75
University of Tennessee, Knoxville Trace: Tennessee Research and Creative Exchange Masters eses Graduate School 12-2009 Development of a Novel Lysophosphatidic Acid Activity Probe to Identify and Characterize New Protein Targets Leah M. Cuthriell University of Tennessee - Knoxville is esis is brought to you for free and open access by the Graduate School at Trace: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Masters eses by an authorized administrator of Trace: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. Recommended Citation Cuthriell, Leah M., "Development of a Novel Lysophosphatidic Acid Activity Probe to Identify and Characterize New Protein Targets. " Master's esis, University of Tennessee, 2009. hps://trace.tennessee.edu/utk_gradthes/520

Development of a Novel Lysophosphatidic Acid Activity Probe to

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Development of a Novel Lysophosphatidic Acid Activity Probe to

University of Tennessee, KnoxvilleTrace: Tennessee Research and CreativeExchange

Masters Theses Graduate School

12-2009

Development of a Novel Lysophosphatidic AcidActivity Probe to Identify and Characterize NewProtein TargetsLeah M. CuthriellUniversity of Tennessee - Knoxville

This Thesis is brought to you for free and open access by the Graduate School at Trace: Tennessee Research and Creative Exchange. It has beenaccepted for inclusion in Masters Theses by an authorized administrator of Trace: Tennessee Research and Creative Exchange. For more information,please contact [email protected].

Recommended CitationCuthriell, Leah M., "Development of a Novel Lysophosphatidic Acid Activity Probe to Identify and Characterize New Protein Targets." Master's Thesis, University of Tennessee, 2009.https://trace.tennessee.edu/utk_gradthes/520

Page 2: Development of a Novel Lysophosphatidic Acid Activity Probe to

To the Graduate Council:

I am submitting herewith a thesis written by Leah M. Cuthriell entitled "Development of a NovelLysophosphatidic Acid Activity Probe to Identify and Characterize New Protein Targets." I haveexamined the final electronic copy of this thesis for form and content and recommend that it be acceptedin partial fulfillment of the requirements for the degree of Master of Science, with a major in Chemistry.

Michael Best, Major Professor

We have read this thesis and recommend its acceptance:

David Baker, Frank Vogt

Accepted for the Council:Carolyn R. Hodges

Vice Provost and Dean of the Graduate School

(Original signatures are on file with official student records.)

Page 3: Development of a Novel Lysophosphatidic Acid Activity Probe to

To the Graduate Council: I am submitting herewith a thesis written by Leah M. Cuthriell entitled “Development of a Novel Lysophosphatidic Acid Activity Probe to Identify and Characterize New Protein Targets.” I have examined the final electronic copy of this thesis for form and content and recommend that it be accepted in partial fulfillment of the requirements for the degree of Master of Science, with a major in Chemistry. Michael Best Major Professor We have read this thesis and recommend its acceptance: David Baker Frank Vogt Accepted for the Council: Carolyn R. Hodges

Vice Provost and Dean of the Graduate School

(Original signatures are on file with official student records)

Page 4: Development of a Novel Lysophosphatidic Acid Activity Probe to

Development of a Novel Lysophosphatidic Acid Activity Probe to Identify and Characterize Protein Targets

A Thesis Presented for The Master of Science Degree

University of Tennessee, Knoxville

Leah Cuthriell December 2009

Page 5: Development of a Novel Lysophosphatidic Acid Activity Probe to

ii

Acknowledgements

First and foremost, I would like to thank God for giving me the ability and

health to complete my undergraduate and graduate career. Secondly, I would

like to thank my parents and sister. Your love and support mean the world to me.

Mom and Dad, without your hard work and sacrifice I would not be where I am

today. I am truly grateful for everything you have done to provide for Delila and

me.

Dr. Best, you are a great advisor. Thank you so much for letting me join

your group and for your patience with me and guidance, especially when

research was not going so well. To the members of the Best group, you guys are

amazing. Thank you so much for your help throughout these two and a half

years. You guys have made graduate school an enjoyable experience. I will

never forget all the fun times, laughs, and frustrations we have had together. I

wish you all the best and will miss you guys.

Page 6: Development of a Novel Lysophosphatidic Acid Activity Probe to

iii

Abstract

Lipids are categorized according to their biological function. Phospholipids,

glycolipids, and cholesterol are bulk membrane lipids that offer structure and

support to the cells and organelle membranes. Signaling lipids include

diacylglycerol, phosphatidic acid, and lysophosphatidic acid (LPA). These are

low abundance lipids that are active in various pathways. LPA acts through

GPCR receptors to activate G or beta mediated stimulation of phospholipase C

leading to phosphatidylinositol-(4,5)-bisphosphate hydrolysis, Gi mediated

inhibition of adenyl cyclase, and Gi mediated stimulation of the mitogenic Ras-

MAP kinase. To further elucidate the biological activity of LPA and potentially

identify new LPA binding receptors, activity based protein profiling (ABPP) can

be utilized to label and identify enzymes. This thesis discusses the development

of an LPA activity probe which incorporates a benzophenone group that can be

photocrosslinked to a protein via ultraviolet radiation and a secondary azide tag

to facilitate purification of the probe and bound protein.

Page 7: Development of a Novel Lysophosphatidic Acid Activity Probe to

iv

Table of Contents Chapter Page Chapter 1: Introduction ........................................................................... 1 Background and Significance................................................................................ 4

LPA and Autotaxin Relationship............................................................................ 6

LPA Signaling........................................................................................................ 7

Activity Based Protein Profiling ........................................................................... 12

Chapter 2: Research Design ................................................................ 19

Experimental Procedure......................................................................................26

References ................................................................................................ 60

Page 8: Development of a Novel Lysophosphatidic Acid Activity Probe to

v

List of Figures

Figure Page Figure 1 General lipid structures 2 Figure 2 Autotaxin production of LPA 5 Figure 3 Illustration of activity based protein profiling 13 Figure 4 Current ABPP probes 15 Figure 5 Fluorophosphonate probes 16 Figure 6 LPA activity 19 Figure 7 Initial head group synthesis 22 Figure 8 Head group synthesis 23 Figure 9 Lipid tail synthesis 24 Figure 10 Lipid synthesis 25

Page 9: Development of a Novel Lysophosphatidic Acid Activity Probe to

vi

List of Abbreviations ABPP Activity Based Protein Profiling ASPP Active Site Peptide Profiling ATX Autotaxin cAMP Cyclic Adenosine Monophosphate DCC N,N-dicyclohexylcarbodiimide DDQ 2,3-dichloro-5,6-dicyanobenzoquinone EDG Endothelial Gene Differentiation EGF Epidermal Growth Factor ENTH Epsin Amino Terminal Homology FP Fluorophosphonate GPCR G protein-coupled receptors GDP Guanosine Diphosphate GTP Guanosine Triphosphate ICAT Isotope Coded Affinity Tags LC-MS Liquid Chromatography Mass Spectrometry LPA Lysophosphatidic Acid LPC Lysophosphatidylcholine LPP Lipid Phosphate Phosphohydrolases MAPK Mitogen-Activated Protein Kinase Mud-PIT Multidimensional Protein Identification Technology PH Pleckstrin Homology PLA1 Phospholipase A1 PLD Phospholipase D PPAR Peroxisome Proliferator-Activated Receptor sPLA2 Secretory Phospholipase A2 TOP Tandem Orthogonal Proteolysis

Page 10: Development of a Novel Lysophosphatidic Acid Activity Probe to

1

Chapter 1: Introduction

Lipids compose the cell membrane and help maintain cell structure. This

was discovered by Gorter and Gendell in 1925 when they performed experiments

with chromocytes and found the cells were surrounded by a lipid layer two

molecules thick.1 For over 50 years, the biological roles of lipids were thought to

be limited to composing membrane bilayers and acting as intermediates in lipid

biosynthesis.2 However, it was later discovered that lipids play more extensive

roles in physiological function than originally thought. It is now known that lipids

play key roles in signal transduction and are intermediates in numerous important

biological pathways.

