containing 0.1

mM DFO to a final concentration of 0.15 mg/mL in 100 L volume. Ten L of 0.5 M sodium

acetate (pH 5.1) and 1 L of 1 M MgCl2 were added to the DNA. To denature genomic DNA,

samples were heated at 100oC for 5 minutes and immediately placed on ice for 5 minutes. One

L of nuclease P1 (4 mg/mL) was added to DNA and incubated at 37oC for 1 hour. Solution pH

was adjusted to 7.8 by adding 1.25 L of 1 M Tris (pH 10.5). Porcine alkaline phosphatase was

prepared by dissolving in water with 0.1 mM DFO (0.1 U/ L). Ten L of alkaline phosphatase

was added to the sample and incubated at 37oC for 1 hour. Enzymes were precipitated by adding

2 L of 5.8 M acetic acid. Samples were transferred to 10,000 NMWL filter units and

centrifuged (12,000 x g for 20 minutes at 4oC), separating digested nucleosides from enzymes.

Page 50: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

38

Samples were stored at –80oC until analysis. Gel electrophoresis was performed to check for

comprehensive DNA digestion.

2.5 Determination of 8-OHdG and 2’-dG levels by high-performance liquid

chromatography with electrochemical detection

To assess the levels of 8-OHdG and 2’-dG in genomic DNA, high-performance liquid

chromatography with multichannel electrochemical detector (HPLC-ECD) (ESA Inc.,

Chelmsford, MA) was used. Compounds were separated on a Waters S-3 4.6 150 mm column

with 0.1% methanol/99.9% 50 mM phosphate buffer (pH 5.5) at a flow rate of 1.0 mL/min (Bolin

et al., 2004; Guindon et al., 2007). The four electrochemical detector channels were set to 0, 250,

475 and 875 mV. The ratio of 8-OHdG to 2’-dG was determined by comparing the concentration

of each nucleoside in a digested DNA sample. The concentration of 8-OHdG and 2’-dG was

quantified by determining the area under the curve (Figure 2.1) and interpolated to standard

curves of 8-OHdG and 2’-dG. It should be noted that DNA isolation, digestion and HPLC

analysis was carried out in one week, in order to prevent prolonged storage and artifactual

oxidation (Kasai, 1997).

2.6 Cell-free whole tissue nuclear protein extract preparation

Cell-free whole tissue nuclear protein extracts were prepared from ~1.0 to 1.5 g of total

tissue from livers of individual mice and from pooled lungs of 4 mice. These extracts, active in

DNA repair synthesis, were prepared as described previously (Wood et al., 1988; Wood et al.,

1995; Bedard et al., 2005). Extracts were stored at –80oC until repair reactions were carried out.

Protein concentration of extracts were determined by the Lowry assay using bovine serum

albumin as a standard (Lowry et al., 1951). Extracts from lung and liver typically contained ~5

and ~15 mg/mL protein, respectively.

Page 51: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

39

Figure 2.1 Chromatograms demonstrating resolution of A) 2’-dG and B) 8-OHdG, isolated from

lung DNA of mice following in vivo treatment with saline. Isocratic chromatography with

electrochemical detection was performed using a Waters S-3 4.6 x 150 mm column and a mobile

phase of 0.1% methanol/99.9% 50 mM phosphate buffer (v/v) (pH 5.5) at 1.0 mL/min at room

temperature. The nucleoside 2’-dG was detected at 875 mV and 8-OHdG at 250mV. The area

under the curve was calculated to determine the concentration of 2’-dG and 8-OHdG in samples.

A = microampere; nA = nanoampere; 2’-dG = 2’-deoxyguanosine; 8-OHdG = 8-

hydroxydeoxyguanosine.

Page 52: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

40

2.7 Preparation of 8-OHdG adducted plasmid

A 2961 bp plasmid derived from pBluescript SK+ (Stratagene, La Jolla, CA) was grown

in E. Coli DH5 cells in 2xYT broth and isolated using a Qiagen Plasmid Mega Kit (Qiagen,

Valencia, CA), as per manufacturer’s instructions with minor modifications. Instead of growing

E. Coli in 500 mL liquid culture, a total of 1 L was grown in 2xYT liquid culture and eventually

applied to a single anion-exchange column to isolate plasmid. The method to prepare DNA

containing 8-OHdG adducts was adapted from Floyd et al. and Schneider et al. (Floyd et al.,

1989; Schneider et al., 1990). The BER pathway can repair 8-OHdG adducts and its activity can

be assessed using the in vitro DNA repair assay. Briefly, 50 L of pBluescript SK+ plasmid (0.4

mg/mL in TE buffer) was placed on a parafilm-covered petri dish on ice. In the dark, 50 L of

methylene blue was added to the plasmid and exposed to white light from a 100 W tungsten lamp

at a distance of 10 cm for 5 minutes. When irradiated with white light, methylene blue generates

singlet oxygen that specifically reacts with guanine residues resulting in 8-OHdG adducts

(Schneider et al., 1990). A range of concentrations of methylene blue (final concentration of 1, 2,

5, 10 and 20 M) was used to optimize the production of oxidized plasmid. Immediately after

light exposure, irradiated DNA was precipitated with 20 L of 5 M NaCl and 300 L 100%

ethanol. DNA was washed with 500 L of 80% ethanol twice, dried down and re-dissolved in TE

buffer. To determine the extent of 8-OHdG adduction, plasmid was thoroughly digested and

subjected to HPLC-ECD. Gel electrophoresis was performed on a 1% agarose gel to determine

extent of nicking damage in methylene blue damaged plasmid. DNA densitometry of the

photograph of the UV-illuminated ethidium bromide gel (ChemImager 4000, Alpha Innotech

Corporation, San Leandro, CA) was performed to quantify the percentage of nicked plasmid (~4

kb band) and intact supercoiled plasmid (~2 kb band) used in the in vitro repair assay.

Page 53: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

41

2.8 In vitro base excision repair assay

The repair synthesis assay (Figure 2.2) was performed as described (Wood et al., 1988;

Wood et al., 1995; Bedard et al., 2005) with slight modifications. The 50 L reaction mixture

contained 400 ng of 8-OHdG damaged plasmid DNA or plasmid containing baseline DNA

oxidation, 40 mM HEPES-KOH (pH 7.8), 0.5 mM dithiothreitol, 5.0 μM dGTP, 20 M of each

dATP, dCTP and dTTP, 23 mM phosphocreatine, 18 g bovine serum albumin (nuclease-free,

Sigma Aldrich, St. Louis, MO), 2.5 g creatine phosphokinase, 2.0 mM ATP, 5.0 mM MgCl2, 0.4

mM EDTA, 100 mM KCl, 7.1 Ci [α32

P] dGTP, and 10 g of lung or 25 g of liver protein

extract (concentrations listed are final concentrations). Untreated plasmid was used as a negative

control and contained background levels of DNA oxidation. Samples were incubated for 3 hours

at 30°C unless otherwise indicated. Reactions were terminated by adding EDTA (final

concentration, 20 mM). Following incubation with 7.3 mg/mL RNase (final concentration, at

37°C for 10 minutes) and 925 g/mL proteinase K with 0.5% SDS (final concentration, at 65°C

for 30 minutes), plasmid DNA was purified by extraction with 250 L of

phenol/chloroform/isoamyl alcohol (25:24:1; v/v/v), precipitated with 1 mL of 100% ice-cold

ethanol, washed in 1 mL of 80% ethanol, dried and linearized with 1 U of EcoR1. Gel

electrophoresis was performed with the linearized plasmid on a 1% agarose gel. Plasmid DNA

recovery was normalized by densitometry of the photograph of the UV-illuminated ethidium

bromide gel (ChemImager 4000, Alpha Innotech Corporation, San Leandro, CA). The extent of

[α32

P] dGTP incorporation was determined by phosphor imaging (Cyclone Plus Phosphor Imager,

Perkin Elmer, Waltham, MA) of the dried gel (Figure 2.3). To determine damage-specific BER

activity in lung and liver of saline control and NNK-treated mice, repair synthesis activity found

in untreated plasmid containing baseline levels of oxidation was subtracted from repair synthesis

Page 54: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

42

Figure 2.2 Schematic of in vitro DNA repair synthesis assay on damaged plasmid DNA by

nuclear extracts isolated from whole tissue. Squares on plasmid represent oxidative DNA lesions.

8-OHdG: 8-hydroxydeoxyguanosine; dGTP: deoxyguanosine triphosphate; EtBr: ethidium

bromide (modified from Salles et al., 1995).

Page 55: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

43

Figure 2.3 UV-illuminated ethidium bromide stained agarose gel of linearized plasmid DNA and

phosphor image demonstrating the ability of cell-free nuclear protein extracts to catalyze in vitro

DNA repair synthesis. DNA in lanes 1 through 4 were incubated with nuclear protein from saline

control lung. Lanes 1 and 2 are replicates and demonstrate background repair synthesis activity

towards untreated plasmid DNA that contains background level of oxidation. Lanes 3 and 4 are

replicates and show repair of 8-OHdG adducted plasmid.

1 2 3 4

Untreated plasmid + + - -

8-OHdG damaged plasmid - - + +

3 kb

2 kb

1kb

Page 56: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

44

activity found in plasmid containing oxidative DNA damage. The in vitro repair assay was

performed in duplicate for each protein extract.

In addition, to optimize the in vitro repair assay, nuclear protein extracts from lungs and

livers of control female A/J mice were prepared and tested. The level of oxidative damage in

plasmid substrate, protein amount and incubation time were investigated to determine the linear

range of each parameter in the in vitro repair assay. Each specific parameter of the BER assay

was tested using separate aliquots from a single liver extract (prepared from 1 animal) or a single

lung extract (prepared from 4 animals).

2.9 Data analysis

Statistically significant differences in levels of DNA oxidation and BER activity between

NNK-treated and control groups were determined by unpaired Student’s t-test (GraphPad Prism 5

software). If heterogeneity of variance was present, data were transformed using the log-log

function prior to statistical analysis. P < 0.05 was considered significant in all cases.

Page 57: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

45

Chapter 3

RESULTS

3.1 Levels of 8-OHdG following in vivo treatment with NNK

The background levels of 8-OHdG in calf thymus DNA and freshly isolated pBluescript

SK+ plasmid were on average 21.5 and 10.1 8-OHdG/105 2’-dG respectively. Gel

electrophoresis revealed thorough digestion of calf thymus DNA and plasmid.

