<<

DEVELOPMENT OF NOVEL SYNTHETIC ROUTES TO THE

EPOXYKETOOCTADECANOIC ACIDS (EKODES) AND THEIR

BIOLOGICAL EVALUATION AS ACTIVATORS OF THE PPAR FAMILY

OF NUCLEAR RECEPTORS

By

ROOZBEH ESKANDARI

Submitted in partial fulfillment of the requirements for

The Degree of Doctor of Philosophy

Thesis Advisor: Gregory P. Tochtrop, Ph.D.

Department of Chemistry

CASE WESTERN RESERVE UNIVERSITY

January, 2016 CASE WESTERN RESERVE UNIVERSITY

SCHOOL OF GRADUATE STUDIES

We hereby approve the thesis/dissertation of

ROOZBEH ESKANDARI

Candidate for the Ph.D degree *.

(signed) Anthony J. Pearson, PhD (Chair of the committee)

Gregory P. Tochtrop, PhD (Advisor)

Michael G. Zagorski, PhD

Blanton S. Tolbert, PhD

Witold K. Surewicz, PhD (Department of and Biophysics)

(date) 14th July, 2015

*We also certify that written approval has been obtained for any proprietary material contained therein.

I dedicate this work to my sister Table of Contents

Table of Contents ...... i

List of Tables ...... vi

List of Figures ...... vii

List of Schemes ...... ix

Acknowledgements ...... xi

List or Abbreviations ...... xiii

Abstract ...... xxiii

CHAPTER 1 General Introduction ...... 1

1.1 Discovery of ...... 1

1.2 The role of synthesis in the advancement of prostaglandins research ...... 3

1.3 Discovery of leukotrienes ...... 6

1.4 The role of synthesis in the advancement of leukotrienes research ...... 6

1.5 Non-enzymatic oxidation of PUFAs ...... 8

1.6 The role of synthesis in the advancement of LPO products ...... 10

1.7 Formation of LPO products ...... 12

1.8 (OS) ...... 16

1.8.1 Formation of /reactive nitrogen species (ROS/RNS)

...... 17

1.8.2 Reactive oxygen species (ROS) ...... 17

i

1.8.2.1 Singlet oxygen ...... 17

1.8.2.2 Superoxide radical ...... 18

1.8.2.3 Hydrogen peroxide ...... 19

1.8.2.4 Hydroxyl radical...... 19

1.8.3 Reactive nitrogen species (RNS) ...... 19

1.8.3.1 Nitric oxide (NO.) ...... 19

1.8.3.2 Peroxynitrite (ONOO-) ...... 19

1.8.4 Reactivity of ROS/RNS...... 20

1.9. Biological roles of oxidized lipids ...... 22

1.9.1 Oxidized lipid and G -coupled receptors (GPCRs) ...... 23

1.9.2 Oxidized lipid and -proliferator-activated receptors

(PPARs) ...... 24

1.10 References ...... 26

CHAPTER 2 Development of novel synthetic routes to the epoxyketooctadecenoic acids (EKODEs) ...... 31

2.1 Introduction ...... 31

2.2 EKODE structure and nomenclature ...... 34

2.3 EKODE-(E)-I family ...... 35

2.3.1 Retrosynthetic analysis of trans-EKODE-(E)- I ...... 36

2.3.2 Synthesis of trans-EKODE-(E)- Ia ...... 37

ii

2.3.3 Synthesis of trans-EKODE-(E)- Ib ...... 38

2.3.4 Retrosynthetic analysis of cis-EKODE-(E)- Ib ...... 40

2.3.5 Synthesis of cis-EKODE-(E)- Ib ...... 40

2.4 EKODE-(E)-II family ...... 42

2.4.1 Retrosynthetic analysis of trans-EKODE-(E)-II ...... 44

2.4.2 Synthesis of bifunctional conjunctive ylide ...... 45

2.4.3 Synthesis of trans-EKODE-(E)-II ...... 47

2.4.4 Divergent synthesis for diverse library of trans-EKODE-(E)-II

...... 49

2.4.4.1 Synthesis of alkyne-end aldehyde ...... 49

2.4.4.2 Synthesis of deuterated labeled aldehyde ...... 49

2.5 Experimental part ...... 52

2.5.1 General experimental details ...... 52

2.5.2 Syntheses ...... 54

2.6 References ...... 88

Chapter 3. Biological evaluation of EKODEs as potential activators of the PPAR family of nuclear receptors ...... 91

3.1 Peroxisome proliferator-activated receptors (PPARs) structure and activity ..... 91

3.2. The physiological functions of the PPARs ...... 95

3.2.1 Functional role of PPARα ...... 95

iii

3.2.1.1 Exogenous ligands (synthetic Xenobiotics) of PPARα ...... 95

3.2.1.2 Endogenous ligands (biological molecules) of PPARα ...... 96

3.2.2 Functional role of PPARɣ ...... 97

3.2.2.1 Exogenous ligands of PPARɣ ...... 98

3.2.2.2 Endogenous ligands of PPARɣ ...... 99

3.2.3. Functional role of PPARβ/δ ...... 101

3.2.3.1 Exogenous ligands of PPARβ/δ ...... 102

3.2.3.2 Endogenous ligands of PPARβ/δ ...... 102

3.3 EKODEs as potential endogenous metabolites ...... 103

3.4 Target reporter assay ...... 105

3.4.1 Bioluminescence ...... 105

3.4.2 Cell-based reporter assay ...... 105

3.4.3 Single luciferase reporter assay ...... 106

3.4.4 Luciferase enzyme mechanism ...... 107

3.4.5 Colorimetric β-galactosidase assay ...... 108

3.5 Experimental ...... 110

3.5.1 Materials ...... 110

3.5.2 Cell transient transfection assay ...... 110

3.5.3 Methods ...... 111

3.5.3.1 Seeding of COS-7 cells ...... 111

3.5.3.2 Transfection of COS-7 cells ...... 111

iv

3.5.2.3 Treatment of transfected cells ...... 112

3.5.2.4 Cell lysis ...... 112

3.5.2.5 Luciferase assay, data normalization ...... 112

3.6 Results and discussion ...... 113

3.7 Conclusion ...... 116

3.8 References ...... 118

Chapter 4. Thesis summary and future directions ...... 122

4.1 Thesis summary ...... 122

4.2 Future directions ...... 125

4.3 References ...... 127

APPENDIX NMR spectra of synthesized molecules ...... 128

BIBLIOGRAPHY ...... 181

v

List of Tables

Table 1.1 Physicochemical properties of reactive species (ROS/RNS), NA: data is not available ...... 20

Table 2.1 Reaction of sulfonium ylide and aldehydes, * yields were calculated based of amount obtained after purification ...... 50

Table 2.2 Reaction of phosphonium ylide and aldehydes.* yields were calculated based of amount obtained after purification ...... 51

vi

List of Figures

Figure 1.1 Non-enzymatic oxidized metabolites of PUFAs ...... 9

Figure 1.2 The molecular structure of linoleate and arachidonate ...... 13

Figure 1.3 Generation of different ROS/RNS and their subsequent reactions ...... 17

.- Figure 1.4 Standard electrode potential for superoxide anion radical (O2 ) and

. hydroperoxyl radical (HO2 ) ...... 21

Figure 1.5 Nernst equations can be used as an estimate to measure potential at known concentration and pH ...... 21

Figure 1.6 Lipid peroxidation products and their signaling through GPCR ...... 24

Figure 1.7 Lipid peroxidation products interact with PPAR and activate their expression machinery ...... 25

Figure 2.1 Linoleic acid oxidation through non-enzymatic reactions ...... 33

Figure 2.2 EKODE I and II structures and nomenclature ...... 34

Figure 3.1 PPARs motifs, conserved region in yellow, variable regions in red ...... 92

Figure 3.2 PPAR binding to ligand along with RXR binding to 9-cis retinoic acid (9cRA) forms a complex which provides an on- off-switch for gene expression ...... 93

Figure 3.3 Three main mechanisms proposed for negative regulation of transcription factors by PPARs ...... 94

Figure 3.4 Synthetic molecules and xenobiotics acting as PPAR-α ligands ...... 96

Figure 3.5 Endogenous (biological molecules) acting as PPAR-α ligands ...... 97

Figure 3.6 Synthetic molecules acting as PPARγ ligands ...... 99

vii

Figure 3.7 Endogenous molecules acting as PPARγ ligands ...... 100

Figure 3.8 Phospholipids which act as endogenous molecules acting as PPARγ ligands

...... 101

Figure 3.9 Synthetic molecules acting as PPARβ/δ ligands ...... 102

Figure 3.10 Endogenous molecules acting as PPARβ/δ ligands ...... 103

Figure 3.11 Illustration of the basic principle of the single luciferase reporter assay for

PPAR ...... 106

Figure 3.12 Reaction catalyzed by firefly luciferase emits light through bioluminescence process ...... 108

Figure 3.13 Colorimetric assay of β-galactosidase using ONPG as the substrate ...... 109

Figure 3.14 Relative light units (RLU) determines activity of PUFAs and lipid metabolites on mouse PPARα by luciferase assay. 1X(5μM), 2X(10μM), 4X(20μM).

Wy14635 is the synthetic ligand ...... 113

Figure 3.15 Relative light units (RLU) determines activity of PUFAs and lipid metabolites on mouse PPARγ by luciferase assay. 1X(5μM), 2X(10μM), 4X(20μM) .

Troglitazone is the synthetic ligand ...... 114

Figure 3.16 Relative light units (RLU) determines activity of PUFAs and lipid metabolites on mouse PPARβ/δ by luciferase assay. 1X(5μM), 2X(10μM), 4X(20μM).

GW0742 is the synthetic ligand ...... 115

Figure 3.17 Relative light units (RLU) determines trans-EKODE-(E)-IIb activation of mouse PPARβ/δ in compare to GW0742 ...... 116

viii

List of Schemes

Scheme 1.1 General synthetic route for Corey lactone aldehyde ...... 4

Scheme 1.2 Synthetic route for E2and F2α ...... 5

Scheme 1.3 Synthetic route for leukotriene A4 (LTA4) ...... 7

Scheme 1.4 Synthetic route for 15-F2c-IsoP ...... 10

Scheme 1.5 Synthetic route for 8-epi-SC-Δ13-9-IsoF ...... 12

Scheme 1.6 Non-enzymatic formation of 15-F2c-IsoP ...... 14

Scheme 1.7 Non-enzymatic formation of 8-epi-SC-Δ13-9-IsoF ...... 15

Scheme 2.1 Non-enzymatic formation of EKODE-I family ...... 35

Scheme 2.2 Retrosynthesis analysis of trans-EKODE-(E)-I ...... 36

Scheme 2.3 Synthetic route for trans-EKODE-(E)-Ia ...... 38

Scheme 2.4 Synthetic route for trans-EKODE-(E)-Ib ...... 39

Scheme 2.5 Retrosynthesis analysis of cis-epoxy aldehyde ...... 40

Scheme 2.6 Synthetic route for cis-EKODE-(E)-Ib ...... 41

Scheme 2.7 Non-enzymatic formation of EKODE-II family ...... 42

Scheme 2.8 Different synthetic strategies in order to make intermediate X ...... 43

Scheme 2.9 Retrosynthesis analysis of trans-EKODE-(E)-II ...... 45

Scheme 2.10 Synthetic rout for bifunctional conjunctive ylide ...... 46

Scheme 2.11 General strategy for synthesis of trans-EKODE-II family ...... 47

Scheme 2.12 Synthetic route for trans-EKODE-(E)-IIa ...... 48

Scheme 2.13 Synthetic route for trans-EKODE-(E)-IIa an b ...... 48

ix

Scheme 2.14 Synthetic route for alkyne-end aldehyde ...... 49

Scheme 2.15 Synthetic route for deuterated-labeled aldehyde ...... 50

Scheme 4.1 At low temperature (0 oC), sulfonium ylide reacts faster than phosphonium

(k1>>k2) ...... 123

Scheme 4.2 Sulfonium ylide reaction is faster than phosphonium one ...... 124

Scheme 4.3 General strategy for phospholipid synthesis ...... 126

x

Acknowledgements

I would like to express my genuine appreciation to my advisor, Prof. Gregory P.

Tochtrop, for his inherent enthusiasm towards science education and sharing with me much of this feeling with me, and also encouraging me to take risks and pursue critical thinking as I grew into an independent scientist in his lab. All of these would not have been achieved without his patience.

I would like to thank the Department of Chemistry, Case Western Reserve

University and the National Science Foundation for financial support.

I also would like to thank my committee member, Prof. Anthony J. Pearson, Prof.

Michael G. Zagorski, Prof. Blanton S. Tolbert and Prof. Witold K. Surewicz, for precious time and effort put into my thesis.

I am so fortunate to study in such a great lab with individuals whose interest in revealing the fundamental aspects of science was inspiring. My best memories in the lab would not happened without: Dr. Emily C. Barker, Dr. Tonibelle Gatbonton-Schwager,

Dr. Yong Han, Jeremy P. Hess , Dr. Vasily A. Ignatenko, Dr. Qingjiang Li, Anna

Owensby, Jeffrey Rabinowitz, Dr. Sushabhan Sadhukhan, Chuan Shi, Elizabeth A.

Stewart, Dr. Brian S. Werry and Dr. Jianye Zhang. Especially, I am grateful to my lifelong friend Mohsen Badiee who happened to work in this lab for most of these years.

I am also thankful to Prof. Noa Noy and her students Dr. Liraz Levi and Mary K. Doud for their insightful comments and the lab space and instruments they provided for me in the Cleveland Clinic Lerner Research Institute. I am also grateful to Prof. Krzysztof

xi

Palczewski and his student Dr. Yuanyuan Chen for their great help when using the instruments in the Department at Case Western Reserve University.

Last but not least, my parents, Masoomeh and Mohammad Hassan, receive the deepest gratitude for teaching me how to enjoy being myself and providing me with love and support through all these years. I also express my sincere gratitude to my dear sister,

Ferdows, and brother-in-law, Jamshid, for being such great friends and inspiring me to do hard work.

xii

List of Symbols and Abbreviations

°C The degree Celsius

AA (arachidonate)

AF1 Activation function 1

AF2 Activation function 2

ANT Adenine nucleotide translocator

ATCC American Type Culture Collection

ATP Adenosine triphosphate

Azelaoyl PAF 1-O-hexadecyl-2-O-(9-carboxyoctanoyl)-sn-glyceryl-3- phosphocholine

α Alpha bp

β Beta

βgal Beta-galactosidase

CBP CREB binding protein

CCD charge-coupled device

CDCl3 Deuterated chloroform

CH2Cl2 Dichloromethane

CH3CN Acetonitrile

10e12zCLA 10(E),12(Z)-Conjugated linoleic

xiii

COUP-TFII chicken ovalbumin upstream promoter-transcription factor

II

CREB cAMP response element binding

CVD Cardiovascular diseases d Doublet

D2O Deuterium oxide

DBD DNA binding domain

DCM Dichloromethane dd Doublet of doublets ddd Doublet of doublets of doublets

DEHA di-(2-ethylhexyl)

DEHP di-(2-ethylhexyl)-

DHA Docosahexaenoic acid (docosahexaenoate)

DHKODE Dihydroxyletooctadecanoic acid

DMEM Dubelco's Modified Eagle Medium

DMSO Dimethyl sulfoxide

DNA Deoxyribonucleic Acid

DODE 9-12,-dioxo-10(E)-dodecenoic acid dt Doublet of triplets

δ delta

E0 Standard electrode potential

xiv

E’ Electrode potential

EI Electron impact

EKODE Epoxyketooctadecenoic acids

EPA Eicosapentaenic acid (Eicosapentaenoate) eq Equivalent

EtOAc Ethyl acetate

ESI Electrospray ionization

FA

FABP Fatty acid binding protein

FAS Fatty acid synthase

Fs Femtosecond

FT-ICR Fourier transform - Ion cyclotron resonance g Gram

16:0/18:1-GPC 1-palmitoyl-2-oleoyl-sn-glycerol-3-phosphocholine

GPCR G-protein coupled receptor

GFP Green fluorescent

γ Gamma h hour(s)

HDAC Histone deacetylase

8(S)-HETE 8(S)-Hydroxy-(5Z,9E,11Z,14Z)-eicosatetraenoic acid

15(S)-HETE 15(S)-Hydroxy-5Z,8Z,11Z,13E-eicosatetraenoic

xv

HKODE Hydroxyketooctadecanoic acid

HMPA Hexamethylphosphoramide

4-HNE 4-Hydroxynonenal

HNF4 Hepatocyte nuclear factor 4

HO. Hydroxyl radical

. HO2 Hydroperoxyl radical

HODA 9-hydroxy-12-oxo-10(E)-dodecenoic acid

9-HODE 9-hydroxy-10E,12Z-octadecadienoic acid

13-HODE 13-hydroxy-9Z,11E-octadecadienoic

HOMO Highest Occupied Molecular Orbital

HPLC High-pressure liquid chromatography

9HpODE 9-hydroperoxy-10E,12Z-octadecadienoic acid

13HpODE 13-hydroperoxy-9Z,11E-octadecadienoic acid

HRMS High-resolution mass spectrometry

Hsp Heat protein

HWE Horner–Wadsworth–Emmons (reaction)

IC50 Half maximal inhibitory concentration

IsoF Isofurane

IsoP Isoprostane

IsoTx Isothromboxane

IsoLG Isolevuglandin

xvi

Keap1 Kelch-like ECH-associated protein 1 kcal/mol Kilocalorie per mole

KODDE Ketooctadecadienoic acid

L Laevorotatory

LA Linoleic acid (linoleate)

LBD Ligand binding domain

LDA Lithium diisopropyl amide

Leukotriene B4 5S,12R-dihydroxy-6Z,8E,10E,14Z-eicosatetraenoic acid lit. Literature

LPA Lysophosphatidic acid

LPO Lipid peroxidation

LT Leukotrienes

LTQ Linear trap quadrupole

Luc Luciferase

M Molar (concentration) m/z Mass-to-charge ratio

MAPK Mitogen-activated protein kinase m-CPBA Meta-chloroperoxybenzoic acid

Me Methyl

MEHP Monoethylhexyl phthalate mg Milligram

xvii

MHz Megahertz min Minute(s) mL Milliliter mm Millimeter mmol Millimole

Mp

Ms Millisecond nm Nanometer n-BuLi n-butyllithium

NCoR corepressor

NeuroK Neuroketal

NeuroP Neuroprostane

NFAT Nuclear factor of activated T-cells

NFκB Nuclear factor-κB

NitroLA 10-Nitrolinoleate

NMR Nuclear magnetic resonance

NO. Nitrogen oxide

. NO2 Nitrogen dioxide

NOS NO synthases

Nrf2 NF-E2 p45-related factor 2

NSAID Nonsteroidal anti-inflammatory drug

xviii

.- O2 Superoxide anion radical

OAP Oxidative addition product

OCP Oxidative cleavage product

4-ONE 4-Oxo Nonenal

ONPG ortho-Nitrophenyl-β-galactoside

ONP ortho-nitrophenol

ONOO− Peroxynitrite

− ONOOCO2 Nitrosoperoxycarbonate

ONOOH Peroxynitrous acid

OS Oxidative Stress

OUEA 11-oxoudec-9(E)-enoic acid oxLDL Oxidized low- lipoproteins

PCC Pyridinium Chlorochromate

Pd Palladium

PG Prostaglandin

PGA1 Prostaglandin A1

PGA2 Prostaglandin A2

PGD2 Prostaglandin D2

PGI2 Prostaglandin I2 pH Potential hydrogen

PFOA

xix

PFOS Perfluorooctanesulfonic acid

PPh3 Triphenylphosphine

PMA Phosphomolybdic acid ppm Parts per million

PPAR Peroxisome proliferator-activated receptors

PPL porcine pancreas lipase

PPRE Peroxisome proliferator response elements

PTM post- translational modification

PUFA Polyunsaturated fatty acid

PVC

RGS4 Regulator of g-protein signaling 4

RLU Relative light units

RNA-seq RNA Sequencing

RNS Reactive nitrogen species

ROS Reactive oxygen species

RT-PCR Reverse transcription polymerase chain reaction

RXR Retinoid X receptor

SMRT Silencing mediator of retinoid and thyroid receptor

SRC1 receptor co-activator 1

SRSA Slow reacting substance of anaphylaxis

xx

STAT Signal transducer and activator of transcription t1/2 Half-life t-EKODE-Ia trans-EKODE-( E)-Ia t-EKODE-Ib trans-EKODE-( E)-Ib t-EKODE-IIa trans-EKODE-( E)-IIa t-EKODE-IIb trans-EKODE-( E)-IIb

THF Tetrahydrofuran

Torr millimetre of mercury

TLC Thin-layer chromatography

TX Thromboxane

TP Thromboxane A2-Prostanoid

TZD

UV Ultraviolet

δ Delta, chemical shift

λmax Lambda maximum (wavelength)

μg Microgram

μm Micrometer (distance)

μs Microsecond

μM Micromolar (concentration)

WADA World Anti-Doping Agency

ω Omega

xxi

X-ray X-radiation

− X³Σg Triplet ground state

xxii

DEVELOPMENT OF NOVEL SYNTHETIC ROUTES TO THE

EPOXYKETOOCTADECANOIC ACIDS (EKODES) AND THEIR

BIOLOGICAL EVALUATION AS ACTIVATORS OF THE PPAR FAMILY

OF NUCLEAR RECEPTORS

Abstract

By

ROOZBEH ESKANDARI

Current drug screening processes help the scientific community develop therapeutics for a variety of specific targets and diseases, but the ability to identify an endogenous molecule or metabolite as a novel pharmacophore still remains an exciting area. In fact, identifying the putative target of endogenous metabolites can provide a deeper layer of understanding regarding cell physiology and regulatory feedback mechanisms. Furthermore, endogenous metabolites play a key role in developing diseases biomarkers and help to develop diagnostic tools.

Moreover, new scaffolds of metabolites can provide possibilities for developing novel therapeutics to treat related diseases, since their action is mostly unique toward a specific target in the cell.

In this regard, oxidized lipid metabolites formed by non-enzymatic reactions have not been systematically studied as signaling molecules. Among the variety of polyunsaturated fatty acids (PUFAs), a family of linoleic acid

xxiii metabolites called EKODEs has shown activities as endogenous metabolites.

Indeed, these molecules exhibit activities such as modulating corticosterone and aldosterone production levels. They also demonstrate antioxidant effects through the Nrf2-Keap1 signaling pathway.

As enthusiasm for understanding these physiological roles increased among different groups, the lack of synthetic methods for preparation of these molecules became more noticeable. This thesis represents a general strategy for providing a highly efficient synthesis for EKODE metabolites. Structurally, this family of metabolites posseses flanking α, β-unsaturated and epoxy functionalities centered on a ketone group. The synthetic method relies on developing a bifunctional intermediate containing two reactive ylides that can be used for generation of epoxy and olefin functionality in a sequential order. In fact, the two ylides kinetically differentiate between their reactivity with various electrophiles, such as aldehydes. This strategy can be used by others as a pipeline to generate a diverse family of active lipid metabolites with similar functionality.

This thesis also contains an initial biological evaluation of EKODEs, along with a number of other lipid peroxidation metabolites on the PPAR family of nuclear receptors. Our data suggest that these endogenous metabolites can regulate PPARβ/δ subtype at low micromolar concentration ranges. These receptors are specifically involved in the development of different chronic diseases, such as , atherosclerosis and cancer. The information presented in

xxiv this thesis can be used as the stepping stone to reveal the chemical physiology of these endogenously formed metabolites.

xxv

Chapter 1: General Introduction

Attempts to understand human physiology date back to at least 420 BC, to the time of Hippocrates when he proposed the idea of the four basic substances or humors. In

19th century, the number of physiological studies started to grow more rapidly, and in the

1850s a French physiologist named Claude Bernard coined the word Milieu intérieur

(internal environment), which was expanded to by an American physiologist

Walter B. Cannon in the 1920s. Cannon suggested that the human body is an open

system, exposed to dynamic changes from the environment, and it functions through

cooperating mechanisms to maintain a steady-state.1,2

Accordingly, scientists started to appreciate describing physiological phenomena

in a more detailed manner from a chemical point of view. In the 1930s, British

physiologist Henry H. Dale proposed the concept of autopharmacology while studying

interaction with muscles.3 Consequently, his discovery provided the foundations for his students to search for endogenous chemical substances that are involved in the regulation of physiology.

1.1 Discovery of prostaglandins

Among Dale’s students, Ulf von Euler, with his expertise and passion for science, discovered a lipid-soluble organic acid with hypotensive and muscle-stimulating actions in human semen. He called these molecules prostaglandin (PG), since the initial thought was that these molecules derived from prostate gland secretions. However, later it was found that the molecules secrete from seminal vesicles. Subsequently, similar molecules were discovered in other tissues with diverse physiological functions.4

1

The study in the field of PGs was partly halted by World War II. Eventually, in

1945 at a meeting at the Physiological Society of Karolinska Institute in Stockholm, von

Euler persuaded Sune Bergström, a lipid chemist, to analyze the structure of PGs. Due to

the lack of sensitive analytical methods, they required a large quantity of vesicular glands

from across the globe to obtain a sufficient amount of PGs to characterize the structures.

Bergström took advantage of stainless steel countercurrent extraction to purify the

samples, and he recognized them as unsaturated hydroxy acids in 1949.5

Improvements in chromatography techniques provided an ideal advantage for

6 purification and obtaining crystal forms of PGE1 and PGF1α. UV-spectroscopy and IR were among the first analytical methods used to determine the structure and quantity of these molecules. Subsequently, more sensitive techniques such as mass spectrometry and its combination with gas chromatography, developed by Ragnar Ryhage, provided the opportunity to characterize the formula and structure of PGs in pico- and nanogram

quantities in a variety of tissues.7

David van Dorp at Unilever research laboratory collaborated with Bergström to

investigate the transformation of isotopically labeled dihomogamma-linoleic acid to

PGE1 and PGF1α with homogenates of sheep glands. This result was critical, because it

confirmed that prostaglandins are oxidized forms of polyunsaturated fatty acids

(PUFAs).8 Subsequently, more detailed studies done with chemical and enzymatic

reactions provided ample information about the main scaffold, functional groups and

their arrangement in the structure of PGs. For example, oxidative ozonolysis mediated

fragmentation of the molecule to smaller pieces that could then be identified with other

2

9 methods, such as mass spectrometry. In addition, X-ray analysis of derivatives of PGF1α

by Sixten Abrahamsson revealed the first absolute configuration of PGs.10

The increasing number of biological applications of PGs, such as being local

, and mediators, vasomotor regulators and neuromodulators

in animals, urged scientists to look for enriched sources. A study of natural products

accidentally discovered that Gorgonia coral contains about 1.0-1.5% of derivative of

11 PGA2. Consequently, the Upjohn Company used coral as the main source to obtain different forms of prostaglandins. Although natural sources provided a variety of PG

structures, in the late 1960s the remarkable work of Elias J. Corey established efficient

synthetic approaches to generate larger quantities of these molecules. In the next section,

the advances in the field of prostaglandins through synthesis are discussed.

1.2 The role of synthesis in the advancement of prostaglandins research

The major hurdles in the synthesis of PGs were that these molecules are quite unstable and they exist in a variety of scaffolds. One of the key discoveries in Corey’s work was the design of a single intermediate, commonly known as the Corey lactone aldehyde.12 In other words, this intermediate was not only used to synthesize PGs, it was

also applied to the generation of PG analogues that were the main focus of the

pharmaceutical industries. This general synthetic route for Corey lactone aldehyde is

3

shown in Scheme 1.1.

Cl CN MeO NaH, THF KOH, H2O/ DMSO Cl o MeOCH Cl OMe Cu(BF4)2, 0 C 2 CN THF, -55 oC

COOH MeO MeO o m-CPBA, NaHCO3 1. NaOH, H2O, 0 C KI3, NaHCO3 o O O 2. CO2 OMe H2O, 0 C O HO

O O O O O O o 1. Ac2O, Py 1. BBr3, CH2Cl2, 0 C I n o OMe 2. ( -Bu)3SnH OMe 2. CrO3, 2Py, CH2Cl2, 0 C O ALBN,Benzene HO AcO AcO

Scheme 1.1 General synthetic route for Corey lactone aldehyde

Next, the Corey lactone aldehyde afforded olefin bonds after going through several steps including Wittig and Horner-Wadworth-Emmons (HWE) coupling.13 In

Scheme 1.2, the procedures for PGE2 and PGF2α are presented. Similar strategies can be applied to other members of PG1’s, PG2’s and PG3’s. These syntheses facilitated the evaluation of the physicochemical properties of PGs and assessment of their biological implications on a worldwide scale.

