257
~Chapter 1~ Introduction 1.1. Biomarkers Biomarkers are a critical component of the drug discovery process and are important in disease diagnosis and treatment. They are so critical, in fact, that the National Institutes of Health formed an Initiative on Biomarkers and Surrogate End Points to define key terms and issues more precisely. According to the initiative committee, a biomarker is described as “a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention 1 .” Biomarkers serve as a bridge between the observed clinical responses of patients and the changes that occur on a molecular level. These can be but are not limited to levels of a metabolite or protein – also included are data such as body weight, organ weight, genotype and urine volume 2 . An ideal and robust biomarker should be noninvasive to sample, selective, sensitive, measured easily, relevant to the condition being studied and serve as an early warning for toxicity or disease 2 . Additionally, interanimal and intraanimal 1

~Chapter 1~ Introduction 1.1. Biomarkers

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: ~Chapter 1~ Introduction 1.1. Biomarkers

~Chapter 1~

Introduction

1.1. Biomarkers

Biomarkers are a critical component of the drug discovery process and are

important in disease diagnosis and treatment. They are so critical, in fact, that the

National Institutes of Health formed an Initiative on Biomarkers and Surrogate End

Points to define key terms and issues more precisely. According to the initiative

committee, a biomarker is described as “a characteristic that is objectively measured

and evaluated as an indicator of normal biological processes, pathogenic processes, or

pharmacologic responses to a therapeutic intervention1.” Biomarkers serve as a

bridge between the observed clinical responses of patients and the changes that occur

on a molecular level. These can be but are not limited to levels of a metabolite or

protein – also included are data such as body weight, organ weight, genotype and

urine volume2. An ideal and robust biomarker should be noninvasive to sample,

selective, sensitive, measured easily, relevant to the condition being studied and serve

as an early warning for toxicity or disease2. Additionally, interanimal and intraanimal

1

Page 2: ~Chapter 1~ Introduction 1.1. Biomarkers

variability should be low and it should change on a timescale congruent with drug

action or disease onset.

The measurement of biomarkers serves three primary purposes in drug

development2: to quantify the dose to which the animal or human was exposed

(biomarkers of exposure), to measure the pathological effect of the drug (biomarkers

of response) and to identify animals or humans that are predisposed to the effects of

the drug (biomarkers of susceptibility). One of the main bottlenecks in the drug

discovery process is the identification of drug candidates that cause toxicity.

Consequently, there is considerable interest from pharmaceutical firms in developing

technologies that increase efficiency in this phase of development3-5. Many

companies have been formed in recent years that solely exist to identify sets of

biomarkers most relevant to various drug candidates or disease conditions in hopes of

identifying a promising drug earlier, saving a vast amount of resources in the process.

Biomarkers are also utilized to study disease onset and treatment. The primary

purpose of biomarker identification in these studies is to discover compounds that can

diagnose a disease earlier than currently established biomarkers or clinical

endpoints3,6. For these diagnostic biomarkers, although a level of the biomarker is

obtained analytically, the value is then categorized as positive or negative. The place

where the line is drawn between positive and negative is extremely important – while

the level should not be low enough that a significant number of false positives results,

it should not be high enough that a large number of false negatives are obtained and

thus the disease remains untreated3.

2

Page 3: ~Chapter 1~ Introduction 1.1. Biomarkers

Traditionally, scientists have searched for a single all-encompassing

biomarker to use in the study of a pathological condition. The use of single

biomarkers can be problematic. A single biomarker likely does not reflect all the

changes induced by the drug or disease, so the information gleaned from single

biomarker assays is incomplete1. For example, many treatments are likely to lower

levels of prostate specific antigen (PSA), but these effects may be unrelated to the

success of the treatment of prostate cancer. Other pathways unrelated to PSA are

tapped by cancer treatment. Biomarker qualification is also complicated by animal-

to-animal variability and differences in response between species5.

Consequently, there is a push in the scientific community to move to profiling

experiments, so that many biomarkers of a condition can be analyzed simultaneously.

It is hypothesized that an increased number of biomarkers increases the probability of

accurately identifying a promising drug candidate or diagnosing a disease. The “-

omics” technologies (genomics, proteomics, and metabonomics) have opened the

door to allow for the simultaneous study of multiple biomarkers. A considerable

amount of research is currently being conducted for gene, protein or metabolite

combinations that collectively serve as more selective and sensitive biomarkers.

While promising, this approach is not without limitations. With an increased number

of variables comes an increased probability that chance (and thus misleading)

correlations will be identified3,7. Even though “-omics” techniques allow for many

biomarkers to be characterized, it is important to ensure that these additional analytes

add value to the study and thus are worth any extra effort or expense2,7. Finally,

3

Page 4: ~Chapter 1~ Introduction 1.1. Biomarkers

profiling techniques do not require that analytes be identified – what matters is their

statistical significance relative to the drug dose or disease onset. This can be

advantageous in that it is more efficient, but can also be disadvantageous. Although it

is not necessarily required that the identity of a biomarker be known or that its exact

role in the changes induced by a drug or disease are fully characterized, this

information does help in the discernment of biomarkers that are most relevant to the

study and thus most likely to give accurate information3,7.

1.2. Metabonomics

1.2.1. History and Context

Although the term “metabonomics” was coined within the last ten years,

scientists have been profiling metabolites in biofluids and tissues for decades. Some

of the earliest studies were in 1965, involving the investigation of energy metabolites

in cancerous tumors8 and livers9. These studies were achieved by individual assays of

the metabolites, requiring a great deal of work per sample. Studies progressed to the

use of gas chromatography-mass spectrometry (GC-MS) and gas chromatography-

flame ionization detection (GC-FID) to analyze organic acids, steroids and other

volatiles in urine10-14 and breath12. Some of the first applications of nuclear magnetic

resonance (NMR) spectroscopy to the study of small endogenous metabolite profiles

in biofluids were in the late 1970s and early 1980s by Dallas Rabenstein15,16, who

examined erythrocyte metabolism. This was followed by work in the mid-1980s by J.

K. Nicholson and P. J. Sadler17. Over the years, a variety of applications have

4

Page 5: ~Chapter 1~ Introduction 1.1. Biomarkers

emerged for metabonomics studies, centered on the identification of biomarkers

(particularly of liver toxicity) and improved detection and diagnosis of disease.

Metabolic profiling of urine by NMR has been used clinically to detect inborn errors

of metabolism in urine18,19. Dietary studies in humans have also found their

application in metabonomics20. Metabolic disturbances following conditions such as

tissue injury21, liver toxicity22-26, liver disease27, liver cancer28 and kidney toxicity29

have all been explored. Disease diagnosis and biomarkers of Type 2 diabetes30,

neurodegenerative disorders31,32, meningitis33, schizophrenia34 and ovarian cancer35

have been characterized. Metabonomics has captured the interest of pharmaceutical

companies interested in expediting the drug development process. In 2001, a

consortium of six major pharmaceutical companies was formed to study the

feasibility of metabonomics methodology in the characterization of toxicity of drug

candidates and to build metabolic profile databases of biofluid samples36.

Metabonomics is based on the principal of a stressor, be it genetic, disease,

pharmaceutical or environmental, causing changes in the levels of endogenous

metabolites. Cells work to maintain homeostasis, so the application of a metabolic

stressor results in changes of intracellular and extracellular metabolite levels to

compensate for the stressor37. The study of endogenous metabolite profiles has many

different terms associated with it. The term “metabonomics” was coined in 1999. It

is defined as “the quantitative measurement of the dynamic multiparametric

metabolic response of living systems to pathophysiological stimuli or genetic

modification38.” The metabolome is defined as the complete set of endogenous small

5

Page 6: ~Chapter 1~ Introduction 1.1. Biomarkers

6

molecules produced by an organism39. Metabolomics is defined as the identification

and quantification of the entire metabolome, to the greatest extent possible39. In

contrast, metabonomics is not concerned with characterizing the entire metabolome,

although it is often used interchangeably in literature with metabolomics. It trades off

the complete characterization of the metabolome with the ability to monitor many

samples over a time course. Historically, metabolomics has primarily centered on the

study of plant or bacterial metabolomes by GC/MS and metabonomics has focused on

the study of animal metabolism by NMR analysis of biofluids.

Metabonomics is part of systems biology, which is a holistic technique that

aims to characterize an organism’s response to a stimulus at the genetic, protein and

metabolite level (Figure 1.1). The measurement of metabolite levels provides an

important insight into both gene expression and protein levels or activity40-42, as the

endpoint of these changes is ultimately in the levels of small molecules. It should be

noted that a stimulus does not necessarily affect change “top down” – that is, starting

at the genetic level. In fact, many stimuli affect change directly at the metabolite

level that is never observed at the genetic or protein level43. Genes and proteins also

respond to stimuli at different timescales than metabolites. Thus, metabolite levels

may reflect a more immediate change from the stressor44.