DNA oxidation was assessed in lungs of NNK-treated and saline control mice at each

time point (Figure 3.1). No changes in 8-OHdG levels were observed between control mice (19.3

± 3.9 8-OHdG/105 2’-dG) and NNK-treated mice (19.5 ± 3.3 8-OHdG/10

5 2’-dG) at the 1 hour

time point (n=4, P = 0.928). At the 2 hour time point, there was an apparent 14% increase in 8-

OHdG levels in the lungs of NNK-treated mice compared to control lungs; however, this was not

statistically significant (n=4, P = 0.238). Lungs of control mice appeared to have a 15% increase

in 8-OHdG levels compared to treated mice at 24 hours, but statistical significance was not

attained (n=4, P = 0.119).

In liver, the levels of 8-OHdG were not statistically different between NNK-treated mice

and control mice at any of the time points examined (Figure 3.2). An apparent 16% increase in 8-

OHdG levels was observed in NNK-treated mice compared to controls at 1 hour, but was not

statistically significant (n=4, P = 0.253). There appeared to be a 27% increase in 8-OHdG levels

in NNK-treated mice compared to control mice at 2 hours; however this was not statistically

significant (n=4, P = 0.166). At 24 hours, control mice (24.8 ± 6.7 8-OHdG/105 2’-dG) had

similar levels of 8-OHdG compared to mice treated with NNK (23.9 ± 6.7 8-OHdG/105 2’-dG)

and was not statistically significant (n=4, P = 0.864).

Page 58: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

46

Lung 1 hr

Saline NNK

0

10

20

30

40

A

Ratio 8

-OH

dG

/10

5 2

'-dG

Lung 2 hr

Saline NNK

0

10

20

30

40

B

Ratio 8

-OH

dG

/10

5 2

'-dG

Lung 24 hr

Saline NNK

0

10

20

30

40

C

Ratio 8

-OH

dG

/10

5 2

'-dG

Figure 3.1 Effect of in vivo treatment with NNK on 8-OHdG levels in lung DNA. DNA was

isolated from mice treated with 10 mol NNK or saline and sacrificed 1 (A), 2 (B) or 24 (C) hr

post-treatment. Results are presented as mean SD of 4 animals performed in triplicate and are

expressed as the ratio of the concentration of 8-OHdG to 2’-dG. No statistically significant

differences were found between NNK and saline control (P > 0.05, Student’s t-test).

Page 59: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

47

Liver 1 hr

Saline NNK

0

10

20

30

40

A

Ratio 8

-OH

dG

/10

5 2

'-dG

Liver 2 hr

Saline NNK

0

10

20

30

40

B

Ratio 8

-OH

dG

/10

5 2

'-dG

Liver 24 hr

Saline NNK

0

10

20

30

40

C

Ratio 8

-OH

dG

/10

5 2

'-dG

Figure 3.2 Effect of in vivo treatment with NNK on 8-OHdG levels in liver DNA. DNA was

isolated from mice treated with 10 mol NNK or saline and sacrificed 1 (A), 2 (B) or 24 (C) hr

post-treatment. Results are presented as mean SD of 4 animals performed in triplicate and are

expressed as the ratio of the concentration of 8-OHdG to 2’-dG. No statistically significant

differences were found between NNK and saline control (P > 0.05, Student’s t-test).

Page 60: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

48

3.2 Characterization of 8-OHdG adducted plasmid DNA substrate for in vitro repair

assay

Photoactivated methylene blue was used to induce 8-OHdG lesions in pBluescript SK+

plasmid (Figure 3.3 A). An increase in 8-OHdG levels was observed between untreated plasmid

and plasmid treated with 1, 2, 5, 10 and 20 M of methylene blue. Untreated plasmid contained a

baseline level of oxidation of 16.1 3.6 8-OHdG/105 2’-dG (n=4, mean SEM), while a

maximum concentration of 20 M methylene blue induced an average of 167 16.4 8-OHdG/105

2’-dG (n=4, mean SEM). Plasmid was also assessed for nicking using gel electrophoresis and

DNA densitometry (Figure 3.3 B). An increase in nicked plasmid was observed when increasing

concentrations of methylene blue was used to induce oxidative DNA damage.

To choose an appropriate substrate for the in vitro repair assay, untreated plasmid and

plasmid treated with 1, 2, 5, 10 and 20 M of methylene blue were incubated with nuclear protein

extracts prepared from control lung and liver to determine extent of repair activity (Figure 3.4).

Oxidative DNA damage in the form of 8-OHdG ranged from ~16 8-OHdG/105 2’-dG lesions in

untreated plasmid to ~170 8-OHdG/105 2’-dG in plasmid treated with 20 M methylene blue.

BER was assessed by normalizing the amount of [32

P] dGTP incorporated to plasmid recovery

( g). As anticipated, incubation of 10 g of lung extract with untreated plasmid containing

baseline oxidation resulted in the lowest levels of BER activity (Figure 3.4 A). Plasmid

containing higher levels of 8-OHdG had increased levels of repair activity compared to untreated

plasmid. A pronounced plateau in repair activity was observed in plasmid containing ~75 to

~175 8-OHdG/105 2’-dG. The incubation of 25 g liver extract with untreated plasmid also

produced the lowest levels of repair activity (Figure 3.4 B). An increase in repair activity was

observed in plasmid containing up to ~75 8-OHdG/105 2’-dG and seemed to plateau at higher

levels of oxidative DNA damage.

Page 61: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

49

0 5 10 15 20 250

50

100

150

200

A

Methylene Blue ( M)

8-O

HdG

/10

5 2

'-dG

0 5 10 15 20 250

10

20

30

40

B

% o

f n

icke

d p

lasm

id

Methylene Blue ( M)

Figure 3.3 Characteristics of 8-OHdG adducted pBluescript SK+ plasmid by photo-oxidation of

methylene blue. Plasmid was grown in and isolated from E. coli DH5 cells. Levels of 8-OHdG

are presented as mean SEM of four independent experiments performed in duplicate and are

expressed as the ratio of the concentration of 8-OHdG to 2’-dG (A). The percentage of nicked

plasmid are expressed as mean SEM of three or four independent experiments (B).

Page 62: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

50

0 25 50 75 100 125 150 175 2000

1000

2000

3000

4000

5000

A Lung, 10 g

8-OHdG/105 2'-dG

am

ol[

32P

]dG

TP

/g D

NA

0 25 50 75 100 125 150 175 2000

2000

4000

6000

8000

10000

12000

B Liver, 25 g

8-OHdG/105 2'-dG

am

ol[

32P

]dG

TP

/g D

NA

Figure 3.4 Optimization of 8-OHdG adducted plasmid for in vitro BER assay. Protein extract

from control lung (10 g, A) or liver (25 g, B) was incubated with plasmid containing varying

amounts of 8-OHdG for 3 hr. Results are presented as mean of duplicates and are expressed as

amol[32

P] dGTP incorporated per g of DNA. Plasmid treated with 5 M methylene blue induced

~40 8-OHdG/105 2’-dG and was chosen to assess NNK’s effect on BER in lung and liver.

Page 63: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

51

Plasmid treated with 5 M methylene blue was selected to assess the effect of NNK on

repair activity in both lung and liver. With this concentration of methylene blue, repair activity

was ~5-fold higher compared to that in untreated plasmid in repair reactions containing lung or

liver extract. This plasmid substrate contained ~40 8-OHdG/105 2’-dG (Figure 3.3) and was used

in subsequent in vitro repair assays.

3.3 Optimization of nuclear protein extract amount for in vitro repair assay

Saline control lung and liver extract was tested with the in vitro repair assay to select an

optimal protein amount to assess BER activity. A range of concentrations of both nuclear protein

extracts were incubated with plasmid treated with 5 M methylene blue for 3 hours. Repair

reactions containing neither lung nor liver extracts resulted in no incorporation of [32

P] dGTP

and thus no BER activity. Incubation reactions containing a maximum of 25 g of lung extract

resulted in an increase in repair activity (Figure 3.5 A). BER activity reached a plateau in

reactions incubated with higher amounts of lung extract. Similarly, repair reactions containing a

maximum of 50 g of liver extract appeared to increase BER activity in a linear fashion (Figure

3.5 B). Higher amounts of liver extract resulted in a plateau of BER activity. Ten g of lung

extract and 25 g of liver extract were chosen to assess BER activity in organs of control and

NNK-treated mice as increases and decreases in BER activity were detectable with these amounts

of protein.

3.4 Optimization of incubation time for in vitro repair assay

To select an optimal incubation time for the repair assay, repair activity was assessed

over time with control lung and liver nuclear protein extracts. Repair reactions contained plasmid

oxidized with 5 M methylene blue and 10 g of lung extract or 25 g of liver extract. An

Page 64: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

52

0 10 20 30 40 50 60 70 800

2000

4000

6000

8000

10000

12000

LungA

Protein ( g)

am

ol[

32P

]dG

TP

/g D

NA

0 10 20 30 40 50 60 70 800

2000

4000

6000

8000

10000

12000

LiverB

Protein ( g)

am

ol[

32P

]dG

TP

/g D

NA

Figure 3.5 Optimization of protein amount for in vitro BER assay. Varying amounts of protein

extract from control lung (A) or liver (B) were incubated with plasmid damaged by 5 M

methylene blue for 3 hr. This parameter was tested using a single lung extract (A) or separate

aliquots from a single liver extract (B). Results are presented as mean of duplicates (A) or mean

SEM of three independent experiments performed in duplicate (B). Ten g of lung protein and 25

g of liver protein was chosen to assess the effect of NNK on BER.

Page 65: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

53

increase in repair activity was observed in repair reactions containing 10 g of lung extract or 25

g of liver extract across all incubation times (Figure 3.6 A and B). The levels of BER activity

appeared to be linear at a maximum incubation time of 4 hours in both lung and liver. An

incubation time of 3 hours was picked to assess BER in lung and liver of control and NNK-

treated mice.

3.5 Effect of in vivo treatment of mice with NNK on BER in lung and liver

Damage-specific BER activity was assessed in lung and liver of mice following in vivo

treatment with NNK using the optimized parameters. No statistically significant differences were

observed between lung extracts prepared from NNK-treated mice and control mice at each time

point (Figure 3.7). At the 1 hour time point, mice treated with NNK appeared to have a 34%

reduction in BER activity in lung tissue compared to controls, however statistical significance

was not attained (n=4, P = 0.114). An apparent 13% decrease in BER activity was observed in

lungs of NNK-treated mice at 2 hours, but was not statistically significant (n=4, P = 0.535). At

24 hours lungs of NNK-treated mice appeared to have a 21% decline in repair activity, but was

not statistically significant (n=4, P = 0.146).

In liver, no statistically significant differences were observed at the 1 hour time point as

repair activities of NNK treatment and control groups were similar (n=4, P = 0.818) (Figure 3.8).