4

O O O O O O P (MeO)2 n-C 5H11 Zn(BH4)2, DME

O NaH,DME n-C5H11 AcO AcO O Corey lactone aldehyde

O O O O 1. of Separation 1. Dibal-H, toluene, -60 oC diastereomers n-C5H11 - , 2. COO 2. K2CO3 MeOH n-C5H11 AcO Ph3P OH 3. DHP,TsOH THPO DMSO CH2Cl2 OTHP

O COOH

HO OH HO PGE2 COOH

THPO OTHP HO COOH

HO OH

PGF2α

Scheme 1.2 Synthetic route for prostaglandin E2 and F2α

Meanwhile, many novel syntheses and purification methodologies were developed in order to achieve a higher yield for the generation of PGs. For example, in the preparation of PGs’ stereocenters, molecular robots opened new areas in the total synthesis. These robot-like assemblers act in multi-step processes, interacting with

5

reactants and reagents to provide a stereoselective environment to obtain the enantiomerically pure products.14,15

1.3 Discovery of leukotrienes

The research on metabolites of PUFAs became one of the major foci at the

Karolinska Institute. In 1977, Beng I. Samuelsson discovered that during the

inflammatory response in leukocytes, arachidonic acid transforms to novel scaffolds

called leukotrienes (LT). More detailed studies by his postdoctoral researcher, Pierre

Borgeat, proved that bioconversion of arachidonic acid to hydroperoxide 5-HPETE is the

16 key step in formation of LTA4. Consequently, LTA4 converts to other forms of LTs: reaction with hydrolase enzyme forms LTB4, reaction with glutathione forms LTC4 and etc. These metabolites play a key role in hypersensitivity reactions, such as asthma and

inflammation.17

1.4 The role of synthesis in the advancement of leukotrienes research

In collaboration with Corey’s lab for synthesizing LTs in large quantities,

Samuelsson mainly focused on studying this newly found family of oxidized lipids. LTA4

was first synthesized as the racemate and soon after, its chiral form generated from D-(-)-

6

ribose.18 (Scheme 1.3)

1. Ph3P COOEt OAc OBz O BzO OH PhCOOH, DME, ∆ BzO Zn/Hg, HCl COOEt Et O 2. Ac O,H SO 2 BzO OBz 2 2 4 OBz

OAc OTs BzO 1. H2, Pd/C. MeOH BzO K2CO3, MeOH COOEt COOMe 2. HCl, MeOH OBz OBz 3. TsCl,Py

1. CrO3.Py,CH2Cl2 Li 2. OEt H H O O THF, -78 oC COOMe COOMe HO o O 3. MsCl, Et3N, CH2Cl2, -45 C H H o 4. pH 7, -45 C to 0 0C

o O 1. HMPA, -78 C COOMe 2. n-BuLi, THF, -78 oC

n-C5H11 Ph3P LTA 4

Scheme 1.3 Synthetic route for leukotriene A4 (LTA4)

The stereocontrolled synthetic strategies for LTs were used to confirm the

structure and the link between 5-HPETE and other metabolites such as LTA4 and LTB4.

In addition, the biogenic components of the slow reacting substance of anaphylaxis

(SRSA) were proved by synthesis of the peptidic LTC4 and etc.

With the aid of novel synthetic approaches, a myriad of oxidized forms of PUFAs

were characterized and studied. Meanwhile, isotope-labeling and advances in molecular biology provided plenty of information for understanding the origin of oxygen atoms in 7

the oxidized lipids.19 Accordingly, the synthetic intermediates were necessary to

unambiguously establish the absolute stereochemistry and study the mechanisms of enzymatic reactions involved in the biosynthesis of PGs, LTs and etc.

1.5 Non-enzymatic oxidation of PUFAs

The long-standing paradigm was that the enzymatic reactions were the only source of oxidized lipids production. In the 1960s, scientists observed examples of oxidized lipids derived from oxidation of PUFAs without the aid of enzymes. 20 Next, in

the late 1970s Robert G. Salomon discovered that as an enzymatic intermediate PGH2 can

rearrange spontaneously to generate different forms of PGs and thromboxanes (TXs).21 In

the 1990s L. Jackson Roberts and coworkers discovered a number of other oxidized

PUFAs called isoprostanes (IsoPs), neuroprostanes (NeuroPs), isothromboxanes (IsoTxs),

isolevuglandin (IsoLGs), neuroketals (NeuroKs) and isofuranes (IsoFs) that were produced through reactive oxygen species (ROS).22-27 (Figure 1.1)

In general, the formation of these oxidized metabolites, either spontaneously or

through ROS, follows the conventional rules of chemistry. These processes allow the

generation of greater numbers of scaffolds while forming racemic mixtures of

stereoisomers. The non-enzymatic reactions on lipids are generally called lipid

peroxidation (LPO).

8

HO HO OH COOH COOH

HO OH HO 7-F4t-NeuroP 15-F2t-IsoP O

COOH O O OHC OH OH

15-A2-IsoTx Iso[4]LGE4(12-E2-IsoK)

OHC COOH HO COOH O OH

O OH OH -NeuroK) IsoFs Iso[9]LGD4(17-D4

Figure 1.1 Non-enzymatic oxidized metabolites of PUFAs

The fact that these metabolites exist in significant amounts in vivo and exhibit

biological activities encouraged the science community to discover their cognate

receptors. Biological activity of LPO products is tissue-specific. Many of these activities are attributed to signaling through Thromboxane A2-Prostanoid (TP) receptor, which is a

G protein-coupled receptor (GPCR). Also, they function through the nuclear receptors,

such as peroxisome proliferator-activated receptors (PPARs).28 In addition to the

biological activities on a variety of lipid targets, LPO products such as F2-IsoPs, F4-

NeuroP and isofurans were used as the gold standard of oxidative stress.29-31

Indeed, the application of LPO products in biological systems triggered a strong

research effort to develop synthetic methods for generating these molecules. Many of the

9

syntheses are similar to the ones developed for products of enzymatic reactions with modified steps towards developing new stereochemistry.32

1.6 The role of synthesis in the advancement of LPO products

One of the well-studied classes of LPO products are IsoPs. The first total syntheses for this family mostly relied on biomimetic routes through radical cyclization reactions. Interestingly, these methods were developed in 1984, even before the discovery of these molecules in the process of developing synthesis for enzymatic reaction products.33 In the 1990s, multiple research groups developed a variety of strategies to generate all-cis-Corey lactone aldehyde for the synthesis of specific stereoisomers.34 One

35 of these strategies for synthesis of 15-F2c-IsoP is shown in Scheme 1.4.

COOMe

O COOMe 1. NaBH4 PhSeCH(COOMe)2 O 2. NaOH/MeOH, DMSO 140 oC Benzene, Sunlamp O 300 W O SePh 3. LiHMDS, PBzCl

COOMe O COOMe O 5 steps O H2O2, acetone-water, PPTS

PBzO SePh O (see Scheme 1.2) PBzO

HO COOH

HO OH

15-F2c-IsoP

Scheme 1.4 Synthetic route for 15-F2c-IsoP 10

At high oxygen concentration in tissues, arachidonic acid oxidation mainly switches to a different class of molecules known as isofurans (IsoF). In Scheme 1.5, synthesis for 8-epi-SC-Δ13-9-IsoF is presented. The key intermediate was a diol epoxide that was obtained through two different Sharpless asymmetric reactions. Next, specific arrangement of the tetrahydrofuran ring and hydroxyl groups was acquired through an epoxide cyclization cascade. In addition, the neat product of 5-exo-tet cyclization was used through Mitsunobu inversion to gain access to the other stereoisomers of isofurans.36

11

Br OH 1. NaI, CuI, K2CO3 Sharpless H asymmetric 1. Et3N, TMSCl OH epoxidation 2. OH 2.LiAlH4 H O O O O O OH

Br2

O 1. TBSCl O O 2. BuLi, BF OEt OH OSO2Ph 3. 2 OH O H HO 1. PhSO2Cl. EtN3 K2CO3 HO CN 2. AD-mix-α O O O O O O

HO HO NC 1. TBDPSCl - 1. P-2 Ni, H2 n O 2. C5H11MgBr + O TBSO 2. H3O HO CN O O CHO

TBDPSO NC HO HOOC

O 2 steps O TBDPSO HO

R2 R2 R 1 R1 R1 = OH, R2 = H

R1 = H, R2 = OH

Scheme 1.5 Synthetic route for 8-epi-SC-Δ13-9-IsoF

1.7 Formation of LPO products

12

PUFAs, as one of the major components of cell membranes, are subject to

modifications with reactive species. The reactions mostly occur through radical mechanisms, building a family of products distinct from enzymatic ones. The radical chain reactions have been categorized in three different major steps: (1) Initiation: the

key event which involves hydrogen-abstraction by radical or oxidant. (2) Propagation:

oxygen or nitrogen addition to carbon-centered radicals, followed by fragmentation and the rearrangement of atoms and bonds. (3) Termination: when radicals combine with each other and stop reacting by forming non-radical products.

7 COOR 11 10 C5H11 (CH2)7COOR 13 Linoleate,LA;18:2 Arachidonate,AA;20:4

COOR COOR

Docosahexanoate,DHA;22:6 Eicosapentaenoate,EPA;20:5

Figure1.2 The molecular structure of major oxidizabale PUFA esters.

In this regard, polyunsaturated fatty acids (PUFAs), such as linoleate (LA), arachidonate (AA), eicosapentaenoate (EPA), docosahexaenoate (DHA) and their esters are predisposed to undergo peroxidation. This is due to the C-H bonds at the bis-allylic positions having lower bond dissociation enthalpy. In other words, the bis-allylic C-H bonds are the weakest bonds in these molecules, and the hydrogen atoms at these positions are favorably abstracted during oxidation.37 Next, the peroxidation reaction in

13 generation for the two nonenzymatic reaction products 15-F2c-IsoP and 8-epi-SC-Δ -9-

IsoF is discussed.

13

O O ROS COOH COOH n n-C5H11 -C5H11 H H H H 3 3

radical attack site

O O O O O O O O COOH COOH n n-C H -C5H11 5 11 3 3 disrotatory (racemic) 11-HpETE cyclic peroxide process

COOH COOH HO O O O O O O O

H H

syn to cyclic peroxide

HO

steps COOH O COOH O HO OH OOH 15-F c-IsoP 15-G2c-IsoP 2 Scheme 1.6 Non-enzymatic formation of 15-F2c-IsoP

The postulated mechanism for formation of the lipid peroxidation product 15-F2c-

IsoP is summarized in Scheme 1.6. The first step involves the conversion of arachidonic acid (AA) to peroxyl form. Following this, the radical reacts internally with the neighboring double bond, producing the short-lived intermediate cyclic peroxide. Next, the two radical π-orbitals combine to form a new σ-bond, thus forming the cyclopentane ring. This process undergoes disrotatory ring formation based on woodward-hoffmann rules for pericyclic reaction. The inward rotation can happen at the syn or anti stereochemistry comparing to the cyclic peroxide. In this case the syn stereoisomer forms and eventually breaks down to F-ring IsoP through spontaneous reactions.38

14

Similarly, at higher level of oxygen 8-epi-SC-Δ13-9-IsoF is derived from arachidonic acid. Once the cyclic peroxide is formed, it undergoes a facile 1,3-SHi reaction to form

diepoxy hydroperoxide. The formation of diepoxide is competing with the formation of

the cyclopentane ring in IsoP.

O O ROS COOH COOH n n-C5H11 -C5H11 H H H H 3 3

radical attack site

O O O O COOH COOH n n-C H -C5H11 5 11 3 3 1,3-SHi (racemic) 11-HpETE

O O O O O O O O COOH H2O COOH n-C5H11 n-C5H11 3 3 diepoxy peroxide

HO HO O HO OH O O O HO OH COOH COOH n-C5H11 n-C5H11 3 3

HO

O COOH OH OH

8-epi-SC-∆13-9-IsoF Scheme 1.7 Non-enzymatic formation of 8-epi-SC-Δ13-9-IsoF

Next, oxygen adds to the molecule on the newly-generated radical to form diepoxy

hydroperoxide. After of the epoxide ring to form the regioisomeric expoxy

diols, an intramolecular nucleophilic ring opening of the epoxide through 3-exo

15

cyclization generates the tetrahydrofuran ring. (Scheme 1.7) The order of some steps may

be different from the mechanism shown, but the products will not change.39,40

Because LPO is one of the downstream reactions of oxidative stress, there has been an increasing focus on understanding the role of pathways related to ROS/RNS.41

1.8 Oxidative stress (OS)

Cells are capable of providing diverse mechanisms to maintain balance between the

production and consumption of the species of redox reactions. The discrepancy in these

regulatory systems can lead to the accumulation of more oxidants, which cause oxidative

stress. In other words, oxidative stress originates either from higher levels of oxidant formation or from the inhibition of antioxidant protective mechanisms.42

Together, oxidative stress and nitrosative stress have been identified as the major

contributors to the pathogenesis and pathophysiology of aging, cancer and chronic

disease.43 They primarily provide reactive species and radicals that are capable of

chemically modifying central components of the cell and producing molecules with

deviated functions. However, oxidative stress has also been recognized as a regulatory mechanism for homoeostasis. Indeed, cells take advantage of these mechanisms to perform necessary radical reactions degrade old machineries and regenerate new constituents. For example, the human utilizes the excessive production of reactive oxygen and nitrogen species to destroy pathogens through a process termed the oxidative burst. 41,44

16

1.8.1 Formation of reactive oxygen species/reactive nitrogen species (ROS/RNS)

Oxidative stress is mainly caused by reactive oxygen species; similarly, nitrosative

stress is attributed to reactive nitrogen species. In physiological conditions, the reactive

species are formed during the mitochondrial electron transport or through different

enzymatic activities and free metals in the cells.44 Once the reactive species are

generated, they can react with each other to form a variety of combinations that can be

more reactive than the original ones. 45(Figure 1.3) As a signaling entity in the cell, their chemical physiology is dictated by different physiochemical factors such as concentration, half-life (t1/2), rate of diffusion across membranes and standard electrode

potential (E0).

1 O2 - ROO / RO RH ν e h RH - - e - e OH ROS O2 O2 H2O2 H+

Fe2+ Fe3+

+ - H NO ONOO ONOOH NO2 RNS CO 2

ONOOCO2

- CO3 Figure1.3 Generation of different ROS/RNS and their subsequent reactions

1.8.2 Reactive oxygen species (ROS)

1.8.2.1 Singlet oxygen

Atmospheric oxygen (O2) exists in a biradical form in which both electrons in the

− open-shell triplet ground state O2 (X³Σg ) have the same spin in two degenerate

17

antibonding πg-orbitals. This feature makes O2 “spin restricted” and not highly

chemically reactive. However, once energy is provided, the spin restriction is

overcome and the molecule is excited to other electronic states of antiparallel spins or singlet spins. In excited forms the spin can be in two different arrangements: O2 (a¹Δg)

+ (non-radical) and O2 (b¹Σg ) (more reactive free radical), the latter of which is less

46 stable and relaxes quickly to the lowest lying excited state O2 (a¹Δg). O2 (a¹Δg) is

considered the most biologically relevant reactive oxygen species. With a half-life of

47 1 μs, O2 (a¹Δg) has sufficient time to diffuse into various targets within the cell.

1.8.2.2 Superoxide radical

Singlet oxygen can lose its spin restriction in a facile process of one-electron

.- reduction to form the superoxide anion radical (O2 ), which is more reactive than

.- triplet oxygen. With a half-life less than 1 μs, O2 has minimal time for diffusion,

.- reaching a few micrometers from the site of generation. O2 acts a Brønsted base and

+ . reacts with H , forming hydroperoxyl radical HO2 . The acidic form is more stable

and hypothetically more permeable through membranes. The pKa for this acid-base

conjugate is around 4.88, which means that compartments with lower pH, such as the

mitochondrial intermembrane space, and lysosomes, would

.- accommodate the acidic molecular form. O2 is primarily a reducing agent. However,

48,49 .- the acidic form acts as an oxidant. Although O2 is not capable of modifying

macromolecules, it acts as a major source of oxidative stress. Specifically, it functions

.- . as a reducing species, which generates many other strong oxidants. O2 / HO2 can

produce hydrogen peroxide (H2O2), which can be decomposed through the Fenton reaction (presence of metal ions such as iron or copper) to form hydroxyl radical

18

. . - (HO ). In addition, superoxide anion radical reacts with NO and forms ONOO , an

extremely reactive RNS.

1.8.2.3 Hydrogen peroxide

Hydrogen peroxide (H2O2) is a weak acid without any unpaired electrons (a non- radical) that has a short half-life 10 μs because of the activities of detoxifying enzymes such as catalase and peroxidase. It has enhanced diffusion across long distances and membranes, although it is likely that this oxidant is less membrane permeate than a gas such as nitric oxide.

1.8.2.4 Hydroxyl radical

Hydroxyl radical (HO.) is the predominant source of oxidative stress that can

damage a wide variety of macromolecules in the cell. It possesses a half-life less than

1 fs. It is mainly biosynthesized through metal centers and is typically referred to as

Fenton-derived, named after Fenton who studied hydrogen peroxide production in

19th century.50

1.8.3 Reactive nitrogen species (RNS)

1.8.3.1 Nitric oxide (NO.)

Nitrogen oxide (NO.) is the product of the enzymes called NO synthases (NOS),

from the reaction of L-arginine and oxygen to produce citrulline and NO. It has a

half-life of 3-5s and is capable of traveling 50–200 μm from its production

source..51,52

1.8.3.2 Peroxynitrite (ONOO-)

Peroxynitrite forms through combining nitrogen oxide and superoxide anion

.- − radical (O2 ). ONOO is a strong nucleophile and exhibits a half-life of about 10 ms.

19

Its protonated form, peroxynitrous acid (ONOOH), is an extremely strong oxidant

(pKa= 6.8). It is capable of directly reacting with organic moieties or undergoing

. . homolysis to nitrogen dioxide (NO2 ) and a hydroxyl radical (HO ). In addition,

− ONOO can react with CO2 (1.3 mM in plasma) to form nitrosoperoxycarbonate

− − (ONOOCO2 ). ONOOCO2 eventually breaks down to two strong and short-lived

.- oxidant radicals, a carbonate radical (OCO2 ) and a nitrogen dioxide as a pair of

caged radicals. 53,54

1.8.4 Reactivity of ROS/RNS

A summary of physicochemical properties of major ROS/RNS is presented in

Table 1.1. These reactive species are formed endogenously through both enzymatic and spontaneous reactions and provide signaling roles in the body. 41

Species In vivo (t1/2)(s) reactivity reaction Standard electrode

molarity potential(v)

1 -6 - 0 O2 NA 10 oxidant 1∆ E =+0.64 gO2 + e O 2

.- -11 -10 -6 - 0 O2 10 -10 10 reductant e E =-0.35 O2 + O 2

-7 -5 -3 - 0 H2O2 10 10 -10 oxidant + E =+0.80, H2O2 + H + e HO + H2O

E0’=+0.39 (pH 7.0)

. -15 -9 + - 0 HO 10 10 oxidant HO + H + e H O E =+2.73, 2 E0’=+2.31 (pH 7.0)

NO. NA 3-5 oxidant + - E0=-0.11 NO + H + e HNO

ONOO- 10-9 10-2 oxidant E0’=+1.4 (pH 7.0) ONOO + 2H + e NO2 + H2O

. 0 NO2 NA NA oxidant - E =+1.04 NO + e NO 2 2

Table1.1 Physicochemical properties of reactive species (ROS/RNS), NA: data is not available.

20

Many of these reactive species are in equilibrium in water. As a result, pH has an

impact on their redox activity. In this regard, the standard electrode potential can provide

valuable information for ROS/RNS species and determine their role in redox biology,

either as oxidants or reductants.

.- For example, under physiological conditions, the superoxide anion radical (O2 ) tends

. to be a reducing agent in comparison to the hydroperoxyl radical (HO2 ). This may not be

readily concluded from the given standard electrode potential (E0) values.

- + - ' + 0 O + 2H + e H O E =+0.97(pH=7) HO2 + H + e H2O2 E =+1.46 2 2 2

- - + e 0 0 O2 + H + HO E =-0.03 O + e O2 E =-0.35 2 2

.- Figure 1.4 Standard electrode potential for superoxide anion radical (O2 ) and hydroperoxyl radical

. (HO2 )

In fact, the Nernst equation can provide a more accurate estimate in which both the

concentration of the reactive species and the pH are necessary components.55(Figures 1.4

& 1.5)

- Ox + mH+ + ne Red

Ox 10 -(m)(pH) E = E0 + 0.059/n log X Nernst equation Red

Figure 1.5 Nernst equations can be used as an estimate to measure redox potential at known concentration and pH

Although the Nernst equation can give an approximate idea about the inherent redox

activity of different species, it can hardly be used to investigate the level of reactivity.

Reactivity is more attributed to kinetics, meaning that the level of reactivity for these

species in the complex network of the cell is mostly dependent on kinetic parameters. 21

The half-life (t1/2) of the reaction for the species provides more valuable information than

thermodynamic properties such as the Nernst equation.56

1.9. Biological roles of oxidized lipids

Although originally scientists in the field were more interested in oxidized lipids derived from enzymatic reactions, non-enzymatic products also became important to the community in the 1980s. The change mainly occurred due to the many LPO products

detected in vivo in both clinical and experimental models. Indeed, the characterization of

adducts to these molecules has been linked to the onset and progression of several

pathological states associated with oxidative stress.43

The reported concentration for oxidized lipids in the cell is often in the nanomolar

range, raising an important question regarding the mechanism by which oxidized lipids

can exert their signaling roles. In fact, one of the crucial features contributing to their function is the existence of reactive functional groups such as α,β-unsaturated carbonyls, epoxy, and aldehyde. These functionalities are mostly formed through radical mechanism of including addition of oxygen or rearrangement of double bonds. It has been suggested that covalent binding of these electrophilic groups to the protein targets provides signal accumulation over time. This phenomenon has been named the covalent advantage. Thus, the full activation of the receptor can be achieved with a low concentration of oxidized lipids. Many targets have been identified as a target for LPO products such as

57-61 Cytochrome c oxidase, Keap1 protein, Hsp70, Hsp90, ANT and ATP synthase. The

key to these modifications is the presence of nucleophilic amino acids such as cysteine,

histidine and lysine that are available in most of the signaling proteins.62 Biological

22

activity of these molecules confirms that downstream of ROS/RNS results in a more specific impact in regulating .

Since fatty acids are one of the major lipid groups that play a role as a source of energy, targets that are related to nutrient sensing were the first to be studied. In the following paragraphs, GPCR and PPAR are presented as examples of targets interacting with oxidized lipids. 63,64

1. 9.1 Oxidized lipid and G protein-coupled receptors (GPCRs)

Like many other oxidized lipids derived from enzymatic reactions such as prostanoids, oxidized free fatty acids derived from polyunsaturated fatty acids (PUFAs) are found to function by reversibly binding to cell G protein-coupled receptors (GPCRs).

These receptors are located on the surface of the cell, with seven domains spanning through the membrane. For example, in mast cells GPR132, as one of the members in proton-sensing GPCRs, has been found to interact with oxidized forms of fatty acids. It functions as a stress-inducible receptor that can recognize lipid overload and oxidative stress. 9-Hydroxyoctadecadienoic acid (9-HODE), an intermediate of LA peroxidation, has been identified as a ligand for this receptor. 65-67 In other studies, 4-HNE has demonstrated modulation of GPCR signaling through adduct formation directly with G

68,69 proteins such as Gαq/11 and RGS4.

23

COOH

LA

lipid peroxidation COOH HO 9-HODE

OH GPCR O

4-HNE

Mast cell

Muscrarinic receptor G protein G protein

Brain Cell

Figure1.6 Lipid peroxidation products and their signaling through GPCR

1. 9.2 Oxidized lipid and peroxisome-proliferator-activated receptors

(PPARs)

Among the intracellular targets, the lipid molecules are capable of promoting

signals via peroxisome-proliferator-activated receptors (PPARs) pathways in both

reversible and irreversible fashions. 70 The PPAR family of nuclear receptors is serving as

a master regulator of cellular differentiation, development and . Upon the binding of oxidized lipids to these receptors, their downstream are up-regulated.

PPARs also regulate the expression of other genes through interaction with coregulators

of gene expression in other gene machineries. (Section 3.2)

24

Cytoplasm

Lipid peroxidation

Co-activators

PPAR RXR9cRA

PPRE

Nucleus

Figure1.7 Lipid peroxidation products interact with PPAR and activate their gene expression machinery.

25

1.10 References

(1) Pennazio, S. Riv Biol-Biol Forum 2009, 102, 253.

(2) Cooper, S. J. Appetite 2008, 51, 419.

(3) Dale, H. H. J Pharmacol Exp Ther 1914, 6, 147.

(4) Rubin, R. P. Pharmacological reviews 2007, 59, 289.

(5) Bergstrom, S. Nord Med 1949, 42, 1465.

(6) Bergstrom, S.; Sjovall, J. Acta Chem Scand 1957, 11, 1086.

(7) Bergstroem, S.; Ryhage, R.; Samuelsson, B.; Sjoevall, J. The Journal of biological

chemistry 1963, 238, 3555.

(8) Vandorp, D. A.; Vonkeman, H.; Nugteren, D. H.; Beerthuis, R. K. Nature 1964,

203, 839.

(9) Bergstrom, S.; Sjovall, J.; Ryhage, R.; Samuelsson, B. Acta Chem Scand 1963, 17,

2271.

(10) Abrahamsson, S.; Bergstrom, S.; Samuelsson, B. P Chem Soc London 1962, 332.

(11) Weinheimer, A. J.; Spraggins, R. L. Tetrahedron letters 1969, 5185.

(12) Corey, E. J.; Weinshen.Nm; Schaaf, T. K.; Huber, W. J Am Chem Soc 1969, 91,

5675.

(13) Corey, E. J.; Nyori, R.; Schaaf, T. K. J Am Chem Soc 1970, 92, 2586.

(14) Corey, E. J.; Link, J. O. Tetrahedron letters 1990, 31, 601.

(15) Corey, E. J.; Imai, N.; Pikul, S. Tetrahedron letters 1991, 32, 7517.

(16) Borgeat, P.; Samuelsson, B. P Natl Acad Sci USA 1979, 76, 3213.

(17) Samuelsson, B. Science 1983, 220, 568.

(18) Corey, E. J.; Clark, D. A.; Goto, G.; Marfat, A.; Mioskowski, C.; Samuelsson, B.;

Hammarstrom, S. J Am Chem Soc 1980, 102, 1436.

26

(19) Samuelsson, B. The Journal of biological chemistry 2012, 287, 10070.

(20) Nugteren, D. H.; Vonkeman, H.; Vandorp, D. A. Recl Trav Chim Pay-B 1967, 86,

1237.

(21) Salomon, R. G.; Salomon, M. F.; Coughlin, D. J. J Am Chem Soc 1978, 100, 660.

(22) Morrow, J. D.; Awad, J. A.; Boss, H. J.; Blair, I. A.; Roberts, L. J., 2nd Proc Natl

Acad Sci U S A 1992, 89, 10721.

(23) Roberts, L. J., 2nd; Montine, T. J.; Markesbery, W. R.; Tapper, A. R.; Hardy, P.;

Chemtob, S.; Dettbarn, W. D.; Morrow, J. D. The Journal of biological chemistry 1998, 273,

13605.

(24) Morrow, J. D.; Awad, J. A.; Wu, A.; Zackert, W. E.; Daniel, V. C.; Roberts, L. J., 2nd

The Journal of biological chemistry 1996, 271, 23185.

(25) Salomon, R. G.; Sha, W.; Brame, C.; Kaur, K.; Subbanagounder, G.; O'Neil, J.;

Hoff, H. F.; Roberts, L. J., 2nd The Journal of biological chemistry 1999, 274, 20271.

(26) Bernoud-Hubac, N.; Davies, S. S.; Boutaud, O.; Montine, T. J.; Roberts, L. J.

Journal of Biological Chemistry 2001, 276, 30964.

(27) Fessel, J. P.; Porter, N. A.; Moore, K. P.; Sheller, J. R.; Roberts, L. J., 2nd Proc Natl

Acad Sci U S A 2002, 99, 16713.