In fact, genomic, transcriptomic and proteomic studies face limitations that

metabonomics studies do not. Both can be expensive – transcriptomic studies are at

least 100 times more expensive than the same number of metabonomic studies41, and

proteomic studies are at least 50 times more expensive45. Gene and protein

Page 7: ~Chapter 1~ Introduction 1.1. Biomarkers

7

Figure 1.1. The study of systems biology, showing the layers of complexity of biological response of a cell to an external stimulus.

Gene (Genomics)

mRNA (Transcriptomics)

Protein (Proteomics)

Metabolite (Metabonomics)

Gene (Genomics)

mRNA (Transcriptomics)

Protein (Proteomics)

Metabolite (Metabonomics)

Page 8: ~Chapter 1~ Introduction 1.1. Biomarkers

expression among species is not as highly conserved as metabolic pathways45.

Transcriptomic studies can be incomplete, as not all genes are transcribed into

mRNA. Therefore, changes in some genes may not be detected by transciptomic

analyses (called “silent mutations”) that are detected by metabonomics41,46.

Additionally, metabonomics studies are typically much simpler to perform45,46.

1.2.2. Sampling Approaches

Sampling is one of the most important considerations for a metabonomics

experiment. The mode of sampling must balance the needs of the experiment with

throughput and the effects sampling will have on the subject (animal or human). For

example, while tissue-specific metabolic information may be desirable, the

throughput of these experiments is notably diminished from the sampling of biofluids,

particularly urine. There are many options available to the researcher for

metabonomics studies. Virtually any biofluid can be studied, as well as tissue

biopsies and extracellular fluid.

1.2.2.1. Blood

Plasma is a popular choice for metabonomics studies, but is considerably

more challenging methodologically than urine, both for sampling and detection.

Plasma sampling is somewhat invasive, but gives a good picture of the organism’s

overall metabolic status. Particularly in the sampling of small animals such as rats or

mice, the volume of blood drawn at a single time is limited to a few hundred

microliters over a set time period. Sampling may also perturb the person’s or

animal’s metabolism through stress experienced from the sampling itself, depending

8

Page 9: ~Chapter 1~ Introduction 1.1. Biomarkers

on the method used to draw the blood. Thus, a high temporal resolution of less than

1-2 hours, which may be desirable for some studies, is extremely difficult to achieve

with plasma sampling. Analysis of plasma by NMR (and MS) is complicated by the

proteins present in the sample. Some choose to precipitate the proteins to clean up

the sample, but this does not completely eliminate the problem. Additionally,

extraction procedures may alter the observed metabolic profile of the biofluid.

Protein precipitation is not always necessary for NMR experiments, although it is in

LC/MS experiments. Specialized NMR pulse sequences are also used to suppress the

signal from macromolecules in solutions such as plasma by capitalizing on the

differences in diffusion and spin relaxation time47-49. A final consideration is the

stability of plasma samples. The stability of plasma samples can cause problems in

analysis, mostly because of the enzymes present in the sample. Although the samples

can be frozen during storage to a point where enzyme activity is minimal, after

thawing for analysis, sample composition can begin to change.

1.2.2.2. Urine

Urine sampling is the most common sampling method for metabonomics

studies. Studies with urine samples tend to focus on metabolic changes in the liver,

kidney and intestinal microflora50, but this biofluid is still considered to reflect the

overall metabolic status of the organism. It is the least invasive biofluid to collect, as

it does not involve any needles. Animal studies can be performed in metabolism

cages, which contain a collection chamber underneath the mesh floor of the cage,

allowing the animal to be disturbed as little as possible. In human studies, urine

9

Page 10: ~Chapter 1~ Introduction 1.1. Biomarkers

sampling has good compliance with test subjects and does not require the assistance

of a medical professional. Time resolution in urine sampling is limited by the length

of time between voids, which is difficult to control or keep constant. Most studies

collect samples continuously for a set length of time, with a typical time resolution of

~8 hours. For some metabolic stressors studied, this may not be sufficient. Urine can

range in pH from 5 to 8, but the pH can increase with bacterial contamination51.

Urine samples are more stable than plasma, as no enzymatic degradation occurs as

long as no bacteria are present. Thus, samples require an antibacterial agent (usually

sodium azide) to prevent bacterial growth. Bacterial growth can cause changes in the

metabolic profile, particularly in the levels of acetate, formate and ethanol52.

1.2.2.3. Other Biofluids

Many other biofluids have been studied by metabonomics, including

cerebrospinal fluid33,34,53-55, seminal fluid56,57, saliva58 and amniotic fluid59,60.

Metabonomics analysis of cerebrospinal fluid has been used to study important

conditions such as dementia53, meningitis33, schizophrenia34, aneurysms54 and spinal

problems such as herniated disks55. Seminal fluid studies have focused on

characterization of male contraceptives56 and prostate cancer57.

Many of these fluids give information on metabolism in a more localized area

than urine or plasma, but are more difficult and invasive to sample. Thus, samples

from human subjects are limited. Samples from laboratory rats (a commonly used

animal in metabonomics studies) are limited in volume (a few microliters, in some

cases) and for some fluids are so invasive to sample that only one time point per rat

10

Page 11: ~Chapter 1~ Introduction 1.1. Biomarkers

can typically be obtained. Additionally, time resolution may be inadequate because

of the limited volumes and invasiveness of sampling, depending on the condition

being studied. As a result of these limitations, studies of these biofluids are less

popular, and many metabonomics studies on tissue metabolism make use of tissue

biopsy samples instead.

1.2.2.4. Tissue Biopsy

The metabonomics community has a great interest in obtaining tissue-specific

metabolic information. To date, the primary sampling method for this is through

tissue biopsy. Tissues biopsied have included kidney, liver, brain and prostate61.

Although interest has centered on these tissues, it is possible to profile virtually any

tissue. Once surgically excised, the biopsy either undergoes an extraction procedure

prior to analysis or is directly analyzed by a specialized NMR magic angle spinning

(MAS) probe. (This will be discussed further in Section 1.3.2.) Both approaches

have been utilized in metabonomics research. Tissue extract NMR has been applied

to the study of traumatic brain injury21 and Batten disease, a genetic

neurodegenerative disorder62. MAS-NMR has been applied to the study of prostate

tumors63, neurodegenerative disorders32, liver toxicity26,64,65 and renal toxicity65.

Although tissue biopsy does give the more specific metabolic information,

there are limitations with this sampling approach. First, only one time point can be

taken per animal. This substantially increases the number of animals resulting in

greater animal-to-animal variability. It could also limit the time resolution obtained

for experiments by requiring a compromise to be made between number of animals

11

Page 12: ~Chapter 1~ Introduction 1.1. Biomarkers

used and time resolution required. Tissue extraction creates selectivity in the

experiment, as not all metabolites may be present in the extract sample depending on

the eluent used. Third, macromolecule interference during analysis is also a problem,

much like plasma samples, as discussed in Section 1.2.2.1.

1.2.2.5. Microdialysis

1.2.2.5.1. History and Basic Concepts

Microdialysis has been in use for decades as a sampling technique to probe

metabolic changes in tissue extracellular fluid66. Probes with a semipermeable

membrane are surgically implanted into the tissue of interest. The probe is perfused

with a solution isotonic to the surrounding tissue, so that there is no net fluid flow

across the membrane. The perfusion of solution through the membrane creates a

concentration gradient between the lumen of the membrane and the surrounding

extracellular fluid. Consequently, molecules below the molecular weight cutoff

(MWCO) of the membrane diffuse into the membrane lumen in accordance with their

concentration gradient. Molecules above the MWCO cannot diffuse across the

membrane and are thus not sampled, making dialysate samples clean and fairly stable

(with regard to enzymatic sample degradation) relative to biofluids such as plasma.

Microdialysis sampling offers several unique advantages for in vivo

experiments. One of the primary advantages lies in the ability to continuously obtain

samples from the same tissue. Many in vivo studies of tissues require the researcher

to sacrifice multiple animals per time point studied to harvest the tissue. Using

microdialysis reduces the number of animals required for experimentation

12

Page 13: ~Chapter 1~ Introduction 1.1. Biomarkers

substantially and reduces the animal-to-animal variability observed in experimental

results. The clean samples tend to make analysis much easier, as sample cleanup is

minimal and any interferences from macromolecules (such as matrix effects in MS)

are virtually eliminated. Also, only free molecules are sampled – those that are

protein-bound are not observed in the sample. For pharmacokinetic studies, this can

be advantageous because only the active drug is analyzed in the samples. A careful

balance must be achieved in these experiments between sample volume, perfusion

rate and time resolution67. The analytical method employed will require a minimum

sample volume, which will place constraints on the achievable time resolution. Time

resolution can be improved by increasing the perfusion rate, but this lowers the

recovery of the compound to perhaps undetectable levels.