At 2 hours NNK-treated mice had an apparent 16% reduction in BER activity in liver; however

this was not statistically significant (n=3 or 4, P = 0.163). BER activity was slightly reduced in

livers of NNK-treated mice at the 24 hour time point but statistical significance was not attained

(n=4, P = 0.675).

Page 66: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

54

Lung

0 1 2 3 4 50

1000

2000

3000

A

Time (hr)

am

ol[

32P

]dG

TP

/g D

NA

Liver

0 1 2 3 4 50

2000

4000

6000

8000

B

Time (hr)

am

ol[

32P

]dG

TP

/g D

NA

Figure 3.6 Optimization of incubation time for in vitro BER assay. Protein extracts from control

lung (10 g, A) or liver (25 g, B) were incubated for varying amounts of time with plasmid

damaged by 5 M methylene blue. This parameter was tested using a single lung extract (A) or

separate aliquots from a single liver extract (B). Results are presented as mean of duplicates (A)

or mean SEM of three independent experiments performed in duplicate (B). A 3 hr incubation

time was chosen to assess NNK’s effect on BER in lung and liver.

Page 67: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

55

Lung 1 hr

Control NNK 0

1000

2000

3000

4000

5000

n=4 n=4

A

am

ol[

32P

]dG

TP

/g D

NA

Lung 2 hr

Control NNK 0

1000

2000

3000

4000

5000

n=4 n=4

B

am

ol[

32P

]dG

TP

/g D

NA

Lung 24 hr

Control NNK 0

2000

4000

6000

8000

n=4 n=4

C

am

ol[

32P

]dG

TP

/g D

NA

Figure 3.7 Effect of in vivo treatment with NNK on in vitro BER in mouse lung extracts towards

8-OHdG adducted plasmid. Extracts were prepared from mice treated with 10 mol NNK or

saline and sacrificed 1 (A), 2 (B) or 24 (C) hr post-treatment. Results are presented as mean SD

and expressed as damage-specific BER. No statistically significant differences were found

between NNK and saline control at each time point (P > 0.05, Student’s t-test).

Page 68: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

56

Liver 1 hr

Control NNK 0

1000

2000

3000

4000

5000

n=4 n=4

A

am

ol[

32P

]dG

TP

/g D

NA

Liver 2 hr

Control NNK 0

1000

2000

3000

4000

5000

n=3 n=4

B

am

ol[

32P

]dG

TP

/g D

NA

Liver 24 hr

Control NNK 0

2000

4000

6000

8000

n=4 n=4

C

am

ol[

32P

]dG

TP

/g D

NA

Figure 3.8 Effect of in vivo treatment with NNK on in vitro BER in mouse liver extracts towards

8-OHdG adducted plasmid. Extracts were prepared from mice treated with 10 mol NNK or

saline and sacrificed 1 (A), 2 (B) or 24 (C) hr post-treatment. Results are presented as mean SD

and expressed as damage-specific BER. No statistically significant differences were found

between NNK and saline control at each time point (P > 0.05, Student’s t-test).

Page 69: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

57

Chapter 4

GENERAL DISCUSSION

In the present study, female A/J mice were treated with a tumourigenic dose of NNK and

the level of DNA oxidation and BER activity was determined in lung and liver of control and

NNK-treated mice.

4.1 In vivo treatment with NNK does not induce oxidative DNA damage

NNK-induced carcinogenesis typically involves metabolic activation to generate reactive

intermediates that methylate or pyridyloxobutylate DNA. In vivo treatment with NNK in rodents,

induces DNA damage in oncogenes and tumour suppressor genes that play a critical role in

carcinogenesis (Hecht, 1999). In addition, in vivo treatment with NNK has been reported to

induce oxidative DNA damage in the form of 8-OHdG in tissues such as lung and liver in mice

(Chung and Xu, 1992; Rosa et al., 1998; Weitberg and Corvese, 1993). The consequences of

excess 8-OHdG, a major oxidative DNA lesion, include GT transversion mutations in

oncogenes and tumour suppressor genes that could contribute to carcinogenesis (Klaunig and

Kamendulis, 2004).

Reported basal levels of DNA oxidation in biological systems have been inconsistent,

with values of 8-OHdG differing by ~2 orders of magnitude (Collins et al., 2004; Ravanat,

2005a). The large variation in 8-OHdG levels can largely be attributed to different methods and

techniques used to isolate and assess DNA oxidation. For example, the levels of 8-OHdG in pig

liver DNA ranged from 0.21 to 44.1 8-OHdG per 105 2’-dG using HPLC-ECD and gas

chromatography-mass spectrometry (GC-MS) (Collins et al., 2004). Guanine is readily

oxidizable during sample preparation and some techniques are conducive to spurious oxidation

that can overestimate the levels of DNA oxidation. GC-MS is prone to high levels of artifactual

Page 70: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

58

oxidation during derivatization which require high temperature and a silylating agent to make

DNA samples volatile for separation (Cadet et al., 1998). Conversely, the lowest levels of basal

8-OHdG have been observed when using enzymes specific for 8-OHdG (Ravanat, 2005a). In

enzymatic nicking assays such as the comet assay, levels of 8-OHdG can range from < 1 to 10 8-

OHdG residues per 106 2’-dG. It is unclear whether enzymes bind all 8-OHdG sites in genomic

DNA. Failure to do so would underestimate the levels of 8-OHdG (Halliwell and Whiteman,

2004; Ravanat, 2005a). When measuring background levels of oxidative DNA damage in control

tissues and cells, HPLC-ECD has generated values as low as several 8-OHdG per 106 2’-dG in

human lymphocytes and as high as 20 8-OHdG per 105 2’-dG in untreated murine liver (Zhang et

al., 2004; ESCODD et al., 2005; Guindon et al., 2007).

In the present study, DNA of lung and liver was isolated, digested and assessed for the

ratio of 8-OHdG per 105 2’-dG using HPLC-ECD. For chromatographic methods including

HPLC-ECD, the addition of metal chelators and antioxidants during sample preparation is

standard practice to reduce spurious oxidation (European Standards Committee on Oxidative

DNA Damage, 2003; Ravanat, 2005a). DFO, an iron chelator that prevents spurious oxidation

via Fenton reactions, and an additional unspecified antioxidant (included in the Wako DNA

Extractor TIS kit) were used when isolating DNA. In addition, a sodium iodide extraction

method was utilized to avoid the use of phenol and chloroform extraction which can artificially

enhance 8-OHdG levels (Claycamp, 1992; Ravanat, 2005a). DNA digestion was also optimized

for thorough digestion to reduce spurious oxidation (Huang et al., 2001). Gel electrophoresis of

the DNA digests revealed absence of high molecular weight DNA, indicating that genomic DNA

was thoroughly digested. DNA isolation, digestion and HPLC analysis of control and NNK-

treated samples were carried within one week to minimize artifactual DNA oxidation. Samples

Page 71: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

59

from control and NNK-treated animals for a given time point were processed at the same time to

minimize the potential impact of inter-day assay variability (Kasai, 1997).

The levels of 8-OHdG were comparable to a number of studies found in the literature.

The background levels of 8-OHdG in calf thymus DNA were 21.5 8-OHdG per 105 2’-dG and

were comparable to optimal levels determined by the European Standards Committee on

Oxidative DNA Damage which ranged from 1.9 to 21.3 8-OHdG per 105 2’-dG when using

HPLC-ECD (Lunec, 1998). These results suggest that artifactual oxidation was minimal when

using the current protocol in the present study. In addition, control levels of oxidative DNA

damage in murine lung and liver were not variable at each time point, and comparable to other

studies that assessed 8-OHdG in such tissues using HPLC-ECD (Zhang et al., 2004; Guindon et

al., 2007).

In the present study, 8-OHdG was assayed in lungs and livers of mice treated with a

tumourigenic dose (10 mol) of NNK after 1, 2 and 24 hours. No statistically significant effects

in 8-OHdG levels were observed in lung or liver of mice treated with NNK at each time point,

indicating in vivo treatment with NNK did not induce oxidative DNA damage. However, a trend

towards elevated 8-OHdG levels was observed in liver and less so in lungs from NNK-treated

mice in the first 2 hours. It is important to recognize that these trends were non-significant, as an

apparent 27% increase in 8-OHdG levels in liver (n=4, P = 0.166) and apparent 14% increase in

8-OHdG levels in lung (n=4, P = 0.238) was observed at 2 hours. Although not statistically

significant this trend towards elevated 8-OHdG levels is similar to other studies that reported an

increase in 8-OHdG levels in lung and liver 2 hours after NNK administration, with a decrease to

basal levels after 24 hours (Chung and Xu, 1992; Xu et al., 1992; Rosa et al., 1998).

Page 72: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

60

Although some studies have reported oxidative DNA damage in experimental animals

after NNK treatment, this is not always the case. In some studies, in vivo treatment with NNK

failed to cause statistically significant changes in DNA oxidation. Treatment with 19.3 mol of

NNK i.p. appeared to increase the levels of 8-OHdG in A/J mouse lung and liver 2 hours after

administration, but the effect was not statistically significant (Chung and Xu, 1992).

Furthermore, treatment of A/J mice with multiple doses of NNK (11.65 mg/kg body weight, via

gavage, 3 times weekly for 10 weeks) did not significantly affect 8-OHdG levels in liver 2 hours

post-treatment (Xu et al., 1992). In such studies, NNK could have altered oxidative DNA

damage in specific cell types of lung and liver but these effects may not have been detected when

analyzing oxidative DNA damage from whole lung and liver. Similar trends were observed in the

current study, with statistically non-significant increases in 8-OHdG levels in lung and liver

following NNK treatment, suggesting NNK may actually have limited effect on DNA oxidation

status.

4.2 In vivo treatment with NNK does not alter overall BER activity

DNA repair systems are critical for protecting and maintaining the genome from

exogenous and endogenous damaging agents. Several DNA repair systems have been identified

and categorized based on the type of damage repaired. A major function of the BER pathway is

to identify and remove bases that have been damaged by ROS. The effects of NNK have

previously been studied on nucleotide excision repair (NER) and direct reversal repair systems,

but have not on BER.

In the present study, an in vitro repair assay was optimized to assess BER in lung and

liver of mice treated with NNK. To assess BER activity, a plasmid substrate containing 8-OHdG

lesions was incubated with cell-free nuclear protein extracts prepared from murine lung and liver.