Diacylglycerol is the first known lipid second messenger. Its role as a

second messenger was proved by Hokin and Hokin in 1950 when they

discovered that acetyl or carbamylcholine caused an early turnover of inositol

phospholipids.2 This discovery, along with Nishizuka uncovering that membrane

lipids activate cyclic nucleotide-independent kinase activity, and the revelation

that negatively charged phospholipids replaced membrane components as

kinase activators, indicated that lipids are more involved in eukaryotic cell

biochemistry than previously thought.2-3 Furthermore, lipids are involved in

organelle biogenesis and can regulate intracellular protein dynamics by binding

protein domains. General lipid structures are given in Figure 1.

Page 11: Development of a Novel Lysophosphatidic Acid Activity Probe to

2

.

Lipids can be broken down into two main categories: bulk membrane lipids

and signaling lipids. Three major types of membrane lipids are phospholipids,

glycolipids, and cholesterol. Examples of phospholipids are phosphatidylcholine,

phosphatidylinositol, phosphatidylserine, and phosphatidylethanolamine. These

lipids, along with cholesterol, compose the bulk of the cell membrane. Low

abundance signaling lipids such as phosphatidic acid, diacylglycerol, the

phosphatidylinositol polyphosphates, and lysophosphatidic acid function as

signaling lipids that regulate biological processes.4 A key role of these lipids is to

recruit peripheral proteins to the cell membrane in a spatiotemporally specific

manner.5

Membranes of cellular components are not uniformly composed of lipids.

Lipid type and concentration are dependent on local lipid metabolism and

regulated lipid transport.5a For example, plasma membranes, endosomes, and

trans-Golgi network membranes maintain transbilayer asymmetry by actively

transporting particular types of lipids from one opposing leaflet to another.5a

Originally, it was thought that specific localization of peripheral proteins would

Page 12: Development of a Novel Lysophosphatidic Acid Activity Probe to

3

require protein–protein interactions because protein–lipid interactions were not

expected to be sufficiently specific.5a However, this idea was refuted when it was

determined that subcellular localization behaviors of peripheral proteins can best

be elucidated by analyzing the kinetics and energetics of their binding to model

membranes, as long as the membranes mimic the cellular membrane to which it

is targeted.5a

Peripheral proteins are composed of one or more modular domains

specialized in lipid binding. These include pleckstrin homology (PH), epsin

amino terminal homology (ENTH), and Tubby domains along with many others.6

Some peripheral proteins, such as phospholipase A2, do not contain separate

protein binding domains, but instead utilize their own molecular surface or an

amphipathic secondary structure.5a Other proteins are post-translationally

modified through the attachment of lipid anchors that embed them in the lipid

bilayer. A prominent example involves the Ras GTPases, which have been

implicated in cancer. Initial protein–lipid binding is mediated by electrostatic

interactions between anionic lipid surfaces and cationic protein surfaces.5a In

addition to the role of recruiting proteins to the plasma membrane, lipids such as

lysophospholipids act as intercellular mediators. Intercellular lipid mediators act

via membrane receptors or nuclear receptors known as corticosteroids in order to

effect biological changes.7

Page 13: Development of a Novel Lysophosphatidic Acid Activity Probe to

4

Background and Significance

The initial indication that lysophosphatidic acid 6a plays a more extensive

role than just being an intermediate in intracellular lipid biosynthesis came when

it was found to induce smooth muscle contraction.8 LPA research began in the

1990s with the discovery that LPA is mitogenic and signals through various G

protein-coupled receptors (GPCR).9 LPA is now known to act as an extracellular

lipid mediator that produces growth factor-like responses in a wide range of cells.

LPA is a lysolipid. Lysolipids possess the distinguishing characteristic in

that they only contain one acyl chain located at either the sn-1 or sn-2 position.

LPA is composed of a single acyl chain located at the sn-1 position, a glycerol

backbone, and a phosphate headgroup (Figure 2). It is the smallest and

structurally simplest glycerophospholipid.10 For a long time, the origins of

extracellular LPA production were unclear until it was determined that autotaxin

(ATX) and phospholipase D (PLD) were the same enzyme that catalyzed LPA

production.10-11 Autotaxin (ATX) was discovered in melanoma cell culture and

was found to increase melanoma cell motility.12 PLD and ATX cleave the choline

group from lysophosphatidylcholine (LPC), producing LPA (Figure 2).

LPA is produced by several cell types including mature neuron cells,

Schwann cells, adipocytes, and fibroblasts.13 Production of the lysolipid by these

cells is the source of LPA found in serum, saliva, and follicular fluid.9 In addition,

LPA is also produced in ovarian cancer cells, glioblastomas, and melanoma

cells, indicating that this lipid may play a role in tumor survival and metastasis.

Page 14: Development of a Novel Lysophosphatidic Acid Activity Probe to

5

LPA is synthesized in the aforementioned cell lines by two methods: the removal

of a fatty acid acyl chain from phosphatidic acid by phospholipase A1 (PLA1) or

phospholipase A2, or the removal of a choline group from phosphatidylcholine,

as shown in Figure 2.8

In phospholipase A1 production of LPA, PLA1 releases the fatty acid chain

from the sn-1 position of membrane phospholipids, producing sn-2 and

polyunsaturated isoforms of LPA that interact with LPA receptor 3.14 Type II

secretory phospholipase A2 (sPLA2) removes an acyl chain from the sn-2

position but is not capable of producing hydrolyzed lipids in intact cell

membranes, resulting in low levels of LPA in the plasma. However, sPLA2 is

capable of selectively hydrolyzing lipids in damaged cells and microvesicles that

are released during apoptosis and by cancer cells, and are found in large

numbers in malignant fluids leading to high levels of LPA expressed in cancer

cells.8

Page 15: Development of a Novel Lysophosphatidic Acid Activity Probe to

6

LPA and Autotaxin Relationship

The protein autotaxin (ATX) was first discovered in melanoma cell culture

and has been found to stimulate cancer cell motility, angiogenesis, and cell

proliferation, indicating that it may be pivotal in cancer biology.15 It is a member

of the nucleotide pyrophosphatase/phosphodiesterase family of enzymes.12a

ATX is a type II integral membrane protein that contains a single transmembrane

domain, two somatomedin B-like domains, and a catalytic domain.16 The highest

levels of ATX/PLD mRNA are expressed in kidney, brain, lung, intestine, and

ovary cells.

ATX production of LPA is of biological importance because it plays key

roles in various pathways. ATX utilizes LPC as a substrate, which exists at high

concentrations in many bodily fluids. The resulting product, LPA, stimulates

cellular responses such as malignant cell proliferation and LPA autocrine control

of adipose tissue. The fact that autotaxin stimulation in malignant cell

proliferation is increased in the presence of LPC provides evidence that LPA is a

signaling component in cell proliferation. The proposed mechanism of cancer

cell motility involves activation through GPCR LPA1.12a In addition to the role it

plays in cell proliferation, ATX-induced LPA production also results in the

differentiation of adipocytes and adipocyte cell lines. ATX has been shown to be

upregulated in genetically obese mice. These two facts, along with studies

showing that LPA exists in extracellular fluid of adipose cells and is released in

Page 16: Development of a Novel Lysophosphatidic Acid Activity Probe to

7

vitro by adipocytes, imply that LPA paracrine control of adipose tissue could be

mediated by ATX.17

LPA signaling

LPA activity is receptor-mediated with the lipid interacting with at least

three G protein-coupled receptors that belong to the endothelial differentiation

gene subfamily, which traverses the membrane seven times.9 The first receptor

to be discovered was LPA1, which is the most widely expressed LPA receptor

with high mRNA levels in the colon, intestine, pancreas, small intestine, and

heart.8-9 High levels of this receptor are also present in the cerebral cortical

ventricular zone during neurogenesis and in adult oligodendrocytes and

Schwann cells.8

After its discovery, two other LPA receptors known as LPA2 and LPA3

were identified. LPA2 and LPA3 are not as widely distributed as LPA1 but are

overexpressed in cancer cells, indicating their role in the disease. Recently,

three other LPA receptors were identified.18 One receptor known as LPA4 is

unique in the fact that it does not belong to the EDG family, but is most similar to

the purinergic family, which means LPA may have a greater biochemical role

than previously thought.9 While there is not enough evidence to support LPA4

activation of phopholipase C and adenylyl cyclase, circumstantial evidence

suggests that the receptor may tentatively mediate the activity of these enzymes.