Much of the work of microdialysis has centered on the brain and sampling of

neurotransmitters. In fact, the first microdialysis experiments were performed in the

brain in 1974 by Ungerstedt68. Since this time, the brain still remains the most

studied tissue by a substantial margin. However, peripheral tissues have also been

studied. To date, a wide variety of tissue types in mammalian organisms have been

sampled by microdialysis, including the muscle, adipose tissue, heart, lung, liver,

kidney, pancreas, eye, placenta and gastrointestinal tract69. Neurochemical and

pharmacokinetic studies are particularly popular for microdialysis experiments, but

many other applications where tissue-specific information is desirable or site-specific

delivery of a compound is needed have been explored.

13

Page 14: ~Chapter 1~ Introduction 1.1. Biomarkers

1.2.2.5.2. Probe Designs

Microdialysis probes require different geometries to achieve successful

implantation and sampling in tissues67,70,71. Spatial resolution must be adequate to

sample from the region of interest. While organs such as the liver are fairly

homogenous, others are heterogeneous, such as the brain. Membrane length and

probe implantation technique must be adjusted accordingly. Other considerations for

probe design include anticipated movement of the rat – awake animals require tubing

long enough to tunnel under the skin and probe implantation must be secure enough

to avoid shifting of the membrane from rat movement. Additionally, the chemical

properties of the analyte must be considered. Hydrophobic analytes can experience

problems with sticking to probe components, particularly the dialysis membrane.

This can lower analyte recovery and introduce variability into the probe performance.

The differing geometries of tissues, their functions and their location in an

organism place differing requirements on microdialysis probe geometries. A

microdialysis probe is composed of the dialysis membrane, which is often actual

membrane used for kidney dialysis patients, tubing and an adapter that allows the

probe to be attached to a syringe for perfusion. The membrane is the most important

component of the probe. Its composition is chosen based on the analytes of interest in

the experiment, as membrane polymers exhibit differing surface chemistries.

Hydrophobic analytes in particular can be difficult to sample quantitatively by

microdialysis, because they tend to stick to the membrane and other probe

components. Commonly used membrane polymers include polyacrylonitrile (PAN),

14

Page 15: ~Chapter 1~ Introduction 1.1. Biomarkers

15

polycarbonate, polyarylethersulfone, regenerated cellulose and cellulose acetate.

Studies have been performed to compare membrane recovery properties – in vitro

results show distinct differences but these differences become much less significant

for in vivo studies72. Most of these membranes have MWCOs ranging from 10-40

kDa. In recent years, a push has been made to increase this MWCO to allow for

localized sampling of proteins to 100-300 kDa73,74. The use of these probes is still

being explored.

Some tissues, such as the brain, are difficult to access and undesirable to

expose. In locations such as this, the concentric cannula style probe is used (Figure

1.2A). This probe design has concentric inlet and outlet tubing on the same side of

the probe, with the membrane on the opposite side. The perfusate flows through the

inner tubing, into the membrane lumen and then is pushed back through the outer

tubing for sample collection. These probes can be implanted through a single point.

Also, these probes tend to be rigid, as they are encased in a stainless steel cannula,

which is advantageous for brain microdialysis experiments but is problematic for

such applications as blood microdialysis in vessels. The rigidity can cause tissue

damage or puncture vessel walls if the tissue moves when the animal moves or can

rupture the membrane, so this probe style is limited to studies in the brain, where the

probe can be secured to the skull. As an alternative to the rigid cannula, our

laboratory designed a flexible cannula-style probe (Figure 1.2C)75 that can be used in

a variety of peripheral tissues and is particularly useful for vessel implantation.

Page 16: ~Chapter 1~ Introduction 1.1. Biomarkers

Figure 1.2. Probe designs for microdialysis sampling. A shows a commercial concentric rigid cannula probe suitable for brain microdialysis, B shows linear probes used in the study of peripheral tissues and C shows a flexible cannula probe used for microdialysis in blood vessels.

A

C

B

A

C

B

16

Page 17: ~Chapter 1~ Introduction 1.1. Biomarkers

Microdialysis in peripheral tissues can be performed with linear probes

(Figure 1.2B), in which the inlet and outlet tubing are on opposite sides of the

membrane. Flow is unidirectional through the probe. The first microdialysis studies

were performed with linear probes implanted transcerebrally68. Since this study,

linear probes have expanded the scope of microdialysis studies to numerous

peripheral tissues, including liver76-78, muscle79-82, stomach83,84, skin80,85-88, heart77,89

and tumors79,90. These probes cause the least amount of damage to the tissue during

implantation of all the microdialysis probe geometries. Also, because they are used

in more homogenous tissues and high spatial resolution is not necessary, the probes

can typically use longer membrane lengths to achieve greater recovery70.

One final probe design that is not commonly used is the shunt probe, which

was designed by our laboratory to sample dialysate from the bile duct91.

Conventional studies of bile involve cannulation of the duct and removal of bile over

the course of the experiment. Rats, the most common animal for this study, have low

amounts of bile, so direct sampling depletes the biofluid substantially, changing the

system being studied. The shunt probe is implanted in the bile duct. The design of

the probe essentially allows for a linear probe to be implanted in the duct while bile

flow is maintained.

1.2.2.5.3. Probe Calibration

Transport across the microdialysis membrane is bidirectional – probes can be

used to both recover compounds from the tissue and deliver compounds to a localized

area (around the implant site). During sampling, microdialysis does not reach a

17

Page 18: ~Chapter 1~ Introduction 1.1. Biomarkers

steady state in the tissues due to the continuous flow of the perfusate. Consequently,

only a fraction of the compound will be delivered or recovered, which is called the

extraction efficiency (EE). EE can be changed by the perfusion rate, membrane and

analyte chemical properties, temperature, probe geometry, active membrane length

and physiological properties such as blood flow70,71,92. The general equation for EE is

as follows93:

EECC

CC

tissueperfusate

dialysateperfusate =−

− (1)

In this equation, Cperfusate represents the analyte concentration entering the probe,

Cdialysate represents the analyte concentration leaving the probe and Ctissue represents

the analyte concentration already present in the tissue.

In order to quantify the analyte in dialysate samples, it is necessary to

determine the probe EE for each experiment in vivo and often, over the course of the

experiment. There are multiple methods available to calibrate probes, which can be

chosen to best suit the needs of the experiment. The no net flux (NNF) method94

involves perfusing the probe with different analyte concentrations, and the analyte

concentration is measured in the dialysate. By plotting the concentration in the

perfusate against the difference in concentration between the perfusate and dialysate,

two pieces of information can be obtained. First, the x intercept shows the

concentration of the compound in the extracellular fluid and second, the slope of the

line gives the recovery for the analyte. Retrodialysis95,96 is based on the principal

that the percentage of analyte lost during delivery is equivalent to the percentage of

18

Page 19: ~Chapter 1~ Introduction 1.1. Biomarkers

analyte recovered from the extracellular fluid. For this calibration, the analyte is

perfused through the probe prior to experimentation and the % delivery of the analyte

is determined, which can then be used to correct for analyte concentrations observed

during the actual experiment. For the studies presented in this dissertation, exact

quantitation of each analyte is not possible due to the large number of analytes

present and lack of prior identification. Also, the data presented examines changes in

metabolite levels, rather than determining exact metabolite concentrations, so

absolute quantitation is not necessary. Therefore, the delivery or internal standard

method71,97 will be used to monitor changes in probe performance. In this method, a

marker compound is chosen with similar chemistry to the analyte. The % delivery of

this standard compound is used to gauge general changes in probe extraction

efficiency over the course of the experiment.

1.2.2.5.4. Tissue Response to Probe Implantation

Membrane choice is important when considering the tissue reaction to the

probe implant. Studies have shown both PAN and polycarbonate experience the

same degree of biofouling over an eight day period98. For long-term awake animal

experiments, cellulose acetate is inappropriate because it dissolves over a 2-3 week

period in the tissue99. Cuprophan is a better choice for these studies, as the membrane

integrity is maintained for at least 12 weeks in vivo99.