Page 73: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

61

Methylene blue irradiated with white light was used to induce oxidative DNA damage in the

plasmid substrate. During photo-oxidation, methylene blue is activated into a transitory excited

state which in turn can react with stable oxygen to generate reactive singlet oxygen (Floyd et al.,

1989). When exposed to DNA, singlet oxygen has high specificity for guanine residues and

usually generates 8-OHdG lesions (Epe, 1991). An increase in 8-OHdG levels was observed

when using a range of methylene blue concentrations to induce oxidative damage. The levels of

8-OHdG were dependent on the final concentration of methylene blue during exposure to light.

These results are consistent with literature that showed increasing 8-OHdG levels in DNA treated

with increasing concentrations of methylene blue irradiated with light (Floyd et al., 1989;

Schneider et al., 1990).

Plasmid was also assessed for nicking status using gel electrophoresis and DNA

densitometry. Methylene blue was selected as an oxidizing agent as it preferentially induces 8-

OHdG over DNA strand breaks that could potentially be repaired by systems other than BER

(Schneider et al., 1990). In the current study, only 3% of the untreated plasmid was nicked,

leaving 97% in supercoiled form. Plasmid treated with 5 M methylene blue and subsequently

chosen to assess BER in lung and liver was 12% nicked, leaving 88% in supercoiled form.

Although a small percentage of both types of plasmid were nicked, DNA single strand nicks have

been shown to stimulate very little incorporation of radiolabelled nucleotides when using a

similar in vitro repair assay that assessed NER (Wood et al., 1988; Brown et al., 2007).

Similarly, no significant differences were observed in NER towards nicked and supercoiled

plasmid that contained POB adducts, indicating no artifactual increase in damage-specific repair

activity due to nicks (Brown et al., 2007). These findings suggest that the limited amounts of

nicked DNA present in the plasmid substrate and used in the current study likely had minimal

impact on the BER activities measured.

Page 74: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

62

When choosing an appropriate plasmid substrate to assess BER, several plasmid

substrates were prepared with varying levels of DNA oxidation. BER activity was dependent on

the amount of 8-OHdG induced by methylene blue within the plasmid substrate. Untreated

plasmid containing background levels of 8-OHdG had the lowest activity compared to plasmid

substrates treated with methylene blue. An upper limit of BER activity was reached in plasmid

substrate treated with high concentrations of methylene blue that induced high levels of 8-OHdG.

The repair system provided by the lung and liver extract could have been limiting, resulting in a

plateau in repair activity. Plasmid treated with 5 M methylene blue induced ~40 8-OHdG per

105 2’-dG and was selected for the assay since repair in this plasmid was ~5-fold higher

compared to untreated plasmid.

BER activity was also protein concentration-dependent and repair duration-dependent.

When assessing protein concentration in the assay, nuclear extract was required for repair activity

as repair reactions that contained no protein had no incorporation of [32

P] dGTP. A plateau in

BER activity was observed when incubations contained > 30 g of lung extract or > 50 g of

liver extract, presumably due to limiting amounts of damaged plasmid substrate. It should be

noted that at higher protein amounts (ranging from 100 to 200 g of liver extract in particular), a

decrease in repair activity was sometimes observed (data not shown). The presence of excessive

amount of proteins may have interfered with recognition and binding of 8-OHdG lesions,

reducing overall efficiency and repair activity. When assessing incubation time, an apparent

linear increase in repair activity was observed in murine lung and liver over 4 hours. Repair

reactions that were terminated immediately had some repair activity. BER proteins may begin to

initiate repair processes upon addition of oxidized plasmid substrate followed by reaction

termination. These results were similar to studies that demonstrated BER activity in extracts

from rat brain was dependent on protein concentration and incubation time (Lan et al., 2003;

Page 75: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

63

Chen et al., 2000). In those studies, an upper limit in BER activity was also observed at protein

amounts > 30 g and incubation times > 8 hours.

When assessing the effects of NNK on BER in liver and lung, selection of an oxidized

plasmid substrate, protein concentration and incubation time was chosen based on the ability to

detect increases and decreases in BER activity. Plasmid treated with 5 M methylene blue, a 3

hour incubation time and 10 g of lung or 25 g of liver extract were chosen for the present in

vitro repair assay. No statistically significant differences in BER were observed in lung or liver

between control and NNK-treated mice at 1, 2 or 24 hours. Hence, administration of a

tumourigenic dose of NNK did not affect BER activity up to 24 hours post treatment. In previous

studies, NNK was found to inhibit NER and O6-methylguanine-DNA methyltransferase (MGMT)

in lung, and increase NER in liver (Belinsky et al., 1988; Liu et al., 1992; Brown and Massey,

2009) but failed to alter BER activity in the current study. As a result, it appears that NNK could

selectively modify DNA repair pathways like NER and MGMT, while sparing others like BER.

However, it is also possible that the previously reported effectiveness of NNK at altering non-

BER DNA repair systems could be due to differences in: doses of NNK; experimental designs;

species and strains of animals; and time points. For example, MGMT inhibition by NNK was

demonstrated in specific lung cell types of rats treated with multiple doses of NNK (Belinsky et

al., 1988). Furthermore, rat hepatocytes treated with OPB, a reactive metabolite of NNK, had

inhibited MGMT activity (Liu et al., 1992). Nonetheless, NNK’s inhibition of NER in lung was

studied using the same dose and strain of mouse and a time course similar to that of the current

BER study (Brown and Massey, 2009).

One mechanism by which NNK could modify specific repair pathways is by forming

specific protein adducts. Treatment with NNK has been shown to pyridyloxobutylate proteins

Page 76: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

64

including hemoglobin and MGMT (Murphy et al., 1990; Mijal et al., 2004). Electrophilic

intermediates generated during NNK metabolism may preferentially bind to major proteins of

NER or MGMT systems modifying their overall repair activity, but may react less with BER

proteins, perhaps due to lower affinity or lack of an available specific amino acid binding motif.

It should be noted that an apparent decreasing trend in BER (by 34% and 21%, at 1 and

24 hours post administration respectively) was observed in lungs of NNK-treated mice, although

it was not statistically significant. A potential explanation for the lack of significant effect of

NNK on repair of the oxidized plasmid is repair of 8-OHdG by additional mechanisms.

Treatment with NNK could have inhibited BER activity in lung, but this inhibition could have

been mitigated by other functional BER glycosylases or non-BER DNA repair mechanisms

present in the nuclear extracts. Humans and mice both have several BER glycosylases that have

overlapping function and redundancy to ensure extra protection against frequently occurring

oxidative DNA lesions, like 8-OHdG (Hazra et al., 2003; Cooke et al., 2003). For example, BER

glycosylases such as OGG1 and Nei-like 1 both repair 8-OHdG adducts in DNA and either

glycosylase could act as a backup in case one has compromised activity (Hazra et al., 2003). In

some cases mismatch repair and NER systems have also been shown to repair 8-OHdG lesions in

DNA, and could alleviate the effect of decreased BER (Reardon et al., 1997; Colussi et al., 2002;

Patel et al., 2007). In the present study, these additional repair mechanisms could have also

played a role in reducing the effect of NNK on BER and protecting the genome from oxidative

DNA in lungs of mice.

A tumourigenic dose of NNK (10 mol) given i.p. also had no significant effect on BER

activity in liver. Interestingly, the same dose and route of administration of NNK increased NER

activity and levels of specific NER proteins in liver of A/J mice 24 hours post administration

(Brown and Massey, 2009). Hence, NNK increases NER activity in liver while having no effect

Page 77: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

65

on BER. These results are consistent with the fact that NER and BER are regulated differently.

For example, post-translational modifications such as phosphorylation, ubiquitylation and

sumoylation occur to varying degrees to proteins in the NER and BER pathways and could affect

enzyme recruitment, DNA-protein interactions, protein-protein interactions and catalytic function

of each DNA repair system (Huang and D'Andrea, 2006; Branzei and Foiani, 2008).

4.3 Conclusions

The research presented in this thesis has demonstrated that a single in vivo administration

of NNK did not induce oxidative DNA damage in A/J mouse lung or liver at 1, 2 or 24 hours.

Only a slight increasing trend in oxidative DNA damage was observed in lung and liver that was

not statistically significant. Additionally, this was the first study to test the effects of NNK on

BER. Using an optimized in vitro repair assay, NNK did not alter overall BER activity in lung

and liver at each time point. A decreasing trend in BER activity was observed in lung that was

not statistically significant.

4.4 Future directions

Although not statistically significant, an increasing trend in 8-OHdG and decreasing trend

in BER activity in lung and liver of mice treated with NNK was observed and should be further

investigated. This was the first study to test the effect of NNK on BER and supplementary

studies should be carried out to verify the present findings. Additional experiments could be

conducted to determine if NNK has effect on BER and DNA oxidation over a daily, weekly or

monthly time course. The current research investigated potential effects of NNK in lung and liver

acutely at 1, 2 and 24 hours post administration. The dose of NNK employed in the current study

(10 mol) has been shown to cause induction of lung adenomas in mice 16 weeks later (Hecht et

al., 1989). The progression of lung adenomas to malignant adenocarcinomas was observed in

Page 78: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

66

lungs 50 weeks post NNK administration (Hecht, 1998). Assessing NNK’s effect on DNA

oxidation and BER in lung and liver over a longer period of time could further elucidate its role

during carcinogenesis. Furthermore, the current research used a single i.p. dose of NNK that does

not accurately model chronic smoking behaviour in humans. While a single 10 mol dose given

i.p. will most likely induce pulmonary carcinogenesis in mice, chronic administration of NNK at

lower doses over several weeks more closely resembles smoking behaviour.

Investigating the effects of NNK is important for understanding tobacco-induced

carcinogenesis. Substantial experimental data has shown NNK to be one of the most potent

pulmonary carcinogens found in cigarettes and tobacco products and most likely plays a role in

tobacco-induced human cancers. Studying NNK in isolation from a chemical agent as complex

as tobacco smoke is helpful in specifically characterizing the molecular mechanism by which a

major component of cigarettes affects DNA repair processes. However, to expand the scope of

this project, a second approach should also be taken that investigates the effect of cigarette smoke

on repair activity in animal models. NNK is only one component of cigarettes and tobacco

smoke that contain over 4000 chemicals including 55 known carcinogens. The current study is

limited by only investigating the effects of NNK on DNA damage and repair. Exposing

experimental animals to cigarette smoke would represent human smoking more accurately by

having inhalation as the primary route of administration as well as treating with agents that have

the same chemical composition as tobacco smoke. BER and other repair processes could be

assessed in lung and liver in these studies.

If statistically significant differences in BER activity were observed in future studies, the

next logical step would be to determine where and how changes were occurring in the pathway.