LPA receptors are known to couple to three subfamilies of G proteins

known as Gα, Gi, and G12/13.9 These three major receptors share high homology

Page 17: Development of a Novel Lysophosphatidic Acid Activity Probe to

8

in amino acid sequences in all areas except for the C-terminal region.9 The

extent of carboxyl tail homology between the LPA1 and LPA2 receptors is 27%

with the LPA2 and LPA3 receptors having only 17% homology.19 Since all other

domains have the same homology, the ability of the receptors to effect biological

change must stem from the different amino acid sequences located in the

carboxyl terminal region.19-20 Evidence to prove the specificity of the carboxyl

terminal region has been found from studies published by Xu et al., in which a

yeast two-hybrid screen was utilized to identify TRIP6 as a LPA2 receptor

interacting protein.19

Major signaling routes for LPA include: G or beta mediated stimulation of

phospholipase C leading to phosphatidylinositol-(4,5)-bisphosphate hydrolysis, Gi

mediated inhibition of adenyl cyclase, Gi mediated stimulation of the mitogenic

Ras-MAP kinase, Gα and G12/13 mediated activation of small GTPase RhoA,

which regulates cytoskeletal movement, and finally Gi activation of

phosphoinositide-3-kinase, Rac, and PKB/Akt to promote cell movement and

suppress cell death.9 LPA also signals through second messenger pathways

and activates the Ras and Rho family of GTPases.9 The Ras and Rho GTPase

cycles include GDP and GTP bound states. GTP binding is promoted by specific

GDP–GTP exchange factors in which GTP bound forms can react with various

downstream effectors producing changes in the cellular environment.8

All mammalian cells, tissues, and organs with the exception of the liver

express LPA receptor subtypes of the EDG family, which indicates that LPA

Page 18: Development of a Novel Lysophosphatidic Acid Activity Probe to

9

signaling occurs in a cooperative manner. However, the identity of the LPA

receptor subtype that couples each known G protein-coupled receptor is

unknown. All three receptors can mediate PLC activation, inhibition of cyclic

adenosine monophosphate (cAMP) accumulation, and activation of mitogen-

activated protein kinase (MAPK) pathway, but different isoforms of LPA have a

different efficacy and potency for each receptor, thus triggering downstream

events. LPA2 exhibits the highest affinity of all the receptors for LPA and

couples more efficiently to neovascularizing factors with LPA1 having a bigger

effect on cell motility events.8

Ras MAP Kinase Signaling

LPA-induced activation of Ras involves the GES, SOS, and intermediate

protein tyrosine kinase dependent activity. According to studies, Ras activation

is not a direct linear pathway such as Gi-tyrosine kinase-Ras-GTP, but instead it

involves exchange factors and cell-type dependent, mutually opposing, GTPase

activating proteins.9 One proposed method of signaling suggests that LPA and

other GPCR agonists act through the epidermal growth factor receptor to trigger

mitogenic signaling. In this transactivation model, a GPCR activated

metalloprotease cleaves the epidermal growth factor (EGF) precursor at the cell

surface to stimulate EGFR signaling. However, the transactivation model is not

widely accepted since most of the studies were conducted with pharmacological

inhibitors.9

Page 19: Development of a Novel Lysophosphatidic Acid Activity Probe to

10

Rho/Rac signaling

Cytoskeletal organization is mediated through Rho GTPases. Three well

studied GTPases are RhoA, Cdc42, and Rac. LPA activation of RhoA proceeds

via G12/13 subunits that bind to three distinct Rho-specific guanine nucleotide

exchange factors, promoting RhoA-GTP accumulation and activation of

downstream Rho-Kinase, which initiates cell rounding, retraction, and endothelial

tight junction opening.9

LPA as an intracellular messenger

It is currently well known that LPA is an intermediate in phospholipid

biosynthesis, which takes place in the endoplasmic reticulum. During this

process, LPA is formed from glycerol-3-phosphate by acyl-CoA and then further

acylated to phosphatidic acid. Evidence may suggest that LPA has another role

in the body besides just being an intermediate in lipid biosynthesis. High

concentrations of LPA can compete with synthetic lipid found within the cell to

bind to the nuclear transcription factor peroxisome proliferator-activated receptor

gamma (PRAR-γ). However, it is unclear how extracellular LPA could cross the

membrane, since it would be rapidly dephosphorylated by membrane-bound lipid

phosphatases. Furthermore, if extracellular LPA found its way to the cytosol, it is

still unclear how the lipid would be able to reach the nucleus. Thus, new studies

need to be conducted in order to determine whether LPA acts as an intracellular

messenger.9

Page 20: Development of a Novel Lysophosphatidic Acid Activity Probe to

11

LPA Degradation

Logically, LPA must be degraded in mammalian cells in order to maintain

cell functionality. Extracellular LPA is degraded by phosphate group removal to

produce inactive monoacylglycerol.9 In addition, a family of membrane ecto

enzymes has been identified whose members mediate dephosphorylation of LPA

and other lipid phosphatases. This group of enzymes is known as lipid

phosphate phosphohydrolases (LPP). LPPs belong to a superfamily of

hydrolases that includes bacterial acid phosphatases and DGPPase that contain

three highly conserved domains. Unlike phospholipase C, LPP does not

dephosphorylate water-soluble phosphate esters and appears to have a

substrate similar to phosphatidic acid, indicating that it competes with

phosphatidic acid (PA) for LPA dephosphorylation. However, more studies are

needed in order to confirm their ability to degrade lysophosphatidic acid.

Activity Based Protein Profiling

As evidenced by the information obtained from the text above, LPA has

extensive biological and pathophysiological roles. Much remains to be learned

about the pathways in which LPA is active and the proteins that it targets. With

the advent of genome sequencing, an array of information about the proteins

present in eukaryotic organisms has been obtained. Genomic techniques such

as chromosomal translocation, transcriptional profiling, and RNA interference–

gene silencing have given insights into the physiological roles of proteins.21

Page 21: Development of a Novel Lysophosphatidic Acid Activity Probe to

12

However, genomic techniques cannot convey other important information

about protein function, including regulation caused by small molecule binding and

post-translational protein modification. To efficiently elucidate protein function at

this level, a range of sophisticated proteomics have been developed. Within the

field of proteomics, basic techniques such as gel electrophoresis and mass

spectrometry have been extended to devise powerful techniques such as

multidimensional protein identification technology (MudPIT) and isotope coded

affinity tags (ICAT), yeast two–hybrid systems, and protein microarrays that

generate more information about quantitative differences in protein abundance

and protein interactions.22 However, these methods are not able to probe

proteins in vivo, which is disadvantageous because variation in activity caused by

small molecule binding and post-translational modification cannot be detected.22b

Probes have been developed that specifically target enzyme families. In addition

to binding already characterized enzyme classes, ABPP probes used in

conjunction with streptavidin chromatography, gel separation, and mass

spectrometry analysis can identify and assign biological function to novel

proteins.22a

The strategy of activity based protein profiling (ABPP) was developed in

order to characterize proteins based on activity rather than abundance. ABPP is

a chemical proteomic method that utilizes active site-directed probes to visualize

changes in protein activity located in cells and tissues. Although recent

advances in genomic sequencing have brought more attention to the utility of

Page 22: Development of a Novel Lysophosphatidic Acid Activity Probe to

13

ABPP, the first stages of ABPP began over a century ago.21 ABPP is ideal for

targeting an array of enzyme families since probes can be constructed that

incorporate mechanism-based inhibitors, protein-reactive natural products, and

general electrophiles.21 Currently, probes have been developed for more than a

dozen enzyme classes, including proteases, kinases, and phosphatases.

General probe design incorporates a reactive group by which the probe is

selectively cross-linked to targets, as well as a secondary tag bound by a linker.

Common reporter tags are rhodamine, azide, alkyne, and biotin. An illustration of

activity based protein profiling is given in Figure 3.