(28) Cracowski, J. L.; Durand, T.; Bessard, G. Trends Pharmacol Sci 2002, 23, 360.

(29) Montuschi, P.; Barnes, P. J.; Roberts, L. J. Faseb J 2004, 18, 1791.

(30) Nourooz-Zadeh, J.; Liu, E. H. C.; Yhlen, B.; Anggard, E. E.; Halliwell, B. J

Neurochem 1999, 72, 734.

(31) Fessel, J. P.; Roberts, L. J. Antioxid Redox Sign 2005, 7, 202.

(32) Jahn, U.; Galano, J. M.; Durand, T. Angew Chem Int Edit 2008, 47, 5894.

(33) Corey, E. J.; Shih, C.; Shih, N. Y.; Shimoji, K. Tetrahedron letters 1984, 25, 5013.

27

(34) Renaud, P.; Vionnet, J. P. J Org Chem 1993, 58, 5895.

(35) Vionnet, J. P.; Renaud, P. Helv Chim Acta 1994, 77, 1781.

(36) Taber, D. F.; Pan, Y. C.; Zhao, X. J Org Chem 2004, 69, 7234.

(37) Porter, N. A.; Wolf, R. A.; Yarbro, E. M.; Weenen, H. Biochem Bioph Res Co 1979,

89, 1058.

(38) Waugh, R. J.; Murphy, R. C. J Am Soc Mass Spectr 1996, 7, 490.

(39) Nixon, J. R.; Cudd, M. A.; Porter, N. A. J Org Chem 1978, 43, 4048.

(40) Porter, N. A.; Zuraw, P. J.; Sullivan, J. A. Tetrahedron letters 1984, 25, 807.

(41) D'Autreaux, B.; Toledano, M. B. Nat Rev Mol Cell Bio 2007, 8, 813.

(42) Davies, K. J. IUBMB life 2000, 50, 279.

(43) Benz, C. C.; Yau, C. Nat Rev Cancer 2008, 8, 875.

(44) Holmstrom, K. M.; Finkel, T. Nat Rev Mol Cell Bio 2014, 15, 411.

(45) Hillard, E. A.; de Abreu, F. C.; Ferreira, D. C.; Jaouen, G.; Goulart, M. O.;

Amatore, C. Chemical communications 2008, 2612.

(46) Ogilby, P. R. Chem Soc Rev 2010, 39, 3181.

(47) Devasagayam, T. P.; Kamat, J. P. Indian journal of experimental biology 2002, 40,

680.

(48) Rao, P. S.; Hayon, E. J Phys Chem-Us 1975, 79, 397.

(49) Ilan, Y. A.; Czapski, G.; Meisel, D. Biochim Biophys Acta 1976, 430, 209.

(50) Prousek, J. Pure Appl Chem 2007, 79, 2325.

(51) Martinez, M. C.; Andriantsitohaina, R. Antioxid Redox Sign 2009, 11, 669.

(52) Ignarro, L. J. Annu Rev Pharmacol 1990, 30, 535.

(53) Szabo, C.; Ischiropoulos, H.; Radi, R. Nat Rev Drug Discov 2007, 6, 662.

28

(54) Denicola, A.; Freeman, B. A.; Trujillo, M.; Radi, R. Arch Biochem Biophys 1996,

333, 49.

(55) Armstrong David, A.; Huie Robert, E.; Lymar, S.; Koppenol Willem, H.; Merényi,

G.; Neta, P.; Stanbury David, M.; Steenken, S.; Wardman, P. In BioInorganic Reaction

Mechanisms 2013; Vol. 9, p 59.

(56) Flohe, L. Biochim Biophys Acta 2013, 1830, 3139.

(57) Chen, J. J.; Schenker, S.; Frosto, T. A.; Henderson, G. I. Bba-Gen Subjects 1998,

1380, 336.

(58) Levonen, A. L.; Landar, A.; Ramachandran, A.; Ceaser, E. K.; Dickinson, D. A.;

Zanoni, G.; Morrow, J. D.; Darley-Usmar, V. M. The Biochemical journal 2004, 378, 373.

(59) Vila, A.; Tallman, K. A.; Jacobs, A. T.; Liebler, D. C.; Porter, N. A.; Marnett, L. J.

Chem Res Toxicol 2008, 21, 432.

(60) Vieira, H. L. A.; Belzacq, A. S.; Haouzi, D.; Bernassola, F.; Cohen, I.; Jacotot, E.;

Ferri, K. F.; El Hamel, C.; Bartle, L. M.; Melino, G.; Brenner, C.; Goldmacher, V.; Kroemer, G.

Oncogene 2001, 20, 4305.

(61) Yarian, C. S.; Rebrin, I.; Sohal, R. S. Biochem Bioph Res Co 2005, 330, 151.

(62) Higdon, A.; Diers, A. R.; Oh, J. Y.; Landar, A.; Darley-Usmar, V. M. Biochemical

Journal 2012, 442, 453.

(63) Blad, C. C.; Tang, C.; Offermanns, S. Nat Rev Drug Discov 2012, 11, 603.

(64) Varga, T.; Czimmerer, Z.; Nagy, L. Bba-Mol Basis Dis 2011, 1812, 1007.

(65) Obinata, H.; Hattori, T.; Nakane, S.; Tatei, K.; Izumi, T. The Journal of biological chemistry 2005, 280, 40676.

(66) Obinata, H.; Izumi, T. Prostag Oth Lipid M 2009, 89, 66.

29

(67) Vangaveti, V.; Shashidhar, V.; Jarrod, G.; Baune, B. T.; Kennedy, R. L. Therapeutic advances in endocrinology and metabolism 2010, 1, 165.

(68) Monroy, C. A.; Doorn, J. A.; Roman, D. L. Chem Res Toxicol 2013, 26, 1832.

(69) Blanc, E. M.; Kelly, J. F.; Mark, R. J.; Waeg, G.; Mattson, M. P. J Neurochem 1997,

69, 570.

(70) Itoh, T.; Fairall, L.; Amin, K.; Inaba, Y.; Szanto, A.; Balint, B. L.; Nagy, L.;

Yamamoto, K.; Schwabe, J. W. R. Nat Struct Mol Biol 2008, 15, 924.

30

Chapter 2 Development of novel synthetic routes to the

epoxyketooctadecenoic acids (EKODEs)

2.1 Introduction

Since the discovery of prostaglandins in the early 1930s by Ulf von Euler and M.

W. Goldblatt, scientists have been struggling for decades to define the mechanism of

action of oxidized lipids.1 In fact, advancements in instrumental methods such as mass

spectrometry and isotope labeling have provided great advantages for researchers to

isolate different oxidized lipids and characterize their structures in the 1960s.2

The broad and potent biological actions of these molecules have driven the

scientific community to investigate them more thoroughly. Further, access to knockout

mouse models for disruption of enzymes involved in lipid biosynthesis pathways and the

availability of genes for production of plausible sites of action have made these

discoveries possible.3

Among different research groups, Sune Bergström, Beng I. Samuelsson and John

R. Vane received the Nobel Prize in physiology or in 1982 for their

“discoveries concerning prostaglandins and related biologically active substances”.

Accordingly, pioneering chemists in total synthesis such as Robert B. Woodward, Gilbert

Stork and Elias J. Corey have shown great interest in developing syntheses of these bioactive lipids.

Although the lipidologists in the field primarily focused on oxidized lipids formed

through enzymatic reactions, non-enzymatic products also became important to the field

in the 1960s. In fact, collaborative work between L. Jackson Roberts II and Robert G.

Salomon resulted in the discovery of lipid peroxidation products such as isoprostane and

31

isolevuglandin. In fact, non-enzymatically produced LPO products have proven to be as important as the enzymatic products in cell signaling.4

The general interest in the field of LPO is focused on biological target modifications with end products of LPO such as 4-HNE and 4-ONE (section1.3). Indeed, a systems analysis conducted by the Lawerence J. Marnett lab yielded interesting results, showing these molecules modulating biological networks. Likewise, his group revealed

4-HNE acts on different targets through the modulation of gene expression or directly on the protein targets.5-7 Recently in our laboratory detailed mechanistic studies showed that

4-HNE modulates the production of nitrogen oxide (NO.) in a concentration-dependent

manner in a macrophage culture cell line. In other words, 4-HNE acts as a second messenger in maintaining normal cell physiology.8

Currently in our laboratory, we are trying to look at lipid peroxidation products of

linoleic acid in a general prospective. In fact, we are examining the entire spectrum of

possible oxidized linoleic acid through lipid peroxidation as potential signaling

molecules. Linoleic acid (LA) is often the most prevalent form of PUFAs present inside

the cell membrane, and the 1,4-diene pattern makes it more vulnerable to oxidation than

other fatty acids.9 As a result, during oxidative stress many products of peroxidation are

derived from linoleic acids. Once ROS / RNS trigger the reaction by abstracting an

electron from LA, then molecule of oxygen adds to the carbon radical to generate

hydrorperoxy intermediate. Following the rearrangement or fragmentation, two classes of

products are formed: (1). Oxidative cleavage products (OCPs) which consist mainly of

the end products of peroxidation, such as 4-hydroxynonenal (4-HNE), and 4-oxononenal

32

(4-ONE). (2). Oxidative addition products (OAPs) resulting from oxygenation of radical intermediates along with rearrangement in double bonds.10 (Figure 2.1)

O O OH COOH COOH O EKODE OH DHKODE O O COOH COOH OH KODDE HKODE

ROS/RNS

COOH

LA

ROS/RNS

OH OH O O COOH

4-HNE HODA O O O O COOH 4-ONE DODE

COOH O O Octenal OUEA Figure 2.1 Linoleic acid oxidation through non-enzymatic reactions

Among these varieties of scaffolds of linoleic acid metabolites, epoxyketooctadecenoic acids (EKODEs) specifically caught our attention. This family of metabolites has been proven to exist in vivo and is involved in pathological states associated with oxidative stress such as asthma, obesity and hypertension.11-13 More detailed physiological studies showed that EKODEs change corticosterone and

33

aldosterone production levels. Evidence points to the latter being conducted through

modulation of Ca2+ concentration inside the cell.14 In addition, these oxidized lipids have

shown antioxidant effects through the Nrf2-Keap1 signaling pathway.15

The interesting physiological activities of EKODEs led us to consider a robust

synthesis for these scaffolds in a broader picture. Likewise, identifying methods of

synthesis for each core can be considered an innovative approach to gain access to an

array of metabolites and their modified forms.

2.2 EKODE structure and nomenclature

There are two series of EKODEs with different arrangements of carbonyl, epoxy and olefin functional groups. Isomers with C=C in the middle, and the others with C=O in the middle were assigned as EKODE-(E)-I and EKODE-(E)-II, respectively. Because the new C=C bonds form in free radical steps, they are more likely to form the trans (E) stereochemistry in the final products of EKODE. Epoxy groups are formed from hydroperoxy intermediates which can lead to both “trans / cis” epoxy, named with the same designation (Scheme 2.2, Scheme 2.7). In addition, “a” and “b” were used to define proximity of epoxy to the carboxylic acid or methyl terminals, respectively. Four isomers for each EKODE-I and EKODE-II can be proposed; however, the previous studies showed the cis-IIa / IIb are not formed (small amount) under peroxidation conditions.16

O O O

R1 R2 R1 R2 R1 R2 O O O trans-IIa/IIb trans-Ia/Ib cis-Ia/Ib

= R1 or R2 C5H11 = R or R (CH2)7COOH 2 1

Figure 2.2 EKODE I and II structures and nomenclature

34

2.3. EKODE-(E)-I family

R2 R1 R1=-(CH2)7COOH R2=-C5H11 LA radicals,

radicals , 2 O O 2

R2 O(O)H O(O)H R1

R1 R2 10E,12Z,9H(p)ODE 9Z,11E,13H(p)ODE

R R R2 R1 2 1 O O O O R R2 1 O2 O2 R2 R1 O O cis-EKODE-(E)-Ia cis-EKODE-(E)-Ib OOH OOH

R R R2 R1 2 O 1 O

O O

R R R2 R1 2 O 1 O trans-EKODE-(E)-Ia trans-EKODE-(E)-Ib Scheme 2.1 Non-enzymatic formation of EKODE-I family

The postulated non-enzymatic formation of both cis- and trans-EKODE-(E)-Ia and -b is summarized in Scheme 2.1. The first step involves the conversion of linoleic acid (LA) to 13 or 9-hydroperoxy (HpODE) intermediates, which is followed by rearrangement to a trans-epoxy and double bonds, producing a second site for oxygenation at the C-10,C-9 positions for Ia and Ib, respectively.

35

The hydroperoxy intermediate undergoes elimination to afford ketone as trans-

EKODE-(E)-Ia and -b. Similarly, if epoxy group is produced as cis stereoisomers, the

intermediates eventually lead to cis-EKODE-(E)-Ia and -b shown in dashed arrows in

Scheme2.1 In the next section, we illustrate a methodology to target total synthesis of

epoxy α, β-unsaturated ketone in the EKODE-(E)-I family of oxidized linoleic acid metabolites.

2.3.1 Retrosynthetic analysis of trans-EKODE-(E)- I

The retrosynthetic analysis for trans-EKODE-(E)-I family is outlined in Scheme

2.2 The trans-EKODE-(E)-I can be obtained from two smaller fragments of trans-epoxy aldehyde and the phosphorane through a Wittig reaction.

O

R1=-(CH2)7COOH R1,2 O R1,2 R2=-C5H11 Wittig olefination

O O Ph P R O + 3 R O 1,2O R Cl 1,2 1,2 R1,2 trans-epoxy aldehyde phosphorane

Wittig olefination

O O R1,2 HO R 1,2

Scheme2.2 Retrosynthesis analysis of trans-EKODE-(E)-I

In order to make the phosphorane; treatment of the carboxylic acid with oxalyl

chloride gave the acid chloride, which was reacted with methyltriphenylphosphorane,

generated by reaction of methyltriphenylphosphonium bromide and stoichiometric

amount of n-butyllithium at -78 oC, to give the phosphorane. For synthesis of the trans-

36

epoxy aldehyde moiety, starting from an aliphatic aldehyde, a one-step Wittig reaction with (triphenylphosphoranylidene)acetaldehyde gave α, β-unsaturated aldehyde, epoxidation of which, with alkaline H2O2, provided the trans-epoxy aldehyde. Finally, a

one-step Wittig reaction afforded the olefin.

2.3.2 Synthesis of trans-EKODE-(E)- Ia

The synthetic procedure for trans-EKODE-(E)-Ia was based on the preparation of trans-epoxy aldehyde (blue) and the phosphorane 7 (red).

In order to make the trans-epoxy aldehyde moiety, ozonolysis of commercially available methyl oleate gave methyl 9-oxononanoate 2 in 75 % yield, which was purified via . A Wittig reaction of 2 with (triphenylphosphoranylidene)acetaldehyde gave methyl (E)-11-oxoundec-9-enoate 4 in a 57 % yield. Next, epoxidation of the α, β- unsaturated aldehyde with alkaline H2O2 afforded the trans-epoxide 5 in a 38 % yield.

In order to make the phosphorene 7, treatment of hexanoic acid with oxalyl chloride gave hexanoyl chloride in a 91 % yield. The latter was reacted with methyltriphenylphosphorane, generated by reaction of methyltriphenylphosphonium bromide and stoichiometric amount of n-butyllithium at -78 oC , to give 1-(triphenyl-λ5-

phosphanylidene) heptan-2-one 7.

37

O Ph P O O 3 3 O H2O2 Methyl oleate MeO O 3 MeO O 5 5 Zn CHCl3, reflux, 48 h NaHCO3, rt, 3h 1 (C19H36O2) 2 75 % 4 57 %

O O 1. (COCl)2, CH2Cl2, reflux,2h Ph3P HOOC MeO O + 5 O PPh Br n -78 o to rt 38 % 5 7 41 % 2. CH3 3 / -BuLi, C 6 Wittig reaction o CH2Cl2, 0 C, 4h

O O

MeO 5 O 47 % 8

Water / Acetone, pH 7 PPL 4h

O O

HO 5 O 93 % trans-EKODE-(E)-Ia Scheme 2.3 Synthetic route for trans-EKODE-(E)-Ia

The reaction of 7 (red) and trans-epoxy aldehyde 5 (blue) moieties at 0 oC

provided the methyl form of trans-EKODE-Ia 8, which was hydrolyzed with porcine pancreas lipase (PPL) to afford the final product trans-EKODE-(E)-Ia in a quantitative yield.

2.3.3 Synthesis of trans-EKODE-(E)-Ib

The synthetic procedure for trans-EKODE-(E)-Ib was based on the preparation of trans-epoxy aldehyde (pink) and the phosphorane 10 (green).

In order to make the phosphorane, methyl 9-oxononanoate 9 obtained from ozonolysis was transformed to its corresponding carboxylic acid in a 95 % yield through Pinnick oxidation. Next, treatment of 9 with oxalyl chloride gave methyl 9-chloro-9-

38

oxononanoate in a 93 % yield. The latter was reacted with methyltriphenylphosphorane,

generated by reaction of methyltriphenylphosphonium bromide and stoichiometric

amount of n-butyllithium at -78 oC , to give methyl 9-oxo-10-(triphenyl-λ5-

phosphanylidene) decanoate 10. In order to produce the trans-epoxy aldehyde 12,

epoxidation of commercially available (E)-oct-2-enal 11 with alkaline H2O2 gave 12 in a

63 % yield.

A reaction of 10 (green) and trans-epoxy aldehyde 12 (pink) at 0 oC provided

methyl ester 13 ,which was hydrolyzed with porcine pancreas lipase (PPL) to afford the

final product trans-EKODE-(E)-Ia in a quantitative yield.

O O COOH1. (COCl)2, CH2Cl2, reflux, 2h NaClO2 MeO MeO O o 5 5 H O ,10 2h n o 2 2 2 C, 95 % 9 2. CH3PPh3Br / -BuLi, -78 C to rt

O O H O PPh3 2 2 MeO +O O O 5 NaHCO 50 % 10 12 63 % 3 11

Wittig reaction o CH2Cl2, 0 C, 4h

O O

MeO 5 O 56 % 13

Water / Acetone,pH 7 PPL 4 h O O

HO 5 O Scheme 2.4 Synthetic route for trans-EKODE-(E)-Ib

39

2.3.4 Retrosynthetic analysis of cis-EKODE-(E)- Ib

cis-EKODE-(E)-I was prepared using a strategy similar to that for the trans analogues, although the cis-epoxy aldehyde was made through a completely different route. Starting from propargyl alcohol, alkylation of the terminal alkyne with bromopentane affords the acetylenic alcohol, which can be hydrogenated over Lindlar catalyst to afford a cis-olefin. Next, epoxidation provides the substrate for further oxidation with PCC in order to produce the cis-epoxy aldehyde.

O HO HO O O

HO HO

Scheme 2.5 Retrosynthesis analysis of cis-epoxy aldehyde

2.3.5 Synthesis of cis-EKODE-(E)- Ib

The synthetic procedure for trans-EKODE-(E)-Ib followed from the preparation of cis-epoxy aldehyde 18 (orange) and the phosphorane 10 (green).

The ylide 10 was made as described in section 2.2.3. In order to make the cis-epoxy aldehyde moiety, propargyl alcohol 14 was reacted with 2 equivalents of n-butyllithium at

-78 oC and then treated with n-bromopentane to give oct-2-yn-1-ol 15 in a 67 % yield.

Subsequent hydrogenation of the acetylenic alcohol intermediate over Lindlar catalyst

afforded (Z)-oct-2-en-1-ol 16 in a 92 % yield. Next, epoxidation with m-

chloroperoxybenzoic acid (m-CPBA) provided cis-(3-pentyloxiran-2-yl)methanol 17 with

40 a 95 % yield. Further oxidation with PCC gave cis-3-pentyloxirane-2-carbaldehyde 18, which is the required cis-epoxy aldehyde moiety.

HO n-bromopentane H2 HO n Lindlar catalyst, 8 h 14 H -BuLi, HMPA, THF 15 67 %

m-CPBA HO HO O CH2Cl2, rt, 2h 16 92 % 17 95 %

PCC CH2Cl2, rt, 2h

O O PPh3 + MeO O 5 O 50 % 10 18 55 %

Wittig reaction o CH2Cl2, 0 C, 4 h

O O

MeO 5 51 % 19 O

Water / Acetone, pH 7 PPL 4 h

O O

HO 5 O 93 % cis-EKODE-(E)-Ib

Scheme 2.6. Synthetic route for cis-EKODE-(E)-Ib

41

Reaction of 10 (green) with cis-epoxy aldehyde 18 (orange) at 0 oC provided the

methyl ester of cis-EKODE-(E)-Ib 19, which was hydrolyzed with porcine pancreas lipase (PPL) to afford the the final product cis-EKODE-(E)-Ib in a quantitative yield.

2.4 EKODE-(E)-II family

R R 2 O(O)H O2 R2 R1 O2 O(O)H 1

R1 R2 10E,12Z,9H(p)ODE Linoleic acid(LA) 9Z,11E,13H(p)ODE

R1=-(CH2)7COOH R2=-C5H11 R R R R 2 O 1 2 O 1

O2 O2

OOH OOH

R R R2 R1 2 O 1 O

O O

R2 R1 R R O 2 O 1 trans-EKODE-(E)-IIb trans-EKODE-(E)-IIa

Scheme 2.7 Non-enzymatic formation of EKODE-II family

The postulated non-enzymatic formation of trans-EKODE-(E)-IIa and-b is summarized in Scheme 2.7. The first step involves the conversion of LA to 13 or 9- hydroperoxy (HpODE) intermediates. Then, rearrangement to an epoxy functionality leads to the second site for oxygenation to a new hydroperoxide at the C-10 position, which is eventually undergoes elimination to afford ketone as trans-EKODE-(E)-IIa and- b.

42

The previous synthetic strategy for making specific arrangement of functional groups in trans-EKODE-(E)-II family involved formation of β-hydroxyenone

intermediate (X) (Scheme 2.8). Starting from an α, β-unsaturated ketone, aldol addition

o of aldehyde (R2CHO) using LDA / THF at -78 C is extremely low yielding. In fact

formation of side-products such as, tetrahydropyran through 6-endo-trig oxa-Michael

cyclization, during aldol reaction or purification on silica gel, make it an inefficient

synthetic route (Scheme 2.8i). 16

O O O OH R2 i. R R1 R2 1 aldol reaction X

O O NOH N R R2 red. 2 X ii. R R R1 1 (3+2) cycloaddition 1

O O O NOH O N O R R red. P 2 2 iii. (RO)2P (RO)2 (RO)2P

O O O OH R1 (RO)2P X R2 Scheme 2.8 Different synthetic strategies in order to make intermediate X.

There are a number of other approaches to avoid aldol addition while generating

intermediate X. For example, the second strategy (Scheme 2.8ii) involves taking

advantage of the reduction of a Δ2-isoxazoline in order to form the β-hydroxyenone. The key step for synthesis through this route is the isoxazoline ring formation.17 Formation of

the five-membered ring through 1,3-dipolar cycloaddition reaction stuffers from poor

regioselecvtivity and makes this approach challenging. In addition, reduction of Δ2-

43

isoxazoline using a variety of methods does not exhibit acceptable regioselectivity, and

there is a noticeable amount of reduced olefin in the final mixture.Finally, a third

approach which would avoid having a double bond in the starting material by using HWE

reagent, also experienced problems with reducing conditions mostly because of side

reactions such as elimination of the hydroxyl group (Scheme 2.8ii).

The previous synthetic routes have several disadvantages and problems: being

laborious, time-consuming, and complicated in terms of number of undesired products.16

Consequently, we sought to exploit a much more efficient tactic to avoid generation of

highly reactive β-hydroxyenone intermediate X.

In the next section, a new procedure has been developed in our laboratory based

on bifunctional conjunctive ylides to overcome the difficulties listed. Specifically, we

describe new, highly chemoselective, sequential reactions of sulfonium and Wittig ylides

with two different aldehydes that can be used for rapid construction of the trans-EKODE-

( E)-II family.

2.4.1 Retrosynthetic analysis of trans-EKODE-(E)-II

Structurally, this family of EKODEs possesses flanking α, β-unsaturated and epoxy functionalities centered on a ketone group. The synthetic method relies on developing a bifunctional intermediate containing two reactive ylides that can be used for generation of epoxy and olefin functionality in a sequential order. Our retrosynthetic analysis for the synthesis of the trans-EKODE-(E)-II family is outlined in Scheme 2.9, and starts from a bifunctional conjunctive ylide. Addition of the first aldehyde through

Johnson-Corey-Chaykovsky reaction produces the triphenylposphine epoxy ketone.

44

Subsequent Wittig olefination with a second aldehyde provided α,β-unsaturated keto

epoxide functionality.

O Wittig olefination O Ph3P R R R 2 O 1 O 1

Johnson–Corey–Chaykovsky reaction

O

Ph3P S(CH3)2

Bifunctional conjunctive ylide Scheme 2.9 Retrosynthesis analysis of trans-Ekode-(E)-II

2.4.2 Synthesis of bifunctional conjunctive ylide

Scheme 2.10 shows the detailed synthesis of bifunctional conjunctive ylide 22.

The synthesis of 22 began with commercially available 1,3-dichloroacetone. Halides in

1,3-dichloroacetone were replaced (Finkelstein reaction, 90 %) by bromide,18 and

substitution of one of the bromides with triphenylphosphine (90 %) followed by base

treatment that provided the Wittig ylide.18 Addition of dimethylsulfide in the presence of sodium tetrafluoroborate afforded the bifunctional conjunctive ylide (77 %). The overall yield was 35 %.

45

O O

Cl Cl 15 eq LiBr Br Br 1. 1.1eq PPh3 / Toluene, rt, overnight

19 Acetone, rt, 3days 20 2. NaHCO3, Methanol/water 90 %

O O Ph P Ph P - 3 Br 5 eq S(CH3)2, 5eq NaBF4 3 S(CH3)2 BF4 Acetone, rt, 24 hrs 21 22 Bifunctional conjunctive ylide 50 % 77 % Scheme 2.10 Synthetic rout for bifunctional conjunctive ylide

A literature review shows that the application of bis-ylides has been known for over a half century. However, their application has been limited to the synthesis of symmetric olefins via Wittig and related types of olefination reactions. 19,20 Recently,

Nagorny et al., developed a series of asymmetric bifunctional ylides in order to make

asymmetric polyene. 21

In this novel approach, the bis-ylides used in the coupling possess different

reactivities toward electrophiles. Appel et al. showed that a triphenylphosphonium ylide

has lower kinetic reactivity compared to the sulfonium version. 22 In fact, in a

phosphonium ylide the carbanion is more stabilized by the adjacent heteroatom than the

corresponding sulfonium derivative. The main cause in the case of phosphonium has been

attributed to the electron withdrawing effect through pπ-dπ which is pulling away

electrons from carbanion. Consequently, this phenomenon exhibits a substantial impact

on the reactivity toward aldehyde electrophiles. This difference in reactivity has enabled

the development of a selective sequence of reactions with respect to the aldehydes.

46

So far there have been only a few examples in which the difference in reaction

rate between ylide reagents has been used. 23,24 However, their application in the total synthesis of natural products has not yet been explored.

2.4.3 Synthesis of trans-EKODE-(E)-II

In the first step, the bifunctional conjunctive ylide was treated with sodium hydride and then with aldehyde (R1CHO) to form an epoxide. Formation of the epoxide

with n-hexanal 24 occurs much faster at lower temperature (at 0 oC for 3 h). However

methyl 9-oxononanoate 2 needs to be warmed to room temperature and requires a longer

time to complete the reaction (rt for 8 h). The yields for the reactions of n-hexanal and

methyl 9-oxononanoate were 72 and 58 percent, respectively. The olefin was formed

from the epoxide intermediate after treatment with the second aldehyde (R2CHO); n-

hexanal and methyl 9-oxononanoate gave yields of 76 and 62 percent, respectively. The

olefination reaction gave primarily trans stereoisomer. Finally, hydrolysis with porcine pancreas lipase (PPL) gave rise to the final product. 25 Starting from intermediate 22, the

overall yield was 60 %.

O O O 1 eq R1CHO Ph3P 1 eq R2CHO Ph3P S(CH3)2 R R2 R1 O 1 O 22

Bifunctional conjunctive ylide Epoxide formation Olefin formation Scheme 2.11 General strategy for synthesis of trans-EKODE-(E)-II family

The advantage of a bifunctional conjunctive ylide is that it represents two

different types of ylides such as sulfonium and phosphonium in the same molecule.