A modified in vitro oligonucleotide repair assay would be carried out to ascertain if changes

occurred during the recognition and excision step, or nucleotide insertion and ligation step of the

Page 79: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

67

BER pathway. If changes in activity occurred at both or either steps, immunoblots of key BER

proteins would be performed on control and NNK-treated animals to determine if changes in

repair activity were due to variation in protein levels. Real-time reverse transcription polymerase

chain reaction could be performed to determine if potential changes in levels of BER proteins

were due to changes in mRNA transcript levels. A nuclear run-on assay could also be used to

determine if NNK alters transcription rates of particular BER genes. Reactive intermediates

generated by metabolism of NNK have been shown to adduct proteins such as hemoglobin

(Murphy et al., 1990) and could methylate or pyridyloxobutylate key BER proteins that could

change protein activity or stability. Adducted BER proteins could be separated using SDS-PAGE

and following immunoblotting, excised, digested and subjected to mass spectrometry for

identification.

Potential changes in the activity of BER proteins could also be due to changes in post-

translational modifications. Phosphorylation, sumoylation and ubiquitylation of repair proteins

are important for regulating several types of DNA repair (Branzei and Foiani, 2008; Huang and

D'Andrea, 2006). For example, sumoylation of certain BER glycosylases can alter catalytic

activity for specific substrates, as well as reduce affinity for the AP site which allows access to

DNA for further processing by additional BER proteins (Hardeland et al., 2002; Steinacher and

Schär, 2005). Enzymatic techniques and mass spectrometric analysis could be used to determine

if NNK-induced changes in BER activity were due to alterations in post-translational

modifications on specific proteins. These types of experiments would ultimately investigate

components of NNK’s mechanism of action and could be helpful in identifying susceptible

individuals and future development of novel preventative therapies or treatments for tobacco-

induced cancer.

Page 80: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

68

REFERENCES

Achanta G and Huang P (2004) Role of p53 in sensing oxidative DNA damage in response to

reactive oxygen species-generating agents. Cancer Res 64:6233-6239.

Auerbach O and Garfinkel L (1991) The changing pattern of lung carcinoma. Cancer 68:1973-

1977.

Bedard LL, Alessi M, Davey S and Massey TE (2005) Susceptibility to aflatoxin B1-induced

carcinogenesis correlates with tissue-specific differences in DNA repair activity in mouse and in

rat. Cancer Res 65:1265-1270.

Belinsky SA (1998) Role of the cytosine DNA-methyltransferase and p16INK4a genes in the

development of mouse lung tumors. Exp Lung Res 24:463-479.

Belinsky SA, Devereux TR, Maronpot RR, Stoner GD and Anderson MW (1989) Relationship

between the formation of promutagenic adducts and the activation of the K-ras protooncogene in

lung tumors from A/J mice treated with nitrosamines. Cancer Res 49:5305-5311.

Belinsky SA, Dolan ME, White CM, Maronpot RR, Pegg AE and Anderson MW (1988) Cell

specific differences in O6-methylguanine-DNA methyltransferase activity and removal of O6-

methylguanine in rat pulmonary cells. Carcinogenesis 9:2053-2058.

Belinsky SA, Nikula KJ, Palmisano WA, Michels R, Saccomanno G, Gabrielson E, Baylin SB

and Herman JG (1998) Aberrant methylation of p16(INK4a) is an early event in lung cancer and

a potential biomarker for early diagnosis. Proc Natl Acad Sci USA 95:11891-11896.

Bertram JS (2000) The molecular biology of cancer. Mol Aspects Med 21:167-223.

Boffetta P, Hecht S, Gray N, Gupta P and Straif K (2008) Smokeless tobacco and cancer. Lancet

Oncology 9:667-675.

Bolin C, Stedeford T and Cardozo-Pelaez F (2004) Single extraction protocol for the analysis of

8-hydroxy-2'-deoxyguanosine (oxo8dG) and the associated activity of 8-oxoguanine DNA

glycosylase. J Neurosci Methods 136:69-76.

Borczuk AC, Gorenstein L, Walter KL, Assaad AA, Wang L and Powell CA (2003) Non-small-

cell lung cancer molecular signatures recapitulate lung developmental pathways. Am J Pathol

163:1949-1960.

Branzei D and Foiani M (2008) Regulation of DNA repair throughout the cell cycle. Nat Rev Mol

Cell Biol 9:297-308.

Brown PJ, Bedard LL and Massey TE (2007) Repair of 4-(methylnitrosamino)-1-(3-pyridyl)-1-

butanone-induced DNA pyridyloxobutylation by nucleotide excision repair. Cancer Lett.

Page 81: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

69

Brown PJ and Massey TE (2009) In vivo treatment with 4-(methylnitrosamino)-1-(3-pyridyl)-1-

butanone (NNK) induces organ-specific alterations in in vitro repair of DNA

pyridyloxobutylation. Mutat Res 663:15-21.

Brunnemann KD, Prokopczyk B, Djordjevic MV and Hoffman D (1996) Formation and analysis

of tobacco-specific N-nitrosamines. Critical Reviews in Toxicology 26:121-137.

Cadet J, D'Ham C, Douki T, Pouget JP, Ravanat JL and Sauvaigo S (1998) Facts and artifacts in

the measurement of oxidative base damage to DNA. Free Rad Res 29:541-550.

Caldecott KW, McKeown CK, Tucker JD, Ljungquist S and Thompson LH (1994) An interaction

between the mammalian DNA repair protein XRCC1 and DNA ligase III. Mol Cell Biol 14:68-

76.

Canadian Cancer Society. (2010). Canadian Cancer Statistics. www.cancer.ca. Accessed: March

2010.

Carmella SG, Borukhova A, Desai D and Hecht SS (1997) Evidence for endogenous formation of

tobacco-specific nitrosamines in rats treated with tobacco alkaloids and sodium nitrite.

Carcinogenesis 18:587-592.

Castonguay A, Lin D, Stoner GD, Radok P, Furuya K, Hecht SS, Schut HA and Klaunig JE

(1983) Comparative carcinogenicity in A/J mice and metabolism by cultured mouse peripheral

lung of N'-nitrosonornicotine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, and their

analogues. Cancer Res 43:1223-1229.

Castonguay A, Pepin P and Briere N (1991) Modulation of 4-(methylnitrosamino)-1-(3-pyridyl)-

1 butanone demethylation and denitrosation by rat liver microsomes. Cancer Lett 59:67-74.

Chen B, Johanson L, Wiest JS, Anderson MW and You M (1994) The second intron of the K-ras

gene contains regulatory elements associated with mouse lung tumor susceptibility. Proc Natl

Acad Sci USA 91:1589-1593.

Chen D, Lan J, Pei W and Chen J (2000) Detection of DNA base-excision repair activity for

oxidative lesions in adult rat brain mitochondria. J Neurosci Res 61:225-236.

Cheng KC, Cahill DS, Kasai H, Nishimura S and Loeb LA (1992) 8-Hydroxyguanine, an

abundant form of oxidative DNA damage, causes G---- T and A----C substitutions. J Biol Chem

267:166-172.

Chmiel NH, Livingston AL and David SS (2003) Insight into the functional consequences of

inherited variants of the hMYH adenine glycosylase associated with colorectal cancer:

complementation assays with hMYH variants and pre-steady-state kinetics of the corresponding

mutated E.coli enzymes. Journal of Molecular Biology 327:431-443.

Christmann M, Tomicic MT, Roos WP and Kaina B (2003) Mechanisms of human DNA repair:

an update. Toxicology 193:3-34.

Page 82: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

70

Chung FL and Xu Y. Increased 8-oxodeoxyguanosine levels in lung DNA of A/J mice and F344

rats treated with the tobacco-specific nitrosamine 4-(methyhiitrosamine)-l-(3-pyridyl)-1-

butanone. 1992. Oxford University Press.

Chute JP, Chen T, Feigal E, Simon R and Johnson BE (1999) Twenty years of phase III trials for

patients with extensive-stage small-cell lung cancer: perceptible progress. Journal of Clinical

Oncology 17:1794-1801.

Claycamp HG (1992) Phenol sensitization of DNA to subsequent oxidative damage in 8-

hydroxyguanine assays. Carcinogenesis 13:1289-1292.

Collins AR, Cadet J, Moller L, Poulsen HE and Viňa J (2004) Are we sure we know how to

measure 8-oxo-7,8-dihydroguanine in DNA from human cells? Archives of Biochemistry and

Biophysics 423:57-65.

Colussi C, Parlanti E, Degan P, Aquilina G, Barnes D, Macpherson P, Karran P, Crescenzi M,

Dogliotti E and Bignami M (2002) The mammalian mismatch repair pathway removes DNA 8-

oxodGMP incorporated from the oxidized dNTP pool. Current Biology 12:912-918.

Cooke MS, Evans MD, Dizdaroglu M and Lunec J (2003) Oxidative DNA damage: mechanisms,

mutation, and disease. FASEB J 17:1195-1214.

Corvalan A and Wistuba II (2010) Molecular pathology of lung cancer, in Lung Cancer:

Prevention, Management, and Emerging Therapies (Stewart DJ ed) pp 1-25.

Cunningham R (1996) A Prescription for Health, in Smoke and mirrors The Canadian Tobacco

War pp 247-265, IDRC International Development Research Centre.

David SS, O'Shea VL and Kundu S (2007) Base-excision repair of oxidative DNA damage.

Nature 447:941-950.

de Laat WL, Jaspers NG and Hoeijmakers JH (1999) Molecular mechanism of nucleotide

excision repair. Genes Dev 13:768-785.

Devesa SS, Shaw GL and Blot WJ (1991) Changing patterns of lung cancer incidence by

histological type. Cancer Epidem Biomark Prev 1:29-34.

Dianov G, Bischoff C, Piotrowski J and Bohr VA (1998) Repair pathways for processing of 8-

oxoguanine in DNA by mammalian cell extracts. J Biol Chem 273:33811-33816.

Ekstrom G and Ingelman-Sundberg M (1989) Rat liver microsomal NADPH-supported oxidase

activity and lipid peroxidation dependent on ethanol-inducible cytochrome P-450 (P-450IIE1).

Biochem Pharmacol 38:1313-1319.

Epe B (1991) Genotoxicity of singlet oxygen. Chem Biol Interact 80:239-260.

Page 83: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

71

ESCODD, Gedik CM and Collins A (2005) Establishing the background level of base oxidation

in human lymphocyte DNA: results of an interlaboratory validation study. The FASEB Journal

19:82-84.

European Standards Committee on Oxidative DNA Damage (2003) Measurement of DNA

oxidation in human cells by chromatographic and enzymic methods. Free Radic Biol Med

34:1089-1099.