Page 23: Development of a Novel Lysophosphatidic Acid Activity Probe to

14

The principal aim of ABPP is the synthesis of small probes that can

covalently label enzyme active sites. Ideally, probes must be able to target a

wide array of enzymes, but since numerous nucleophilic residues without

catalytic activity exist within the proteome, cross-reactivity with such residues

must be avoided in order to prevent jeopardizing ABPP studies.23 This can be

achieved by combining reactive and binding groups that target conserved

mechanistic or structural features in enzyme active sites.23 Reactive groups can

be either electrophilic and modify active sites or photoaffinity groups that label

targets upon UV irradiation.23

Once the probe has labeled target proteins via reaction with active site

residues, purification of the successfully labeled proteins is facilitated by the

secondary reporter tag on the probe. Reporter tags such as alkynes and azides

can undergo click reactions that allow the probe to be biotinylated and then

purified via a streptavidin column. Examples of probes that have been currently

utilized in activity based protein profiling are given in Figure 4. In addition to the

probes drawn in Figure 4, Cravatt and colleagues have designed a series of

probes that target the serine hydrolases. These probes, termed

fluorophosphonate reagents (shown in Figure 5) have been derivatized with a

number of linkers and a reporter tag such as rhodamine or biotin in order to

visualize and characterize catalytically active serine hydrolases in proteomes.22b

These probes have shown great selectivity for labeling active serine hydrolases

but not the inactive inhibitor bound forms.

Page 24: Development of a Novel Lysophosphatidic Acid Activity Probe to

15

O O

OO

N3

O

N3

PO

OO

N

O

Bifunctional lipid probe

Benzophenone photoaffinity tag

Azide secondary tag

Page 25: Development of a Novel Lysophosphatidic Acid Activity Probe to

16

S

HNNH

O

OHN

O

HN

ONH O

N

O OH

ONH

PO

OO

OHO

Arylphosphonate probe

O OO

N

NO

HN

HN

OP

O

FO

Fluorophosphonate probe

Rhodamine tag

The great reactivity of these probes stems from an electrophilic center that

is primed for covalent reaction with a highly nucleophilic oxygen atom of serine

and the near-tetrahedral structure of the fluorophosphonate (FP) groups, which

mimic the enzyme substrate tetrahedral intermediate. Craik and Mahrus

synthesized a series of probes with complementary reactivity to the

fluorophosphonate probes.22b These probes incorporate a diphenyl phosphonate

electrophile and substrate mimicking recognition groups that promote selective

recognition.

Once probes have been synthesized, they can be utilized to study protein

interactions through a variety of different methods. One platform that exists is gel

electrophoresis. This method incorporates the use of fluorescent probes in

conjunction with biotinylated probes. Fluorescent probe labeled proteomes are

distinguished by 1D or 2D polyacrylamide gel electrophoresis. Then, biotinylated

probes can be used along with streptavidin affinity chromatography, gel

separation, and mass spectrometry in order to purify the labeled probes. Another

Page 26: Development of a Novel Lysophosphatidic Acid Activity Probe to

17

method that offers higher resolution than gel electrophoresis is liquid

chromatography–mass spectrometry (LC–MS).23

LC–MS can be divided into two categories. The first category consists of

experiments that analyze protein targets of probes. The second deals with those

that specifically analyze probe-labeled peptides derived from these targets. The

first method is essentially a combination of ABPP and MudPIT. The procedure

involves incubating biotinylated probes with streptavidin beads to enrich probe

labeled proteins. Incubation is then followed by trypsin digestion and the

enriched proteins are analyzed by LC–MS. This process is capable of detecting

50–100+ enzyme activities but does not offer a straightforward way of identifying

probe labeled peptides of enzyme targets. To overcome this disadvantage,

active site peptide profiling (ASPP) can be used. ASPP involves digesting probe

treated proteomes with trypsin prior to incubation. Probe labeled peptides are

enriched using either streptavidin or antibody resins followed by analysis with

LC–MS.21

In order to obtain the information that ABPP and ASPP offers, Speers and

Cravatt developed tandem orthogonal proteolysis–ABPP (TOP–ABPP).21 Even

though advancements in resolution and information gleaned from ABPP have

occurred, methods that involve LC–MS are still not desirable due to the large

sample volume that is required in order to run an analysis. Ideally, an ABPP

procedure will eventually be produced that offers the high resolution of LC–MS

along with the minimal sample requirement of gel electrophoresis. Progress has

Page 27: Development of a Novel Lysophosphatidic Acid Activity Probe to

18

been made towards this goal by combining capillary electrophoresis coupled with

laser-induced fluorescence. However, more work needs to be done in order to

achieve a method which produces high resolution with minimal sample

requirement.21

ABPP is currently utilized in biological experiments such as target

discovery, inhibitor discovery, and enzyme active site characterization. ABPP is

a suitable method for target discovery because it can account for protein

regulation via small molecule binding and post-translational modification.

Inhibitor discovery involves utilizing the probe as a competitor in which inhibitors

are then identified by their ability to block probe labeling of enzymes. This

strategy negates the need for recombinant expression and purification because

enzyme tests can be conducted within the proteome. In addition, the probe acts

as a substitute for substrate analysis resulting in the ability to detect enzymes

that may not be detectable via other methods. Finally, with the ability to label

new enzyme active sites, ABPP is an avenue to characterize new enzymes that

have not yet been documented.21

Page 28: Development of a Novel Lysophosphatidic Acid Activity Probe to

19

Chapter 2: Research Design

The main focus of my research has been the development of the LPA

activity probe shown in Figure 6. The synthetic route chosen for production of

the activity probe involved the incorporation of a benzophenone group and an

azide tag. Proteins that bind the LPA backbone can be cross-linked to the probe

through irradiation of the photoaffinity group with ultra-violet light. The azide can

then be used to label cross-linked adducts via click chemistry. The probe, along

with bound proteins, can then be purified via a streptavidin column. The activity

probe 23 in Figure 6 was synthesized from (S)-glycerolacetonide 1 in 16 steps.

HO OO

O

NHO

HNO

O

N3

POOH

OH

Benzophenone group

Azide tag

23

Figure 6. LPA activity probe.

Page 29: Development of a Novel Lysophosphatidic Acid Activity Probe to

20

The first synthetic route (shown in Figure 7) to the target compound began

by protecting the free alcohol of (S)-glycerolacetonide with tert-butyldiphenylsilyl

chloride to produce 2 (Figure 7). After acetal deprotection with 90% acetic acid

to 3, a monomethoxytrityl protecting group was selectively installed on the

primary hydroxyl group to yield 4. This was followed by incorporation of a p-

methoxybenzyl chloride protecting group to generate 5. However, 6 was not

obtained in high yield, and reproducibility was problematic. A better synthetic

route (shown in Figure 8) was then developed in which the order of protecting

group installation was switched. The first step of the synthesis involved

protecting the free alcohol of 1 with a p-methoxybenzyl protecting group, followed

by acetal deprotection to yield 9. After this step, the primary alcohol of 9 was

selectively protected by monomethoxytrityl chloride (MMT) followed by

incorporation of a tert-butyldiphenylsilyl protecting group to yield 11. Finally, the

MMT group was deprotected producing the free alcohol 12.

Incorporation of the benzophenone cross-linking group and azide tag

(shown in Figure 9) began with the coupling of 4-benzoylbenzoic acid to Z-N-

Boc-L-lysine methyl ester to produce 14. Trifluoroacetic acid deprotection yielded

the free amine 15, which was converted into azide 16 using 0.01 M copper

sulfate solution, 0.804 M potassium carbonate solution and imidazole-1-sulfonyl

azide. Once the azide was purified, the methyl ester was hydrolyzed using 2 M

sodium hydroxide and methanol to yield 17. Coupling of the acid to methyl 6-

aminohexanoate yielded 19 upon ester hydrolysis with 2 M sodium hydroxide

Page 30: Development of a Novel Lysophosphatidic Acid Activity Probe to

21

and ethanol (Figure 10). Following this reaction, 19 and 12 underwent a coupling

reaction to produce 20. After purification, the benzyl-protected phosphoramidite

was installed following p-methoxybenzyl deprotection with DDQ to yield 22. The

free phosphate was then produced with trimethylsilyl bromide. Finally, the lipid

intermediate was deprotected with tetrabutylammonium fluoride to produce the

target LPA probe 23.