Immune reactions are unavoidable with microdialysis probe implantations,

particularly those implanted for extended periods of time. These reactions have been

well-characterized in the literature and tend to be consistent between tissue

19

Page 20: ~Chapter 1~ Introduction 1.1. Biomarkers

types67,76,100. After implantation, a small amount of hemorrhaging occurs around the

implantation site from damage incurred, but good contact is still maintained between

the tissue and membrane. Inflammation is the initial immune response of the tissue to

the foreign body, occurring three to four hours after implantation. Blood flow

increases at the implantation site and neutrophils infiltrate, surrounding the probe.

Eight hours after implantation, neutrophil infiltration continues to increase and some

necrosis is evident. After 1-3 days, macrophages infiltrate and begin to remove the

necrotic tissue and any bacteria present. For extended implants (1 week or more), the

tissue eventually begins to generate a capsule of connective tissue around the probe to

isolate it from the rest of the tissue. Angiogenesis also occurs with the fibrous

capsule.

1.2.2.5.5. Application to Metabonomics Studies

Very few microdialysis experiments have been studied by metabonomics.

These have primarily been proof of concept studies examining neurotoxicity induced

by tetradotoxin and liver toxicity induced by potassium and paracetamol77,78,101-103.

Our laboratory is the first to explore and refine the analytical methodology required

for these tissue-targeted metabonomics studies.

1.3. Analytical Methods for Metabonomics

Metabonomics is extremely demanding on the analytical method. The desire

is for nonselective detection in a very complex sample with a wide range of analyte

concentrations. Analytical methods must achieve a balance between the degree of

20

Page 21: ~Chapter 1~ Introduction 1.1. Biomarkers

21

nonselectivity and detection limits. Increasing selectivity improves detection limits,

but narrows the scope of the compounds detected. Most metabonomics methods aim

for nonselectivity while sacrificing detection limits – consequently, most of the

compounds profiled are energy compounds, which are typically highest in abundance.

Because of the large number of samples, the method should require little sample

preparation and be high throughput with respect to analysis time. Analytically, the

method should be robust and have a wide dynamic range. The complexity of the

composition of a biofluid sample is illustrated in Figure 1.3, which depicts NMR

detection of rat urine.

As discussed in the following sections, different analytical techniques bring

different advantages to the metabolic profiling of biofluid samples. Although using

multiple analytical techniques would give a broader picture of the metabolites in the

sample, typically only one is employed to achieve higher throughput. Unlike

metabolomics, metabonomics is more concerned with obtaining a snapshot of the

detectable metabolites in a sample and observing how these change in response to a

metabolic stressor.

1.3.1. Nuclear Magnetic Resonance (NMR) Spectroscopy

Proton NMR (1H-NMR) spectroscopy is by far the most popular analytical

method for metabonomics studies. It is based on the property of nuclear spin. Some

nuclei, including 1H, 2H, 13C and 31P, have nuclear spin and are thus NMR active.

The spinning nucleus generates a magnetic moment, which (in the simplest case)

aligns in one of two directions when placed in an external magnetic field (Bo). This

Page 22: ~Chapter 1~ Introduction 1.1. Biomarkers

22

Figure 1.3. 1H-NMR spectrum of rat urine in 10% D2O (A). B and C show close-ups of sections of the original spectrum in A.

9 8 7 6 5 4 3 2 1 0

4.0 3.5 3.0 2.5 2.0 1.5 1.0

A

B

C

9 8 7 6 5 4 3 2 1 09 8 7 6 5 4 3 2 1 09 8 7 6 5 4 3 2 1 0

4.0 3.5 3.0 2.5 2.0 1.5 1.04.0 3.5 3.0 2.5 2.0 1.5 1.04.0 3.5 3.0 2.5 2.0 1.5 1.0

8.5 8.0 7.5 7.0 6.58.5 8.0 7.5 7.0 6.5

A

B

C

8.5 8.0 7.5 7.0 6.58.5 8.0 7.5 7.0 6.5

Page 23: ~Chapter 1~ Introduction 1.1. Biomarkers

results in two populations of nuclei in different energy states – one state aligned

parallel to Bo (higher energy) and one aligned antiparallel to Bo (lower energy). In

the NMR experiment, a pulse of energy is applied at the resonant frequency of the

nuclei in the form of radio waves, causing nuclei to change spin states after

absorption of the energy. (The resonant frequency of the nucleus is its Larmor

frequency, which is the rate that the nucleus precesses about the Bo axis.) The

difference in spin state population is what generates the NMR signal. This is

relatively small in NMR compared to other types of spectroscopy, causing NMR’s

known problems with sensitivity. The change in spin state generates a signal at a

specific frequency that is related to the chemical environment around the proton of

interest. Neighboring protons can also cause splitting patterns in the proton

resonance signal. These two factors combined make it possible to learn a great deal

about the structure of the compound of interest. NMR detection is inherently

quantitative, as all protons will generate the same signal/nucleus, so only one known

compound is required to obtain quantitative information about all the analytes

detected, providing that the spectrum is acquired with suitable care.

1H-NMR is one of the most nonselective analytical techniques available and is

uniquely suited to measurements of endogenous metabolites, which generally contain

protons. The proton has nearly the highest sensitivity, generating the most

signal/nucleus of all NMR-active nuclei, except for tritium. No sample derivatization

or a priori decisions are required for analyte detection. Additionally, the sample is

not destroyed during analysis. However, 1H-NMR suffers from poor sensitivity, and

23

Page 24: ~Chapter 1~ Introduction 1.1. Biomarkers

is generally able to detect approximately a 10 μM concentration (depending on

experimental conditions and amount of signal averaging). Some limitations in

sensitivity can be overcome using cryoprobes, which are specialized NMR probes

designed to reduce thermal or Johnson noise through the cooling of probe

components. Although these improve signal-to-noise and reduce the required

acquisition time, they are very expensive to purchase and maintain. Resonance

overlap can also cause problems, reducing the number of analytes detected and

causing quantitation problems. Moving to higher field magnets improves signal

dispersion, and as a result generates many more resonances. In a single transient

from NMR analysis on a urine sample on a high-field magnet (800 MHz), 5000 lines

can be resolved52.

Sample preparation is generally simple52. All samples should be centrifuged

to pellet any particulates. Urine samples are generally diluted by D2O to a 10%

deuterium concentration. The D2O provides a lock signal for the NMR experiment,

and an internal standard such as trimethylsilyl propionic acid (TSP) is often included

as a chemical shift and quantitation standard. Sometimes buffer is used as well in the

dilution to control the pH and thus reduce resonance shifting. Plasma sample

preparation is also fairly straightforward. Although buffer is generally not added,

samples are diluted in saline and then D2O is added for a lock signal. Some

researchers choose to further clean up the sample by precipitating the protein, but this

step is generally eliminated because of pulse sequences like the Carr-Purcell-

Meiboom-Gill (CPMG) sequence that can suppress the macromolecule signal.

24

Page 25: ~Chapter 1~ Introduction 1.1. Biomarkers

One of the main obstacles to be overcome in the NMR experiment is the large

water signal from the aqueous samples, which causes resonance overlap, baseline

distortion, radiation damping and dynamic range problems52,104,105. In aqueous

samples, water occurs at a concentration of 55 M. This is much greater than the

analytes of interest, which are in the micromolar to millimolar range48,106. The water

signal can be reduced by removing the aqueous solvent through speed vacuuming or

lyophilization and reconstituting in a deuterated solution, but even with this, some

signal will remain. Water suppression pulse sequences are generally utilized to

reduce the intensity of the water resonance. A variety of pulse sequences are

available that achieve suppression from different mechanisms, including saturation of

the resonance, pulsed field gradient resonance destruction and resonance zero net

excitation105,106. Saturation of the water resonance is one of the simplest ways to

reduce water signal. It is accomplished by applying a long, low intensity pulse at the

frequency of the water signal, equalizing or saturating the spin populations so that

there is no net spin population to detect. This approach can cause saturation of

exchangeable protons and can deplete signal around the HOD resonance105. The

second approach uses pulsed field gradients or soft pulses. These sequences, such as

WATERGATE and WET, cause zero net inversion and dephasing of the water

resonance, destroying its signal while solute signal is maintained. We routinely use

WET on samples reconstituted in a deuterated solution. Also, the experiments are

typically shorter in length and show few effects on the solute resonances104. Finally,

water signal can be suppressed by manipulation of the water resonance to place it out

25

Page 26: ~Chapter 1~ Introduction 1.1. Biomarkers

of the axis of detection (z axis) while placing solute signal in the xy plane for

detection. These sequences have a lower efficiency of suppression of the water

signal104.