Consequently, this intermediate provides an opportunity to perform corresponding types of reactions with aldehydes, such as Corey-epoxidation and Wittig olefination. The presented structure of the trans-EKODE-(E)-II family in Figure 2.1 shows a palindromic

47

sequence of functional groups. In other words, by using two different aldehydes and

changing the order of addition to the ylides, both products can be obtained. The synthetic

route for trans-EKODE-(E)-IIa is presented in Scheme 2.12. The reaction of aldehyde 2

with the sulfonium ylide forms epoxy intermediate 23 (58 %), after which the

phosphonium ylide reacts with n-hexanal 24 and forms the olefin 25 (76

%).

O

1eq O OMe ,1eq NaH O O O O 5 1eq Ph3P Ph P 2 OMe 24 3 S(CH3)2 5 o O 22 Acetonitrile, -30 C to rt, overnight 23 Benzene, reflux, 48 h

O O O O PPL OMe OH O 5 Acetone, water, 3 h O 5 trans-EKODE-(E)-IIa 25

Scheme2.12 Synthetic route for trans-EKODE-(E)-IIa

Similarly, the synthetic route for trans-EKODE-(E)-IIa is presented in Scheme

2.12. The reaction of aldehyde 24 with sulfonium ylide formed an epoxy intermediate 26

(72%), after which the phosphonium ylide reacted with n-hexanal 24 and formed the olefin 27 (62 %).

48

O O O O NaH 1eq O OMe 1eq ,1eq Ph P 5 3 2 Ph3P S(CH3)2 24 o O 22 Acetonitrile, -30 C to rt, 3 h 26 Benzene, reflux, 48 h

O O O O PPL HO MeO 5 O 5 O Acetone, water, 3 h 27 trans-EKODE-(E)-IIb Scheme 2.13 Synthetic route for trans-EKODE-(E)-IIa an b

The described method was applied to the synthesis of the trans-EKODE-(E)-II family. Our synthesis commenced from bifunctional conjunctive ylide with the addition of two known aldehydes. Finally, hydrolysis with porcine pancreas lipase (PPL) gave rise to the final product in quantitative yield.25 By changing the order of addition for two

aldehydes, both trans-EKODE-(E)-IIa and -b were obtained. Starting from intermediate

22, the overall yield was 41 % for trans-EKODE-(E)-IIa and –b.

2.4.4 Divergent synthesis for the varied library of trans-EKODE-(E)-II

Since our initial intention was studying physiological roles of the trans-EKODE-

(E)-II family, we envisioned that the synthesis of modified alkyne-terminal and deuterated-labeled lipids could be useful for our purpose.

2.4.4.1 Synthesis of alkyne-end aldehyde

To make the alkyne-end aldehyde, propargyl alcohol 14 was reacted with 2eq amount of n-butyllithium at -78 oC and treated with n-bromopropane to give hex-2-yn-1-

ol 28 (57 % ). Next, the alkyne-zipper reaction provided by a strong base moved the triple

bond to the terminal end of the molecule to form terminal alkyne in product 29.

Oxidation of primary alcohol with PCC provided an aldehyde 30 (68 %).

49

n-BuLi, 1-bromopropane HO Li, , 70 o 1h HO (CH2)3(NH2)2 C, o H THF / HMPA -78 C to rt / overnight 28 t-BuOK, rt, 3h 14

PCC HO O DCM, 3h 30 29

Scheme 2.14 Synthetic route for alkyne-end aldehyde

2.4.4.2. Synthesis of deuterated labeled aldehyde

To make the deuterated-labeled aldehyde, d11-carboxylic acid 31 was reduced with LiAlH4, followed by oxidation of alcohol 32 with PCC to obtain an aldehyde.

D D D D D D D D D D D D PCC LiAlH4,ether, 2h HOH C HOOC 2 CD3 O CD3 CD3 D D D D CH2Cl2, 3h D D D D D D D D

32 33 31

acid Hexanoic-d11

Scheme 2.15 Synthetic route for deuterated-labeled aldehyde

Bifunctional conjunctive ylide was treated with sodium hydride and then aldehyde

(R1CHO) to form an epoxide. Yields are between. 58-72%.

50

O - O Ph3P 1eq R1CHO, 1eq NaH S(CH3)2 BF4 Ph3P o R Acetonitrile, -30 C to rt, overnight O 1 Bifunctional conjuctive ylide

entry aldehyde product yield(%)*

O O O 1 OMe Ph3P 58 O 5 OMe O 5 2 23 O 2 Ph3P O 72 O 24 26

O Ph P 3 3 67 O O 34 30

D D D D O D D D D 4 Ph3P 70 O CD3 CD D O 3 D D D D D D D

33 35

Table 2.1 Reaction of sulfonium ylide and aldehydes, * yields were calculated based of amount obtained after purification

Epoxide intermediate was treated with aldehyde (R2CHO) and formed the olefin.

This reaction gave primarily trans stereoisomer. The yields were between 55- 76 %.

51

O O 1eq R CHO Ph3P 2 R R2 R1 O 1 Benzene, reflux, 48h O

entry substrate aldehyde product yield(%)*

O O 1 23 O O 76 24 O 5 25 O O

O O 71 2 23 30 O 5 36

D D D D D D D D O O 63 D C O O CD3 3 O 5 3 23 D D D D D D D D 37 33 O O O 62 O O 5 OMe 27 4 26 O 5 2 O O O O 5 55 OMe O 5 34 O 5 38 2 O O O D D D D D C O OMe 3 5 59 6 35 O O 5 D D D D 39 2

Table 2.2 Reaction of phosphonium ylide and aldehydes.* yields were calculated based of amount obtained after purification

In summary, we have described a divergent synthetic route to gain access to the

library of the trans-EKODE-(E)-II family and their alkyne-end and deuterium-labeled homologues. The route we present allowed us to prepare gram quantities of these molecules. Such a strategy might also be useful for the rapid generation of other lipid metabolites derived from other PUFAs with a similar combination of functionalities that are valuable as biologically active molecules.

52

2.5 Experimental

2.5.1 General experimental details

All reactions were performed in oven-dried glassware, under an argon atmosphere

with rigid exclusion of moisture from reagents and solvents. Hexanoic-d11 acid (98 atom

% 2H) and PPL (porcine pancreas lipase, Type II) were purchased from Sigma-Aldrich.

All other reagents were used as supplied by chemical manufacturers. The exact molarity of n-butyllithium was determined each time by titration prior to use with 2-propanol,

Et2O at 0 °C and 1,10-phenanthroline as the indicator. Tetrahydrofuran was distilled from a purple solution of sodium benzophenone ketyl. All other solvents were used as purchased.

Ozonolysis was performed on the ozone generator lab series (L21) (Pacific

Ozone), using industrial grade compressed oxygen (purity 99.5 %). Liquid

chromatography was performed using forced air-flow (flash chromatography) on silica

gel (230- 400 mesh), by eluting solvent (reported as V: V ratio mixture). Analytical thin

layer chromatography (TLC) was performed on Partisil K6F Silica Gel 60A 0.25 mm

plates with fluorescent indicator. Visualization of the developed chromatogram was

accomplished with UV light (254 nm) and stained with either ethanolic phosphomolybdic

acid (PMA) or ceric ammonium molybdate. 1H and 13C NMR spectra were either

recorded on a Varian Inova spectrometer operating at 400 MHz and 100 MHz for the 1H and 13C respectively or Bruker Ascend Avance III HDTM operating at 500 MHz and 125

MHz for the 1H and 13C respectively (Department of Chemistry, Case Western Reserve

University). The internal references were CDCl3 (δ 7.26) and CD3OD (δ 3.31) for 1H and

(δ 77.36) and CD3OD (δ 49.00) for 13C spectra, respectively. NMR data are presented in

53

the following order: chemical shift, peak multiplicity (b = broad, s = singlet, d = doublet,

t = triplet, q = quartet, m = multiplet, dd = doublet of doublet, ddd = doublet of doublet of

doublet, ddt = doublet of doublet of triplet, dq = doublet of quatrate, dm = doublet of

multiplet, br = broad), coupling constant (in Hz). Mass spectra were acquired on a

Thermo Scientific LTQ-FT hybrid mass spectrometer (Rieveschl Laboratories for Mass

Spectrometry, University of Cincinnati) using electrospray ionization (positive mode).

54

2.5.2 Syntheses

Methyl 9-oxononanoate (2) (C10H18O3)

COOMe O

An oven-dried 0.5-L, two-necked round-bottomed flask was equipped with a mechanical stirrer. The flask was charged with methyl oleate 1 (20.8 g, 70 mmol) in dichloromethane (250 mL) and methanol (100 mL). The vessel was cooled to -78 °C and ozone was bubbled through the solution using a Pasteur pipet until a persistent dark blue

color appeared. The solution was purged with argon gas until the color dissipated, and

then the cold bath was removed. The ozonide was decomposed by adding glacial acetic

acid (25ml) in one portion, followed by powdered Zn (9 g, 0.14 mol) in small portions to

control heat evolution. The mixture continued to stir for an additional half an hour and

then filtered through Celite to remove unreacted Zn in the mixture. Water (100 mL) was

added to the mixture to prevent acetal formation. The solution was concentrated to about

one-half the initial volume in vacuo and slowly added to saturated NaHCO3 (150 mL).

The mixture was extracted with CH2Cl2 (3 × 200 mL). The organic phases were dried and

concentrated. The crude product was distilled to produce nonanal (bp 44 oC, 1 Torr) and

9.82 (75 %) of methyl 9-oxononanoate 2 (bp 103 oC / 1 Torr) as a colorless oil. (lit.26 bp

105-108 oC / 1 Torr)(Caution: Ozone is highly toxic and can react explosively with

numerous oxidizable materials.) Identity and purity of the product were confirmed by 1H

26 1 NMR. Spectroscopic data were found to match lit. data. H NMR (400 MHz, CDCl3) δ

1.28-1.36 (m, 6H) 1.58-1.66 (m, 4), 2.42 (td, 2H, J = 7.2, 2.0 Hz), 3.66 (s, 3H), 9.76 (t,

3H, J = 2 Hz).

55

Methyl (E)-11-oxoundec-9-enoate (4) (C12H20O3)

COOMe O

An oven-dried 50-mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with methyl 9-oxononanoate 3 (1.90 g, 10.22 mmol) and (triphenylphosphoranylidene)acetaldehyde 3 (3.72 g, 12.26 mmol) in dry chloroform

(20 mL). The mixture was heated under reflux at 70 oC for 48 h. Next, water (30 mL) was

added to the mixture, which was extracted with CH2Cl2 (3 × 30 mL).The combined

organic phases were dried with anhydrous magnesium sulfate and concentrated under

reduced pressure. Purification by column chromatography (Hexanes: ether / 80: 20)

afforded 1.23 g (57 %) of methyl (E)-11-oxoundec-9-enoate 4 as a yellow oil. Identity and purity of the product were confirmed by 1H NMR. Spectroscopic data were found to

1 match lit. data.16 H NMR (400 MHz, CDCl ) δ 1.28-1.39 (6H), 1.45-1.69 (4H), 2.28- 3

2.37(m, 4H), 3.67 (s, 3H), 6.08 (ddt, 1H, J = 15.6, 8.0, 1.6 Hz), 6.85 (dt, 1H, J = 15.6, 6.8

Hz), 9.50 (d, 1H, J = 8.0 Hz).

trans-methyl 8-(3-formyloxiran-2-yl)octanoate (5) ( C12H20O4)

COOMe O O

An oven-dried 50-mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with methyl (E)-11-oxoundec-9-enoate 4 (1.20 g, 5.66 mmol) in MeOH (15 mL). The flask was cooled in an ice-bath and NaHCO3 (570 mg,

6.78 mmol) was added. The reaction mixture was cooled between 0 °C, and H2O2

solution (1.7 mL, 16.7 mmol, 30% solution) was added dropwise via syringe. The

56

resulting mixture was vigorously stirred at ambient temperature for 3 h. The resulting

suspension was cooled between 0-5 °C in an ice bath and the excess hydrogen peroxide

was quenched with a saturated solution of sodium thiosulfate (2.0 mL dropwise). The

mixture was concentrated by rotary evaporation at room temperature. The aqueous phase

was extracted with EtOAc (3 × 10 mL) the combined organic phases dried with

anhydrous magnesium sulfate and concentrated under reduced pressure. Purification by

column chromatography (Hexanes: ether / 75: 25) afforded 488 mg (38 %) of trans-

methyl 8-(3-formyloxiran-2-yl)octanoate 5 as yellow oil. Identity and purity of the

product were confirmed by 1H NMR. Spectroscopic data were found to match lit. data.16

1 H NMR (400 MHz, CDCl3) δ 1.22-1.52 (8H), 1.52-1.70 (4H), 2.32 (t, 2H, J = 7.2 Hz),

3.15 (dd, 1H, J = 6.4 Hz and 2.0 Hz), 3.23 (td, 1H, J = 5.6 Hz and 2.0 Hz), 3.68 (s, 3H),

9.02 (d, 1H, J = 6.4 Hz).

5 1-(triphenyl-l λ -phosphanylidene)heptan-2-one (7) ( C25H27OP)

O Ph P 3 An oven-dried 50-mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with hexanoic acid 6 (2.09 g, 18 mmol). Then oxalyl

chloride (27 mmol) was added slowly via syringe under argon at room temperature. Once

evolution of gas had stopped (ca. 30 min), the mixture was heated under reflux for 2 h

and then cooled to room temperature. The excess oxalyl chloride was distilled at

atmospheric pressure and the remaining material was distilled to produce 2.20 g (91 %) hexanoyl chloride (bp 145 oC / 750 Torr) as a colorless oil. (lit.27 bp 151-153 oC / 1 Torr).

It was directly used for the next reaction.

57

Next, an oven-dried 100-mL, two-necked round-bottomed flask was equipped

with a magnetic stirring bar and sealed under argon with two rubber septa, one of which

contained a needle adapter, to an argon-inlet. The flask was charged with methyltriphenyl-phosphonium bromide (3.66 g, 10.26 mmol) in THF (20 mL). The solution was cooled in a dry ice-acetone bath at -78 °C. Then n-butyllithium (2.12 mL,

2.42 M in hexane, 5.13 mmol) was added, causing a red color to develop. The reaction

mixture continued to stir for an additional 1 h, whereupon hexanoyl chloride (690 mg,

5.13 mmol) was added dropwise slowly via syringe. The mixture was then allowed to stir

at ambient temperature for 1 h. The reaction mixture was neutralized with aqueous

saturated NH4Cl (10 mL) and evaporated the THF and resulted a viscous oil. The oil was extracted with EtOAc( 3 × 20 mL) and washed with NaOH (2 N, 50 mL).The combined organic phases were dried with anhydrous magnesium sulfate and concentrated under reduced pressure. Purification by column chromatography (DCM: Methanol / 98: 2) afforded 860 mg (45 %) of 1-(triphenyl-l λ5-phosphanylidene)heptan-2-one 7 as brown oil. Identity and purity of the product were confirmed by 1H NMR. 1H NMR (500 MHz,

2 CDCl3) δ 0.88 (bs, 3H,), 1.1-1.89 (m, 4H), 2.3 (bs, 2H), 3.72(d, 1H, JHP = 26.0 Hz), 7.42-

7.71 (m, 15H).

Methyl (E)-8-(3-(3-oxooct-1-en-1-yl)oxiran-2-yl)octanoate (8) ( C19H32O4)

O COOMe O

An oven-dried 50-mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with 1-(triphenyl-l λ5-phosphanylidene)heptan-2-one

58

7 (749mg, 2 mmol) and CH2Cl2 (10 mL). The resulting mixture was cooled to 0 °C in an ice bath. Then trans-methyl 8-(3-formyloxiran-2-yl)octanoate 5 (456mg, 2 mmol) was

added in CH2Cl2 (10 mL) via syringe. The reaction mixture continued to stir for an

additional 4 h at 0 °C. Next water (10 mL) was added to the mixture and extracted with

CH2Cl2 (3 × 20 mL). The combined organic phases were dried with anhydrous

magnesium sulfate and concentrated under reduced pressure. Purification by column

chromatography (Hexanes: ether / 85: 15) afforded 305 mg (47%) of methyl (E)-8-(3-(3-

oxooct-1-en-1-yl)oxiran-2-yl) octanoate 8 as a white oil. Identity and purity of the

product were confirmed by 1H NMR and 13CNMR. Spectroscopic data were found to

16 1 match lit. data. H NMR ( 500 MHz, CDCl3) δ 0.89 (t, 3H, J = 7.0 Hz), 1.22-1.69 (m,

18H), 2.30 (t, 2H, J = 7.5 Hz), 2.53 (t, 1H, J = 7.5 Hz), 2.89 (td, 1H, J = 5.5, 2.0 Hz),

3.20 (dd, 1H, J = 7.0, 2.0 Hz), 3.67 (s, 1H), 6.38 (d, 1H, J = 16.0 Hz), 6.51 (dd, 1H, J =

13 16.0, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 14.26, 22.79, 24.06, 25.20, 26.09, 29.31,

29.43, 29.46, 31.74, 32.22, 34.37, 41.00, 51.82, 56.97, 61.88, 131.69, 142.77, 174.60,

200.09.

General procedure: PPL hydrolysis

An oven-dried 25-mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with 0.1 mmol of the desired methyl ester EKODE-

(E) and PPL (220 mg) in acetone (1 ml) and phosphate buffer (pH 7, 5 mL, 0.2 M). The

mixture was stirred vigorously at ambient temperature for 4h and filtered through a pad

of Celite with EtOAc. The two phases separated and the organic phase was concentrated

59

under reduced pressure. Purification by column chromatography (Hexane: EtOAc / 50:

50) afforded the final product of carboxylic acids.

(E)-8-(3-(3-oxooct-1-en-1-yl)oxiran-2-yl)octanoic acid (trans-EKODE-(E)-Ia)

(C18H30O4)

O COOH O

Following the general procedure for PPL hydrolysis of methyl (E)-8-(3-(3- oxooct-1-en-1-yl)oxiran-2-yl)octanoate 8 afforded 29mg(93 %) of trans-EKODE-(E)-Ia as white solid. Identity and purity of the product were confirmed by 1H NMR and

13CNMR. Spectroscopic data were found to match lit. data.16 1H NMR ( 500 MHz,

CDCl3) δ 0.88 (t, 3H, J = 7.0 Hz), 1.23-1.69 (m, 18H), 2.35 (t, 2H, J = 7.5 Hz), 2.53 (t,

1H, J = 7.0 Hz), 2.91 (td, 1H, J = 5.5, 2.0 Hz), 3.21 (dd, 1H, J = 7.0, 2.0 Hz), 6.39 (d, 1H,

13 J = 15.5 Hz), 6.51 (dd, 1H, J = 15.5, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 14.26,

22.79, 24.07, 24.94, 26.08, 29.22, 29.41, 29.44, 31.75, 32.21, 34.09, 40.96, 56.99, 61.89,

131.70, 142.80, 180.50, 200.18.

9-methoxy-9-oxononanoic acid (9) (C10H18O4)

HOOC COOMe

An oven-dried 100-mL, two-necked round-bottomed flask containing a magnetic

stirring bar was equipped with an equalizing dropping funnel and an argon inlet. The flask was charged with methyl 9-oxononanoate 2 (2.8 g, 13.8 mmol) in acetonitrile (15 mL) and NaH2PO4 (480 mg) in water (6 mL) and H2O2 (1.74 mL, 17.1 mmol, 30 %). A

solution of NaClO2 (2.4 g, 21 mmol, 80 % purity) in water (20 mL) was added drop-wise

60

over 2 h to a stirred mixture while keeping the temperature at 10 oC with cooling. A small

amount (0.2 g) of Na2SO3 was added to destroy the unreacted HOCl and H2O2. The

mixture was acidified with 10 % aqueous HCl to pH 1 and concentrated by rotary

evaporation at room temperature. The aqueous phase was extracted with EtOAc (3 × 10

mL), the combined organic phases dried with anhydrous magnesium sulfate and

concentrated under reduced pressure afforded 2.7 g (95 %) of 9-methoxy-9-oxononanoic

acid 9 as a colorless oil. Identity and purity of the product were confirmed by 1H and 13C

28 1 NMR. Spectroscopic data were found to match lit data. H NMR (400 MHz, CDCl3) δ

1.25-1.38 (m, 6H), 1.56-1.70 (m, 4H), 2.3 (t, 2H, J = 17.6 Hz), 2.34 (t, 2H, J = 17.6 Hz),

13 3.66 (s, 3H); C NMR (100 MHz, CDCl3) δ 24.86, 25.13, 29.12, 29.16, 29.19, 34.33,

51.83, 174.68, 180.43.

Methyl 9-oxo-10-(triphenyl-l5-phosphanylidene)decanoate (10) (C29H33O3P)

O Ph P COOMe 3 An oven-dried 50-mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with 9-methoxy-9-oxononanoic acid 9 (2.43 g, 12 mmol). Then oxalyl chloride (18 mmol) was added slowly via syringe under argon at room temperature. Once evolution of gas had stopped (ca. 30 min), the mixture was heated under reflux for 2 h and then cooled to room temperature. The excess oxalyl chloride was distilled at atmospheric pressure and the remaining material was distilled to produce 2.50 mg (94 %) of methyl 9-chloro-9-oxononanoate (bp 125 oC / 1 Torr) as a

colorless oil. (lit.29 bp 125-127 oC / 1 Torr) Identity and purity of the product were

confirmed by 1H NMR. Spectroscopic data were found to match lit. data.16 1H NMR (500

61

MHz, CDCl3) δ 1.25-1.39 (m, 1H), 1.58-1.75 (m, 1H), 2.30 (t, 2H, J = 7.0 Hz), 2.88 (t,

2H, J = 7.0 Hz), 3.67 (s, 3H).

Next, an oven-dried 100-mL, two-necked round-bottomed flask was equipped

with a magnetic stirring bar and was sealed under argon with two rubber septa, one of

which contained a needle adapter, to an argon-inlet. The flask was charged with methyltriphenyl-phosphonium bromide (3.66 g, 10.26 mmol) in THF (20 mL). The solution was cooled in a dry ice-acetone bath at -78 °C. Then n-butyllithium (2.20 mL,

2.33 M in hexane, 10.26 mmol) was added, causing a red color to develop. The reaction mixture continued to stir for an additional 1 h, whereupon methyl 9-chloro-9- oxononanoate (1.13 g, 5.13 mmol) was added dropwise slowly via syringe, the mixture was then allowed to stir at ambient temperature for an additional1 h. The reaction mixture was neutralized with aqueous saturated NH4Cl (10 mL) and evaporated the THF,

resulting in a viscous oil. The oil was extracted with EtOAc (3 × 20 mL) and washed

with NaOH (2 N, 100 mL).The combined organic phases were dried with anhydrous

magnesium sulfate and concentrated under reduced pressure, affording 2.5 g (53 %) of

methyl 9-oxo-10-(triphenyl-l5-phosphanylidene)decanoate 10 as a brown oil. The

product was used as crude in its crude form in the Wittig reaction.

trans-3-pentyloxirane-2-carbaldehyde (12) (C8H14O2)

O O An oven-dried, 50-mL, round-bottomed flask was equipped with a magnetic stirring bar. The flask was charged with (E)-oct-2-enal 11 (714 mg, 5.66 mmol) in MeOH

(15 mL). The flask was cooled in an ice-bath and NaHCO3 (570 mg, 6.78 mmol) was

62

added. The reaction mixture was cooled to 0 °C, and H2O2 solution (1.7 mL, 16.7 mmol,

30 %solution) was added dropwise via syringe. The resulting mixture was vigorously stirred at ambient temperature for 1.5 h. The resulting suspension was cooled between 0-

5 °C in an ice bath and the excess hydrogen peroxide was quenched with a saturated

solution of sodium thiosulfate (2.0 mL dropwise). The mixture was concentrated by

rotary evaporation at room temperature. The aqueous phase was extracted with EtOAc (3

× 10 mL), and the combined organic phases were dried with anhydrous magnesium sulfate and concentrated under reduced pressure. Purification by column chromatography

(Hexanes: ether / 75: 25) afforded 510mg (63 %) of trans-3-pentyloxirane-2- carbaldehyde 12 as yellow oil. Identity and purity of the product were confirmed by 1H

and 13C NMR. Spectroscopic data were found to match lit. data.16 1H NMR (400 MHz,

CDCl3) δ 0.89 (t, 3H, J = 7.2 Hz), 1.27-1.70 (m, 8H), 3.13 (dd, 1H, J = 6.4, 2.0 Hz), 3.21-

3.24 (m, 1H), 9.01 (d, 1H, J = 6.4 Hz).

Methyl (E)-9-oxo-11-(3-pentyloxiran-2-yl)undec-10-enoate (13) (C19H32O4)

O O

MeO O

An oven-dried 50- mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with methyl 9-oxo-10-(triphenyl-l5-phosphanylidene) decanoate 10 (921 mg, 2 mmol) and CH2Cl2 (10 mL). The resulting mixture was cooled to 0 °C in an ice bath. Trans-3-pentyloxirane-2-carbaldehyde 5 (284 mg, 2 mmol) was added in CH2Cl2 (10 mL) via syringe. The reaction mixture continued to stir for an

additional4 h at 0 °C. Next, water (10 mL) was added to the mixture and extracted with

CH2Cl2 (3 × 20 mL). The combined organic phases were dried with anhydrous

63

magnesium sulfate and concentrated under reduced pressure. Purification by column

chromatography (Hexanes: ether / 85: 15) afforded 363 mg (56 %) of methyl (E)-9-oxo-

11-(3-pentyloxiran-2-yl)undec-10-enoate 13 as a yellow oil. Identity and purity of the product were confirmed by 1H and 13C NMR. Spectroscopic data were found to match lit.

16 1 data. H NMR (500 MHz, CDCl3) δ 0.89 (t, 3H, J = 7.0 Hz), 1.24-1.67 (m, 18H), 2.34

(t, 2H, J = 7.5 Hz), 2.53 (td, 1H, J = 7.0, 2.0 Hz), 2.91 (td, 1H, J = 7.0, 2.0 Hz), 3.21 (dd,

1H, J = 7.0, 2.0 Hz), 3.63 (s, 3H), 6.35 (d, 1H, J = 16.0 Hz), 6.45(dd, 1H, J = 15.5, 7.0

13 Hz); C NMR (125 MHz, CDCl3) δ 14.31, 22.87, 24.24, 25.20, 25.84, 29.27, 29.35(2C),

31.86, 32.24, 34.39, 40.90, 51.83, 56.99, 61.99, 131.62, 142.92, 174.63, 200.02.

(E)-9-oxo-11-(3-pentyloxiran-2-yl)undec-10-enoic acid (trans-EKODE-(E)-

Ib)(C18H30O4)

O O

HO O

Following the general procedure for PPL hydrolysis of methyl (E)-9-oxo-11-(3- pentyloxiran-2-yl)undec-10-enoate 13 afforded 28.0 mg (90 %) of trans-EKODE-(E)-Ib

as a yellow solid. Identity and purity of the product were confirmed by 1H and 13C NMR.

16 1 Spectroscopic data were found to match lit. data. H NMR (500 MHz, CDCl3) δ 0.89 (t,

3H, J = 7.0 Hz), 1.24-1.67 (m, 18H), 2.34 (t, 2H, J = 7.5 Hz), 2.53 (td, 1H, J = 7.0, 2.0

Hz), 2.91 (td, 1H, J = 7.0, 2.0 Hz), 3.21 (dd, 1H, J = 7.0, 2.0 Hz), 6.42(d, 1H, J = 15.5

13 Hz), 7.06(dd, 1H, J = 16.0, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 14.40, 22.87, 24.23,

24.92, 25.83, 29.16, 29.31(2C), 31.85, 32.23, 34.14, 40.86, 57.02, 62.00, 131.17, 142.93,

179.18, 200.04.