Floyd RA, West MS, Eneff KL and Schneider JE (1989) Methylene blue plus light mediates 8-

hydroxyguanine formation in DNA. Arch Biochem Biophys 273:106-111.

Foiles PG, Akerkar SA, Carmella SG, Kagan M, Stoner GD, Resau JH and Hecht SS (1991)

Mass spectrometric analysis of tobacco-specific nitrosamine-DNA adducts in smokers and

nonsmokers. Chem Res Toxicol 4:364-368.

Fortini P and Dogliotti E (2007) Base damage and single-strand break repair: mechanisms and

functional significance of short- and long-patch repair subpathways. DNA Repair 6:398-409.

Fortini P, Parlanti E, Sidorkina OM, Laval J and Dogliotti E (1999) The type of DNA glycosylase

determines the base excision repair pathway in mammalian cells. J Biol Chem 274:15230-15236.

Fraga CG, Shigenaga MK, Park JW, Degan P and Ames BN (1990) Oxidative damage to DNA

during aging: 8-hydroxy-2'-deoxyguanosine in rat organ DNA and urine. Proc Natl Acad Sci USA

87:4533-4537.

Giovino GA (2002) Epidemiology of tobacco use in the United States. onc 21:7326-7340.

Guindon KA, Bedard LL and Massey TE (2007) Elevation of 8-hydroxydeoxyguanosine in DNA

from Isolated Mouse Lung Cells following In Vivo Treatment with aflatoxin B1. Toxicol Sci.

Halliwell B and Aruoma OI (1991) DNA damage by oxygen-derived species: Its mechanism and

measurement in mammalian systems. FEBS Lett 281:9-19.

Halliwell B and Whiteman M (2004) Measuring reactive species and oxidative damage in vivo

and in cell culture: how should you do it and what do the results mean? British Journal of

Pharmacology 142:231-255.

Hanahan D and Weinberg RA (2000) The hallmarks of cancer. Cell 100:57-70.

Hanrahan EO and Glisson B (2010) Small cell carcinoma, in Lung Cancer: Prevention,

Management, and Emerging Therapies (Stewart DJ ed) pp 395-434.

Hardeland U, Steinacher R, Jiricny J and Schar P (2002) Modification of the human thymine-

DNA glycosylase by ubiquitin-like proteins facilitates enzymatic turnover. EMBO J 21:1456-

1464.

Page 84: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

72

Hazra TK, Izumi T, Kow YW and Mitra S (2003) The discovery of a new family of mammalian

enzymes for repair of oxidatively damaged DNA, and its physiological implications.

Carcinogenesis 24:155-157.

Health Canada. (2010). Tobacco Health Concerns. www.hc-sc.gc.ca/hc-ps/tobac-tabac/index-

eng.php. Accessed: October 2010.

Hecht SS (1998) Biochemistry, biology, and carcinogenicity of tobacco-specific N-nitrosamines.

Chem Res Toxicol 11:559-603.

Hecht SS (1999) Tobacco smoke carcinogens and lung cancer. J Natl Cancer Inst 91:1194-1210.

Hecht SS (2002) Cigarette smoking and lung cancer: chemical mechanisms and approaches to

prevention Lancet Oncol 3:461-469.

Hecht SS (2003) Tobacco carcinogens, their biomarkers and tobacco-induced cancer. Nat Rev

Cancer 3:733-744.

Hecht SS, Chen CB, Hirota N, Ornaf RM, Tso TC and Hoffmann D (1978) Tobacco-specific

nitrosamines: formation from nicotine in vitro and during tobacco curing and carcinogenicity in

strain A mice. J Natl Cancer Inst 60:819-824.

Hecht SS and Hoffmann D (1988) Tobacco-specific nitrosamines, an important group of

carcinogens in tobacco and tobacco smoke. Carcinogenesis 9:875-884.

Hecht SS, Morse MA, Amin S, Stoner GD, Jordan KG, Choi CI and Chung FL (1989) Rapid

single-dose model for lung tumor induction in A/J mice by 4-(methylnitrosamino)-1-(3-pyridyl)-

1-butanone and the effect of diet. Carcinogenesis 10:1901-1904.

Hecht SS, Trushin N, Castonguay A and Rivenson A (1986) Comparative tumorigenicity and

DNA methylation in F344 rats by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and N-

nitrosodimethylamine. Cancer Res 46:498-502.

Hecht SS, Adams JD, Numoto S and Hoffmann D (1983) Induction of respiratory tract tumors in

Syrian golden hamsters by a single dose of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone

(NNK) and the effect of smoke inhalation. Carcinogenesis 4:1287-1290.

Hecht SS, Chen Ch, Ohmori T and Hoffmann D (1980) Comparative Carcinogenicity in F344

Rats of the Tobacco-specific Nitrosamines, N Ǧ-Nitrosonornicotine and 4-(N-Methyl-N-

nitrosamino)-1-(3-pyridyl)-1-butanone. Cancer Res 40:298-302.

Heidelberger C (1977) Chemical carcinogenesis. Cancer 40:Suppl-3.

Herzog CR, Noh S, Lantry LE, Guan KL and You M (1999) Cdkn2a encodes functional variation

of p16INK4a but not p19ARF, which confers selection in mouse lung tumorigenesis. Mol

Carcinog 25:92-98.

Page 85: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

73

Hoffmann D, Hoffmann I and El Bayoumy K (2001) The less harmful cigarette: a controversial

issue. a tribute to Ernst L. Wynder. Chem Res Toxicol 14:767-790.

Hoffmann D, Rivenson A, Amin S and Hecht SS (1984) Dose-response study of the

carcinogenicity of tobacco-specific N-nitrosamines in F344 rats. J Cancer Res Clin Oncol

108:81-86.

Holzle D, Schlobe D, Tricker AR and Richter E (2007) Mass spectrometric analysis of 4-

hydroxy-1-(3-pyridyl)-1-butanone-releasing DNA adducts in human lung. Toxicology 232:277-

285.

Horio Y, Chen A, Rice P, Roth JA, Malkinson AM and Schrump DS (1996) Ki-ras and p53

mutations are early and late events, respectively, in urethane-induced pulmonary carcinogenesis

in A/J mice. Mol Carcinog 17:217-223.

Huang TT and D'Andrea AD (2006) Regulation of DNA repair by ubiquitylation. Nat Rev Mol

Cell Biol 7:323-334.

Huang X, Powell J, Mooney LA, Li C and Frenkel K (2001) Importance of complete DNA

digestion in minimizing variability of 8-oxo-dG analyses. Free Radic Biol Med 31:1341-1351.

Hung RJ, Brennan P, Canzian F, Szeszenia-Dabrowska N, Zaridze D, Lissowska J, Rudnai P,

Fabianova E, Mates D, Foretova L, Janout V, Bencko V, Chabrier A, Borel S, Hall J and Boffetta

P (2005a) Large-scale investigation of base excision repair genetic polymorphisms and lung

cancer risk in a multicenter study. J Natl Cancer Inst 97:567-576.

Hung RJ, Hall J, Brennan P and Boffetta P (2005b) Genetic polymorphisms in the base excision

repair pathway and cancer risk: a HuGE review. Am J Epidemiol 162:925-942.

Hussain SP, Aguilar F, Amstad P and Cerutti P (1994) Oxy-radical induced mutagenesis of

hotspot codons 248 and 249 of the human p53 gene. onc 9:2277-2281.

Igarashi M, Watanabe M, Yoshida M, Sugaya K, Endo Y, Miyajima N, Abe M, Sugano S and

Nakae D (2009) Enhancement of lung carcinogenesis initiated with 4-(N-

hydroxymethylnitrosamino)-1-(3-pyridyl)-1-butanone by Ogg1 gene deficiency in female, but not

male, mice. Journal of Toxicological Sciences 34:163-174.

Jackman DM and Johnson BE (2005) Small-cell lung cancer. [Review] [159 refs]. Lancet

366:1385-1396.

Jackson JH (1994) Potential molecular mechanisms of oxidant-induced carcinogenesis. Environ

Health Perspect 102:Suppl-7.

Jalas JR, Ding X and Murphy SE (2003) Comparative metabolism of the tobacco-specific

nitrosamines 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and 4-(methylnitrosamino)-1-(3-

pyridyl)-1-butanol by rat cytochrome P450 2A3 and human cytochrome P450 2A13. Drug Metab

Dispos 31:1199-1202.

Page 86: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

74

Jalas JR, Hecht SS and Murphy SE (2005) Cytochrome P450 enzymes as catalysts of metabolism

of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, a tobacco specific carcinogen. Chem Res

Toxicol 18:95-110.

Kasai H (1997) Analysis of a form of oxidative DNA damage, 8-hydroxy-2'-deoxyguanosine, as a

marker of cellular oxidative stress during carcinogenesis. Mutation Research/Reviews in Mutation

Research 387:147-163.

Kim PM and Wells PG (1996) Genoprotection by UDP-glucuronosyltransferases in peroxidase-

dependent, reactive oxygen species-mediated micronucleus initiation by the carcinogens 4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone and benzo[a]pyrene. Cancer Res 56:1526-1532.

Kinoshita A, Wanibuchi H, Morimura K, Wei M, Nakae D, Arai T, Minowa O, Noda T,

Nishimura S and Fukushima S (2007) Carcinogenicity of dimethylarsinic acid in Ogg1-deficient

mice. Cancer Sci 98:803-814.

Klaunig JE and Kamendulis LM (2004) The role of oxidative stress in carcinogenesis. Annu Rev

Pharmacol Toxicol 44:239-267.

Klungland A and Lindahl T (1997) Second pathway for completion of human DNA base

excision-repair: reconstitution with purified proteins and requirement for DNase IV (FEN1).

EMBO Journal 16:3341-3348.

Klungland A, Rosewell I, Hollenbach S, Larsen E, Daly G, Epe B, Seeberg E, Lindahl T and

Barnes DE (1999) Accumulation of premutagenic DNA lesions in mice defective in removal of

oxidative base damage. Proc Natl Acad Sci USA 96:13300-13305.

Lan J, Henshall DC, Simon RP and Chen J (2000) Formation of the base modification 8-

hydroxyl-2'-deoxyguanosine and DNA fragmentation following seizures induced by systemic

kainic acid in the rat. Journal of Neurochemistry 74:302-309.

Lan J, Li W, Zhang F, Sun FY, Nagayama T, O'Horo C and Chen J (2003) Inducible repair of

oxidative DNA lesions in the rat brain after transient focal ischemia and reperfusion. J Cereb

Blood Flow Metab 23:1324-1339.