1H-NMR in metabonomics studies has faced challenges in the analysis of low

volume biofluid and microdialysis samples. Two primary choices are available to

reduce the volume requirement for these analyses – specialized NMR tubes or

specialized NMR probes. Typical 5 mm NMR tubes require a few hundred

microliters of sample. This volume requirement can be reduced by using smaller

NMR tubes, which can go down to 1 mm or by using Shigemi tubes, which reduce

sample volume requirements by only requiring enough sample to fill the active region

of the probe. Specialized NMR probes, such as nanoprobes or capillary probes107-109

can also be used. These probes have a flow cell volume of a few microliters, with an

observe volume of 1-2 μL or less. These probes can be advantageous for the analysis

of these samples, but do require access to an NMR magnet either dedicated to

analysis with the capillary probe or that allows for routine probe changes.

Solid samples are also challenging for NMR analysis, because of restricted

analyte motion in the sample matrix and heterogeneity within the sample, causing

resonance broadening106. Most solid tissue samples undergo tissue extraction

procedures prior to NMR analysis, but these procedures are time-consuming and

introduce selectivity, thus reducing the scope of metabolites that can be profiled. An

increasingly popular alternative to tissue extraction is magic angle spinning (MAS)

NMR26,63-65,110-113, which directly analyzes small molecules in solid tissue samples.

26

Page 27: ~Chapter 1~ Introduction 1.1. Biomarkers

The tissue sample is inserted into a small rotor, which is spun in the core of the

magnet (~4-6 KHz spin rate) at the magic angle of 54.78°, so that molecular motion is

similar to solution phase. Spinning at this angle reduces the line broadening observed

in these solid samples, improving resolution and signal.

1.3.2. Mass Spectrometry (MS)

Next to NMR, most metabonomics studies are performed by LC/MS. Most

metabolomic studies, which focus on quantitation of the entire metabolome, have

been performed on the GC/MS, showing good sensitivity and resolution. However,

GC/MS experiments have been limited in metabonomic studies114,115, which focus on

the analysis of multiple samples to determine an organism’s response to a metabolic

stimulus. The throughput of GC/MS in these studies is low due to the need for

derivatization of polar nonvolatile analytes such as amino acids and sugars, which

comprise a large portion of the metabolome52,114.

Therefore, LC/MS has been much more popular for metabonomics studies.

This work was pioneered by the lab of Robert Plumb at Waters Corporation, with

initial studies focusing on the detection of metabolites in urine samples using LC-

ESI-MS62,116. Sample preparation is much less involved than with GC/MS

methodology – most samples can be either directly injected or undergo a short protein

precipitation procedure. Throughput is lower than NMR, as experiment times can be

upwards of 1 hour. The MS detection itself has improved detection limits compared

with NMR (provided the molecule ionizes efficiently) and has a good dynamic range.

The use of smaller diameter LC columns (1-2 mm) has improved resolution and

27

Page 28: ~Chapter 1~ Introduction 1.1. Biomarkers

lowered sample volume requirements. Others have explored increasing the resolution

through the use of ultra-performance LC (UPLC)117, or the use of columns with

smaller particle sizes. However, differences in ionization efficiencies between

compounds introduce selectivity into this detection mode. Also, changes in

ionization efficiency can occur during the actual MS experiment, when coeluting

compounds affect the ionization of the other compounds118. Analysis is generally

performed using a gradient LC system with a reversed phase column and ionization is

accomplished by electrospray in positive and negative mode. Those developing

analytical methodology focus on increasing peak capacity. A peak capacity of 1500

has been achieved, but at the expense of throughput – this separation took

approximately 20 hours119! Improved resolution has also been explored through new

column stationary phases, including monolithic120 and hydrophilic interaction

chromatography (HILIC) phases121.

While MS detection with separation does allow for lower detection limits,

several new issues are introduced by this analytical technique. First, the technique

itself is more selective – different classes of compounds will be detected depending

on the column, mobile phase, ionization technique, mass analyzer and other

conditions. Second, quantitation is difficult in mass spectrometry studies. One of the

primary ideas behind metabonomics is no a priori identification of metabolites of

interest. However, the best quantitation in mass spectrometry studies comes using

deuterated standards of analytes. Not only is this difficult to do in metabonomics

studies, it is impractical based on the number of metabolites detected in a single

28

Page 29: ~Chapter 1~ Introduction 1.1. Biomarkers

experiment. Additionally, drift in retention times can make comparison between runs

difficult122.

A small number of studies have also been performed by CE/MS123-127. Some

applications include characterization of enzyme activity123 These dramatically reduce

sample volume requirements, which could potentially improve time resolution or

allow for the concentration of samples. CE/MS also has the potential to give high

resolution separations. Separate run conditions need to be employed for anionic and

cationic analytes. In the initial study, 1692 metabolites were detected in 20 hours, but

only 150 were identified124. This raises a couple of issues: first, it can be difficult to

identify compounds and second, quantitation must be performed by running

standards, making it difficult to not identify compounds of interest before analysis.

CE/MS studies to date have primarily focused on more metabolomic applications in

plants and bacteria, but some metabonomics studies on urine samples have been

performed126.

1.3.3. Other Techniques

UV detection is not ideal for metabonomics studies, for two primary

reasons37. First, the data generated gives little information about the molecule

generating it, and second, the linewidth of the signal generated is large, lowering peak

resolution. Nevertheless, a few studies have been performed with this detection

mode, including CE/UV128 and LC/UV129 analyses.

Fourier transform-infrared spectroscopy (FT-IR) has been utilized in limited

studies to characterize biofluid samples130. Its main advantages lie in the speed of

29

Page 30: ~Chapter 1~ Introduction 1.1. Biomarkers

sample acquisition (several seconds) and its nonselectivity. However, it has found

limited use in metabonomics studies because of the difficulty in signal assignment

and less overall information present in a single spectrum. It is possible to distinguish

different functional groups present in the sample, but individual metabolites typically

cannot be distinguished.

Electrochemical (EC) analytical techniques have been applied to

metabonomics studies, including studies with coulometric arrays131 and microchip

technology132. This offers excellent detection limits and good dynamic range.

However, detection is limited to redox-active compounds. Compound classes such as

hormones, antioxidants, neurotransmitters and oxidative stress markers can be

profiled using this technique133.

Combining detection modes in metabonomics is being explored, in an attempt

to gain the advantages from two orthogonal analytical techniques and increase the

number of metabolites detected. NMR and MS have been combined, primarily by

splitting flow from the LC to each detection scheme134. An LC-EC-Array-MS

parallel detection method has also been described133.

1.4. Statistics for Metabonomics

1.4.1 Multivariate Statistics Overview

The complexity of the data generated in metabonomics analyses, be it from

mass spectrometry or NMR spectroscopy, makes it very difficult to evaluate visually

and very cumbersome to analyze in a univariate manner. It is also desirable to

30

Page 31: ~Chapter 1~ Introduction 1.1. Biomarkers

monitor patterns of biomarkers, or how multiple biomarkers change simultaneously

with respect to each other. Consequently, pattern recognition techniques are used on

metabonomics data sets through multivariate statistical techniques. Generally, two

types of pattern recognition methods exist: unsupervised and supervised.

Unsupervised methods, including principal components analysis (PCA), examine

patterns and clustering in the metabolite data without a priori identification of

groupings or trends. These are typically used to simply describe trends occurring in

the data. Supervised methods, such as discriminant analysis, are used to classify the

data into groups. In these methods, “training sets” of data are used to define the

boundaries of groups (i.e., control and dosed) and then the model generated is used to

classify unknown metabolic profiles into their respective groups with a certain degree

of confidence. There are many statistical techniques available to the researcher. This

discussion will focus on the two most relevant to this research, principal components

analysis and partial least squares regression.

These statistical techniques are well-suited to metabonomics data analysis for

several reasons. First, they are adaptable to many different types of data – MS, NMR,

or data from any other analytical method can be translated into the matrices needed

for these analyses135. Also, the quality of these analyses tends to improve with an

increasing number of variables, because the new or latent variables are a weighted

average of the original variables135.

Metabonomics data sets are incredibly complex, with a large number of

variables. What the variables represent is dependent on the analytical method

31

Page 32: ~Chapter 1~ Introduction 1.1. Biomarkers

employed. (For simplicity and because it is most pertinent to this research, the

following discussion will be limited to NMR data. Variables from an NMR data set

represent integral regions in the NMR spectra.) Data matrices for these variables

contain two types of data – noise (or undesirable variation) and systematic variation

directly related to the metabolic perturbation135. It is the goal of the researcher to

minimize the noise so that systematic variation can be studied without interference,

which can be done through a variety of mechanisms, including scaling136. This can

be done in many ways and is discussed further in Chapter 2.