Conclusion

The best synthethic route to produce a novel LPA activity probe involved

protecting the free alcohol of (S)-glycerolacetonide with p-methoxybenzyl

chloride. After acetal deprotection, MMT and TBDPS groups were selectively

installed. This was necessary in order to produce 12 with a free alcohol at the

sn-1 position. The next synthetic strategy involved producing an acyl chain

which incorporated a benzophenone cross-linking group and an azide label that

would facilitate purification of the probe and bound protein. Synthesis of the

derivatized acyl chain began with a lysine analog and was achieved in seven

steps yielding 19. Next, 12 and 19 were coupled together to produce 20. After

installing a dibenzyl protected phospharamidite, 23 was achieved in 61% yield

following TMSBr and TBAF deprotection to produce an LPA activity probe that

can be utilized to identify and characterize new protein targets.

Page 31: Development of a Novel Lysophosphatidic Acid Activity Probe to

22

Page 32: Development of a Novel Lysophosphatidic Acid Activity Probe to

23

OMMTHO

PMBOTBDPSClimidazole

N

OMMTTBDPSO

PMBO

OHTBDPSO

PMBOCSA

MeOH,CH2Cl210 11 12

99% 31%

Page 33: Development of a Novel Lysophosphatidic Acid Activity Probe to

24

OHN

O

N3

O

O

N N SO

ON3

CuSO4, K2CO3MeOH

1675% over two steps

NaOHMeOH

HOHN

O

N3

O

O

17

99%

Page 34: Development of a Novel Lysophosphatidic Acid Activity Probe to

25

TBDPSO OO

O

NHO

HNO

O

N3

NH

HN

O

N3

O

O

DCC, DMAP

CH2Cl2TBDPSO OH

OO

12 19

20

O

HO

99%

Figure 10. Lipid Synthesis

NH

HN

O

N3

O

O

O

OH2N

DMAP, EDCl,NMM, CH2Cl2

18

O

O

88%

O

NaOHEtOH

19

NH

HN

O

N3

O

O

HO

O

93%

Page 35: Development of a Novel Lysophosphatidic Acid Activity Probe to

26

Page 36: Development of a Novel Lysophosphatidic Acid Activity Probe to

27

TBDPSO OO

HO

NHO

HNO

O

N3

TBDPSO OO

O

NHO

HNO

O

N3

POO

OPO

ON

m-CPBACH2Cl2

21 22

Figure 10. Lipid synthesis continued.

50%

Page 37: Development of a Novel Lysophosphatidic Acid Activity Probe to

28

TBDPSO OO

O

NHO

HNO

O

N3

POO

O HO OO

O

NHO

HNO

O

N3

POOH

OH1. TMSBrCH2Cl2

2. TBAFTHF

22 23

Figure 10. Lipid synthesis continued.

61%

Page 38: Development of a Novel Lysophosphatidic Acid Activity Probe to

29

Experimental Procedures

General. All reagents were purchased from Aldrich, Acros, or ChemImpex and

used as received. Dry solvents were obtained from a Pure Solv solvent delivery

system purchased from Innovative Technology, Inc. Column chromatography

was performed using 230–400 mesh silica gel purchased from Sorbent

Technologies. NMR spectra were obtained using a Varian Mercury 300

spectrometer and a Bruker Avance 400 spectrometer. Mass spectra were

obtained with a MALDI-TOF spectrometer. Optical rotation values were obtained

using a Perkin-Elmer 241 polarimeter.

(S)-4-((4-methoxybenzyloxy)methyl)-2,2-dimethyl-1,3-dioxolane (8)

Sodium hydride (518 mg, 12.1 mmol) was dissolved in 40 mL N,N-

dimethylformamide at 0 °C under nitrogen. Compound 7 (1 mL, 8.1 mmol) was

dissolved in 40 mL N,N-dimethylformamide and added slowly over 10 minutes to

the sodium hydride solution. After complete addition of 7, the reaction was

stirred at 0 °C for 1 h, then p-methoxybenzyl chloride (1.64 mL, 12.1 mmol) was

added. The mixture was stirred for 18 h at room temperature. After the reaction

was complete, water (50 mL) was added to quench the reaction. The mixture

was then extracted with dichloromethane and saturated sodium chloride (2 x 50

mL). The organic phase was then dried with magnesium sulfate, filtered, and

Page 39: Development of a Novel Lysophosphatidic Acid Activity Probe to

30

then concentrated. Column chromatography on silica gel and a gradient solvent

system of 10–50% ethyl acetate/hexanes afforded 8 as a clear oil (1.51 g, 74%).

NMR data matched that in the literature.24

(R)-3-(4-methoxybenzyloxy)propane-1,2-diol (9)

Compound 8 (1.51 g, 5.9 mmol) was dissolved in methanol (59 mL), and p-

toluenesulfonic acid (181 mg, 0.95 mmol) was added to the solution. The

reaction was stirred at room temperature for 24 h. The reaction was quenched

with sodium bicarbonate (110 mg, 1.3 mmol) and extracted with water and

dichloromethane (2 x 50 mL). The organic phase was dried with magnesium

sulfate, filtered, and concentrated. Column chromatography on silica gel and a

gradient solvent system of 50% methanol/ethyl acetate and 100% methanol

produced 9 (661 mg, 52%) as a clear oil.

NMR data matched that in the literature.24

(S)-1-((4-methoxybenzyl)oxy)-3-((4-methoxyphenyl)diphenylmethoxy)

propan-2-ol (10)

Diol 9 (661 mg, 3.1 mmol) was dissolved in pyridine (31 mL) under nitrogen. 4-

methoxytrityl chloride (1.91 g, 6.2 mmol), 4-dimethylaminopyridine (381 mg, 0.31

mmol), and molecular sieves were added. The reaction was stirred at room

Page 40: Development of a Novel Lysophosphatidic Acid Activity Probe to

31

temperature for 24 h. After the reaction was complete, the mixture was filtered

over Celite to remove the molecular sieves and concentrated. Column

chromatography on silica gel and a gradient solvent system of 10–50% ethyl

acetate/hexanes and 100% ethyl acetate afforded 10 as a clear oil (1.33 g, 88%).

NMR data matched that in the literature.24b

(S)-tert-butyl((1-((4-methoxybenzyl)oxy)-3-((4-

methoxyphenyl)diphenylmethoxy)propan-2-yl)oxy)diphenylsilane (11)

Alcohol 10 (408 mg, 0.84 mmol) was dissolved in pyridine (8 mL) under nitrogen.

To the mixture was added imidazole (114 mg, 1.7 mmol), t-butyldiphenyl

chlorosilane (437 µL, 1.7 mmol), and molecular sieves. The reaction was stirred

at room temperature for 18 h. The mixture was then filtered over celite to remove

the molecular sieves and concentrated. Chromatography on silica gel with a

gradient solvent system of 10–25% ethyl acetate/hexanes afforded 11 (600 mg,

99%). NMR data matched that in the literature.24b

Page 41: Development of a Novel Lysophosphatidic Acid Activity Probe to

32

(R)-2-((tert-butyldiphenylsilyl)oxy)-3-((4-methoxybenzyl)oxy)propan-1-ol

(12)

Compound 11 (599 mg, 0.83 mmol) was dissolved in methanol:dichloromethane

(33 mL 2:1). Camphorsulfonic acid (96 mg, 0.414 mmol) was added. The

reaction was stirred at room temperature for 3.5 h after which it was neutralized

by triethylamine and concentrated. Chromatography on silica gel with a gradient

solvent system of 25–35% ethyl acetate/hexanes afforded 12 (113 mg, 31%).

NMR data matched literature.24b

(S)-methyl2-(4-benzoylbenzamido)-6-((tert-

butoxycarbonyl)amino)hexanoate (14)

Compound 13 (400 mg, 1.0 mmol) was dissolved in methanol (11 mL), and 10%

palladium hydroxide (40 mg) by weight was added. The mixture was placed

under hydrogen and stirred for 24 h at room temperature. After the 24-h period,

the reaction was filtered over Celite. The filtrate was concentrated, and the

remaining traces of solvent were removed. The residue was then dissolved in

Page 42: Development of a Novel Lysophosphatidic Acid Activity Probe to

33

N,N-dimethylformamide (6.7 mL) and placed under nitrogen. N-

methylmorpholine (390 µL, 3.5 mmol), along with 4-benzylbenzoic acid (229 mg,

1.1 mmol), EDCl (233 mg, 1.2 mmol), and 4-dimethylaminopyridine (149 mg, 1.2

mmol) was added. The reaction was stirred for 5 h at room temperature. After

completion, the reaction was concentrated. Column chromatography on silica

gel with a solvent gradient system of 50% ethyl acetate/hexanes afforded 14

(351 mg, 74%). NMR data matched that in the literature.25

OHN

O

NHO

O

O

OHN

O

N3

O

O

N N SO

ON3

CuSO4, K2CO3MeOHO

2.