LaVoie EJ, Prokopczyk G, Rigotty J, Czech A, Rivenson A and Adams JD (1987) Tumorigenic

activity of the tobacco-specific nitrosamines 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone

(NNK), 4-(methylnitrosamino)-4-(3-pyridyl)-1-butanol (iso-NNAL) and N'-nitrosonornicotine

(NNN) on topical application to Sencar mice. Cancer Lett 37:277-283.

Lijinsky W, Thomas BJ and Kovatch RM (1991) Local and Systemic Carcinogenic Effects of

Alkylating Carcinogens in Rats Treated by Intravesicular Administration. Cancer Science

82:980-986.

Lindahl T and Wood RD (1999) Quality control by DNA repair. Science 286:1897-1905.

Page 87: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

75

Liu BQ, Peto R, Chen ZM, Boreham J, Wu YP, Li JY, Campbell TC and Chen JS (1998)

Emerging tobacco hazards in China: 1. Retrospective proportional mortality study of one million

deaths. BMJ 317:1411-1422.

Liu L, Castonguay A and Gerson SL (1992) Lack of correlation between DNA methylation and

hepatocarcinogenesis in rats and hamsters treated with 4-(methylnitrosamino)-1-(3-pyridyl)-1-

butanone. Carcinogenesis 13:2137-2140.

Loft S and Poulsen HE (1996) Cancer risk and oxidative DNA damage in man. Journal of

Molecular Medicine 74:297-312.

Lowry OH, Rosebrough NJ, Farr AL and Randall RJ (1951) Protein measurement with the folin

phenol reagent. J Biol Chem 193:265-275.

Luch A (2005) Nature and nurture - lessons from chemical carcinogenesis. Nature Reviews

Cancer 5:113-125.

Lunec J (1998) ESCODD: European Standards Committee on Oxidative DNA Damage. Free

Radic Res 29:601-608.

Malkinson AM (1998) Molecular comparison of human and mouse pulmonary adenocarcinomas.

Exp Lung Res 24:541-555.

Maser E (2004) Significance of reductases in the detoxification of the tobacco-specific

carcinogen NNK. Trends in Pharmacological Sciences 25:235-237.

McBride TJ, Schneider JE, Floyd RA and Loeb LA (1992) Mutations induced by methylene blue

plus light in single-stranded M13mp2. Proc Natl Acad Sci USA 89:6866-6870.

Meunier B, de Visser SlP and Shaik S (2004) Mechanism of Oxidation Reactions Catalyzed by

Cytochrome P450 Enzymes. Chemical Reviews 104:3947-3980.

Meuwissen R and Berns A (2005) Mouse models for human lung cancer. Genes Dev 19:643-664.

Mijal RS, Thomson NM, Fleischer NL, Pauly GT, Moschel RC, Kanugula S, Fang Q, Pegg AE

and Peterson LA (2004) The Repair of the Tobacco Specific Nitrosamine Derived Adduct O6-[4-

Oxo-4-(3-pyridyl)butyl]guanine by O6-Alkylguanine-DNA Alkyltransferase Variants. Chem Res

Toxicol 17:424-434.

Miller EC and Miller JA (1981) Mechanisms of chemical carcinogenesis. Cancer 47:Suppl-64.

Minowa O, Arai T, Hirano M, Monden Y, Nakai S, Fukuda M, Itoh M, Takano H, Hippou Y,

Aburatani H, Masumura K, Nohmi T, Nishimura S and Noda T (2000) Mmh/Ogg1 gene

inactivation results in accumulation of 8-hydroxyguanine in mice. Proc Natl Acad Sci USA

97:4156-4161.

Page 88: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

76

Murphy SE, Spina DA, Nunes MG and Pullo DA (1995) Glucuronidation of 4-

((hydroxymethyl)nitrosamino)-1-(3-pyridyl)-1-butanone, a metabolically activated form of 4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone, by phenobarbital-treated rats. Chem Res Toxicol

8:772-779.

Murphy SE, Palomino A, Hecht SS and Hoffman D (1990) Dose-Response Study of DNA and

Hemoglobin Adduct Formation by 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone in F344 Rats.

Cancer Res 50:5446-5452.

Mustonen R, Schoket B and Hemminki K (1993) Smoking-related DNA adducts: 32

P-

postlabeling analysis of 7-methylguanine in human bronchial and lymphocyte DNA.

Carcinogenesis 14:151-154.

Nademanee K (1992) The amiodarone odyssey. J Am Coll Cardiol 20-5:1063-1065.

Nakamura J, La DK and Swenberg JA (2000) 5'-nicked apurinic/apyrimidinic sites are resistant to

beta-elimination by beta-polymerase and are persistent in human cultured cells after oxidative

stress. J Biol Chem 275:5323-5328.

Osterdahl BG and Slorach S (1988) Tobacco-specific N-nitrosamines in the saliva of habitual

male snuff dippers. Food Additives & Contaminants 5:581-586.

Patel PR, Bevan RJ, Mistry N and Lunec J (2007) Evidence of oligonucleotides containing 8-

hydroxy-2'-deoxyguanosine in human urine. Free Radical Biology and Medicine 42:552-558.

Patten CJ, Smith TJ, Murphy SE, Wang M-H, Lee J, Tynes RE, Koch P and Yang CS (1996)

Kinetic analysis of the activation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone by

heterologously expressed human P450 enzymes and the effect of P450-specific chemical

inhibitors on this activation in human liver microsomes. Arch Biochem Biophys 333:127-138.

Paz-Elizur T, Ben-Yosef R, Elinger D, Vexler A, Krupsky M, Berrebi A, Shani A, Schechtman E,

Freedman L and Livneh Z (2006) Reduced repair of the oxidative 8-oxoguanine DNA damage

and risk of head and neck cancer. Cancer Res 66:11683-11689.

Paz-Elizur T, Krupsky M, Blumenstein S, Elinger D, Schechtman E and Livneh Z (2003) DNA

repair activity for oxidative damage and risk of lung cancer. J Natl Cancer Inst 95:1312-1319.

Paz-Elizur T, Sevilya Z, Leitner-Dagan Y, Elinger D, Roisman LC and Livneh Z (2008) DNA

repair of oxidative DNA damage in human carcinogenesis: potential application for cancer risk

assessment and prevention. Cancer Lett 266:60-72.

Petermann E, Ziegler M and Oei SL (2003) ATP-dependent selection between single nucleotide

and long patch base excision repair. DNA Repair 2:1101-1114.

Peterson LA and Hecht SS (1991) O6-Methylguanine is a critical determinant of 4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone tumorigenesis in A/J mouse lung. Cancer Res

51:5557-5564.

Page 89: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

77

Peterson LA, Thomson NM, Crankshaw DL, Donaldson EE and Kenney PJ (2001) Interactions

between methylating and pyridyloxobutylating agents in A/J mouse lungs: implications for 4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced lung tumorigenesis. Cancer Res 61:5757-

5763.

Peterson LA, Ng DK, Stearns RA and Hecht SS (1994) Formation of NADP(H) Analogs of

Tobacco-Specific Nitrosamines in Rat Liver and Pancreatic Microsomes. Chem Res Toxicol

7:599-608.

Pitot HC, Goldsworthy T and Moran S (1981) The natural history of carcinogenesis: implications

of experimental carcinogenesis in the genesis of human cancer. Journal of Supramolecular

Structure & Cellular Biochemistry 17:133-146.

Prokopczyk B, Cox JE, Hoffmann D and Waggoner SE (1997) Identification of tobacco-specific

carcinogen in the cervical mucus of smokers and nonsmokers. J Natl Cancer Inst 89:868-873.

Prokopczyk B, Rivenson A and Hoffmann D (1991) A study of betel quid carcinogenesis IX.

Comparative carcinogenicity of 3-(methylnitrosamino)propionitrile and 4-(methylnitrosamino)-1-

(3-pyridyl)-1-butanone upon local application to mouse skin and rat oral mucosa. Cancer Lett

60:153-157.

Prokopczyk B, Trushin N, Leszczynska J, Waggoner SE and El Bayoumy K (2001) Human

cervical tissue metabolizes the tobacco-specific nitrosamine, 4- (methylnitrosamino)-1-(3-

pyridyl)-1-butanone, via alpha-hydroxylation and carbonyl reduction pathways. Carcinogenesis

22:107-114.

Puntarulo S and Cederbaum AI (1998) Production of Reactive Oxygen Species by Microsomes

Enriched in Specific Human Cytochrome P450 Enzymes. Free Radical Biology and Medicine

24:1324-1330.

Ravanat JL (2005a) Measuring Oxidized DNA Lesions as Biomarkers of Oxidative Stress : An

Analytical Challenge. FABAD Journal of Pharmaceutical Sciences 30:100-113.

Ravanat JL (2005b) Oxidized DNA Lesions as Biomarkers of Oxidative Stress : An Analytical

Challenge. FABAD Journal of Pharmaceutical Sciences 30:100-113.

Reardon JT, Bessho T, Kung HC, Bolton PH and Sancar A (1997) In vitro repair of oxidative

DNA damage by human nucleotide excision repair system: possible explanation for

neurodegeneration in xeroderma pigmentosum patients. Proc Natl Acad Sci U S A 94:9463-9468.

Rice JM, Diwan BA, Hu H, Ward JM, Nims RW and Lubet RA (1994) Enhancement of

hepatocarcinogenesis and induction of specific cytochrome P450-dependent monooxygenase

activities by the barbiturates allobarbital, aprobarbital, pentobarbital, secobarbital and 5-phenyl-

and 5-ethylbarbituric acids. Carcinogenesis 15:395-402.

Rivenson A, Hoffmann D, Prokopczyk B, Amin S and Hecht SS (1988) Induction of lung and

exocrine pancreas tumors in F344 rats by tobacco-specific and Areca-derived N-nitrosamines.

Cancer Res 48:6912-6917.

Page 90: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

78

Robertson AB, Klungland A, Rognes T and Leiros I (2009) DNA repair in mammalian cells:

Base excision repair: the long and short of it. Cellular & Molecular Life Sciences 66:981-993.

Ronai ZA, Gradia S, Peterson LA and Hecht SS (1993) G to A transitions and G to T

transversions in codon 12 of the Ki-ras oncogene isolated from mouse lung tumors induced by 4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and related DNA methylating and

pyridyloxobutylating agents. Carcinogenesis 14:2419-2422.

Rosa JG, Prokopczyk B, Desai DH, Amin SG and el-Bayoumy K (1998) Elevated 8-hydroxy-2'-

deoxyguanosine levels in lung DNA of A/J mice and F344 rats treated with 4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone and inhibition by dietary 1,4-

phenylenebis(methylene)selenocyanate. Carcinogenesis 19:1783-1788.