64

oct-2-yn-1-ol (15) (C8H14O)

HO

An oven-dried 500 mL, two-necked round-bottomed flask equipped with a magnetic stirring bar was sealed under argon with two rubber septa, one of which contained a needle adapter attached to an argon-filled balloon. The flask was charged with propargyl alcohol 14 (5.61 g, 0.1 mol), tetrahydrofurane (THF, 150 mL) and hexamethyl phosphoric triamide ( HMPA, 40 mL). The solution was cooled in a dry ice- acetone bath at -78 °C, and n-butyllithium (81.6 mL, 2.45 M in hexane, 0.2mol) was added slowly via syringe. The resulting solution was stirred for 5 min at -78 °C, after which it was slowly warmed to -30 °C. Following, 1-bromopentane (9.23 g, 75 mmol) was added slowly to the mixture via syringe. The temperature was slowly increased to ambient temperature overnight while the mixture stirred. The reaction mixture was neutralized with aqueous saturated NH4Cl (100 mL) and extracted with EtOAc (3 × 100 mL), the combined organic phases were dried with anhydrous sodium sulfate and

concentrated under reduced pressure. Purification by column chromatography (EtOAc:

Hexanes / 85: 15) afforded 6.30 g (67% yield) of oct-2-yn-1-ol 15 as a colorless oil.

Identity and purity of the product were confirmed by 1H and 13C NMR. Identity and

purity of the product were confirmed by 1H NMR. Spectroscopic data were found to

30 1 match lit. data. H NMR (400 MHz, CDCl3) δ 0.90 (t, 1H, J = 7.2 Hz), 1.28-1.39 (m,

4H), 1.51 (quint, 2H, J = 7.2 Hz), 2.21 (tt, 2H, J = 7.2, 2.0 Hz), 4.25 (t, 2H, J = 2.0 Hz);

13 C NMR (100 MHz, CDCl3) δ 14.29, 19.02, 22.53, 28.62, 31.36, 51.71, 78.56, 86.95.

65

(Z)-oct-2-en-1-ol (16) ( C8H16O)

HO

An oven-dried 1L round-bottomed flask was equipped with a magnetic stirring bar. The flask was charged with oct-2-yn-1-ol 15 (3.03 g, 24.0 mmol) and hexane (350

mL) under an argon atmosphere. To this solution, distilled quinoline (0.9 mL) and

Lindlar catalyst (0.36 g, 12% by weight) were added. The argon was evacuated by water aspirator and then filled with hydrogen from the balloon by adjusting the three-way valve. The reaction was monitored by 1H NMR and completed within 8 hours. The hydrogen atmosphere was replaced with argon and the mixture was filtered using a pad of

Celite. (Note: The filter cake must be kept wet with solvent because the palladium saturated with hydrogen is pyrophoric in air when dry.) The hexane was evaporated and the residue was purified by flash column chromatography (silica, EtOAc: Hexanes / 15:

85) affording 2.82 g (92 % yield) of (Z)-oct-2-en-1-ol 16 as a colorless oil. Identity and purity of the product were confirmed by 1H and 13C NMR. Spectroscopic data were found

31 1 to match lit. data. H NMR (400 MHz, CDCl3) δ 0.88 (t, 1H, J = 6.8 Hz), 1.23-1.41 (m,

6H), 2.07 (q, 2H, J = 7.2 Hz), 4.19 (d, 2H, J = 6.4 Hz), 5.47-5.68 (m, 2H); 13C NMR (100

MHz, CDCl3) δ 14.40, 22.86, 27.73, 29.62, 31.75, 58.96, 128.58, 133.67.

(Z)--(3-pentyloxiran-2-yl)methanol (17) (C8H16O2)

HO O

An oven-dried 250 mL, round-bottomed flask was equipped with a magnetic stirring bar. The flask was charged with m-chloroperoxybenzoic acid (m-CPBA) (7.9 g,

35.2 mmol, 77 % purity) and CH2Cl2 (80 mL). The reaction mixture was cooled to 0 °C

66

and (Z)-oct-2-en-1-ol (4.50 g, 35.2 mmol) in CH2C12 (5 mL) was added.The solution was warmed to room temperature and stirred for 2 h. The excess oxidant was quenched with a saturated solution of sodium thiosulfate (5.0 mL dropwise) and continued stirring for an additional 1 h. The reaction mixture was extracted with CH2C12 (3 × 30 mL), the

combined organic phases dried with anhydrous magnesium sulfate and concentrated

under reduced pressure. The crude product was distilled to give 4.82 g (95%) of (Z)-(3-

pentyloxiran-2-yl)methanol 17 (bp 60 oC, 0.2 Torr) as a colorless oil. Identity and purity of the product were confirmed by 1H and 13C NMR. Spectroscopic data were found to

32 1 match lit. data. H NMR (400 MHz, CDCl3) δ 0.90 (t, 3H, J = 7.2 Hz), 1.30-1.35 (m,

4H), 1.45-1.60 (m, 4H), 3.00-3.06 (m, 1H), 3.16 (dt, 1H, J = 7.2, 4.0 Hz), 3.68 (dd, 1H, J

13 = 7.2, 12.0 Hz), 3.86 (dd, 1H, J = 1.6, 12.0 Hz); C NMR (100 MHz, CDCl3) δ 14.32,

22.89, 26.66, 28.27, 31.92, 57.17, 57.70, 61.29.

(Z)-3-pentyloxirane-2-carbaldehyde (18) (C8H14O2)

O O

An oven-dried 250-mL, round-bottomed flask was equipped with a magnetic stirring bar and a rubber septum. The flask was charged with pyridinium chlorochromate

(12.26 g, 57 mmol) and 85 mL of dichloromethane. A solution of (3-pentyloxiran-2- yl)methanol 17 ( 4.10 g, 28.5 mmol) in 15 mL of dichloromethane was transferred into the reaction mixture via syringe over 5 min. The reaction mixture turned dark brown. The resulting mixture was stirred at ambient temperature for 3 h. The reaction mixture was then charged with 60 mL of diethyl ether and silica gel (30 g). The solution was decanted, and the remaining dark brown resinous was thoroughly washed with three 30-

67

mL portions of diethyl ether. The combined dark brown / black ether was filtered and

concentrated by rotary evaporation at room temperature. Purification by column

chromatography (Hexanes: ether / 70: 30) afforded 2.23 g (55 % yield) of (Z)-3- pentyloxirane-2-carbaldehyde 18 as colorless oil. Identity and purity of the product were

1 13 1 confirmed by H and C NMR. H NMR (400 MHz, CDCl3) δ 0.89 (t, 3H, J = 7.2 Hz),

1.29-1.36 (m, 6H), 1.60-1.80 (m, 2H), 3.23-3.28 (m, 1H), 3.34 (t, 1H, J = 4.8 Hz), 9.46

13 (d, 1H, J = 5.2 Hz); C NMR (100 MHz, CDCl3) δ 14.24, 22.78 , 26.59, 28.42, 31.66,

58.26, 59.56, 199.55.

Methyl (E)-9-oxo-11-(3-pentyloxiran-2-yl)undec-10-enoate (19) (C19H32O4)

O O

O O

An oven-dried 50-mL, round-bottomed flask was equipped with a magnetic

stirring bar. The flask was charged with methyl 9-oxo-10-(triphenyl-l5-

phosphanylidene)decanoate 10 (1.11 g, 2.41 mmol) and CH2Cl2 (12 mL). The resulting

mixture was cooled to 0 °C in an ice bath. Then, cis-3-pentyloxirane-2-carbaldehyde 18

(291 mg, 2.41 mmol) was added in CH2Cl2 (12 mL) via syringe. The reaction mixture

continued to stir for an additional 4 h at 0 °C. Water (12 mL) was added to the mixture

and extracted with CH2Cl2 (3 × 25 mL).The combined organic phases were dried with

anhydrous magnesium sulfate and concentrated under reduced pressure. Purification by

column chromatography (Hexanes: ether / 85: 15) afforded 398 mg (51 %) of methyl (E)-

9-oxo-11-(3-pentyloxiran-2-yl)undec-10-enoate 19 as a yellow oil. Identity and purity of

the product were confirmed by 1H and 13C NMR. Spectroscopic data were found to match

68

16 1 lit. data. H NMR (500 MHz, CDCl3) δ 0.89 (t, 3H, J = 7.0 Hz), 1.25-1.68 (m, 18H),

2.34 (m, 2H), 2.53 (t, 1H, J = 7.0 Hz), 3.19 (m, 1H), 3.51 (dd, 1H, J = 6.0, 5.0 Hz), 3.65

(s, 1H), 6.41 (d, 1H, J = 16.0 Hz), 6.62 (dd, 1H, J = 16.0, 7.0 Hz); 13C NMR (125 MHz,

CDCl3) δ 14.26, 22.83, 24.30, 25.21, 26.30, 27.93, 29.26, 29.36, 29.38, 31.79, 34.64,

41.18, 55.81, 60.19, 133.18, 140.04, 174.15, 199.64.

(E)-9-oxo-11-(3-pentyloxiran-2-yl)undec-10-enoic acid( cis-EKODE-(E)-

Ib)(C18H30O4)

O O

HO O

Following the general procedure for PPL hydrolysis of methyl (E)-9-oxo-11-(3- pentyloxiran-2-yl)undec-10-enoate 19 afforded 29.0 mg (93 %) of cis-EKODE-(E)-Ib as a yellow solid. Identity and purity of the product were confirmed by 1H and 13C NMR.

16 1 Spectroscopic data were found to match lit. data. H NMR (500 MHz, CDCl3) δ 0.89 (t,

3H, J = 7.0 Hz), 1.25-1.68 (m, 18H), 2.34 (t, 2H, J = 7.5 Hz), 2.54(t, 1H, J = 7.5 Hz),

3.20 (m, 1H), 3.52 (ddd, 1H, J = 6.0, 4.5, 0.5 Hz), 6.40 (dd, 1H, J = 15.5, 0.5 Hz),

13 6.65(dd, 1H, J = 15.5, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 14.28, 22.85, 24.30,

24.93, 26.31, 27.94, 29.18, 29.32, 29.35, 31.81, 34.17, 41.17, 55.85, 60.24, 133.20,

140.09, 179.28, 199.71.

69

1,3-dibromopropan-2-one (20) (C3H4Br2O)

O Br Br

An oven-dried 2.0-L, round-bottomed flask was equipped with a mechanical stirrer. The flask was charged with 1,3-dichloro-2-propanone (25 g, 196 mmol) in

acetone (750 mL). Powdered lithium bromide (150 g, 1.73mol) was added slowly using a funnel. The reaction mixture was stirred at ambient temperature for 48 h. Additional

lithium bromide (100 g, 1.15 mol) and acetone (250 mL) were added and stirring

continued for an extra 24 h. Evaporation of the solvent gave a white solid that was

transferred to a 2.0 L separatory funnel charged with 1.0 L of cold water. The aqueous

phase was extracted with methylene chloride (3 × 500 mL), the combined organic phases

were dried with anhydrous magnesium sulfate and concentrated under reduced pressure

to give 42.1 g (99 % crude yield) of the 1,3-dibromopropan-2-one as a yellow syrup.

(Caution: The product has a low boiling point. The water bath in rota-vap needs to be

cold to avoid losing the product. bp79.5-805. oC at 9 Torr).33 Analysis of the crude by 1H

NMR indicates conversion was > 90 %. Identity and purity of the product were

confirmed by 1H NMR and 13CNMR. Spectroscopic data from 1HNMR were found to

33 1 13 match lit. data. H NMR (400 MHz, CDCl3) δ 4.13; C NMR (100 MHz, CDCl3) δ

31.18, 194.13.

70

1-bromo-3-(triphenylphosphoranylidene)propan-2-one (21) (C21H18BrOP)

O Ph P Br 3

An oven-dried 500-mL, two-necked round-bottomed flask containing a magnetic

stirring bar was equipped with an equalizing dropping funnel and an argon inlet. The flask was charged with triphenylphosphine (49.8 g, 190 mmol) in toluene (125 mL). The solution of crude 1,3-dibromopropan-2-one (41.0 g, 190 mmol) in toluene (125 mL) was added through the dropping funnel. Stirring continued overnight at ambient temperature.

The white precipitate formed during the reaction was collected by filtration, washed with toluene, and concentrated under reduced pressure. Powdered sodium bicarbonate (42 g,

0.5 mol) was added to a stirred solution of dried in 60% aqueous methanol (800 mL)

was added. Stirring continued for an additional 30 min and more water (200 mL) was

added to the mixture. After being stirred for another 30 min, the solid was collected by

filtration, and thoroughly washed with water. The solid was transferred to a 2.0 L

separatory funnel charged with 1.0 L of cold water. The aqueous phase was extracted with methylene chloride (3 × 500 mL), the combined organic phases were dried with

anhydrous sodium sulfate and concentrated under reduced pressure to give 62.6 g (83%

crude yield) of the 1-bromo-3-(triphenylphosphoranylidene)propan-2-one as a white solid. Analysis of the crude by 1H NMR indicates conversion was >60% (contaminated

with 1-chloro-3-(triphenylphosphoranylidene)propan-2-one). Identity and purity of the

1 13 1 product were confirmed by H and C NMR and HRMS. H NMR (400 MHz, CDCl3) δ

4 2 13 3.91 (d, 2H, JHP = 1.6), 4.26 (d, 1H, JHP = 24.0), 7.42-7.70 (15H,m); C NMR (100

3 1 1 MHz, CDCl3) δ 35.77 (d, JCP = 17.4 Hz), 52.67 (d, JCP = 109.0 Hz), 125.95 (d, JCP = 56

3 4 2 Hz, C-1’), 129.22 (d, JCP = 12.3 Hz, C-3’) 132.65 (d, JCP = 2.8 Hz, C-4’) 133.36 (d, JCP

71

2 = 10.2 Hz, C-2’), 184.99 (d, JCP = 4.3 Hz), HRMS (ESI) m/z calcd for C21H19BrOP+

(M+1)+ 397.03514, found 397.03520.

Dimethyl(2-oxo-3-(triphenylphosphoranylidene)propyl)sulfonium

tetrafluoroborate [Bifunctional conjunctive ylide] (22) (C23H24BF4OPS)

O Ph P - 3 S(CH3)2 BF4

An oven-dried 1.0L, round-bottomed flask was equipped with a magnetic stirring bar. The flask was charged with sodium tetrafluoroborate (27.5 g, 0.25 mol), methyl

sulfide (15.5 g, 0.25 mol), 1-bromo-3-(triphenylphosphoranylidene)propan-2-one (19.86 g, 50 mmol) and acetone (500 mL). The reaction mixture continued to stir at ambient temperature for 48h. The filtrate was concentrated under reduced pressure. Purification by column chromatography (DCM: MeOH / 95: 5) afforded 17.95 g (77 % yield) of bifunctional conjunctive ylide as a white solid. Identity and purity of the product were confirmed by 1H and 13C NMR and HRMS.13C NMR data did not duplicate lit. data.23 1H

2 NMR (400 MHz, CDCl3) δ 2.91 (s,6H), 4.16 (d, 1H, JHP = 21.2 Hz), 4.42 (s, 2H), 7.47-

13 3 7.64 (15H, m); C NMR (100 MHz, CDCl3) δ 25.01, 52.43 (d, JCP = 20.5 Hz), 56.71 (d,

1 1 3 JCP = 104 Hz), 125.20 (d, JCP = 90.8 Hz, C-1’), 129.45 (d, JCP = 12.4 Hz, C-3’), 133.09

2 2 (bs, C-4’), 133.21 (d, JCP = 10.3 Hz, C-2’), 177.06 (d, JCP = 4.0 Hz), HRMS (ESI) m/z

+ + calcd for C23H24OPS (M+1) 379.12800, found 379.12807.(confirms the cation)

72

Hex-2-yn-1-ol (28) (C6H10O)

HO

An oven-dried 500-mL, two-necked round-bottomed flask equipped with a

magnetic stirring bar was sealed under argon with two rubber septa, one of which contained a needle adapter attached to an argon-filled balloon. The flask was charged with propargyl alchohol (5.61 g, 0.1 mol), tetrahydrofurane (THF, 150 mL) and hexamethyl phosphoric triamide (HMPA, 40 mL). The solution was cooled in a dry ice- acetone bath at -78 °C, and n-butyllithium (84.4 mL, 2.37 M in hexane, 0.2mol) was added slowly via syringe. The resulting solution was stirred for 5 min at -78 °C, after which slowly warmed to -30 °C. Next, 1-bromopropane (9.23 g, 75 mmol) was added slowly to the mixture via syringe. The temperature was slowly increased to ambient temperature overnight while the mixture stirred. The reaction mixture was neutralized with aqueous saturated NH4Cl (100 mL) and extracted with EtOAc (3 × 100 mL), and the combined organic phases were dried with anhydrous sodium sulfate and concentrated under reduced pressure. Purification by column chromatography (EtOAc: Hexanes / 85:

15) afforded 6.50 g (57 %) of product as a colorless oil. Identity and purity of the product

were confirmed by 1H and 13C NMR. Spectroscopic data were found to match lit. data.34

1 H NMR (400 MHz, CDCl3) δ 0.96 (t, 1H, J = 7.2 Hz), 1.51 (sextet, 2H, J = 7.2 Hz), 1.99

(bs, 1H), 2.17 (tt, 2H, J = 7.2, 2.0 Hz), 4.24 (dt, 2H, J = 5.6, 2.0 Hz); 13C NMR (100

MHz, CDCl3) δ 13.72, 20.96, 22.35, 51.41, 78.75, 86.44.

73

Hex-5-yn-1-ol 29 (C6H10O)

HO

An oven-dried 500-mL, two-necked round-bottomed flask equipped with a

magnetic stirring bar was sealed under argon with two rubber septa, one of which contained a needle adapter attached to an argon inlet. The flask was charged with Li (4.48 g, 0.64mol) and 1,3- diaminopropane (240 mL). The mixture was stirred while heating in an oil bath at 70 °C until the blue color discharged (1 h). The prepared lithium amide

suspension was cooled to room temperature. Next, potassium tert-butoxide (42.4 g, 384 mmol), was added to the flask using a powder funnel. The resulting pale yellow solution was stirred for 20 min at room temperature, and then Hex-2-yn-1-ol (6.27 g, 64 mmol) was added over 10 min via syringe. The reddish-brown mixture was stirred for 3h and then poured into ice-water (1L) and extracted with Et2O (3 × 300 mL). The ether extracts

were combined and successively washed with 1 L of water, 10 % hydrochloric acid and

saturated sodium chloride solution. The ether solution was dried over anhydrous

magnesium sulfate, filtered and concentrated under reduced pressure. Purification by

column chromatography (EtOAc: Hexanes / 80: 20) afforded 5.33 g (84 %) of product as

a colorless oil. Identity and purity of the product were confirmed by 1H and 13C NMR.

35 1 Spectroscopic data were found to match lit. data. H NMR (400 MHz, CDCl3) δ 1.54-

1.68 (m, 4H), 1.93 (t, 1H, J = 2.8 Hz), 2.19 (td, 2H, J = 6.8, 2.8 Hz), 2.56 (bs, 1H), 3.60

13 (t, 2H, J = 6.4 Hz); C NMR (100 MHz, CDCl3) δ 18.40, 24.95, 31.82, 62.31, 68.82,

84.58.

74

Hex-5-ynal (30) ( C6H12O)

O

An Oven-dried 500-mL, three-necked, round-bottomed flask was equipped with a

magnetic stirring bar, a rubber septum, a glass stopper and an argon inlet. The flask was

charged with pyridinium chlorochromate (21.5 g, 100 mmol) and dichloromethane (150

mL). A solution of Hex-5-yn-1-ol (4.9 g, 50 mmol) in 20 mL of dichloromethane was

transferred into the reaction mixture via syringe over 5 min, and the resulting mixture

was stirred at ambient temperature for 3 h. The reaction mixture was then charged with

125 mL of diethyl ether and silica gel (50 g), the solution was decanted and the remaining

dark brown resinous polymer was thoroughly washed with three 50-mL portions of

diethyl ether. The combined dark brown / black ether was filtered and concentrated by

rotary evaporation at room temperature. The crude product was distilled to give 2.49 g

(52 %) of product (bp 29 oC, 1 Torr) as a colorless oil. Identity and purity of the product were confirmed by 1H and 13C NMR. Spectroscopic data were found to match lit. data.36

1 H NMR (400 MHz, CDCl3) δ 1.85 (quintet, 2H, J = 7.2 Hz), 1.97 (t, 1H, J = 2.8 Hz),

2.26 (td, 2H, J = 7.2, 2.8 Hz), 2.60 (td, 2H, J = 7.2, 1.2 Hz), 9.80 (1H, bs); 13C NMR (100

MHz, CDCl3) δ 18.07, 21.09, 42.82, 69.68, 83.48, 202.13.

Hexan-2,2,3,3,4,4,5,5,6,6,6-d11-1-ol (32) (C6H3D11O)

D D D D

HO CD3 D D D D

An oven-dried 250-mL, three-necked round-bottomed flask was equipped with a

glass stopper, a dropping funnel, a reflux condenser topped with an drying tube and a

75

magnetic stirring bar. The flask was charged with LiAlH4 (1.4 g, 37 mmol) and

anhydrous ether (50 mL). A solution of hexanoic-d11 acid (3.2 g, 25 mmol) in ether (50 mL) was transferred to the addition funnel and added over a 10-min period. The reaction mixture was heated under reflux for another 2h, diluted with ether (50 mL) and quenched by addition of water (25 mL) slowly via syringe. Next, the mixture was extracted with

EtOAc (3 × 50 mL). The combined organic phases were dried with anhydrous sodium sulfate and concentrated under reduced pressure affording 2.75 g (97 %) of product as a colorless oil. Identity and purity of the product were confirmed by 1H and 13C NMR. 1H

13 NMR (400 MHz, CDCl3) δ 1.81 (s, 1H, OH), 3.60 (s, 2H); C NMR (100 MHz, CDCl3)

δ 62.90.

Hexanal-2,2,3,3,4,4,5,5,6,6,6-d11 (33) (C6HD11O)

D D D D

O CD3 D D D D

An oven-dried 250-mL, three-necked round-bottomed flask was equipped with a magnetic stirring bar, a rubber septum, a glass stopper and an argon inlet. The flask was charged with pyridinium chlorochromate (8.6 g, 40 mmol) and dichloromethane (60 mL).

A solution of hexan-2,2,3,3,4,4,5,5,6,6,6-d11-1-ol ( 2.26 g, 20 mmol) in dichloromethane

(10mL) was transferred into the reaction mixture via syringe over 5 min, and the resulting

mixture was stirred at ambient temperature for 3 h. The reaction mixture was then charged with diethyl ether (50 mL) and silica gel (20 g). The solution was decanted, and the remaining dark brown resinous polymer thoroughly washed with three 20-mL portions of diethyl ether. The combined dark brown / black ether was filtered and

76

concentrated by rotary evaporation at room temperature to give 1.51 g (68 %) of the

crude product as a yellow oil. The product was used with no further purification. Identity

and purity of the product were confirmed by 1H and 13C NMR. 1H NMR (400 MHz,

13 CDCl3) δ 9.74 (1H, CHO); C NMR (100 MHz, CDCl3) δ 203.66.

General procedure: Johnson–Corey–Chaykovsky reaction

An oven-dried 50-mL, two-necked round-bottomed flask containing a magnetic stirring bar was sealed under argon with two rubber septa, one of which contained a needle adapter attached to an argon-inlet. The solution was cooled in a dry ice-ethanol bath at -30 °C. The flask was charged with bifunctional conjunctive ylide (2.0 mmol) and sodium hydride (80 mg, 2.0 mmol, 60 % in mineral oil) and desired aldehyde (2.0 mmol) and acetonitrile (30 mL). The reaction mixture continued to stir while bring heat to room temperature over 3 h. The reaction mixture was neutralized with aqueous saturated

NH4Cl (20 mL) and extracted with EtOAc (3 × 20 mL), the combined organic phases

were dried with anhydrous sodium sulfate and concentrated under reduced pressure.

Purification by column chromatography (DCM: Methanol / 98: 2) afforded desired epoxy

products.

77

Methyl 8-(3-(2-(triphenyl- λ 5-phosphanylidene)acetyl)oxiran-2-yl)octanoate

(23) (C31H35O4P)

O O Ph3P O O

Following the general procedure for Johnson–Corey–Chaykovsky reaction and

using aldehyde 2 (372 mg, 2.0 mmol) and completion in 8 h. Purification by column

chromatography afforded 583 mg (58%) of product as a yellow oil. Identity and purity of

1 13 1 the product were confirmed by H, C NMR and HRMS. H NMR (400 MHz, CDCl3) δ

1.20-1.79 (m, 12H), 2.28 (t, 2H, J = 7.6 Hz), 3.05 (bm, 1H), 3.14 (t, 1H, J = 2 Hz), 3.65

2 13 (s, 3H), 3.94 (d, 1H, JHP = 25.6 Hz), 7.41-7.71 (m, 15H); C NMR (100MHz, CDCl3) δ

1 25.03, 26.11, 29.18, 29.26, 29.30, 32.23, 34.19, 48.26 (d, 1H, JCP = 98 Hz), 51.58, 59.90,

1 3 60.04 (d, J = 17 Hz), 126.56 (d, JCP = 91 Hz, C-1’), 129.05 (d, JCP = 12.3 Hz, C-3’),

4 2 2 132.40 (d, JCP = 2.7 Hz, C-4’), 133.17 (d, JCP = 10.3 Hz, C-2’), 174.40, 188.11 (d, JCP =

+ + 4.0 Hz), HRMS (ESI) m/z calcd for C31H36O4P (M+1) 503.23457, found 503.23469.

1-(3-pentyloxiran-2-yl)-2-(triphenyl-λ5-phosphanylidene)ethan-1-one (26)

(C27H29O2P)

O Ph3P O

Following the general procedure for Johnson–Corey–Chaykovsky reaction and

using aldehyde 24 (200 mg, 2.0 mmol) and purification by column chromatography

afforded 600 mg (72 %) of product as a yellow oil. Identity and purity of the product

1 13 1 were confirmed by H, C NMR and HRMS. H NMR (500 MHz, CDCl3) δ 0.88 (t, 3H,

78

2 J = 6.5 Hz), 1.22-1.82 (m, 8H), 3.05 (bm, 1H), 3.13 (t, 1H, J = 2.0 Hz), 3.94 (d, 1H, JHP=

13 24 Hz), 7.35-7.73 (m, 15H); C NMR (125 MHz, CDCl3) δ14.24, 22.80, 25.95, 31.82,

1 1 32.35, 48.86 (d, JCP = 110 Hz), 60.07, 60.17 (d, J = 16.6 Hz), 126.36 (d, JCP = 90.6 Hz,

3 4 2 C-1’), 129.15 (d, JCP = 12.3 Hz, C-3’), 132.49 (d, JCP = 2.8 Hz, C-4’), 133.31 (d, JCP =

2 + + 10.2 Hz, C-2’), 188.31 (d, JCP = 3.5 Hz), HRMS (ESI) m/z calcd for C27H30O2P (M+1)

417.19779, found 417.19787.

1-(3-(pent-4-yn-1-yl)oxiran-2-yl)-2-(triphenyl- λ5-phosphanylidene)ethan-1-

one (34) (C27H25O2P)

O Ph3P O

Following the general procedure for Johnson–Corey–Chaykovsky reaction and

using aldehyde 30 (192 mg, 2.0 mmol) and purification by column chromatography

afforded 553 mg (67 %) of product as a yellow oil. Identity and purity of the product

1 13 1 were confirmed by H, C NMR and HRMS. H NMR (500 MHz, CDCl3) δ 1.49-1.71

(m, 4H), 1.89 (t, 1H, J = 2.0 Hz), 2.16-2.24 (m, 2H), 3.05 (ddd, 1H, J = 8.5, 4, 2.0 Hz),

2 13 3.13 (t, 2H, J = 2.0 Hz), 3.99 (d, 1H, JHP = 23.5 Hz), 7.34-7.63 (m, 15H); C NMR (125

1 MHz, CDCl3) δ 18.25, 25.06, 31.06, 49.07 (d, 1H, JCP = 110 Hz), 59.10, 59.72 (d, J = 17

1 3 Hz), 68.70, 84.03, 126.44 (d, JCP = 91 Hz, C-1’), 129.01 (d, JCP = 12.4 Hz, C-3’), 132.38

4 2 2 (d, JCP = 2.8 Hz, C-4’), 133.15 (d, JCP = 10.3 Hz, C-2’), 187.65 (d, JCP = 4.0 Hz).