1. TFACH2Cl2

(S)-methyl 6-amino-2-(4-benzoylbenzamido)hexanoate (16)

Compound 14 (343 mg, 0.71 mmol) was dissolved in dichloromethane (5.9 mL).

Trifluoroacetic acid (5.9 mL) was added, and the solution was stirred at room

temperature for 2.5 h. After completion, the solvent was removed, and the

residue (15) was placed under vacuum for 24 h. The residue was then dissolved

in methanol (7.5 mL), and imidazole-1-sulfonyl azide hydrochloride (177 mg, 0.8

mmol), 0.804 M potassium carbonate solution (1.4 mL, 1.2 mmol), and 0.01 M

copper(II) sulfate pentahydrate solution (707 µL, 0.007 mmol) were added. The

reaction was stirred at room temperature for 3.5 h. After completion, the reaction

Page 43: Development of a Novel Lysophosphatidic Acid Activity Probe to

34

was extracted with ethyl acetate and 2 M hydrochloric acid. The organic phase

was dried with magnesium sulfate and concentrated. Column chromatography

on silica gel with a solvent gradient of 50% ethyl acetate/hexanes afforded 16

(208 mg, 75%). NMR data matched that in the literature.25

(S)-6-azido-2-(4-benzoylbenzamido)hexanoic acid (17)

Compound 16 (276 mg, 0.7 mmol) was dissolved in methanol (12.3 mL), and 2 M

sodium hydroxide (12.3 mL) was added. The reaction was stirred at room

temperature for 1 h. After completion, the reaction was extracted with 2 M

hydrochloric acid. The organic layer was dried with magnesium sulfate and

concentrated to yield 17 (266 mg, 99%). The residue was passed on without

further purification. NMR data matched that in the literature.25

(R)-methyl 6-(6-azido-2-(4-benzoylbenzamido)hexanamido)hexanoate (18)

Compound 17 (239 mg, 0.628 mmol) was dissolved in dichloromethane (11.4

mL), placed under nitrogen, and 4-dimethylaminopyridine (99 mg, 0.817 mmol)

Page 44: Development of a Novel Lysophosphatidic Acid Activity Probe to

35

was added. Methyl 6-aminohexanoate hydrochloride (148 mg, 0.817 mmol) was

dissolved in dichloromethane (11.4 mL) and N-methylmorpholine (691 mL, 6.28

mmol) was added. The solution was combined with 17 dissolved in

dichloromethane, and EDCl (156 mg, 0.817 mmol) was added. The reaction was

stirred at room temperature for 18 h. Column chromatography on silica gel with a

solvent gradient of 35–75% ethyl acetate/hexanes afforded 18 (273 mg, 88%).

1H NMR (300 MHz, CDCl3 ) δ 1.37 (dd, J = 14.94, 7.91 Hz, 2H), 1.61 (m, 8H),

1.83 (d, J = 7.32 Hz, 1H), 2.0 (m, 1H), 2.30 (t, J = 7.32 Hz, 2H), 3.28 (m, 4H),

3.65 (s, 3H), 4.68 (q, J = 7.04 Hz, 1H), 6.62 (m, 1H), 7.50 (t, J = 7.46 Hz, 2H),

7.62 (t, J = 7.40 Hz, 1H), 7.78 (d, J=8.30 Hz, 2H), 7.83 (d, J=8.12 Hz, 2H), 7.94

(dd, J = 8.17 Hz, 2H); 13C NMR (300 MHz CDCl3) δ 191.58, 170.86, 168.26,

166.05, 166.01, 136.55, 132.54, 129.71, 129.67, 128.35, 128.07, 126.73, 53.11,

51.14, 50.71, 38.96, 33.34, 31.88, 28.58, 28.55, 28.16, 25.84, 25.82, 23.91,

22.37; MALDI-HRMS [M + Na]+ calcd: 530.2374 found: 530.2394; [α]D296K =

+0.21 (c 6.5, 1:4 methanol/chloroform)

(R)-6-(6-azido-2-(4-benzoylbenzamido)hexanamido)hexanoic acid (19)

Compound 18 (303 mg, 0.598 mmol) was dissolved in ethanol (3 mL) and 2 M

sodium hydroxide (3 mL) was added. The solution was stirred at room

Page 45: Development of a Novel Lysophosphatidic Acid Activity Probe to

36

temperature for 1 h. Once the reaction was complete, the solution was extracted

with dichloromethane and 2 M hydrochloric acid (2 x 30 mL). The organic phase

was dried with magnesium sulfate and concentrated to yield 19 (272 mg, 93%).

The residue was used in the next reaction.

Compound 12 (320 mg, 0.6 mmol) was dissolved in dichloromethane (11 mL)

under nitrogen. N,N-dicyclohexylcarbodiimide (173 mg, 0.84 mmol) and 4-

dimethylaminopyridine (103 mg, 0.84 mmol) were added. The mixture was

stirred at room temperature for 20 minutes. Compound 19 (396 mg, 0.84 mmol)

was then added in 10 mL dichloromethane, and the reaction was stirred at room

temperature for 24 hours. Once the reaction was complete, the mixture was

concentrated. Column chromatography on silica gel with a solvent gradient

system of 35–75% ethyl acetate/hexanes afforded 20 as a clear oil (596 mg,

99%).

1H NMR (300 MHz, CDCl3) δ 1.04 (s, 9H), 1.28 (m, 2H), 1.51 (m, 8H), 1.94 (m,

2H), 2.11 (m, 2H), 3.26 (m, 4H), 3.40 (d, J = 4.86 Hz, 2H), 3.79 (s, 3H), 4.11 (m,

3H) 4.69 (s,2H), 6.74 (s, 1H), 6.82 (d, J = 8.30 Hz, 2H), 7.11 (d, J = 8.35 Hz, 2H),

Page 46: Development of a Novel Lysophosphatidic Acid Activity Probe to

37

7.40 (m, 6H), 7.50 (d, J=7.90 Hz, 2H), 7.59 (d, J=7.61 Hz, 1H), 7.65 (t, J = 7.68

Hz, 4H), 7.78 (d, J =8.30 Hz, 2H), 7.83 (d, J=8.12 Hz, 2H), 7.94 (d, J = 8.17 Hz,

2H); 13C NMR (300 MHz CDCl3) δ 195.80, 173.25, 171.18, 168.59, 166.37,

146.02, 140.31, 136.84, 135.80, 135.78, 133.61, 132.87, 130.06, 129.61, 129.15,

127.63, 127.51, 127.47, 127.09, 113.62, 72.80, 70.86, 70.19, 66.06, 55.21,

53.45, 51.04, 39.38, 33.67, 32.28, 29.00, 28.51, 26.80, 26.24, 24.16, 22.71,

19.25; MALDI-HRMS [M + Na]+ calcd: 948.4338 found: 948.4330; [α]296KD =

+0.041 (c 1.9, 1:4 methanol/chloroform)

TBDPSO OO

OO

NHO

HNO

O

N3

TBDPSO OO

HO

NHO

HNO

O

N3

DDQCH2Cl2

21

Compound 20 (147 mg, 0.159mmol) was dissolved in dichloromethane (5 mL)

and 2,3-dichloro-5,6-dicyanobenzoquinone (DDQ) (90 mg, 0.398 mmol) was

added. Following completion (4.5 h), the reaction was extracted with 10%

sodium carbonate and dichloromethane (2 x 30 mL). The organic phase was

Page 47: Development of a Novel Lysophosphatidic Acid Activity Probe to

38

dried with magnesium sulfate and concentrated. Column chromatography on

silica gel with a solvent gradient of 40–75% ethyl acetate/hexanes and 100%

ethyl acetate/hexanes afforded 21 as a clear oil (91.7 mg, 78%).