1.4.2. Principal Components Analysis

Principal components analysis (PCA) is by far the most prevalent statistical

tool in metabonomics studies135,137. It is fairly easy and efficient to obtain results and

gives an accurate view of the data set. It can be used to point the way for further

statistical tests or important variables to probe further, or can be used to describe the

data on its own. PCA allows for visualization of trends in the data, both groupings

and time trends and can identify data outliers136.

PCA reduces complex sets of data (with a high number of correlated

variables) to a model with fewer variables but containing the same amount of

variance137-139. This is called data reduction, making the data set more manageable

and easier to study. Mechanistically, PCA accomplishes data reduction through the

generation of new variables via an eigenanalysis of the original data set. The

eigenanalysis is actually performed on a matrix of correlation coefficients between

each variable. A set of new variables, called principal components, are generated by

32

Page 33: ~Chapter 1~ Introduction 1.1. Biomarkers

creating linear combinations of the original variables (for example, NMR integral

regions). Each principal component is orthogonal to the others135. The set of new

variables describes the same amount of variance in the original data set, with the first

principal component explaining the most amount of variance and each subsequent

principal component explaining successively less variance. PCA generates two

matrices of information – a scores matrix and a loadings matrix. The loadings matrix

describes the new variables/principal components by giving the coefficients used in

the linear combination of original variables to calculate the new variable’s value. The

scores matrix contains a data point for each sample in the original data matrix, which

is calculated from the linear combination described in the loadings matrix. A PCA

model can contain the same numbers of principal components as there are variables in

the original data set, but this is typically not necessary. Most trends in the data can be

visualized in the first few principal components. The PCA model is examined by

generating scores plots, or by plotting the new principal components against each

other. Clustering in the data will illustrate which samples have similarities (and

differences). Time trends will also emerge in these plots. By observing which

axis/principal component the trend is on, loadings for that principal component can be

used to determine which of the original variables were important in defining that

trend.

1.4.3. Partial Least Squares Regression

Partial least squares regression (PLS) is used to correlate complex

metabonomics data sets (called predictors) to another set of data (called

33

Page 34: ~Chapter 1~ Introduction 1.1. Biomarkers

responses)140,141. Responses could correspond to quantitative variables, such as time

or drug dosage or to qualitative variables, such as groupings. These data sets are

represented as separate matrices in the analysis. A model is created that maximizes

the covariance between the matrices135,136,142 by developing latent variables to

describe the data set143. The scores and loadings for the model can give information

about the strength of metabolite correlation to the response matrix. PLS models are

typically tested through cross-validation135. In this test, part of the data set is

removed and the model is recalculated. This is compared to the original model, and

the difference between the models provides a gauge of the model robustness.

1.5. Tissue-Targeted Metabonomics Application: Oxidative Stress

Metabonomics has seen many applications over the years, as reviewed in

Section 1.2.1. This research is primarily concerned with the application of

metabonomics to study oxidative stress. Oxidative stress is a potentially devastating

occurrence in the human body. It plays an important role in the pathogenesis of many

debilitating or fatal conditions, including myocardial infarction (heart attack), stroke,

atherosclerosis, Parkinson’s disease and Alzheimer’s disease1-3. The societal impact

of oxidative stress damage is widespread. In the United States alone, 8.1 million

people suffered a heart attack and 5.8 million people suffered a stroke in 2005144.

Coronary heart disease is the leading cause of death in the United States, with stroke

the third leading cause of death. Combined yearly healthcare costs associated with

coronary heart disease and stroke are over 500 billion dollars144.

34

Page 35: ~Chapter 1~ Introduction 1.1. Biomarkers

Oxidative stress can occur from a variety of sources. Conditions such as

stroke or heart attack cause oxidative damage to tissue resulting from

ischemia/reperfusion, or the cessation and subsequent restoration of blood flow.

Oxidative stress can also be induced chemically, such as through redox cycling of the

quinone moiety on doxorubicin, a common chemotherapy agent145. The resulting

oxidative damage is often irreversible and could cause organ failure. Oxidative stress

will result from any situation in which the body is unable to mediate free radical

levels.

The agents causing oxidative damage, reactive oxygen species (ROS) and

reactive nitrogen species (RNS), are highly reactive compounds, including free

radicals and peroxides, produced in aerobic organisms146. The ROS include

superoxide (O2-), hydrogen peroxide (H2O2), peroxynitrite (ONOO-) and the

hydroxyl radical (OH). Organisms constantly generate a low level of ROS under

basal metabolic conditions. In the liver, the concentration of H2O2 ranges from 10-7 to

10-9 M, and the concentration of O2- is approximately 10-11 M147. ROS play a role in

a few normal cellular processes, including catalysis by ribonucleotide reductase and

phagocyte immune response148. Free radicals mostly originate from the mitochondria

(during cellular respiration)146 and the endoplasmic reticulum148. There is a balance

in the body between basal free radical production and levels of antioxidants to

mediate the radicals. Many of these free radicals are dangerous, as they are highly

reactive, with the capability to destroy virtually any cellular component if left

unchecked due to insufficient antioxidant levels146,149.

35

Page 36: ~Chapter 1~ Introduction 1.1. Biomarkers

36

On a molecular level, oxygen free radicals can be generated through a variety

of pathways (Figure 1.4), in addition to cellular respiration in the mitochondria. First,

enzymatic formation is common. Xanthine oxidase, mitochondrial cytochrome

oxidase, cyclooxygenase, lipoxygenase and NADPH oxidases all can cause the

transfer of a single electron to molecular oxygen148,150. The autooxidation of

compounds such as the catecholamines can also result in ROS production148,150.

Finally, reperfusion causes an insurgence of neutrophils to the site of ischemia which

release proteolytic enzymes that generate free radicals149. Reduction of molecular

oxygen by one electron forms the superoxide anion, while two electron reduction of

molecular oxygen results in the formation of hydrogen peroxide148. Hydrogen

peroxide is also formed through a dismutation reaction of superoxide, typically

catalyzed by superoxide dismutase, although the reaction can slowly occur

spontaneously148.

O2- + O2

- + 2H+ → H2O2 + O2 (1)

Superoxide can go on to form other products such as the highly reactive hydroxyl

radical and peroxides. The Haber-Weiss reaction forms hydroxyl radicals and

molecular oxygen from superoxide and hydrogen peroxide substrates149. This is a

two step reaction. The first step is the reaction of superoxide with Fe3+, mediated by

superoxide dismutase, forming Fe2+ and molecular oxygen148. The Fenton reaction

then forms the hydroxyl radical from hydrogen peroxide, as catalyzed by Fe2+

generated from the first reaction149,151.

H2O2 + Fe2+ → Fe3+ + OH + OH- (2)

Page 37: ~Chapter 1~ Introduction 1.1. Biomarkers

O2-

Supe

roxi

de

Dis

mut

ase

H2O

, O2

Cat

alas

e

Lipi

d pe

roxi

datio

nD

NA

stra

nd b

reak

age

Enzy

me

inac

tivat

ion

H2O

2

OH Fe

2+

(O2-

+ Fe

3+→

Fe2+

+ O

2)

H2O

GSH

GSS

G

Glu

tath

ione

Pe

roxi

dase

Glu

tath

ione

R

educ

tase

NA

DPH

, H+

NA

DP+

Ant

ioxi

dant

s :Th

iols

Asc

orba

teV

itam

ine

Eβ-

caro

tene

Oxi

dativ

e D

amag

e

O2

1 e-

2 e-

NO

ON

OO

-A

popt

osis

Nec

rosi

s

151.

Figu

re 1

.4.

Path

way

s of f

orm

atio

n of

oxy

gen

free

radi

cals

. A

dapt

ed fr

om re

fere

nce

O2-

Supe

roxi

de

Dis

mut

ase

H2O

, O2

Cat

alas

e

Lipi

d pe

roxi

datio

nD

NA

stra

nd b

reak

age

Enzy

me

inac

tivat

ion

H2O

2

OH Fe

2+

(O2-

+ Fe

3+→

Fe2+

+ O

2)

H2O

GSH

GSS

G

Glu

tath

ione

Pe

roxi

dase

Glu

tath

ione

R

educ

tase

NA

DPH

, H+

NA

DP+

Ant

ioxi

dant

s :Th

iols

Asc

orba

teV

itam

ine

Eβ-

caro

tene

Oxi

dativ

e D

amag

e

O2

1 e-

2 e-

NO

ON

OO

-A

popt

osis

Nec

rosi

s

O2-

Supe

roxi

de

Dis

mut

ase

H2O

, O2

Cat

alas

e

Lipi

d pe

roxi

datio

nD

NA

stra

nd b

reak

age

Enzy

me

inac

tivat

ion

H2O

2

OH Fe

2+

(O2-

+ Fe

3+→

Fe2+

+ O

2)

H2O

GSH

GSS

G

Glu

tath

ione

Pe

roxi

dase

Glu

tath

ione

R

educ

tase

NA

DPH

, H+

NA

DP+

Ant

ioxi

dant

s :Th

iols

Asc

orba

teV

itam

ine

Eβ-

caro

tene

Oxi

dativ

e D

amag

e

O2

1 e-

2 e-

NO

ON

OO

-A

popt

osis

Nec

rosi

s

151.