79

1-(3-(pentyl-d11)oxiran-2-yl)-2-(triphenyl-l5-phosphanylidene)ethan-1-one

(35) (C27H18D11O2P)

O D D D D Ph3P CD O 3 D D D D

Following the general procedure for Johnson–Corey–Chaykovsky reaction and

using aldehyde 33 (223 mg, 2.0 mmol) and purification by column chromatography

afforded 599 mg (70 %) of product as a brown oil. Identity and purity of the product were

1 13 1 confirmed by H, C NMR and HRMS. H NMR ( 500 MHz, CDCl3) δ 3.04 (b, 1H),

2 13 3.04 (t, 1H, J = 2.0 Hz), 3.94 (d, 1H, JHP = 24.0 Hz), 7.39-7.72 (m, 15 H); C NMR (125

1 1 MHz, CDCl3) δ 48.90 (d, JCP = 110.0 Hz), 60.09, 60.13 (d, J = 16.5 Hz), 126.79 (d, JCP

3 4 = 90.6 Hz, C-1’), 129.48 (d, JCP = 12.3 Hz, C-3’) 132.37 (d, JCP = 2.8 Hz, C-4’) 133.36

2 2 (d, JCP = 10.3 Hz, C-2’), 188.44 (d, JCP = 4.0 Hz), HRMS (ESI) m/z calcd for

+ + C27H19D11O2P (M+1) 428.267, found 428.266.

General procedure: Wittig reaction

An oven-dried 50-mL, round-bottomed flask, containing a magnetic stirring bar

charged with desired epoxy (1 mmol) from Johnson–Corey–Chaykovsky reaction and

desired aldehyde (1 mmol) and toluene (10 mL). The mixture was heated under reflux overnight and allowed to cool to room temperature. The reaction mixture was concentrated under reduced pressure. Purification by column chromatography (Hexane:

Et2O / 85: 15) afforded α, β-unsaturated keto epoxide products.

80

Methyl (E)-8-(3-(oct-2-enoyl)oxiran-2-yl)octanoate (25) (C19H32O4)

O O

O O

Following the general procedure for Wittig reaction between 23 and 24 and

purification by column chromatography afforded 278 mg (86%) of product as a yellow

oil. Identity and purity of the product were confirmed by 1H, 13C NMR and HMRS.

16 1 Spectroscopic data were found to match lit. data. H NMR (400 MHz, CDCl3) δ 0.87 (t,

3H, J = 7.2 Hz), 1.25-1.69 (m, 18H), 2.18 (dd, 2H, J = 15.0, 7.0 Hz), 2.30 (t, 2H, J = 7.5

Hz), 3.02(t, 1H, J = 6 Hz), 3.32 (bs, 1H), 3.67(s, 3H), 6.23 (d, 1H, J = 15.5 Hz), 7.08

13 (1H, dt, J = 15.5, 7.0 Hz); C NMR (100 MHz, CDCl3) δ 14.32, 22.76, 25.11, , 26.07,

27.97, 29.31, 29.41, 29.42, 31.71, 32.14, 33.08, 34.38, 51.83, 58.66, 59.36, 124.27,

+ + 151.02, 174.61. 196.05; HRMS (ESI) m/z calcd for C19H32O4Na (M+Na) 347.21928,

found 347.21943.

(E)-8-(3-(oct-2-enoyl)oxiran-2-yl)octanoic acid (trans-EKODE-(E)-

IIa)(C18H30O4)

O O

OH O

Following the general procedure for PPL hydrolysis of methyl (E)-8-(3-(oct-2-

enoyl)oxiran-2-yl)octanoate 25 afforded 146 mg (94 %) of product as a yellow oil.

Identity and purity of the product were confirmed by 1H, 13C NMR and HRMS.

16 1 Spectroscopic data were found to match lit. data. H NMR (500 MHz, CDCl3) δ 0.89 (t,

3H, J = 7.0 Hz), 1.21-1.74 (m, 18H), 2.17-2.24 (m, 2H), 2.34 (t, 2H, J = 7.0 Hz), 3.04

81

(ddd, 1H, J = 6.0, 5.0, 2.0 Hz), 3.33 (d, 1H, J = 2.0 Hz), 6.23 (dt, 1H, J = 15.5, 1.5 Hz),

13 7.07 (1H, dt, J = 15.5, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 14.27, 22.73, 24.91,

26.08, 27.94, 29.20, 29.36, 29.38, 31.68, 32.11, 33.06, 34.25, 58.66, 59.33, 124.26,

- - 151.06, 179.81. 196.08; HRMS (FAB) calcd for C18H29O4 (M-H) 309.427, found

309.207.

Methyl (E)-8-(3-(oct-2-en-7-ynoyl)oxiran-2-yl)octanoate (36) (C19H28O4)

O O

O O

Following the general procedure for Wittig reaction between 23 and 30 and

purification by column chromatography afforded 260 mg (81 %) of product as a yellow oil. Identity and purity of the product were confirmed by 1H, 13C NMR and HRMS. 1H

NMR (500 MHz, CDCl3) δ 1.26-1.66 (m, 12H), 1.70 (quintet, 2H, J = 7.0 Hz), 1.98 (t,

1H, J = 2.5 Hz), 2.22 (td, 2H, J = 7.0, 2.5 Hz), 2.30 (t, 2H, J = 7.0 Hz), 2.36 (t, 2H, J =

7.0 Hz), 3.04 (td, 1H, J = 5.5, 2.0 Hz), 3.33 (d, 1H, J = 2 Hz), 3.66 (s, 3H), 6.27 (d, 1H, J

13 = 16 Hz), 7.06 (1H, dt, J = 16.0, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 18.27, 25.20,

26.09, 29.30 (2C), 29.39, 29.41, 31.81, 32.12, 34.38, 51.82, 58.61, 59.42, 69.49, 83.716,

+ + 124.76, 149.27, 174.59, 195.93; HRMS (ESI) m/z calcd for C19H28O4Na (M+Na)

343.18798, found 343.18812.

(E)-8-(3-(oct-2-en-7-ynoyl)oxiran-2-yl)octanoic acid (40) (C18H26O4)

O O

OH O

82

Following the general procedure for PPL hydrolysis of methyl (E)-8-(3-(oct-2-en-

7-ynoyl)oxiran-2-yl)octanoate 36 afforded 139 mg (91 %) of product as yellow oil.

Identity and purity of the product were confirmed by 1H, 13C NMR and HRMS. 1H NMR

(500 MHz, CDCl3) δ 1.22-1.88 (m, 12H), 1.70 (quintet, 2H, J = 7.0 Hz), 1.98 (t, 1H, J =

2.5 Hz), 2.17-2.29 (m, 4H), 2.30-240 (m, 2H), 3.05 (t, 1H, J = 5.0 Hz), 3.33 (bs, 1H),

13 6.27 (d, 1H, J = 15.5 Hz), 7.06 (1H, dt, J = 15.5, 7.0 Hz); C NMR (125 MHz, CDCl3) δ

18.27, 24.92, 26.08, 26.96, 29.20, 29.36, 29.38, 31.82, 32.09, 34.15, 58.62, 59.41, 69.50,

- - 83.72, 124.75, 149.33, 179.28, 195.97; HRMS (ESI) m/z calcd for C18H25O4 (M-H)

305.395, found 305.176.

Methyl (E)-8-(3-(oct-2-enoyl-4,4,5,5,6,6,7,7,8,8,8-d11)oxiran-2-yl)octanoate

(37) (C19H21D11O4)

D D D D O O D C O 3 O D D D D

Following the general procedure for Wittig reaction between 23 and 33 and

purification by column chromatography afforded 245 mg (73 %) of product as a yellow oil. Identity and purity of the product were confirmed by 1H and 13C NMR and HMRS.

1 H NMR (500 MHz, CDCl3) δ 1.27-1.71 (m, 12H), 2.30 (t, 2H, J = 7.5 Hz), 3.04 (t, 1H, J

= 5.0 Hz), 3.33 (bs, 1H), 3.66 (s, 3H), 6.23 (d, 1H, J = 16.0 Hz), 7.07 (d, 1H, J = 16.0

13 Hz); C NMR (125 MHz, CDCl3) δ 25.20, 26.09, 29.30, 29.39, 29.41, 32.13, 34.37,

51.81, 58.64, 59.34, 124.34, 150.95, 174.59, 196.02; HRMS (ESI) m/z calcd for

+ + C19H25D11NO4 (M+NH4) 353.567, found 353.333.

83

(E)-8-(3-(oct-2-enoyl-4,4,5,5,6,6,7,7,8,8,8-d11)oxiran-2-yl)octanoic acid (41)

(C18H19D11O4)

D D D D O O D C OH 3 O D D D D

Following the general procedure for PPL hydrolysis of methyl (E)-8-(3-(oct-2-

enoyl-4,4,5,5,6,6,7,7,8,8,8-d11)oxiran-2-yl)octanoate 37 afforded 150 mg (93 %) of product as yellow oil. Identity and purity of the product were confirmed by 1H, 13C NMR

1 and HMRS. H NMR (500 MHz, CDCl3) δ 1.29-1.71 (m, 12H), 2.28 (t, 2H, J = 7.5 Hz),

3.01 (m, 1H), 3.52 (bs, 1H), 6.28 (d, 1H, J = 16 Hz), 7.10 (d, 1H, J = 16.0 Hz); 13C NMR

(125 MHz, CDCl3) δ 26.82, 26.03, 30.10, 30.23, 30.24, 32.82, 34.91, 59.43, 60.01,

- - 126.55, 152.05, 177.68, 197.45; HRMS (ESI) m/z calcd for C18H18D11O4 (M-H)

320.493, found 320.276.

Methyl (E)-11-oxo-11-(3-pentyloxiran-2-yl)undec-9-enoate (27) (C19H32O4)

O O

O O

Following the general procedure for Wittig reaction between 26 and 24 and

purification by column chromatography afforded 234 mg (72 %) of product as a yellow

oil. Identity and purity of the product were confirmed by 1H, 13C NMR and HMRS.

16 1 Spectroscopic data were found to match lit. data. H NMR (500 MHz, CDCl3) δ 0.87 (t,

3H, J = 7.2 Hz), 1.19-1.73 (m, 18H), 2.21 (dd, 2H, J = 7.0, 2 Hz), 2.30 (t, 2H, J = 7.5

Hz), 3.05 (td, 1H, J = 5.5, 2.0 Hz), 3.34 (d, 1H, J = 2.0 Hz), 3.67 (s, 3H), 6.23 (d, 1H, J =

13 15.5 Hz), 7.07 (1H, dt, J = 15.5, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 14.27, 22.83,

84

25.19, 25.83, 28.19, 29.22, 29.31 (2C), 31.76, 32.13, 33.04, 34.37, 51.83, 58.75, 59.38,

+ + 124.32, 150.82, 174.64, 196.11; HRMS (ESI) m/z calcd for C19H32O4Na (M+Na)

347.21928, found 347.21942.

(E)-11-oxo-11-(3-pentyloxiran-2-yl)undec-9-enoic acid(trans-EKODE-(E)-

IIb) (C18H28O4)

O O

OH O

Following the general procedure for PPL hydrolysis of methyl (E)-11-oxo-11-(3- pentyloxiran-2-yl)undec-9-enoate 27 afforded 143 mg (92 %) of product as yellow oil.

Identity and purity of the product were confirmed by 1H, 13C NMR and HRMS.

16 1 Spectroscopic data were found to match lit. data. H NMR (400 MHz, CDCl3) δ 0.88 (t,

3H, J = 7.2 Hz), 1.17-1.67 (m, 18H), 2.21 (dq, 2H, J = 6.8, 1.6 Hz), 2.33 (t, 2H, J = 7.6

Hz), 3.05 (ddd, 1H, J = 6.0, 5.0, 2.0 Hz), 3.34 (d, 1H, J = 2.0 Hz), 6.25 (dt, 1H, J = 15.5,

13 1.5 Hz), 7.06 (1H, dt, J = 15.5, 7.0 Hz); C NMR (100 MHz, CDCl3) δ 14.28, 22.84,

24.93, 25.83, 28.20, 29.22, 29.30 (2C), 31.77, 32.14, 33.04, 34.20, 58.74, 59.40, 124.32,

- - 150.76, 179.44, 196.09, HRMS (FAB) calcd for C18H29O4 (M-H) 309.427, found

309.207.

85

Methyl (E)-11-oxo-11-(3-(pent-4-yn-1-yl)oxiran-2-yl)undec-9-enoate (38) (

C19 H28O4)

O O

O O

Following the general procedure for Wittig reaction between 34 and 2 and

purification by column chromatography afforded 208 mg (65 %) of product as a yellow oil. Identity and purity of the product were confirmed by 1H and 13C NMR and HRMS.

1 H NMR (500 MHz, CDCl3) δ 1.24-1.90 (m, 16H), 1.97 (t, 1H, J = 2.5 Hz), 2.18-2.25 (m,

2H), 2.30 (t, 2H, J = 7.5 Hz), 3.07 (dt, 1H, J = 5.0, 1.5 Hz), 3.38 (d, 1H, J = 1.5 Hz), 3.66

(s, 3H), 6.23 (d, 1H, J = 16 Hz), 7.08 (1H, dt, J = 16.0, 7.0 Hz); 13C NMR (125 MHz,

CDCl3) δ 18.39, 24.99, 25.20, 28.19, 29.22, 29.31, 30.97, 33.06, 34.36, 51.80, 58.08,

59.06, 69.47, 83.72, 124.41, 151.03, 174.53, 195.76; HRMS (ESI) m/z calcd for

+ + C19H28O4Na (M+Na) 343.18798, found 343.18812.

(E)-11-oxo-11-(3-(pent-4-yn-1-yl)oxiran-2-yl)undec-9-enoic acid (42) ( C18

H26O4)

O O

OH O

Following the general procedure for PPL hydrolysis of methyl (E)-11-oxo-11-(3-

(pent-4-yn-1-yl)oxiran-2-yl)undec-9-enoate 38 afforded 144 mg (94 %) of product as a yellow oil. Identity and purity of the product were confirmed by 1H, 13C NMR and

1 HRMS. H NMR (500 MHz, CDCl3) δ 1.22-1.88 (m, 12H), 1.97 (t, 1H, J = 2.5 Hz), 2.17-

2.32 (m, 4H,), 2.35 (t, 2H, J = 7.5 Hz), 3.08 (bs, 1H), 3.29(d, 1H, J = 5 Hz), 6.26 (d, 1H,

86

13 J = 15.5 Hz), 7.08 (1H, dt, J = 15.5, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 18.39,

24.93, 24.99, 28.18, 29.21, 29.28, 29.35, 30.97, 33.06, 34.09, 58.11, 59.11, 69.49, 83.74,

- - 124.43, 151.00, 178.90, 195.76; HRMS (ESI) m/z calcd for C18H25O4 (M-H) 305.395,

found 305.176.

Methyl (E)-11-oxo-11-(3-(pentyl-d11)oxiran-2-yl)undec-9-enoate (39) (C19

H21D11O4)

D D D D O O D C O 3 O D D D D

Following the general procedure for Wittig reaction between 35 and 2 and

purification by column chromatography afforded 146 mg (62 %) of product as yellow oil.

Identity and purity of the product were confirmed by 1H and 13C NMR and HRMS. 1H

NMR (500 MHz, CDCl3) δ 1.23-1.67 (m, 10H), 2.20 (q, 2H, J = 7.0 Hz), 2.27 (t, 2H, J =

7.0 Hz), 3.01 (s, 1H), 3.31 (s, 1H), 3.64 (s, 3H), 6.21 (d, 1H, J = 15.5 Hz), 7.05 (dt, 1H, J

13 = 15.5, 7.0 Hz); C NMR (125 MHz, CDCl3) δ 25.15, 28.15, 29.26, 29.27 (2C), 32.89,

34.31, 51.75, 58.58, 59.30, 124.28, 150.66, 174.51, 196.00 ,HRMS (ESI) m/z calcd for

+ + C19H25D11NO4 (M+NH4) 353.567, found 353.333.

(E)-11-oxo-11-(3-(pentyl-d11)oxiran-2-yl)undec-9-enoic acid (43)

D D D D O O D C OH 3 O D D D D Following the general procedure for PPL hydrolysis of methyl (E)-11-oxo-11-(3-

(pentyl-d11)oxiran-2-yl)undec-9-enoate 39 afforded 144 mg (94 %) of product as a

87

yellow oil. Identity and purity of the product were confirmed by 1H and 13C NMR and

1 HRMS. H NMR (500 MHz, CDCl3) δ 1.20-1.71 (m, 10H), 2.22 (q, 2H, J = 6.5 Hz), 2.35

(t, 2H, J = 6.5 Hz), 3.04 (s, 1H), 3.34 (s, 1H), 6.23 (d, 1H, J = 15.5 Hz), 7.07 (dt, 1H, J =

13 15.5, 6.0 Hz); C NMR (125 MHz, CDCl3) δ 24.95, 28.20, 29.23, 29.30, 29.36, 33.04,

34.17, 58.67, 59.36, 124.40, 150.79, 179.08, 196.16 HRMS (ESI) m/z calcd for

- - C18H18D11O4 (M-H) 320.493, found 320.276.

88

2. 6 References

(1) Samuelsson, B. The Journal of biological chemistry 2012, 287, 10070.

(2) Abrahamsson, S.; Bergstrom, S.; Samuelsson, B. P Chem Soc London 1962, 332.

(3) Austin, S. C.; Funk, C. D. Prostag Oth Lipid M 1999, 58, 231.

(4) Roberts, L. J.; Salomon, R. G.; Morrow, J. D.; Brame, C. J. Faseb J 1999, 13, 1157.

(5) Jacobs, A. T.; Marnett, L. J. Accounts Chem Res 2010, 43, 673.

(6) West, J. D.; Marnett, L. J. Chem Res Toxicol 2005, 18, 1642.

(7) Vila, A.; Tallman, K. A.; Jacobs, A. T.; Liebler, D. C.; Porter, N. A.; Marnett, L. J.

Chem Res Toxicol 2008, 21, 432.

(8) Gatbonton-Schwager, T. N.; Sadhukhan, S.; Zhang, G. F.; Letterio, J. J.; Tochtrop,

G. P. Redox Biol 2014, 2, 755.

(9) Yin, H.; Xu, L.; Porter, N. A. Chemical reviews 2011, 111, 5944.

(10) Zhu, X.; Tang, X.; Anderson, V. E.; Sayre, L. M. Chem Res Toxicol 2009, 22, 1386.

(11) Lundstrom, S. L.; Levanen, B.; Nording, M.; Klepczynska-Nystrom, A.; Skold, M.;

Haeggstrom, J. Z.; Grunewald, J.; Svartengren, M.; Hammock, B. D.; Larsson, B. M.; Eklund, A.;

Wheelock, A. M.; Wheelock, C. E. PloS one 2011, 6, e23864.

(12) Bruder, E. D.; Ball, D. L.; Goodfriend, T. L.; Raff, H. American journal of physiology. Regulatory, integrative and comparative physiology 2003, 284, R1631.

(13) Goodfriend, T. L.; Ball, D. L.; Egan, B. M.; Campbell, W. B.; Nithipatikom, K.

Hypertension 2004, 43, 358.

(14) Payet, M. D.; Goodfriend, T. L.; Bilodeau, L.; Mackendale, C.; Chouinard, L.;

Gallo-Payet, N. American journal of physiology. Endocrinology and metabolism 2006, 291,

E1160.

89

(15) Wang, R.; Kern, J. T.; Goodfriend, T. L.; Ball, D. L.; Luesch, H. Prostaglandins,

leukotrienes, and essential fatty acids 2009, 81, 53.

(16) Lin, D.; Zhang, J.; Sayre, L. M. The Journal of organic chemistry 2007, 72, 9471.

(17) Curran, D. P. J Am Chem Soc 1983, 105, 5826.

(18) Kim, K. S.; Szarek, W. A. Can J Chem 1981, 59, 878.

(19) Minami, T.; Harui, N.; Taniguchi, Y. Journal of Organic Chemistry 1986, 51, 3572.

(20) Hercouet, A.; Lecorre, M. Tetrahedron 1977, 33, 33.

(21) Cichowicz, N. R.; Nagorny, P. Organic letters 2012, 14, 1058.

(22) Appel, R.; Mayr, H. Chemistry 2010, 16, 8610.

(23) Magdesieva, N. N.; Chovnikova, N. G.; Brunovlenskaya, I. I. Zh Org Khim+ 1984,

20, 2097.

(24) Magdesieva, N. N.; Kyandzhetsian, R. A.; Chovnikova, N. G.; Emelyanova, N. N.

Zh Org Khim+ 1981, 17, 340.

(25) Acharya, H. P.; Kobayashi, Y. Angewandte Chemie 2005, 44, 3481.

(26) Sun, M. J.; Deng, Y. J.; Batyreva, E.; Sha, W.; Salomon, R. G. Journal of Organic

Chemistry 2002, 67, 3575.

(27) Bally, I.; Gard, E.; Ciornei, E.; Biltz, M.; Balaban, A. T. J Labelled Compd 1975, 11,

63.

(28) Kusukawa, T.; Tanaka, S.; Inoue, K. Tetrahedron 2014, 70, 4049.

(29) Huisgen, R.; Rietz, U. Tetrahedron 1958, 2, 271.

(30) Marino, J. P.; Nguyen, H. N. The Journal of organic chemistry 2002, 67, 6291.

(31) Gansauer, A.; Fan, C. A.; Keller, F.; Keil, J. J Am Chem Soc 2007, 129, 3484.

(32) Li, X.; Borhan, B. J Am Chem Soc 2008, 130, 16126.

(33) Cox, R. A.; Warkenti.J Can J Chemistry 1972, 50, 3242.

90

(34) Fox, R. J.; Lalic, G.; Bergman, R. G. J Am Chem Soc 2007, 129, 14144.

(35) Bedard, A. C.; Collins, S. K. J Am Chem Soc 2011, 133, 19976.

(36) Moodie, L. W. K.; Larsen, D. S. Eur J Org Chem 2014, 2014, 1684.

91

Chapter 3. Biological evaluation of EKODEs as potential activators of the

PPAR family of nuclear receptors

3.1 Peroxisome proliferator-activated receptors (PPARs) structure and

activity

Increasing dietary fat and sugar consumption in the Western diet along with the

sedentary lifestyle has expanded a number of pathological states such as , obesity and cardiovascular diseases (CVD).1 In fact, high caloric intake plays an important role in

limiting human lifespan through modulation of , insulin sensitivity,

glucose homeostasis, etc.2 It is well-known that most species of higher multicellular

organisms such as humans have developed homeostatic mechanisms to be able to control

fatty acids intake and metabolism. A family of nuclear receptors, the peroxisome

proliferator-activated receptors (PPARs), makes a prominent contribution to regulate

lipid metabolism, catabolism and storage. In humans, the PPARs are a family of three receptors PPARα (NR1C1) (nuclear receptor subfamily 1, group C, member 1), PPARβ/δ

(NR1C2) and PPARγ (NR1C3) which are products of three different genes.3

Comparing to other types of nuclear receptors, PPARs share a domain structural feature. The most conserved regions are C and E in the protein sequence. The sequence homology in C shows the DNA binding domain (DBD) is highly conserved. However, in

region E, which represents the ligand binding-domains (LBD), moderate level of

conservation has been observed between subtypes. Interestingly, the differences in this

region have been contributed to distinct pharmacological effects of each subtype of

PPAR. The N-terminal A/B domain holds the AF1 ligand-independent activation domain,

and an AF2 ligand-dependent activation domain is situated in the F region. The numbers

92

shown in the LBD and DBD are the percentage amino-acid identity of human PPARβ/δ and PPARγ1 compared to human PPARα.4 (Figure 3.1)

A/B C D E F

Human PPARα N AF1 DBD LBD AF2 C

Human PPARβ/δ N 86 70 C

Human PPARγ N 83 68 C

Figure 3.1 PPARs motifs, conserved region in yellow, variable regions in red

The PPARs form a heterodimer with the retinoid X receptorα (RXRα) (NR2B)

and binds to peroxisome proliferator response elements (PPRE). These elements are

composed of two hexameric nucleotide recognition motifs of AGGTCA spaced by one

nucleotide. They reside on the promoter region of PPAR target genes. In addition, the

PPRE element consists of an additional AAACT motif at the upstream region. The

variable region D, or hinge region of PPAR, provides a broad interaction space with the

upstream AAACT element.5

In the absence of the ligand, the PPAR-RXR heterodimer remains bound to the

nuclear receptor corepressor (NCoR) and the silencing mediator of retinoid and thyroid

hormone receptor (SMRT), which are mostly present in the corepressor complex. SMRT

functions as a platform protein and facilitates the recruitment of histone deacetylases

(HDACs). In fact, the corepressor protein complex reduces target gene transcription by

causing the deacetylation of histones. Unlike the PPARβ/δ isoform, the PPARα and

PPARγ isoforms are not capable of binding to DNA while they are associated with the co-repressor complex. Similar to other types of nuclear receptors, the activity of these receptors can be enhanced by ligands; this process called “transactivation.”

93

Upon the binding of ligands to PPARs, they detach from the co-repressor complex. Next, the PPARs recruit specific co-activator complexes, such as steroid receptor co-activator 1 (SRC1) and cAMP response element binding (CREB)-binding protein (CBP)/p300. These co-activator complexes, through their histone- acetyltransferase activity, are capable of reorganizing the chromatin packaging and driving the transcriptional machinery to the promoter region. (Figure 3.2)

CBP/p300 CBP/p300 SRC1 SRC1

PPAR RXR 9cRA PPAR RXR 9cRA PPAR RXR 9cRA

NCoR/SMRT

Ligand PPRE PPRE PPRE

Figure3.2 PPAR binding to ligand along with RXR binding to 9-cis retinoic acid (9cRA) forms a complex which provides an on- off-switch for gene expression

The expression of PPARs is tissue-specific and regulated at different levels, including post-translational modification (PTM). In addition, their gene expression activity depends on the response elements’ position to which they bind. Furthermore, the availability of active chromatin can determine the level of target gene transcription.

Various types of fatty acids and related oxidized forms (eicosanoids) can bind to PPARs with variable degrees of specifity between isoforms.6

94

1 2 3 PPAR RXR 9cRA Co-activators Co-activators

PPAR RXR 9cRA PPAR RXR 9cRA

transcription factor

MAPK

Co-activators

transcription transcription transcription factor factor factor

Figure 3.3 Three main mechanisms proposed for negative regulation of transcription factors by

PPARs

Regulation of gene transcription with PPARs extends far beyond their effect on

transactivation of specific genes in an agonist-dependent manner. PPARs like other types

of nuclear receptors are also known to interact with various expression machineries and

act through transrepression. In fact, PPARs are capable of suppressing several important

signaling cascades. There are three main mechanisms: (1). PPARs compete on the common coactivators binding and consuming their limited supply for other genes, (2). they can bind to the transcription factors (for example, AP1, NF-κB, NFAT or STATs)

and make them unavailable for certain response elements, (3). they can inhibit mitogen-

activated protein kinase (MAPK) phosphorylation ability and inhibit transcription factor activity. For instance, PPARs can inhibit nuclear factor-κB (NFκB) and AP1 in

macrophages and deteriorate the pro-inflammatory signaling by decreasing the

expression of pro-inflammatory cytokines, chemokines and cell adhesion molecules.

(Figure 3.3)

95

3.2. The physiological functions of the PPARs

3.2.1 Functional role of PPARα

PPARα was the first member of the family discovered. Since it has a diverse

tissue distribution, it exhibits different physiological activities among organisms. In

humans, it mainly exists in metabolically active organs such as the , heart and

. PPARα shows a central role in fatty acid catabolism and ketone body synthesis in the liver. Its expression is controlled by other transcription factors and nuclear receptors such as hepatocyte nuclear factor 4 (HNF4) and chicken ovalbumin upstream promoter- transcription factor II (COUP-TFII).