Russo MT, De Luca G, Degan P, Parlanti E, Dogliotti E, Barnes DE, Lindahl T, Yang H, Miller

JH and Bignami M (2004) Accumulation of the Oxidative Base Lesion 8-Hydroxyguanine in

DNA of Tumor-Prone Mice Defective in Both the Myh and Ogg1 DNA Glycosylases. Cancer

Res 64:4411-4414.

Sakamoto K, Tominaga Y, Yamauchi K, Nakatsu Y, Sakumi K, Yoshiyama K, Egashira A, Kura

S, Yao T, Tsuneyoshi M, Maki H, Nakabeppu Y and Tsuzuki T (2007) MUTYH-null mice are

susceptible to spontaneous and oxidative stress induced intestinal tumorigenesis. Cancer Res

67:6599-6604.

Sakumi K, Tominaga Y, Furuichi M, Xu P, Tsuzuki T, Sekiguchi M and Nakabeppu Y (2003)

Ogg1 knockout-associated lung tumorigenesis and its suppression by Mth1 gene disruption.

Cancer Res 63:902-905.

Salles B, Frit P, Provot C, Jaeg JP and Calsou P (1995) In vitro eukaryotic DNA excision repair

assays: an overview. Biochimie 77:796-802.

Sattler U, Frit P, Salles B and Calsou P (2003) Long-patch DNA repair synthesis during base

excision repair in mammalian cells. EMBO Reports 4:363-367.

Schneider JE, Price S, Maidt L, Gutteridge JM and Floyd RA (1990) Methylene blue plus light

mediates 8-hydroxy 2'-deoxyguanosine formation in DNA preferentially over strand breakage.

Nucleic Acids Research 18:631-635.

Schwartz AG (2004) Genetic predisposition to lung cancer. Chest 125:Suppl-9S.

Sekido Y, Fong KM and Minna JD (1998) Progress in understanding the molecular pathogenesis

of human lung cancer. Biochim Biophys Acta 1378:F21-F59.

Sekido Y, Fong KM and Minna JD (2003) Molecular genetics of lung cancer. Annual Review of

Medicine 54:73-87.

Slebos RJC, Hruban RH, Dalesio O, Mooi WJ, Offerhaus GJA and Rodenhuis S (1991)

Relationship between K-ras oncogene activation and smoking in adenocarcinoma of the human

lung. J Natl Cancer Inst 83:1024-1027.

Page 91: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

79

Smith GBJ, Bend JR, Bedard LL, Reid KR, Petsikas D and Massey TE (2003) Biotransformation

of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in peripheral human lung microsomes.

Drug Metab Dispos 31:1134-1141.

Smith TJ, Guo Z, Guengerich FP and Yang CS (1996) Metabolism of 4-(methylnitrosamino)-1-

(3-pyridyl)-1-butanone (NNK) by human cytochrome P450 1A2 and its inhibition by phenethyl

isothiocyanate. Carcinogenesis 17:809-813.

Smith TJ, Stoner GD and Yang CS (1995) Activation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-

butanone (NNK) in human lung microsomes by cytochromes p450, lipoxygenase, and

hydroperoxides. Cancer Res 55:5566-5573.

Spiegelhalder B and Bartsch H (1996) Tobacco-specific nitrosamines. Eur J Cancer Prev

5:Suppl-8.

Srivastava DK, Berg BJ, Prasad R, Molina JT, Beard WA, Tomkinson AE and Wilson SH (1998)

Mammalian abasic site base excision repair. Identification of the reaction sequence and rate-

determining steps. J Biol Chem 273:21203-21209.

Steinacher R and Schär P (2005) Functionality of Human Thymine DNA Glycosylase Requires

SUMO-Regulated Changes in Protein Conformation. Current Biology 15:616-623.

Stoner GD (1998) Introduction to mouse lung tumorigenesis. Exp Lung Res 24:375-383.

Stucki M, Pascucci B, Parlanti E, Fortini P, Wilson SH, Hubscher U and Dogliotti E (1998)

Mammalian base excision repair by DNA polymerases delta and epsilon. onc 17:835-843.

Su T, Bao Z, Zhang QY, Smith TJ, Hong JY and Ding X (2000) Human cytochrome P450

CYP2A13: predominant expression in the respiratory tract and its high efficiency metabolic

activation of a tobacco-specific carcinogen, 4-(methylnitrosamino)-1-(3-pyridyl)-1- butanone.

Cancer Res 60:5074-5079.

Subramanian J, and Govindan R (2007) Lung cancer in never smokers: a review. J Clin Oncol

25:561-70.

Tjaelve H (1991) The tissue distribution and the tissue specificity of bioactivation of some

tobacco-specific and some other N- nitrosamines. CRC Crit Rev Toxicol 21:265-294.

Travis WD, Brambilla E, Muller-Hermelink HK and Harris CC (2004) Pathology and genetics of

tumours of the lung, pleura, thymus and heart . Lyon : IARC Press, Oxford : Oxford University

Press .

Vogel U, Nexo BA, Wallin H, Overvad K, Tjonneland A and Raaschou-Nielsen O (2004) No

association between base excision repair gene polymorphisms and risk of lung cancer.

Biochemical Genetics 42:453-460.

Page 92: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

80

Wang L, Spratt TE, Liu X-K, Hecht SS, Pegg AE and Peterson LA (1997) Pyridyloxobutyl

adduct O6-[4-oxo-4-(3-pyridyl)butyl]guanine is present in 4-(acetoxymethylnitrosamino)-1-(3-

pyridyl)-1-butanone-treated DNA and is a substrate for O6-alkylguanine-DNA alkyltransferase.

Chem Res Toxicol 10:562-567.

Wei Q and Spitz MR (1997) The role of DNA repair capacity in susceptibility to lung cancer: a

review. Cancer Metastasis Rev 16:295-307.

Weitberg AB and Corvese D (1993) Oxygen radicals potentiate the genetic toxicity of tobacco-

specific nitrosamines. Clin Genet 43:88-91.

Westra WH, Slebos RJ, Offerhaus GJ, Goodman SN, Evers SG, Kensler TW, Askin FB,

Rodenhuis S and Hruban RH (1993) K-ras oncogene activation in lung adenocarcinomas from

former smokers. Evidence that K-ras mutations are an early and irreversible event in the

development of adenocarcinoma of the lung. Cancer 72:432-438.

Wilson VL, Weston A, Manchester DK, Trivers GE, Roberts DW, Kadlubar FF, Wild CP,

Montesano R, Willey JC and Mann DL (1989) Alkyl and aryl carcinogen adducts detected in

human peripheral lung. Carcinogenesis 10:2149-2153.

Wiseman H and Halliwell B (1996) Damage to DNA by reactive oxygen and nitrogen species:

role in inflammatory disease and progression to cancer. Biochem J 313 ( Pt 1):17-29.

Wood AJJ, Spira A and Ettinger DS (2004) Multidisciplinary Management of Lung Cancer. New

Eng J Med 350:379-392.

Wood RD, Biggerstaff M and Shivji MKK (1995) Detection and Measurement of Nucleotide

Excision Repair Synthesis by Mammalian Cell Extracts in Vitro. Methods: A Companion to

Methods in Enzymology 7:163-175.

Wood RD, Robins P and Lindahl T (1988) Complementation of the xeroderma pigmentosum

DNA repair defect in cell- free extracts. Cell 53:97-106.

World Health Organization. (2010). Tobacco Free Initiative. www.who.int/tobacco/en/.

Accessed: March 2010.

Wynder EL and Hoffmann D (1994) Smoking and lung cancer: scientific challenges and

opportunities. Cancer Res 54:5284-5295.

Xie Y, Yang H, Cunanan C, Okamoto K, Shibata D, Pan J, Barnes DE, Lindahl T, McIlhatton M,

Fishel R and Miller JH (2004) Deficiencies in mouse Myh and Ogg1 result in tumor

predisposition and G to T mutations in codon 12 of the K-ras oncogene in lung tumors. Cancer

Res 64:3096-3102.

Xu Y, Ho CT, Amin SG, Han C and Chung FL (1992) Inhibition of tobacco-specific nitrosamine-

induced lung tumorigenesis in A/J mice by green tea and its major polyphenol as antioxidants.

Cancer Res 52:3875-3879.

Page 93: DNA OXIDATION AND BASE EXCISION REPAIR IN LUNG AND …

81

Yang CS and Smith TJ (1996) Mechanisms of nitrosamine bioactivation and carcinogenesis.

Advances in experimental medicine and biology 387:385-394.

You M, Wang D, Liu P, Vikis H, James M, Lu Y, Wang Y, Wang M, Chen Q, Jia D, Liu Y, Wen

W, Yang P, Sun Z, Pinney SM, Zheng W, Shu XO, Long J, Gao YT, Xiang YB, Chow WH,

Rothman N, Petersen GM, de Andrade M, Wu Y, Cunningham JM, Wiest JS, Fain PR, Schwartz

AG, Girard L, Gazdar A, Gaba C, Rothschild H, Mandal D, Coons T, Lee J, Kupert E, Seminara

D, Minna J, Bailey-Wilson JE, Amos CI and Anderson MW (2009) Fine Mapping of

Chromosome 6q23-25 Region in Familial Lung Cancer Families Reveals RGS17 as a Likely

Candidate Gene. Clinical Cancer Research 15:2666-2674.

You M, Wang Y, Nash B and Stoner GD (1993) SHORT COMMUNICATION: K-ras mutations

in benzotrichloride-induced lung tumors of A/J mice. Carcinogenesis 14:1247-1249.

Yu BP (1994) Cellular defenses against damage from reactive oxygen species. Physiological

Reviews 74:139-162.

Zhang H, Xu Y, Kamendulis LM and Klaunig JE (2000) Morphological transformation by 8-

hydroxy-2'-deoxyguanosine in Syrian hamster embryo (SHE) cells. Toxicological Sciences

56:303-312.

Zhang YT, Zheng QS, Pan J and Zheng RL (2004) Oxidative damage of biomolecules in mouse

liver induced by morphine and protected by antioxidants. Basic Clin Pharmacol Toxicol 95:53-

58.

Zhang Z, Wang Y, Herzog CR, Liu G, Lee HW, DePinho RA and You M (2002) A strong

candidate gene for the Papg1 locus on mouse chromosome 4 affecting lung tumor progression.

onc 21:5960-5966.

Zhou J, Ahn J, Wilson SH and Prives C (2001) A role for p53 in base excision repair. EMBO J

20:914-923.