1H NMR (300 MHz, CDCl3) δ 1.07 (s, 9H), 1.29 (m, 2H), 1.52 (m, 8H), 1.78 (m,

1H), 1.97 (m, 1H), 2.15 (t, J = 7.05, 2H), 2.43 (s, 1H), 3.27 (m, 4H), 3.53 (d, J =

3.66 Hz, 2H), 3.91 (m, 1H), 4.10 (m, 3H), 4.64 (m, 1H), 7.39 (m, 6H), 7.50 (d,

J=7.90 Hz, 2H), 7.59 (d, J=7.61 Hz, 1H), 7.65 (d, J = 7.68 Hz, 4H), 7.78 (d, J

=8.30 Hz, 2H), 7.83 (d, J=8.12 Hz, 2H), 7.94 (d, J = 8.17 Hz, 2H) 13C NMR (300

MHz CDCl3) δ 145.80 145.86, 136.98, 136.84, 136.64, 135.49, 132.69, 129.89,

128.22, 127.59, 127.45, 126.88, 71.25, 53.22, 50.86, 33.47, 32.06, 28.30, 26.66,

22.51, 19.05, 16.62 MALDI-HRMS [M + Na]+ calcd: 828.3766, found 828.3763

[α]296KD = +0.095 (c 0.41, 1:4 methanol/chloroform).

Page 48: Development of a Novel Lysophosphatidic Acid Activity Probe to

39

TBDPSO OO

HO

NHO

HNO

O

N3

TBDPSO OO

O

NHO

HNO

O

N3

POO

OPO

ON

m-CPBACH2Cl2

22

Compound 21 (100 mg, 0.124 mmol) was dissolved in dichloromethane (10 mL).

Dibenzyl diisopropylphosphoramidite (122 µL, 0.373 mmol) and 1H-tetrazole

(828 µL, 0.373 mmol) were added. The reaction was stirred at room temperature

under nitrogen for 24 h. Then the reaction was cooled to 0 °C and mCPBA (64

mg, 0.373 mmol) was added. The reaction was stirred for 1 h at room

temperature. After completion, the solvent was concentrated and the residue

was purified via chromatography on silica gel with a solvent gradient of 30–75%

ethyl acetate/hexanes and 100% ethyl acetate to yield 22 (14.1 mg, 50%).

1H NMR (300 MHz, CDCl3) δ 1.03 (s, 9H), 1.27 (m, 2H), 1.50 (m, 8H), 1.79 (m,

1H), 1.94 (m, 1H), 3.23 (m, 4H), 3.97 (m, 5H), 4.71 (m, 1H), 4.93 (s, 4H), 7.30

Page 49: Development of a Novel Lysophosphatidic Acid Activity Probe to

40

(m, 14H), 7.48 (m, 3H), 7.62 (m, 6H), 7.78 (m, 4H), 7.96 (m, 2H) 13C NMR (300

MHz CDCl3) δ 195.97, 173.68, 171.49, 171.48, 166.40, 140.36, 135.89, 133.21,

133.06, 130.98, 130.16, 128.68, 128.54, 127.965, 127.949, 127.859, 127.799,

127.28, 69.78, 69.65,, 69.49, 69.39, 67.74, 64.31, 60.50, 53.56, 51.19, 39.52,

33.79, 32.51, 29.12, 28.66, 28.79, 26.89, 26.39, 24.42, 22.81, 21.15, 19.36,

14.30, MALDI-HRMS [M + Na]+ calcd: 1088.4365, found 1088.4398 [α]296KD = -

1.16 (c 0.26, 1:4 methanol/chloroform).

23

Compound 22 (0.033 mmol) was dissolved in dichloromethane under nitrogen,

and trimethylsilyl bromide (309 µL) was added, and the reaction was run until

completion. After the disappearance of starting material, the solvent was

Page 50: Development of a Novel Lysophosphatidic Acid Activity Probe to

41

removed, and the residue was dissolved in methanol (5 mL). The reaction was

stirred at room temperature for 1 h. The methanol was then removed. After

residue solvent had been removed, the product was dissolved in 5.0 mL

tetrahydrofuran. Tetrabutylammonium fluoride trihydrate (0.327 mmol) was

added, and the reaction was allowed to run overnight. The next day the solvent

was removed, and the product was purified via a reversed-phase column with a

solvent gradient of 90–10% methanol to produce 23 (12.1 mg, 61%).

1H NMR (300 MHz, CDCl3) δ 1.05 (m, 9H), 1.29 (m, 7H), 1.65 (m, 1H), 1.95 (m,

1H), 2.33 (m, 1H), 3.35 (m, 4H), 3.68 (m, 1H), 4.12 (m, 1H), 7.39 (m, 2H), 7.59

(m, 1H), 7.73 (m,2H), 7.80 (m, 2H), 8.00 (m, 2H) 13C NMR (300 MHz, CDCl3)

196.44, 196.41, 173.86, 171.71,140.62, 137.25, 135.18, 133.53, 130.46, 130.45,

128.90, 127.86, 127.74, 63.96, 59.15, 53.80, 39.70, 34.43, 33.36, 32.54, 29.65,

29.14, 27.61, 24.85, 24.23, 20.07, 13.82, MALDI-HRMS [M+ Na]+ calcd:

691.2040, found: 691.1531

Page 51: Development of a Novel Lysophosphatidic Acid Activity Probe to

42

Page 52: Development of a Novel Lysophosphatidic Acid Activity Probe to

43

Page 53: Development of a Novel Lysophosphatidic Acid Activity Probe to

44

Page 54: Development of a Novel Lysophosphatidic Acid Activity Probe to

45

Page 55: Development of a Novel Lysophosphatidic Acid Activity Probe to

46

Page 56: Development of a Novel Lysophosphatidic Acid Activity Probe to

47

Page 57: Development of a Novel Lysophosphatidic Acid Activity Probe to

48

Page 58: Development of a Novel Lysophosphatidic Acid Activity Probe to

49

Page 59: Development of a Novel Lysophosphatidic Acid Activity Probe to

50

Page 60: Development of a Novel Lysophosphatidic Acid Activity Probe to

51

Page 61: Development of a Novel Lysophosphatidic Acid Activity Probe to

52

Page 62: Development of a Novel Lysophosphatidic Acid Activity Probe to

53

Page 63: Development of a Novel Lysophosphatidic Acid Activity Probe to

54

Page 64: Development of a Novel Lysophosphatidic Acid Activity Probe to

55

Page 65: Development of a Novel Lysophosphatidic Acid Activity Probe to

56

Page 66: Development of a Novel Lysophosphatidic Acid Activity Probe to

57

Page 67: Development of a Novel Lysophosphatidic Acid Activity Probe to

58

Page 68: Development of a Novel Lysophosphatidic Acid Activity Probe to

59

Page 69: Development of a Novel Lysophosphatidic Acid Activity Probe to

60

Page 70: Development of a Novel Lysophosphatidic Acid Activity Probe to

61

Page 71: Development of a Novel Lysophosphatidic Acid Activity Probe to

62

References

Page 72: Development of a Novel Lysophosphatidic Acid Activity Probe to

63

1. Gorter, E.; Grendel, F., On Biomolecular Layers of Lipoids on the Chromocytes of the Blood. J. Exp. Med. 1925, 41 (4), 439-443.

2. Ghosh, S.; Strum, J.; Bell, R., Lipid biochemistry: functions of glycerolipids and sphingolipids in cellular signaling. FASEB J. 1997, 11 (1), 45-50.

3. TAKAI, Y.; KISHIMOTO, A.; IWASA, Y.; KAWAHARA, Y.; MORI, T.; NISHIZUKA, Y.; TAMURA, A.; FUJII, T., A Role of Membranes in the Activation of a New Multifunctional Protein Kinase System. J Biochem 1979, 86 (2), 575-578.

4. Haucke, V.; Di Paolo, G., Lipids and lipid modifications in the regulation of membrane. Curr. Opin. Cell Biol. 2007, 19 (4), 426-435.