3.2.1.1 Exogenous ligands (synthetic Xenobiotics) of PPARα

Synthetic ligands such as (, , GW7647) and Wy-

14,643 (pirinixic acid) are being used in the treatment of dyslipidemias. In addition, industrial chemicals and environmental pollutants exert their toxic effect through this receptor. For example, the , such as di-(2-ethylhexyl)-phthalate (DEHP) and its related metabolite monoethylhexyl phthalate (MEHP), and di-(2-ethylhexyl) adipate

(DEHA) are activators of PPARα.7-9 Furthermore, certain perfluorinated compounds such

as perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) also function

as PPARα ligands.10,11 (Figure 3.4)

96

O O S O O OH O OH N N O H O Cl Cl O Clofibrate Fenofibrate GW7647

O H O O N N S OH O O N OH Cl O O O Wy-14,643 DEHA MEHP O O F F F F F F F F F F F F F F O HO CCl3 HO CF S CF3 3 HO F F F F F F Trichloroacetic acid F F F F F F O PFOA PFOS

Figure 3.4 Synthetic molecules and xenobiotics acting as PPAR-α ligands

3.2.1.2 Endogenous ligands (biological molecules) of PPARα

The Gustafsson laboratory reported for the first time that endogenous fatty acids

can activate PPARα.12Interestingly, PPARα is the only subtype in the family which binds

to a variety of fatty acids with a high micromolar range of affinity.13 In human , the

concentration of free fatty acids is between 0.02-20 μM.14 Therefore, it is not well-

understood whether free fatty acid can be endogenous ligands for PPARα receptors. This

idea encouraged scientists to search for other molecules with interesting phenotypic

effects related to PPARs. In fact, they have discovered a number of oxidized metabolites,

such as eicosanoids, function as endogenous ligands in a tissue-specific manner. For

example, 8(S)-Hydroxy-(5Z,9E,11Z,14Z)-eicosatetraenoic acid(8-HETE) specifically activates PPARα.15 In addition, in a separate study on oxidized lipids, 9-hydroxy-

10E,12Z-octadecadienoic acid(9-HODE) and 13-hydroxy-9Z,11E-octadecadienoic 97

acid(13-HODE) derived from oxidized low-density lipoproteins (oxLDL) have shown

activation of PPARα.16 Another group interested in fatty acid catabolism working on hepatocytes has shown that when the fatty acid synthase (FAS) enzyme actively presents, a phospholipid, 1-palmitoyl-2-oleoyl-sn-glycerol-3-phosphocholine (16:0/18:1-GPC) acts as an endogenous ligand for PPARα.17 Recently, it has been demonstrated that in kupffer

cells, stimulation of the enzyme 5-lipoxygenase leads to activation of PPARα through production of intracellular 5S,12R-dihydroxy-6Z,8E,10E,14Z-eicosatetraenoic

18 acid(). (Figure 3.5)

OH COOH COOH

HO

8(S)-HETE 9-HODE

OH OH COOH COOH

OH

Leukotriene B4 13-HODE

O O P N O O O O O H

O

16:0/18:1-GPC

Figure 3.5 Endogenous (biological molecules) acting as PPAR-α ligands

3.2.2 Functional role of PPARγ

PPARγ has been highly studied among different species so far. It mostly plays a

key role in the differentiation of adipocyte tissues. It also modulates the expression of

genes involved in energy storage and utilization. The agonist of this receptor has been 98

demonstrated to improve the sensitivity of target tissues to insulin and to reduce plasma

glucose, lipid and insulin levels in mellitus.

3.2.2.1 Exogenous ligands of PPARγ

The major synthetic molecules for PPARγ belong to a family known as (TZDs) or “glitazones” used as antidiabetic agents. Their mechanism of action was not revealed until the mid-1990s, when their link to the differentiation of adipocyte tissues through PPARγ was proposed.19 Later, Spiegelman was able to show

that upon the binding of the ligand to PPARγ, the complex binds to the promoter region

of target genes.20 Two of the members of this family ( and ) are

shown in Figure 3.6. Since their discovery, extensive work has been performed to

expand the scaffolds targeting one or a combination of PPARs. Because of this, a number

of other drugs acting from nonrelated targets have shown activation of PPARγ at the

micromolar range, including LTD4 receptor antagonist LY171883 (tomelukast).

Furthermore, several nonsteroidal anti-inflammatory drugs (NSAIDs) such as

indometacin have shown PPARγ activation.21 (Figure3.6)

99

S S O O N N NH O NH O O O O HO Rosiglitazone Troglitazone

O O OH O OH N N N O N N H O Cl

Tomelukast Indometacin

Figure 3.6 Synthetic molecules acting as PPARγ ligands

3.2.2.2 Endogenous ligands of PPARγ

Unlike the other subtypes, PPARγ has a strong preference for polyunsaturated acids. 13 PPARγ also binds to their oxidized metabolites from enzymatic or non- enzymatic reactions. For example, the lipoxygenase products of linoleic acid and arachidonic acid are 13(S)-HODE and 15(S)-HETE , respectively.22 In addition, the

12,14 prostaglandins 15-deoxy-Δ -prostaglandin J2 and 15-ketoprostaglandinE2 have shown activity at PPARγ.23,24 Furthermore, nitroalkene fatty acids 10-Nitrolinoleate and 9-

25,26 Nitrooleate have demonstrated PPARγ activity. (Figure 3.7)

100

COOH COOH

OH OH 13(S)-HODE 15(S)-HETE

O COOH COOH

O HO O ∆12,14 15-deoxy- J2 -Prostaglandin 15-keto Prostaglandin E2

NO2 COOH COOH

NO2 10-Nitrolinoleate 9-Nitrooleate

Figure 3.7 Endogenous molecules acting as PPARγ ligands

Phospholipids also exhibit affinity towards PPARγ. (Figure.3.8). Species of lysophosphatidic acid (LPA) such as 1-arachidonoyl-2-hydroxy-sn-glycero-3- phosphate(20:4 Lyso PA) have been proposed to act via PPARγ.27 However, LPAs

mediate their function mainly through G-protein coupled receptors on the cell surface. In addition, azelaoyl PAF derived from oxidized low-density lipoprotein (oxLDL) particles promote the differentiation of monocytes through the nuclear receptor PPARγ.28

(Figure.3.8)

101

O O P O O OH O HO H 20:4 Lyso PA

O P N O O O O O H

O Azelaoyl PAF

Figure 3.8 Phospholipids which act as endogenous molecules acting as PPARγ ligands

3.2.3. Functional role of PPARβ/δ

PPARβ/δ has been identified as a regulatory factor in the development of several chronic diseases, such as obesity, atherosclerosis and cancer. Similar to PPARγ, activating PPARβ/δ can increase insulin sensitivity and improve glucose uptake in diabetic models. It is known that PPARβ/δ activates the expression of fatty acid- catabolizing enzymes in skeletal muscles. Research from the Evans laboratory has shown overexpression of this protein in mice muscles enhances physical strength and endurance.

In fact, these mice which are called “marathon mice” are able to run twice as far as normal mice. Although research on the development of drugs that can empower muscles is still under investigation, athletes have been reported abusing PPARβ/δ agonists to improve their performance in competitions. Consequently, the World Anti-Doping

Agency (WADA) has issued an alert to consumers about the health hazard of the available agonists on the black market.

102

3.2.3.1 Exogenous ligands of PPARβ/δ

The initial efforts towards finding novel agonists for PPARβ/δ were mainly

focused on modification of natural fatty acids. For example L-631033 developed by

Merck had low micromolar binding affinity toward PPARβ/δ. Eventually, research

groups have found high-affinity ligands such as L-165041, GW501516 and GW 0742,

which have shown PPARβ/δ activation. (Figure.3.9)

COOH O COOH O

OH HO O O

O L-631033 L-165041

N N

F3C S F C S 3 S S F O COOH O COOH GW501516 GW0742

Figure 3.9 Synthetic molecules acting as PPARβ/δ ligands

3.2.3.2 Endogenous ligands of PPARβ/δ

It is known that PPARβ/δ exhibits a relatively high affinity for many saturated

13,29 and unsaturated fatty acids and eicosanoids. Eicosanoids PGA1, PGA2, PGD2, and

prostacyclin (PGI2) have been found to act as high affinity endogenous molecules towards the PPARβ/δ receptor.15,30,31 (Figure 3.10)

103

O O COOH COOH

OH OH PGA 1 PGA 2 COOH

OH O COOH

O HO OH OH PGD PGI 2 2

Figure 3.10 Endogenous molecules acting as PPARβ/δ ligands

3.3 EKODEs as potential endogenous metabolites

Although different research groups discovered a variety of fatty acids or related metabolites with high affinity towards PPARs, most of these findings were limited to a specific tissue. Therefore, there has been an interest towards finding endogenous metabolites which activate PPAR nuclear receptors in a global way.

So far, among different lipid metabolites that discussed in literature, the nitro form of fatty acid such as 10-Nitrolinoleate and 9-Nitrooleate can activate PPARγ subtype in human cells. In other words, the nitroalkene fatty acids which are products of nitrosative stress demonstrated a sub-micromolar range affinity to PPARs section 3.2.2.2.

These findings were quite important since they present how non-enzymatic pathways can regulate cell physiology in a concentration-dependent manner. Accordingly, our laboratory was sought the physiological role of lipid peroxidation products in cell

104

regulatory pathway feedback. In fact, the experiments designed here aimed to measure

the affinity of these molecules to the three PPAR nuclear receptors.

Among different molecules within the PUFA family that are known to be

modified during lipid peroxidation as a downstream of oxidative stress, we are focusing on molecules derived from linoleic acid called EKODEs. These molecules have shown interesting roles in regulating hormones, such as corticosterone and aldosterone production levels. They also demonstrate antioxidant effects through the Nrf2-Keap1 signaling pathway.32 In other words, the result from the Nrf2-Keap1 study was quite

remarkable because the products of peroxidation exhibit an anti-inflammatory effect by

triggering the detoxifying pathway of Nrf2. This idea raises the question whether the metabolites from this family can stimulate signals from other major known pathways

related to inflammation. In this regard, PPARs act as one of the major gene expression

machineries in the cell that are known to exhibit anti-inflammatory effects.33 As discussed

earlier, PPARs play a major role in regulating metabolism and catabolism in the body in

different tissues.

Along with other global techniques that are used to identify natural ligand for

PPAR, reporter assays still remain quite robust and reliable.34 In this study, a

heterologous expression setting of PPAR nuclear receptor in COS-7 cells was used. In

order to quantify the affinity of binding, a luciferase reporter system was employed. A

detail of fundamentals of the reporter assay will be presented in the following section.

105

3.4 Target reporter assay

3.4.1 Bioluminescence

Bioluminescence has become more prevalent among researchers due to its

sensitivity and accuracy. In fact, luciferase-luciferin reaction provides a convenient reporter system to measure activities such as gene expression, post-translational modification (PTM), protein–protein interaction in a cell-based assay set up. Although fluorescence imaging techniques with the aid of green fluorescent proteins (GFPs) and synthetic dyes is still among the most common methods to study biological systems, the new discoveries in bioluminescence as an ultrasensitive approach is opening new doors to the field of cell physiology.

3.4.2 Cell-based reporter assay

Cell-based assays utilizing reporter enzymes are widely used widely to investigate promoters, interaction between promoters and transcription factors, , and other cellular activities. In fact, they provide great advantages such as being rapid and relatively inexpensive making them ideal for drug screening to study drug targets and signaling pathways. Among the reporter genes, luciferase, an enzyme which catalyzes the bioluminescence reaction has been used more frequently. In fact, comparing to the other typical enzymes such as chloramphenicol acetyltransferase, β-glucuronidase, luciferase is highly sensitive and it gives a linear response over at least eight orders of magnitude. In addition, another advantage of this method is the ease of measuring light intensity with photomultiplier of a charge-coupled device (CCD) camera. Accordingly, luciferase is a suitable reporter system for measuring gene expression quantitavily.35

106

3.4.3 Single luciferase reporter assay

All types of bioluminescence luciferase systems follow the same principle of

chemical reaction: at the end the electronically exited state product will release light as a bioluminescence signal. The high emission intensity depends on the extraordinary quantum yield of the reaction (ΦBL). Indeed, the quantum yield in the case of the firefly

luciferase reaction is >0.40 which means that four photons can be obtained from ten

reacting molecules.36 In addition, the intensity of light is dependent on the amount of

reagents (ATP, Mg2+, and oxygen) and concentration of the enzyme. In order to measure

transactivation of PPAR nuclear receptors with their ligand, the response element (PPRE)

attached to a luciferase gene in the reporter vector along with another vector containing

the PPAR receptor gene are transfected into target cells.

Luciferase Living cell Ligand PPRE reporter gene

PPAR transfection

βGal gene

PPAR

transfection

PPAR gene

mRNA transfection

Luciferase protein

βGal protein

PPAR

Luciferin

cell extract

Figure 3.11 Illustration of the basic principle of the single luciferase reporter assay for PPAR receptor

107

Because luciferase expression is controlled by the fused genetic elements, reporter

expression is directly correlated with the activity of the PPRE. After transfection of the

plasmid into target cells, the PPAR vector provides the protein, which can interact with

the ligand and eventually move toward the PPRE. In other words, transactivation of

PPARs with the ligand leads to expression of luciferase and emitting the bioluminescence signal. The amount of signal is a good estimation of the activity of luciferase protein, which indicates the binding affinity of ligand to the PPAR nuclear receptor in the cells.

3.4.4 Luciferase enzyme mechanism

Luciferase acts as an enzyme and in the presence of cofactors (ATP, Mg2+ and

oxygen) it conducts an oxidative reaction on the substrate luciferin. At the beginning it

proceeds as an acyl-adenylate intermediate from a carboxylic acid substrate and ATP.

Next, luciferase as a monooxygenase adds molecular oxygen to luciferyl acyl-adenylate

complex creating a highly strained dioxentanone. At this stage the enzyme promotes

cleavage of CO2, forming oxyluciferin through [2+2] retero-cycloaddition, which is

forbidden by orbital symmetry law.37 Then the electronic excited state emits light while

returning to the ground state.38,39 As a result, this method can give a quantitative

measurement of ligand-PPAR interaction.40

108

O N N COOH MgATP PPi N N AMP O2 O S S luciferase O S S D-Luciferin D-Luciferyl Adenylate

O O O O AMP N N N N O O O S S O S S AMP CO2 dioxentanone * O N N N N O S O S S O S

excited oxyluciferin Light

Figure 3.12 Reaction catalyzed by firefly luciferase emits lights through bioluminescence process

3.4.5 Colorimetric β-galactosidase assay

To eliminate intrinsic experimental error, such as differences in the number and

viability of cells used, as well as the efficacy of the cell transfection, and lysis, etc. The

beta-galactosidase (βgal) gene is transfected to the cells in a separate vector at the same time. This enzyme is resistant to protease degradation and is stable in physiological condition. The enzyme which is the product of LacZ gene recognizes terminal nonreducing β-D-galactose residue in β -D-galactopyranosides. For example, hydrolysis of lactose provides β -D-galactose and glucose. Interestingly the enzyme is capable of hydrolysis of a broad range of substrates as long as they possess the β-D-galactose 109

residue. As a result, scientists took advantage of this property and designed substrates

that eventually broke into colored products. One of the widely used substrates is the ortho-Nitrophenyl-β-galactoside (ONPG) molecule which can be hydrolyzed to β -D-

galactose and ortho-nitrophenol(ONP). The latter absorbs light at 420 nm (seen as intense

yellow color) while the precursor molecule ONPG does not. Thus, by using ONPG as a

substrate, the activity of the enzyme can be quantified by measuring the amount of

absorption of ONP at 420 nm using a spectrophotometer or a microplate reader.

OH OH OH NO OH H O 2 NO2 H H O O β HO HO -gal H OH H OH HO + H H H OH H H

ONPG β-D-galactose ONP

chromophore

Figure3.13 Colorimetric assay of β-galactosidase using ONPG as the substrate

110

3.5 Experimental

3.5.1 Materials

Each compound was prepared in DMSO at 1000x prior the experiment.

Specifically, compounds and concentrations were as follows: 4-Chloro-6-(2,3-xylidino)-

2-pyrimidinylthioacetic acid (Wy14643; Sigma) prepared at 5mM concentration, 4-[2-(3-

Fluoro-4-trifluoromethyl-phenyl)-4-methyl-thiazol-5-ylmethylsulfanyl]- 2-methyl- phenoxy}-acetic acid(GW0742; Sigma) prepared at 5mM concentration, 5-[4-[(6-

Hydroxy-2,5,7,8-tetramethylchroman-2-yl)methoxy]benzyl]-2,4-thiazolidinedione,

(troglitazone )prepared at 5mM linoleic acid 4-hydroxynonenal(4-HNE), 4- oxononenal(4-ONE), 10-nitrolinoleate, 10(E),12(Z)-conjugated linoleic (Cayman

Chemical). EKODE molecules were synthesized, as described in Chapter 2, and purified by HPLC. EKODE amount were gravimetrically quantified using an AD-6 microbalance

(Perkin Elmer Corporation, Norwich, CT). All the lipids and oxidized metabolites stock solutions were prepared at 20.0mM. Cell Culture Dishes were 100mmx20mm, 12-well plate (Corning).

3.5.2 Cell transient transfection assay

COS-7 cells from American Type Culture Collection (ATCC) were grown to 60% confluence in Dubelco's Modified Eagle Medium (DMEM, Corning, Manassas, VA) supplemented with 10% FBS and 1% Antibiotic-Antimycotic (Life Technologies). Cells

were incubated at 37°C and 5% CO2 in a Forma™ Series II 3110 Water-Jacketed CO incubator (Thermo Scientific). Then COS-7 cells were transiently transfected with a plasmid containing the luciferase gene under the control of three tandem PPAR response elements (PPRE) (PPRE X3 -TK-luciferase) and mousePPARα, mousePPARγ, or

111 mousePPARβ/δ expression plasmids(Addgene, Cambridge, MA), respectively. pSV-β-

Galactosidase plasmid (Promega, Madison,WI) was co-transfected to monitor transfection efficiency. Plasmids were transiently transfected using PolyFect (Qiagen

301107). Cell lysates were prepared and luciferase and β-galactosidase activities were assayed using kits as described in methods section 3.5.2.5. Readings were taken with a

Flex Station 3 plate reader (Molecular devices) (Pharmacology department, Case Western

Reserve University).

3.5.3 Methods

3.5.3.1 Seeding of COS-7 cells

10 5 cells were seeded to each well in a 12-well plate in DMEM (supplemented with 10% FBS, 1% Antibiotic-Antimycotic streptomycin) as described in 3.6.1.

3.5.3.2 Transfection of COS-7 cells

After 24 hours of seeding and letting cells grow to 60% confluence, the growth media was replaced with a fresh media and transfection occurred. Cells were transfected with 200 ng PPRE X3 -TK-luciferase, 20 ng PPAR, PPAR, PPARγ, 100 ng internal reporter plasmid pSV-β-Galactosidaseand. A mix of these plasmid constructs and 4 μL

Polyfect reagent reached to the final volume of 40 μL with charcoal-treated DMEM.

Transfection mixture was incubated at room temperature for 10 minutes. Adding 40 μL of the media (DMEM, supplemented with 10% FBS, 1% Antibiotic-Antimycotic streptomycin) to the mixture brought the volume to 80 μL, which was dispensed into each well. Transfected cells were incubated for overnight prior to the treatment.

112

3.5.2.3 Treatment of transfected cells

The growth media was replaced with a charcoal-treated media for 4 hours.

Subsequently 1 μL of the ligands from 1000x stock solution in DMSO was added to each well. For the control experiment, activity of vehicle (DMSO) was tested by adding 1 μL

to each well. Ligand-treated cells were incubated for 24 hours prior to the readout.

3.5.2.4 Cell lysis

The media were removed from each well and cells washed once with 1 x PBS.

Then a 150 μL lysis buffer was added to each well. After scraping the cell remnants,

plates were kept at 4oC.

3.5.2.5 Luciferase assay, data normalization

In order to measure the luciferase activity in ligand- treated cells, the manufacturer protocol was followed. From each well 20 μL of cell lysate was placed in a

96-well opaque plate (white bottom) and treated with 100 μL of luciferase mixture.

(Caution: the addition needs to be quick to eliminate the error in signal collection). In order to normalize the data, internal reporter plasmid pSV-β-Galactosidase was used.

Following the instructions of the pSV-β-Galactosidase manufacturer, 30 μL of cell lysate from each well was placed in a 96-well plate, and a mixture containing ONPG added.

After one hour of incubation at 37 oC, the signal measured at 420 nm. Data were

collected as the relative light units (RLU) obtained from the luciferase assay divided by

the corresponding absorbance value obtained at 420 nm in the β-galactosidase assays

(Luc/β-galactosidase). Each compound was repeated in triplicate in a single experiment.

Therefore, values from the replicate were then averaged and expressed as mean normalized firefly luciferase activity.

113

3.6 Result and discussion

As we discussed earlier, transactivation of PPAR receptors with ligands, activates

luciferase expression and the signal obtained can be used as a quantitative measurement

for affinity of the ligand to PPAR. The result shown in Figure 3.14 is the luciferase read

out normalized by βgal which was used as internal reporter assay. In the case of

mPPARα, synthetic ligand Wy14635 was used at 5 μM. Among different lipids and

related metabolites of EKODEs, trans-EKODE-(E)-IIa and trans-EKODE-(E)-IIb showed a better activity, however this activation is not comparable to the synthetic ligands.

Figure3.14 Relative light units (RLU) determines activity of PUFAs and lipid metabolites on mouse PPARα by luciferase assay. 1X(5μM), 2X(10μM), 4X(20μM). Wy14635 is the synthetic ligand

114

Similarly the data for mPPARγ presented in Figure 3.15. Among different lipids and related metabolites of EKODE, trans-EKODE-(E)-IIa and trans-EKODE-(E)-IIb show a slightly better activity.

Figure3.15 Relative light units (RLU) determines activity of PUFAs and lipid metabolites on mouse PPARγ by luciferase assay. 1X(5μM), 2X(10μM), 4X(20μM). Troglitazone is the synthetic ligand

Interestingly, trans-EKODE-(E)-IIb showed a tremendous activation towards mPPARβ/δ which can be interpreted as specific interaction of this metabolite with mPPARβ/δ receptor. In this case activation by 10 μM of this metabolite is comparable with GW0742

at 5μM Figure 3.15.

115

Figure3.16 Relative light units (RLU) determines activity of PUFAs and lipid metabolites on mouse PPARβ/δ by luciferase assay. 1X(5μM), 2X(10μM), 4X(20μM). GW0742 is the synthetic ligand

In order to understand this exceptional activation, another experiment with lower

concentration of trans-EKODE-(E)-IIb was performed. Interestingly, this metabolite shows activity in a dose-response manner between 2.5-20 μM of concentration Figures

3.16 & 3.17.

116

Figure3.17 Relative light units (RLU) determines trans-EKODE-(E)-IIb activation of mouse

PPARβ/δ in compare to GW0742

3.7 Conclusion

Since the PPAR nuclear receptors has been intervened in different metabolic

networks and related diseases. Finding a high affinity endogenous ligand can shed light

on the PPAR role in the maintenance of cell physiology and therapeutics for variety of

diseases.41 Interestingly, nitrosative products of fatty acids have been known as

endogenous ligand for PPARγ, which emphasizes on the fact that downstream signaling

of reactive nitrogen species (RNS) can be important in the maintaining of cell

physiology. In the case of nitration products, they exhibit phenotypic effect as it has been

observed by PPARγ activation through synthetic ligands. Indeed, this observation along

with binding studies to the receptor suggested that the nitrated fatty acids function

through PPARγ.25

Similarly our result indicates that trans-EKODE-(E)-IIb activates PPARβ/δ in a dose dependent manner provides the evidence that this molecule can be nominated as 117

possible endogenous activator for PPARβ/δ. However, more detail studies need to be

performed in order to reveal their mode of interaction. The outcome of this finding will

definitely have a great impact on understanding of diseases which are associated with

PPARβ/δ such as chronic metabolic diseases and muscle dystrophy. In addition, at this

point designing specific molecules with the core structure of trans-EKODE-(E)-II can be found beneficial.

118

3.8 References

(1) Cordain, L.; Eaton, S. B.; Sebastian, A.; Mann, N.; Lindeberg, S.;

Watkins, B. A.; O'Keefe, J. H.; Brand-Miller, J. Am J Clin Nutr 2005, 81, 341.

(2) Wallace, D. C. Annu Rev Genet 2005, 39, 359.

(3) Michalik, L.; Desvergne, B.; Wahli, W. Nat Rev Cancer 2004, 4, 61.

(4) Berger, J.; Moller, D. E. Annual review of medicine 2002, 53, 409.

(5) Daynes, R. A.; Jones, D. C. Nat Rev Immunol 2002, 2, 748.

(6) Harmon, G. S.; Lam, M. T.; Glass, C. K. Chemical reviews 2011, 111,

6321.

(7) Peters, J. M.; Taubeneck, M. W.; Keen, C. L.; Gonzalez, F. J.

1997, 56, 311.

(8) Venkata, N. G.; Robinson, J. A.; Cabot, P. J.; Davis, B.; Monteith, G. R.;

Roberts-Thomson, S. J. Toxicol Lett 2006, 163, 224.

(9) Hamano, T.; Kobayashi, K.; Sakairi, T.; Hayashi, M.; Mutai, M. The

Journal of toxicological sciences 2001, 26, 275.

(10) Intrasuksri, U.; Rangwala, S. M.; O'Brien, M.; Noonan, D. J.; Feller, D. R.

Gen Pharmacol 1998, 31, 187.

(11) Shipley, J. M.; Hurst, C. H.; Tanaka, S. S.; DeRoos, F. L.; Butenhoff, J.

L.; Seacat, A. M.; Waxman, D. J. Toxicol Sci 2004, 80, 151.

(12) Gottlicher, M.; Widmark, E.; Li, Q.; Gustafsson, J. A. P Natl Acad Sci

USA 1992, 89, 4653.

119

(13) Xu, H. E.; Lambert, M. H.; Montana, V. G.; Parks, D. J.; Blanchard, S. G.;

Brown, P. J.; Sternbach, D. D.; Lehmann, J. M.; Wisely, G. B.; Willson, T. M.; Kliewer,

S. A.; Milburn, M. V. Mol Cell 1999, 3, 397.

(14) Jungling, E.; Kammermeier, H. Analytical biochemistry 1988, 171, 150.

(15) Yu, K.; Bayona, W.; Kallen, C. B.; Harding, H. P.; Ravera, C. P.;

Mcmahon, G.; Brown, M.; Lazar, M. A. Journal of Biological Chemistry 1995, 270,

23975.

(16) Delerive, P.; Furman, C.; Teissier, E.; Fruchart, J. C.; Duriez, P.; Staels, B.

Febs Lett 2000, 471, 34.

(17) Chakravarthy, M. V.; Lodhi, I. J.; Yin, L.; Malapaka, R. R. V.; Xu, H. E.;

Turk, J.; Semenkovich, C. F. Cell 2009, 138, 476.

(18) Narala, V. R.; Adapala, R. K.; Suresh, M. V.; Brock, T. G.; Peters-Golden,

M.; Reddy, R. C. Journal of Biological Chemistry 2010, 285, 22067.

(19) Lehmann, J. M.; Moore, L. B.; Smitholiver, T. A.; Wilkison, W. O.;

Willson, T. M.; Kliewer, S. A. Journal of Biological Chemistry 1995, 270, 12953.

(20) Tontonoz, P.; Hu, E.; Graves, R. A.; Budavari, A. I.; Spiegelman, B. M.

Gene Dev 1994, 8, 1224.

(21) Jaradat, M. S.; Wongsud, B.; Phornchirasilp, S.; Rangwala, S. M.; Shams,

G.; Sutton, M.; Romstedt, K. J.; Noonan, D. J.; Feller, D. R. Biochem Pharmacol 2001,

62, 1587.

(22) Nagy, L.; Tontonoz, P.; Alvarez, J. G. A.; Chen, H. W.; Evans, R. M. Cell

1998, 93, 229.

120

(23) Forman, B. M.; Tontonoz, P.; Chen, J.; Brun, R. P.; Spiegelman, B. M.;

Evans, R. M. Cell 1995, 83, 803.

(24) Hammarstrom, S.; Trinks, C.; Wigren, J.; Surapureddi, S.; Soderstrom,

M.; Glass, C. K. Adv Exp Med Biol 2002, 507, 343.

(25) Cui, T.; Schopfer, F. J.; Zhang, J.; Chen, K.; Ichikawa, T.; Baker, P. R.;

Batthyany, C.; Chacko, B. K.; Feng, X.; Patel, R. P.; Agarwal, A.; Freeman, B. A.; Chen,

Y. E. The Journal of biological chemistry 2006, 281, 35686.

(26) Baker, P. R. S.; Lin, Y. M.; Schopfer, F. J.; Woodcock, S. R.; Groeger, A.

L.; Batthyany, C.; Sweeney, S.; Long, M. H.; Iles, K. E.; Baker, L. M. S.; Branchaud, B.

P.; Chen, Y. Q. E.; Freeman, B. A. Journal of Biological Chemistry 2005, 280, 42464.

(27) Zhang, C. X.; Baker, D. L.; Yasuda, S.; Makarova, N.; Balazs, L.;

Johnson, L. R.; Marathe, G. K.; McIntyre, T. M.; Xu, Y.; Prestwich, G. D.; Byun, H. S.;

Bittman, R.; Tigyi, G. J Exp Med 2004, 199, 763.