Figu

re 1

.4.

Path

way

s of f

orm

atio

n of

oxy

gen

free

radi

cals

. A

dapt

ed fr

om re

fere

nce

37

Page 38: ~Chapter 1~ Introduction 1.1. Biomarkers

The hydroxyl radical is highly toxic, as it is one of the most reactive species

known148. It can cause lipid peroxidation, inactivate enzymes and induce DNA strand

breakage151. Although not as toxic as the hydroxyl radical, superoxide itself can also

instigate oxidative damage directly through reaction with FeS enzyme centers,

resulting in enzyme inactivation. However, it is more likely that it contributes to

oxidative stress indirectly through its reduction of transition metal ions and

conversion to hydrogen peroxide148. Superoxide can also react nonenzymatically

with nitric oxide, producing peroxynitrite (ONOO-)148,149, another causative agent of

oxidative damage. This ROS is also highly reactive and has been shown to increase

superoxide formation by inhibition of superoxide dismutase and interference with the

electron transport chain148.

The effects of oxidative damage are felt by every cellular component.

Oxidative damage at DNA bases primarily occurs through oxidation of the guanine

base. This causes an 8-hydroxyguanine lesion, leading to G to T transversions in

DNA replication. These lesions are repaired by the body through base excision

repair, which removes the damaged base, preventing further mutagenic damage from

occurring. The extent of DNA damage from ROS is often measured by the levels of

excreted base excision repair products in plasma or urine, such as the marker 8-

hydroxy-2’-deoxyguanosine (8-OHdG)152. Oxidative damage can also occur on

lipids, where ROS react with polyunsaturated fatty acids, forming hydroperoxides,

conjugated dienes and aldehydes. The hydroxyl or peroxynitrite radical produces an

alkyl radical that produces a peroxyl radical (ROO) after reaction with molecular

38

Page 39: ~Chapter 1~ Introduction 1.1. Biomarkers

oxygen. This instigates a cycle of lipid peroxidation, as the peroxyl radical can gain a

hydrogen from the neighboring fatty acid, which creates a hydroperoxide and another

peroxyl radical150. Common markers of lipid peroxidation include malondialdehyde

and other dialdehydes, as well as 4-hydroxynonenal (4-HNE)153. Lipid peroxidation

also produces formaldehyde, acetaldehyde, acetone and propionaldehyde from β-

cleavage of lipid peroxides153.

Free radicals are neutralized in vivo by a variety of sources. Some proteins

can indirectly mediate oxidative damage by chelation of metal ions148. The enzymes

catalase and glutathione peroxidase both inactivate hydrogen peroxide, converting it

to water151. The hydroxyl radical is not trapped in vivo, but repair mechanisms are in

place to mediate the damage caused, such as base excision repair discussed

previously. Free radicals are also mediated nonenzymatically, through compounds

such as thiols, ascorbate, uric acid, vitamin E and β-carotene147,150. Thiols are the

small molecule antioxidant defense system in the body – the most predominant being

glutathione (GSH), a tripeptide consisting of glycine, glutamate and cysteine. Other

endogenous thiols include cysteine, homocysteine and cysteinylglycine. The

sulfhydryl group on the cysteine interacts with ROS, producing a thiyl radical (GS)

through one electron oxidation. The thiyl radical reacts with GS- and superoxide to

form glutathione disulfide (GSSG) and H2O2. Thiols act as a redox buffer by

maintaining a balance between reduced and oxidized forms – the relative amounts of

these can give some indication as to the oxidative stress status of the tissue.

39

Page 40: ~Chapter 1~ Introduction 1.1. Biomarkers

Because oxidative stress is so closely linked to mitochondrial activity, there is

a natural transitive relationship between oxidative stress and energy metabolism148.

Problems in the electron transport chain may result in ROS formation, which can then

create a cycle of increasing ROS production. During ischemia, energy is generated

anaerobically from the conversion of glucose to lactate154,155. Energy stores are also

utilized rapidly, including creatine phosphate, glycogen, and adenosine

compounds154. Without an energy supply, ions leak across the cell membrane,

causing depolarization150. Adenosine monophosphate (AMP) is metabolized to

hypoxanthine, xanthine and uric acid by xanthine oxidase, producing the superoxide

radical150. A decrease in pH is observed from anaerobic metabolism, resulting in

activation of lysosomal enzymes. The combined effects of these metabolic changes

result in dysfunction for the organ, such as contractile problems in the heart156.

Although ischemia causes extreme changes in metabolism, studies suggest that most

of the damage to the tissue occurs during reperfusion, when most of the ROS are

formed149. This is know as the oxygen paradox – although oxygen is essential for the

survival of organisms, a resurgence of it after ischemia can injure the tissue through

the formation of reactive oxygen species149.

Oxidative stress can also be induced by the administration of doxorubicin

(also known as adriamycin), a common chemotherapy drug used to treat a variety of

cancers. Doxorubicin exerts its anticancer effects through intercalation into and

cross-linking with DNA bases, causing problems with strand separation and

interfering with the activity of helicase, topoisomerase II and DNA and RNA

40

Page 41: ~Chapter 1~ Introduction 1.1. Biomarkers

polymerase157,158. It also induces apoptosis158-160. The doxorubicin dose that can be

given to a patient is limited due to its cardiotoxic side effects observed above a

threshold dose, leading to irreversible changes to the heart and congestive heart

failure161.

The mechanism for this cardiotoxicity is unclear, but has been shown to be

linked to free radical production, mitochondrial dysfunction, excess calcium levels,

activity changes in membrane-bound enzymes and abnormal fatty acid oxidation162-

165. The evidence for free radicals causing cardiotoxicity is strong145 because studies

have demonstrated the mediation of cardiomyopathy through administration of

antioxidants166. Heart tissue is uniquely susceptible to doxorubicin toxicity, due to its

high rate of oxidative metabolism, lowered levels of antioxidants, and the affinity of

doxorubicin to cardiolipin (a phospholipid in cardiac mitochondrial membranes)165.

It is thought that cardiac cells are more susceptible to damage from doxorubicin

because of a different enzyme in the mitochondrial membrane that reduces

doxorubicin and causes it to displace CoQ10, a critical element in the electron

transport chain145. Although doxorubicin is notorious for its cardiotoxic side effects,

its toxicity is observed in other tissues as well, such as the liver167-169. One of the

primary mechanisms by which doxorubicin generates its toxic side effects in the liver

is through lipid peroxidation ultimately ending in membrane disruption, as discussed

previously145,167,169,170. Doxorubicin has been shown to increase levels of superoxide

dismutase, catalase and glutathione peroxidase in liver tissue, while decreasing levels

of cytochrome P450 and glutathione168,169,171.

41

Page 42: ~Chapter 1~ Introduction 1.1. Biomarkers

Doxorubicin initiates oxidative damage in tissues through redox cycling. It

contains a tetracyclic group, with a daunosamine sugar linked by a glycosidic bond.

The tetracyclic moiety contains adjacent quinone and hydroquinone structures that

allow for reduction to the semiquinone145. The semiquinone in turn will give up the

electron to molecular oxygen, creating the superoxide free radical that can go on to

cause cellular damage or undergo conversion to the more highly reactive hydroxyl

radical, causing damage to lipids, DNA and proteins. (This redox cycling is

illustrated in Figure 1.5.) Doxorubicin can be reduced by enzymes, such as NADH

dehydrogenase, xanthine oxidase or NADPH-cytochrome P450 reductase145,159.

Doxorubicin can also be reduced through complexation with metal ions, typically

iron159,167,168,170. In this reaction, Fe3+ accepts an electron from doxorubicin, forming

an Fe2+-doxorubicin complex, which goes on to reduce oxygen to ROS168. Thus,

doxorubicin can be a chemical inducer of oxidative stress, allowing oxidative stress to

be induced much more easily and reproducibly than with ischemic mechanisms.

42

Page 43: ~Chapter 1~ Introduction 1.1. Biomarkers

43

Figure 1.5. Doxorubicin redox cycling. Adapted from reference 145.