5. (a) Wonhwa, C.; Stahelin, R. V., Membrane-Protein Interactions in Cell Signaling and Membrane Trafficking. Annu. Rev. Biophys. Biomol. Struct. 2005, 34 (1), 119-C-2; (b) Sprong, H.; van der Sluijs, P.; van Meer, G., How proteins move lipids and lipids move proteins. Nat. Rev. Mol. Cell Biol. 2001, 2 (7), 504-513.

6. (a) Carroll, K.; Gomez, C.; Shapiro, L., Tubby proteins: the plot thickens. Nat. Rev. Mol. Cell Biol. 2004, 5 (1), 55-64; (b) De Camilli, P.; Chen, H.; Hyman, J.; Panepucci, E.; Bateman, A.; Brunger, A. T., The ENTH domain. FEBS Lett. 2002, 513 (1), 11-18; (c) Ferguson, K. M.; Kavran, J. M.; Sankaran, V. G.; Fournier, E.; Isakoff, S. J.; Skolnik, E. Y.; Lemmon, M. A., Structural Basis for Discrimination of 3-Phosphoinositides by Pleckstrin Homology Domains. Mol. Cell 2000, 6 (2), 373-384.

7. Im, D.-S., Discovery of new G protein-coupled receptors for lipid mediators. J. Lipid Res. 2004, 45 (3), 410-418.

8. Mills, G. B.; Moolenaar, W. H., The emerging role of lysophosphatidic acid in cancer. Nat. Rev. Cancer 2003, 3 (8), 582-591.

9. Moolenaar, W. H.; van Meeteren, L. A.; Giepmans, B. N. G., The ins and outs of lysophosphatidic acid signaling. BioEssays 2004, 26 (8), 870-881.

10. van Meeteren, L. A.; Moolenaar, W. H., Regulation and biological activities of the autotaxin-LPA axis. Prog. Lipid Res. 2007, 46 (2), 145-160.

11. Noguchi, K.; Herr, D.; Mutoh, T.; Chun, J., Lysophosphatidic acid (LPA) and its receptors. Curr. Opin. Pharmacol. 2009, 9 (1), 15-23.

12. (a) Kishi, Y.; Okudaira, S.; Tanaka, M.; Hama, K.; Shida, D.; Kitayama, J.; Yamori, T.; Aoki, J.; Fujimaki, T.; Arai, H., Autotaxin Is Overexpressed in Glioblastoma Multiforme and Contributes to Cell Motility of Glioblastoma by Converting Lysophosphatidylcholine TO Lysophosphatidic Acid. J. Biol. Chem. 2006, 281 (25), 17492-17500; (b) van Meeteren, L. A.; Ruurs, P.; Christodoulou, E.; Goding, J. W.; Takakusa, H.; Kikuchi, K.; Perrakis, A.; Nagano, T.; Moolenaar, W. H., Inhibition of Autotaxin by Lysophosphatidic Acid and Sphingosine 1-Phosphate. J. Biol. Chem. 2005, 280 (22), 21155-21161.

Page 73: Development of a Novel Lysophosphatidic Acid Activity Probe to

64

13. Fukushima, N.; Weiner, J. A.; Chun, J., Lysophosphatidic acid (LPA) is a novel extracellular regulator of cortical neuroblast morphology. Dev. Biol. 2000, 228 (1), 6-18.

14. (a) Bandoh, K.; Aoki, J.; Hosono, H.; Kobayashi, S.; Kobayashi, T.; Murakami-Murofushi, K.; Tsujimoto, M.; Arai, H.; Inoue, K., Molecular cloning and characterization of a novel human G-protein-coupled receptor, EDG7, for lysophosphatidic acid. J. Biol. Chem. 1999, 274 (39), 27776-27785; (b) Bandoh, K.; Aoki, J.; Taira, A.; Tsujimoto, M.; Arai, H.; Inoue, K., Lysophosphatidic acid (LPA) receptors of the EDG family are differentially activated by LPA species - Structure-activity relationship of cloned LPA receptors. FEBS Lett. 2000, 478 (1-2), 159-165.

15. (a) Murata, J.; Lee, H. Y.; Clair, T.; Krutzsch, H. C.; Arestad, A. A.; Sobel, M. E.; Liotta, L. A.; Stracke, M. L., cDNA cloning of the human tumor motility-stimulating protein, autotaxin, reveals a homology with phosphodiesterases. J. Biol. Chem. 1994, 269 (48), 30479-30484; (b) Nam, S. W.; Clair, T.; Kim, Y. S.; McMarlin, A.; Schiffmann, E.; Liotta, L. A.; Stracke, M. L., Autotaxin (NPP-2), a metastasis-enhancing motogen, is an angiogenic factor. Cancer Res. 2001, 61 (18), 6938-6944.

16. Stracke, M. L.; Clair, T.; Liotta, L. A. In Autotaxin, tumor motility-stimulating exophosphodiesterase, Weber, G., Ed. pp 135-144.

17. Valet, P.; Pagès, C.; Jeanneton, O.; Daviaud, D.; Barbe, P.; Record, M.; Saulnier-Blache, J. S.; Lafontan, M., Alpha2-adrenergic receptor-mediated release of lysophosphatidic acid by adipocytes. A paracrine signal for preadipocyte growth. J. Clin. Invest. 1998, 101 (7), 1431-1438.

18. Prestwich, G. D.; Gajewiak, J.; Zhang, H.; Xu, X.; Yang, G.; Serban, M., Phosphatase-resistant analogues of lysophosphatidic acid: Agonists promote healing, antagonists and autotaxin inhibitors treat cancer. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2008, 1781 (9), 588-594.

19. Xu, J.; Lai, Y.-J.; Lin, W.-C.; Lin, F.-T., TRIP6 Enhances Lysophosphatidic Acid-induced Cell Migration by Interacting with the Lysophosphatidic Acid 2 Receptor. J. Biol. Chem. 2004, 279 (11), 10459-10468.

20. Lin, F.-T.; Lai, Y.-J., Regulation of the LPA2 receptor signaling through the carboxyl-terminal tail-mediated protein-protein interactions. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2008, 1781 (9), 558-562.

21. Cravatt, B. F.; Wright, A. T.; Kozarich, J. W., Activity-Based Protein Profiling: From Enzyme Chemistry to Proteomic Chemistry. Annu. Rev. Biochem. 2008, 77 (1), 383-414.

22. (a) Uttamchandani, M.; Li, J. Q.; Sun, H.; Yao, S. Q., Activity-based protein profiling: New developments and directions in functional proteomics. ChemBioChem 2008, 9 (5), 667-675; (b) Evans, M. J.; Cravatt, B. F., Mechanism-Based Profiling of Enzyme Families. Chem. Rev. 2006, 106 (8), 3279-3301.

Page 74: Development of a Novel Lysophosphatidic Acid Activity Probe to

65

23. Cravatt, B. F.; Wright, A. T.; Kozarich, J. W., Activity-based protein profiling: From enzyme chemistry. Annu. Rev. Biochem. 2008, 77, 383-414.

24. (a) Perly, B.; Dufourc, E. J.; Jarrell, H. C., Facile and high yielding syntheses of phosphatidylcholines and phosphatidylethanolamines containing 2H-labeled acyl chains. J. Labelled Compd. Radiopharm. 1984, 21 (1), 1-13; (b) Rowland, M. M.; Best, M. D., Modular synthesis of bis(monoacylglycero)phosphate for convenient access to analogues bearing hydrocarbon and perdeuterated acyl chains of varying length. Tetrahedron 2009, 65 (34), 6844-6849.

25. Gong, D.; Bostic, H. E.; Smith, M. D.; Best, M. D., Synthesis of Modular Headgroup Conjugates Corresponding to All Seven Phosphatidylinositol Polyphosphate Isomers for Convenient Probe Generation. Eur. J. Org. Chem. 2009, 2009 (24), 4170-4179.

Page 75: Development of a Novel Lysophosphatidic Acid Activity Probe to

66

Vita

Leah M. Cuthriell was born in Opp, Alabama in 1984 and grew up in the

Flat Creek community near Samson, AL. She is the daughter of Peadon and

Barbara Cuthriell. In May of 2003, she graduated from Samson High School in

Samson, AL. After graduation, she attended Huntingdon College in Montgomery,

AL where she received a B.S. degree in chemistry in 2007. She completed a

Master of Science degree in chemistry in December 2009.