(28) Davies, S. S.; Pontsler, A. V.; Marathe, G. K.; Harrison, K. A.; Murphy,

R. C.; Hinshaw, J. C.; Prestwich, G. D.; St Hilaire, A.; Prescott, S. M.; Zimmerman, G.

A.; McIntyre, T. M. Journal of Biological Chemistry 2001, 276, 16015.

(29) Forman, B. M.; Chen, J.; Evans, R. M. P Natl Acad Sci USA 1997, 94,

4312.

(30) Lim, H.; Gupta, R. A.; Ma, W. G.; Paria, B. C.; Moller, D. E.; Morrow, J.

D.; DuBois, R. N.; Trzaskos, J. M.; Dey, S. K. Gene Dev 1999, 13, 1561.

(31) Hatae, T.; Wada, M.; Yokoyama, C.; Shimonishi, M.; Tanabe, T. Journal of Biological Chemistry 2001, 276, 46260.

121

(32) Wang, R.; Kern, J. T.; Goodfriend, T. L.; Ball, D. L.; Luesch, H.

Prostaglandins, leukotrienes, and essential fatty acids 2009, 81, 53.

(33) Rizzo, G.; Fiorucci, S. Current opinion in pharmacology 2006, 6, 421.

(34) Montanez, J. E.; Peters, J. M.; Correll, J. B.; Gonzalez, F. J.; Patterson, A.

D. Toxicologic pathology 2013, 41, 410.

(35) Fan, F.; Wood, K. V. Assay and drug development technologies 2007, 5,

127.

(36) Ando, Y.; Niwa, K.; Yamada, N.; Enomot, T.; Irie, T.; Kubota, H.;

Ohmiya, Y.; Akiyama, H. Nat Photonics 2008, 2, 44.

(37) Van Vranken, D. L.; Weiss, G. A. Introduction to bioorganic chemistry and chemical biology; Garland Science: , 2013.

(38) Marques, S. M.; Esteves da Silva, J. C. Iubmb Life 2009, 61, 6.

(39) Almeida de Oliveira, M.; Bartoloni, F. H.; Augusto, F. A.; Ciscato, L. F.;

Bastos, E. L.; Baader, W. J. The Journal of organic chemistry 2012, 77, 10537.

(40) Denburg, J. L.; Mcelroy, W. D. Biochemistry-Us 1970, 9, 4619.

(41) Schupp, M.; Lazar, M. A. The Journal of biological chemistry 2010, 285,

40409.

122

Chapter 4 Thesis summary and future directions

4.1 Thesis summary

In Chapter 1, the discovery of oxidized lipids as endogenous metabolites

has been discussed. First, prostaglandins (PGs) and leukotrienes (LT) with a short

overview of their history were introduced. The role of stereocontrolled synthesis

in characterization and understanding of the biosynthetic pathways associated

with these metabolites was introduced. Next, the discovery of lipid peroxidation

products (LPO) derived from non-enzymatic reactions was discussed. The

synthesis of LPOs along with their physiological significance was then introduced. The interplay between oxidative stress and modification of lipids was deliberated. Next, the background of oxidative stress was introduced. Based on a

comprehensive literature review, the different types of ROS/RNS were discussed

and how their physicochemical properties can determine their reaction kinetics

and thermodynamics. The discussion continued with the chemical basis of how

lipid peroxidation products (LPO) can activate diverse biotargets.

In Chapter 2, one of the main products of linoleic acid peroxidation, or

EKODEs, was introduced. These molecules have been found in pathological

states with an oxidative stress background. Following, the postulate mechanism

was shown regarding how ROS/RNS initiates radical formation, and bonds

rearrange themselves to form a variety of scaffolds. The fact that these molecules

possess an α, β-unsaturated ketone structure makes them more interesting to

investigate to define their role in cell physiology. The detailed synthesis for

123

different combinations of this family of metabolites was also discussed. Our

challenges during the synthesis of EKODE-(E)-II encouraged us to implement a novel strategy through a divergent approach to synthesize these metabolites and related molecules. In other words, we have shown how this proposed synthetic strategy can help us to build different libraries of EKODE-like molecules. The fact that we were able to design a core reactive “bifunctional conjunctive ylide”

22 helped us to obtain these metabolites in high yields in a rapid timeframe.

O

Ph3P S(CH3)2 22

Bifunctional conjunctive ylide

RCHO RCHO

k2 k1

O O Ph3P R S(CH3)2 R O

Scheme.4.1 At low temperature (0oC), sulfonium ylide reacts faster than phosphonium

(k1>>k2)

As shown in Scheme 4.1 the bifunctional molecule takes advantage of the

difference in reactivity between two ylides attached to the main scaffold of

EKODE-(E)-II. The importance of this methodology is that the ylides exhibit

different nucleophilicity while reacting with aldehydes providing an ideal

opportunity for substrate discrimination based on inherent reactivity. In this

example, the sulfonium ylide reacts faster than phosphonium, providing an

advantage when performing sequential reactions with the aldehydes of interest.

124

O O 1eq R1CHO 1eq R2CHO Ph P 3 S(CH3)2 R R Epoxide formation Olefin formation 2 O 1 22

Bifunctional conjunctive ylide EKODE-(E)-II

Scheme.4.2 Sulfonium ylide reaction is faster than phosphonium one

The use of the bifunctional conjunctive ylide resulted in production of EKODE-

(E)-II and their alkyne-terminal bioorthogonal forms and 2H-labeled derivatives that can be used for further mechanistic studies. The strategy outlined here establishes a general approach that can be applied to a wide range of PUFA metabolites, to develop libraries of reactive lipid metabolites to study their possible biological functions.

In Chapter 3, the biological evaluation of PPAR nuclear receptors was performed. The previous kinetic studies on these metabolites, along with other end products of LPO, suggested that they are acting through formation of Michael adducts in a comparable rate.1 The result provided a stepping-stone toward our

ambition of revealing biological targets for these metabolites.

A luciferase reporter assay was utilized to measure the transactivation of

these lipid metabolites toward PPAR nuclear receptors. In a heterologous

expression setting of PPAR nuclear receptor in COS-7 cells, transactivation of the

PPAR nuclear receptor with the respective ligands was measured. Interesting

results were observed showing trans-EKODE-(E)-IIb activating PPARβ/δ nuclear receptors. These receptors are specifically involved in the development of different chronic diseases, such as obesity, atherosclerosis and cancer.The result from this assay indicated that this specific metabolite can regulate cell signaling

125

through PPARβ/δ. However, the results here raise a fundamental question

regarding whether these molecules can regulate chronic diseases, which obviously

needs further investigation. In addition, the detailed of the mode of interaction needs to be revealed via more thorough structural studies, such as X-ray crystallography.

4.2 Future directions

To facilitate a deeper understanding of the function of these metabolites, our future investigation is going to follow on two parallel directions: first, our laboratory is currently designing a set of global experiments such as RNA-seq or

microarray experiments to characterize functional targets in the cell. The outcome of this experiment will be critical in order to provide us with preliminary data.

Consequently, the specific targets will be confirmed through RT-PCR experiments, and quantification of RNA expression levels will be used to evaluate data obtained from global studies. The information will help us build models for proposing networks regulated by EKODEs. In addition, understanding how these specific metabolites are interacting with proteins on metabolic or signaling networks will provide us deeper insight toward revealing their mechanisms of action. By understanding how these metabolites are interacting with cellular lipase enzymes and intracellular carrier proteins such as fatty acid binding proteins (FABPs), research can determine how these interactions can affect signaling and the metabolism of other fatty acids. In this regard, the bioorthogonal surrogate and deuterated-labeled molecules, which were developed through our

126

novel synthetic approach, will likely be very beneficial. Structural studies such as

X-ray crystallography can be employed to investigate the interaction modes at the

level of molecular detail.

Our second focus will be on employing the synthetic strategy represented

in this thesis for developing a variety of related molecules from other PUFAs and

investigating their biological implications. In addition, these oxidized fatty acids

can be conjugated to the structure of phospholipids such as phosphatidylcholine

and lysophosphatidic acids, which have shown interesting results in activating G-

protein coupled receptors (GPCR) and nuclear receptors with other oxidized

lipids.2-4 Synthesis of phospholipids can be achieved with different commercially

available phosphorous containing heads through synthetic or chemoenzymatic

esterification as shown in Scheme 4.3.5-7

O OR OR esterification Oxidized fatty acids O Oxidized fatty acids COOH + HO Phsophorous Phsophorous head head

R: acylgroup or H Scheme 4.3 General strategy for phospholipid synthesis

Overall, the versatile synthetic approach which has been presented in this thesis

provides the synthesis of the EKODE family and similar analogues to facilitate

the biological evaluation for the future.

127

4.3 References

(1) Lin, D.; Zhang, J.; Sayre, L. M. J Org Chem 2007, 72, 9471.

(2) McIntyre, T. M.; Pontsler, A. V.; Silva, A. R.; St Hilaire, A.; Xu, Y.;

Hinshaw, J. C.; Zimmerman, G. A.; Hama, K.; Aoki, J.; Arai, H.; Prestwich, G. D. P Natl

Acad Sci USA 2003, 100, 131.

(3) Choi, J. W.; Herr, D. R.; Noguchi, K.; Yung, Y. C.; Lee, C. W.; Mutoh,

T.; Lin, M. E.; Teo, S. T.; Park, K. E.; Mosley, A. N.; Chun, J. Annual review of

Pharmacology and Toxicology 2010, 50, 157.

(4) Hammond, V. J.; Morgan, A. H.; Lauder, S.; Thomas, C. P.; Brown, S.;

Freeman, B. A.; Lloyd, C. M.; Davies, J.; Bush, A.; Levonen, A. L.; Kansanen, E.;

Villacorta, L.; Chen, Y. E.; Porter, N.; Garcia-Diaz, Y. M.; Schopfer, F. J.; O'Donnell, V.

B. J Biol Chem 2012, 287, 41651.

(5) Jung, M. E.; Berliner, J. A.; Angst, D.; Yue, D. W.; Koroniak, L.; Watson,

A. D.; Li, R. S. Org Lett 2005, 7, 3933.

(6) Xu, Y.; Prestwich, G. D. J Org Chem 2002, 67, 7158.

(7) Duclos, R. I. Chem Phys Lipids 2010, 163, 102.

128

APPENDIX NMR spectra of synthesized molecules

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

BIBLIOGRAPHY

182

(1) Acharya, H. P.; Kobayashi, Y. Angewandte Chemie 2005, 44, 3481.

(2) Ando, Y.; Niwa, K.; Yamada, N.; Enomot, T.; Irie, T.; Kubota, H.; Ohmiya,

Y.; Akiyama, H. Nat Photonics 2008, 2, 44.

(3) Appel, R.; Mayr, H. Chemistry 2010, 16, 8610.

(4) Armstrong David, A.; Huie Robert, E.; Lymar, S.; Koppenol Willem, H.;

Merényi, G.; Neta, P.; Stanbury David, M.; Steenken, S.; Wardman, P. In BioInorganic

Reaction Mechanisms 2013; Vol. 9, p 59.

(5) Austin, S. C.; Funk, C. D. Prostag Oth Lipid M 1999, 58, 231.

(6) Baker, P. R. S.; Lin, Y. M.; Schopfer, F. J.; Woodcock, S. R.; Groeger, A.

L.; Batthyany, C.; Sweeney, S.; Long, M. H.; Iles, K. E.; Baker, L. M. S.; Branchaud, B. P.;

Chen, Y. Q. E.; Freeman, B. A. Journal of Biological Chemistry 2005, 280, 42464.

(7) Bedard, A. C.; Collins, S. K. J Am Chem Soc 2011, 133, 19976.

(8) Benz, C. C.; Yau, C. Nat Rev Cancer 2008, 8, 875.

(9) Berger, J.; Moller, D. E. Annual review of medicine 2002, 53, 409.

(10) Blad, C. C.; Tang, C.; Offermanns, S. Nat Rev Drug Discov 2012, 11, 603.

(11) Blanc, E. M.; Kelly, J. F.; Mark, R. J.; Waeg, G.; Mattson, M. P. Journal of neurochemistry 1997, 69, 570.

(12) Bruder, E. D.; Ball, D. L.; Goodfriend, T. L.; Raff, H. American journal of physiology. Regulatory, integrative and comparative physiology 2003, 284, R1631.

(13) Chakravarthy, M. V.; Lodhi, I. J.; Yin, L.; Malapaka, R. R. V.; Xu, H. E.;

Turk, J.; Semenkovich, C. F. Cell 2009, 138, 476.

(14) Chen, J. J.; Schenker, S.; Frosto, T. A.; Henderson, G. I. Bba-Gen Subjects

1998, 1380, 336.

183

(15) Choi, J. W.; Herr, D. R.; Noguchi, K.; Yung, Y. C.; Lee, C. W.; Mutoh, T.;

Lin, M. E.; Teo, S. T.; Park, K. E.; Mosley, A. N.; Chun, J. Annu Rev Pharmacol Toxicol

2010, 50, 157.

(16) Cichowicz, N. R.; Nagorny, P. Organic letters 2012, 14, 1058.

(17) Cordain, L.; Eaton, S. B.; Sebastian, A.; Mann, N.; Lindeberg, S.; Watkins,

B. A.; O'Keefe, J. H.; Brand-Miller, J. Am J Clin Nutr 2005, 81, 341.

(18) Cox, R. A.; Warkenti.J Can J Chemistry 1972, 50, 3242.

(19) Cui, T.; Schopfer, F. J.; Zhang, J.; Chen, K.; Ichikawa, T.; Baker, P. R.;

Batthyany, C.; Chacko, B. K.; Feng, X.; Patel, R. P.; Agarwal, A.; Freeman, B. A.; Chen,

Y. E. The Journal of biological chemistry 2006, 281, 35686.

(20) D'Autreaux, B.; Toledano, M. B. Nat Rev Mol Cell Bio 2007, 8, 813.

(21) Davies, K. J. Iubmb Life 2000, 50, 279.

(22) Davies, K. J. A. Iubmb Life 2000, 50, 279.

(23) Davies, S. S.; Pontsler, A. V.; Marathe, G. K.; Harrison, K. A.; Murphy, R.

C.; Hinshaw, J. C.; Prestwich, G. D.; St Hilaire, A.; Prescott, S. M.; Zimmerman, G. A.;

McIntyre, T. M. Journal of Biological Chemistry 2001, 276, 16015.

(24) Daynes, R. A.; Jones, D. C. Nat Rev Immunol 2002, 2, 748.

(25) Delerive, P.; Furman, C.; Teissier, E.; Fruchart, J. C.; Duriez, P.; Staels, B.

Febs Lett 2000, 471, 34.

(26) Denburg, J. L.; Mcelroy, W. D. Biochemistry-Us 1970, 9, 4619.

(27) Denicola, A.; Freeman, B. A.; Trujillo, M.; Radi, R. Arch Biochem Biophys

1996, 333, 49.

(28) Devasagayam, T. P.; Kamat, J. P. Indian journal of experimental biology

184

2002, 40, 680.

(29) Duclos, R. I. Chem Phys Lipids 2010, 163, 102.

(30) Fan, F.; Wood, K. V. Assay and drug development technologies 2007, 5,

127.

(31) Flohe, L. Biochim Biophys Acta 2013, 1830, 3139.

(32) Forman, B. M.; Chen, J.; Evans, R. M. P Natl Acad Sci USA 1997, 94, 4312.

(33) Forman, B. M.; Tontonoz, P.; Chen, J.; Brun, R. P.; Spiegelman, B. M.;

Evans, R. M. Cell 1995, 83, 803.

(34) Fox, R. J.; Lalic, G.; Bergman, R. G. J Am Chem Soc 2007, 129, 14144.

(35) Gansauer, A.; Fan, C. A.; Keller, F.; Keil, J. J Am Chem Soc 2007, 129,

3484.

(36) Gatbonton-Schwager, T. N.; Sadhukhan, S.; Zhang, G. F.; Letterio, J. J.;

Tochtrop, G. P. Redox Biol 2014, 2, 755.

(37) Goodfriend, T. L.; Ball, D. L.; Egan, B. M.; Campbell, W. B.;

Nithipatikom, K. Hypertension 2004, 43, 358.

(38) Gottlicher, M.; Widmark, E.; Li, Q.; Gustafsson, J. A. P Natl Acad Sci USA

1992, 89, 4653.

(39) Hamano, T.; Kobayashi, K.; Sakairi, T.; Hayashi, M.; Mutai, M. The

Journal of toxicological sciences 2001, 26, 275.

(40) Hammarstrom, S.; Trinks, C.; Wigren, J.; Surapureddi, S.; Soderstrom, M.;

Glass, C. K. Adv Exp Med Biol 2002, 507, 343.

(41) Hammond, V. J.; Morgan, A. H.; Lauder, S.; Thomas, C. P.; Brown, S.;

Freeman, B. A.; Lloyd, C. M.; Davies, J.; Bush, A.; Levonen, A. L.; Kansanen, E.;

185

Villacorta, L.; Chen, Y. E.; Porter, N.; Garcia-Diaz, Y. M.; Schopfer, F. J.; O'Donnell, V.

B. The Journal of biological chemistry 2012, 287, 41651.

(42) Harmon, G. S.; Lam, M. T.; Glass, C. K. Chemical reviews 2011, 111,

6321.

(43) Hatae, T.; Wada, M.; Yokoyama, C.; Shimonishi, M.; Tanabe, T. Journal of

Biological Chemistry 2001, 276, 46260.

(44) Henry, K. M.; Townsend, C. A. J Am Chem Soc 2005, 127, 3300.

(45) Hercouet, A.; Lecorre, M. Tetrahedron 1977, 33, 33.

(46) Higdon, A.; Diers, A. R.; Oh, J. Y.; Landar, A.; Darley-Usmar, V. M.

Biochemical Journal 2012, 442, 453.

(47) Hillard, E. A.; de Abreu, F. C.; Ferreira, D. C.; Jaouen, G.; Goulart, M. O.;

Amatore, C. Chemical communications 2008, 2612.

(48) Holmstrom, K. M.; Finkel, T. Nat Rev Mol Cell Bio 2014, 15, 411.

(49) Huisgen, R.; Rietz, U. Tetrahedron 1958, 2, 271.

(50) Ignarro, L. J. Annu Rev Pharmacol 1990, 30, 535.

(51) Ilan, Y. A.; Czapski, G.; Meisel, D. Biochim Biophys Acta 1976, 430, 209.

(52) Intrasuksri, U.; Rangwala, S. M.; O'Brien, M.; Noonan, D. J.; Feller, D. R.

Gen Pharmacol 1998, 31, 187.

(53) Itoh, T.; Fairall, L.; Amin, K.; Inaba, Y.; Szanto, A.; Balint, B. L.; Nagy, L.;

Yamamoto, K.; Schwabe, J. W. R. Nat Struct Mol Biol 2008, 15, 924.

(54) Jacobs, A. T.; Marnett, L. J. Accounts Chem Res 2010, 43, 673.

(55) Jaradat, M. S.; Wongsud, B.; Phornchirasilp, S.; Rangwala, S. M.; Shams,

G.; Sutton, M.; Romstedt, K. J.; Noonan, D. J.; Feller, D. R. Biochem Pharmacol 2001, 62,

186

1587.

(56) Jung, M. E.; Berliner, J. A.; Angst, D.; Yue, D. W.; Koroniak, L.; Watson,

A. D.; Li, R. S. Organic letters 2005, 7, 3933.

(57) Jungling, E.; Kammermeier, H. Analytical biochemistry 1988, 171, 150.

(58) Kim, K. S.; Szarek, W. A. Can J Chem 1981, 59, 878.

(59) Kusukawa, T.; Tanaka, S.; Inoue, K. Tetrahedron 2014, 70, 4049.

(60) Lehmann, J. M.; Moore, L. B.; Smitholiver, T. A.; Wilkison, W. O.;

Willson, T. M.; Kliewer, S. A. Journal of Biological Chemistry 1995, 270, 12953.

(61) Levonen, A. L.; Landar, A.; Ramachandran, A.; Ceaser, E. K.; Dickinson,

D. A.; Zanoni, G.; Morrow, J. D.; Darley-Usmar, V. M. The Biochemical journal 2004,

378, 373.

(62) Li, X.; Borhan, B. J Am Chem Soc 2008, 130, 16126.

(63) Lim, H.; Gupta, R. A.; Ma, W. G.; Paria, B. C.; Moller, D. E.; Morrow, J.

D.; DuBois, R. N.; Trzaskos, J. M.; Dey, S. K. Gene Dev 1999, 13, 1561.

(64) Lin, D.; Zhang, J.; Sayre, L. M. The Journal of organic chemistry 2007, 72,

9471.

(65) Lundstrom, S. L.; Levanen, B.; Nording, M.; Klepczynska-Nystrom, A.;

Skold, M.; Haeggstrom, J. Z.; Grunewald, J.; Svartengren, M.; Hammock, B. D.; Larsson,

B. M.; Eklund, A.; Wheelock, A. M.; Wheelock, C. E. PloS one 2011, 6, e23864.

(66) Magdesieva, N. N.; Chovnikova, N. G.; Brunovlenskaya, I. I. Zh Org

Khim+ 1984, 20, 2097.

(67) Magdesieva, N. N.; Kyandzhetsian, R. A.; Chovnikova, N. G.;

Emelyanova, N. N. Zh Org Khim+ 1981, 17, 340.

187

(68) Marino, J. P.; Nguyen, H. N. The Journal of organic chemistry 2002, 67,

6291.

(69) Marques, S. M.; Esteves da Silva, J. C. Iubmb Life 2009, 61, 6.

(70) Martinez, M. C.; Andriantsitohaina, R. Antioxid Redox Sign 2009, 11, 669.

(71) McIntyre, T. M.; Pontsler, A. V.; Silva, A. R.; St Hilaire, A.; Xu, Y.;

Hinshaw, J. C.; Zimmerman, G. A.; Hama, K.; Aoki, J.; Arai, H.; Prestwich, G. D. P Natl

Acad Sci USA 2003, 100, 131.

(72) Michalik, L.; Desvergne, B.; Wahli, W. Nat Rev Cancer 2004, 4, 61.

(73) Minami, T.; Harui, N.; Taniguchi, Y. Journal of Organic Chemistry 1986,

51, 3572.

(74) Monroy, C. A.; Doorn, J. A.; Roman, D. L. Chemical research in toxicology

2013, 26, 1832.

(75) Montanez, J. E.; Peters, J. M.; Correll, J. B.; Gonzalez, F. J.; Patterson, A.

D. Toxicologic pathology 2013, 41, 410.

(76) Moodie, L. W. K.; Larsen, D. S. Eur J Org Chem 2014, 2014, 1684.

(77) Morrow, J. D.; Hill, K. E.; Burk, R. F.; Nammour, T. M.; Badr, K. F.;

Roberts, L. J. P Natl Acad Sci USA 1990, 87, 9383.

(78) Nagy, L.; Tontonoz, P.; Alvarez, J. G. A.; Chen, H. W.; Evans, R. M. Cell

1998, 93, 229.

(79) Narala, V. R.; Adapala, R. K.; Suresh, M. V.; Brock, T. G.; Peters-Golden,

M.; Reddy, R. C. Journal of Biological Chemistry 2010, 285, 22067.

(80) Obinata, H.; Hattori, T.; Nakane, S.; Tatei, K.; Izumi, T. The Journal of biological chemistry 2005, 280, 40676.

188

(81) Obinata, H.; Izumi, T. Prostag Oth Lipid M 2009, 89, 66.

(82) Ogilby, P. R. Chem Soc Rev 2010, 39, 3181.

(83) Payet, M. D.; Goodfriend, T. L.; Bilodeau, L.; Mackendale, C.; Chouinard,

L.; Gallo-Payet, N. American journal of physiology. Endocrinology and metabolism 2006,

291, E1160.

(84) Peters, J. M.; Taubeneck, M. W.; Keen, C. L.; Gonzalez, F. J. Teratology

1997, 56, 311.

(85) Porter, N. A.; Wolf, R. A.; Yarbro, E. M.; Weenen, H. Biochem Bioph Res

Co 1979, 89, 1058.

(86) Prousek, J. Pure Appl Chem 2007, 79, 2325.

(87) Rao, P. S.; Hayon, E. J Phys Chem-Us 1975, 79, 397.

(88) Rizzo, G.; Fiorucci, S. Current opinion in pharmacology 2006, 6, 421.

(89) Roberts, L. J.; Salomon, R. G.; Morrow, J. D.; Brame, C. J. Faseb J 1999,

13, 1157.

(90) Salomon, R. G. Accounts Chem Res 1985, 18, 294.

(91) Samuelsson, B. The Journal of biological chemistry 2012, 287, 10070.

(92) Schupp, M.; Lazar, M. A. The Journal of biological chemistry 2010, 285,

40409.

(93) Shipley, J. M.; Hurst, C. H.; Tanaka, S. S.; DeRoos, F. L.; Butenhoff, J. L.;

Seacat, A. M.; Waxman, D. J. Toxicol Sci 2004, 80, 151.

(94) Sun, M. J.; Deng, Y. J.; Batyreva, E.; Sha, W.; Salomon, R. G. Journal of

Organic Chemistry 2002, 67, 3575.

(95) Szabo, C.; Ischiropoulos, H.; Radi, R. Nat Rev Drug Discov 2007, 6, 662.

189

(96) Tontonoz, P.; Hu, E.; Graves, R. A.; Budavari, A. I.; Spiegelman, B. M.

Gene Dev 1994, 8, 1224.

(97) Vangaveti, V.; Shashidhar, V.; Jarrod, G.; Baune, B. T.; Kennedy, R. L.

Therapeutic advances in endocrinology and metabolism 2010, 1, 165.

(98) Varga, T.; Czimmerer, Z.; Nagy, L. Bba-Mol Basis Dis 2011, 1812, 1007.

(99) Venkata, N. G.; Robinson, J. A.; Cabot, P. J.; Davis, B.; Monteith, G. R.;

Roberts-Thomson, S. J. Toxicol Lett 2006, 163, 224.

(100) Vieira, H. L. A.; Belzacq, A. S.; Haouzi, D.; Bernassola, F.; Cohen, I.;

Jacotot, E.; Ferri, K. F.; El Hamel, C.; Bartle, L. M.; Melino, G.; Brenner, C.; Goldmacher,

V.; Kroemer, G. Oncogene 2001, 20, 4305.

(101) Vila, A.; Tallman, K. A.; Jacobs, A. T.; Liebler, D. C.; Porter, N. A.;

Marnett, L. J. Chemical research in toxicology 2008, 21, 432.

(102) Wallace, D. C. Annu Rev Genet 2005, 39, 359.

(103) Wang, R.; Kern, J. T.; Goodfriend, T. L.; Ball, D. L.; Luesch, H.

Prostaglandins, leukotrienes, and essential fatty acids 2009, 81, 53.

(104) West, J. D.; Marnett, L. J. Chemical research in toxicology 2005, 18, 1642.

(105) Xu, H. E.; Lambert, M. H.; Montana, V. G.; Parks, D. J.; Blanchard, S. G.;

Brown, P. J.; Sternbach, D. D.; Lehmann, J. M.; Wisely, G. B.; Willson, T. M.; Kliewer, S.

A.; Milburn, M. V. Mol Cell 1999, 3, 397.

(106) Xu, Y.; Prestwich, G. D. The Journal of organic chemistry 2002, 67, 7158.

(107) Yarian, C. S.; Rebrin, I.; Sohal, R. S. Biochem Bioph Res Co 2005, 330,

151.

(108) Yin, H.; Xu, L.; Porter, N. A. Chemical reviews 2011, 111, 5944.

190

(109) Yu, K.; Bayona, W.; Kallen, C. B.; Harding, H. P.; Ravera, C. P.;

Mcmahon, G.; Brown, M.; Lazar, M. A. Journal of Biological Chemistry 1995, 270,

23975.

(110) Zagorski, M. G.; Salomon, R. G. J Am Chem Soc 1982, 104, 3498.

(111) Zhang, C. X.; Baker, D. L.; Yasuda, S.; Makarova, N.; Balazs, L.; Johnson,

L. R.; Marathe, G. K.; McIntyre, T. M.; Xu, Y.; Prestwich, G. D.; Byun, H. S.; Bittman, R.;

Tigyi, G. J Exp Med 2004, 199, 763.

(112) Zhu, X.; Tang, X.; Anderson, V. E.; Sayre, L. M. Chemical research in toxicology 2009, 22, 1386.

191