O2

O2-

e-

ROS, RNSROS, RNS

O2

O2-

e-

Page 44: ~Chapter 1~ Introduction 1.1. Biomarkers

1.6. Focus of This Research

This research focuses on the development of tissue-targeted metabonomics, an

analytical approach to examining tissue-specific metabolism. There is a need in the

metabonomics community for additional technologies that can provide insight into

tissue metabolism, particularly if the technology is noninvasive and allows for

continuous sampling from the same animal. Microdialysis sampling is used to collect

the small endogenous metabolites in the interstitial fluid of the tissue of interest. (In

this study, focus was primarily directed to the liver.) These samples were then

analyzed by proton NMR and subjected to multivariate data analysis to find patterns

in the metabolic data.

Initially, focus was placed on the development of the analytical methodology.

Several issues had to be addressed before application of this technique to biological

problems. First, the analysis of small volume dialysate samples by NMR had to be

optimized. Variance in the data from experimental conditions had to be identified,

characterized and then removed from the metabolic data by normalization. These

factors included probe extraction efficiency, variation from the NMR analysis and

animal-to-animal variability. Data pretreatment steps were examined for their impact

on the results of the statistical analyses.

Next, inherent biological variation in the metabolic information was

characterized. Processes such as animal-to-animal variability, circadian rhythms,

animal activity and anesthesia effects can all cause changes in the basal metabolism

44

Page 45: ~Chapter 1~ Introduction 1.1. Biomarkers

of the animal. It is important to be able to identify these changes so that they can be

separated from metabolic changes induced by an experimental stressor.

Finally, the developed tissue-targeted metabonomics method was applied to

the study of hepatic oxidative stress, as induced by the chemotherapy agent

doxorubicin. This was a proof of concept study used to determine if the technique

could successfully detect and follow metabolic changes over time. These studies

were performed in conjunction with the quantitation of a biomarker of oxidative

stress, glutathione, to determine if data analysis was more successful using a

previously established marker to guide interpretation.

1.7. References

1. Biomarkers Definition Working Group. (2001). Biomarkers and surrogate endpoints: preferred definitions and conceptual work. Clin. Pharmacol. Ther. 69, 89-95.

2. Timbrell, J. (2005). Overview of Biomarkers in Metabonomics in Toxicity

Assessment. Robertson, D. G., Lindon, J., Nicholson, J. K. and Holmes, E. (Eds.). Taylor and Francis Group, New York, 27-74.

3. LaBaer, J. (2005). So, you want to look for biomarkers (introduction to the

special biomarkers issue). J. Proteom. Res. 4, 1053-1059. 4. Frank, R. and Hargreaves, R. (2003). Clinical biomarkers in drug discovery

and development. Nat. Rev. Drug Discov. 2, 566-580. 5. Bailey, W. J. and Ulrich, R. (2004). Molecular profiling approaches for

identifying novel biomarkers. Expert Opin. Drug Saf. 3, 137-151. 6. Colburn, W. A. (2003). Biomarkers in drug discovery and development:

from target identification through drug marketing. J. Clin. Pharmacol. 43, 329-341.

7. Baker, M. (2005). In biomarkers we trust? Nat. Biotech. 23, 297-304.

45

Page 46: ~Chapter 1~ Introduction 1.1. Biomarkers

8. Gold, J. (1966). Metabolic profiles in human solid tumors. I. A new technic

for the utilization of human solid tumors in cancer research and its application to the anaerobic glycolysis of isologous benign and malignant colon tissues. Cancer Res. 26, 695-705.

9. Burke, W. T. and Gangloff, E. C. (1965). Metabolic profiles during

carcinogenesis and related conditions in hepatic tissue. Cancer Res. 25, 833-839.

10. Horning, E. C. and Horning, M. G. (1971). Metabolic profiles: gas-phase

methods for analysis of metabolites. Clin. Chem. 17, 802-809. 11. Thompson, J. A. and Markey, S. P. (1975). Quantitative metabolic profiling

of urinary organic acids by gas chromatography-mass spectrometry: comparison of isolation methods. Anal. Chem. 47, 1313-1321.

12. Teranishi, R., Mon, T. R., Robinson, A. B., Cary, P. and Pauling, L. (1972).

Gas chromatography of volatiles from breath and urine. Anal. Chem. 44, 18-20.

13. Reimendal, R. and Sjovall, J. B. (1973). Computer evaluation of gas

chromatographic-mass spectrometric analyses of steroids from biological materials. Anal. Chem. 45, 1083-1089.

14. Jellum, E., Stokke, O. and Eldjarn, L. (1972). Combined use of gas

chromatography, mass spectrometry, and computer in diagnosis and studies of metabolic disorders. Clin. Chem. 18, 800-809.

15. Brown, F. F., Campbell, I. D., Kuchel, P. W. and Rabenstein, D. L. (1977).

Human erythrocyte metabolism studies by 1H spin echo NMR. FEBS Lett. 82, 12-16.

16. Rabenstein, D. L. (1984). 1H NMR methods for the noninvasive study of

metabolism and other processes involving small molecules in intact erythrocytes. J. Biochem. Biophys. Meth. 9, 277-306.

17. Nicholson, J. K., O'Flynn, M. P. and Sadler, P. J. (1984). Proton-nuclear-

magnetic-resonance studies of serum, plasma and urine from fasting normal and diabetic subjects. Biochem. J. 217, 365-375.

18. Moolenaar, S. H., Engelke, U. F. H. and Wevers, R. A. (2003). Proton

nuclear magnetic resonance spectroscopy of body fluids in the field of inborn errors of metabolism. Ann. Clin. Biochem. 40, 16-24.

46

Page 47: ~Chapter 1~ Introduction 1.1. Biomarkers

19. Iles, R. A., Hind, A. J. and Chalmers, R. A. (1985). Use of proton nuclear

magnetic resonance spectroscopy in detection and study of organic acidurias. Clin. Chem. 31, 1795-1801.

20. Solanky, K. S., Bailey, N. J. C., Beckwith-Hall, B. M., Davis, A., Bingham,

S., Holmes, E., Nicholson, J. K. and Cassidy, A. (2003). Application of biofluid 1H nuclear magnetic resonance-based metabonomic techniques for the analysis of the biochemical effects of dietary isoflavones on human plasma profile. Anal. Biochem. 323, 197-204.

21. Viant, M. R., Lyeth, B. G., Miller, M. G. and Breman, R. F. (2005). An

NMR metabonomic investigation of early metabolic disturbances following traumatic brain injury in a mammalian model. NMR Biomed. 18, 507-516.

22. Azmi, J., Griffin, J. L., Shore, R. F., Holmes, E. and Nicholson, J. K. (2005).

Chemometric analysis of biofluids following toxicant induced hepatotoxicity: a metabonomic approach to distinguish the effects of 1-naphthylisothiocyanate from its products. Xenobiotica 35, 839-852.

23. Espina, J. R., Shockcor, J. P., Herron, W. J., Car, B. D., Contel, N. R.,

Ciaccio, P. J., Lindon, J. C., Holmes, E. and Nicholson, J. K. (2001). Detection of in vivo biomarkers of phospholipidosis using NMR-based metabonomic approaches. Magn. Reson. Med. 39, 559-565.

24. Ishihara, K., Katsutani, N. and Aoki, T. (2006). A metabonomics of study of

the hepatotoxicants galactosamine, methylene dianiline and clofibrate in rats. Basic Clin. Pharmacol. 99, 251-260.

25. Lafaye, A., Junot, C., Ramounet-Le Gall, B., Fritsch, P., Tabet, J.-C. and

Ezan, E. (2003). Metabolite profiling in rat urine by liquid chromatography/electrospray ion trap mass spectrometry. Application to the study of heavy metal toxicity. Rapid Commun. Mass Spectrom. 17, 2541-2549.

26. Mortishire-Smith, R. J., Skiles, G. L., Lawrence, J. W., Spence, S., Nicholls,

A. W., Johnson, B. A. and Nicholson, J. K. (2004). Use of metabonomics to identify impaired fatty acid metabolism as the mechanism of a drug-induced toxicity. Chem. Res. Toxicol. 17, 165-173.

27. Yang, J., Zhao, X., Liu, X., Wang, C., Gao, P., Wang, J., Li, L., Gu, J., Yang,

S. and Xu, G. (2006). High performance liquid chromatography-mass spectrometry for metabonomics: potential biomarkers for acute deterioration of liver function in chronic hepatitis B. J. Proteom. Res. 5, 554-561.

47

Page 48: ~Chapter 1~ Introduction 1.1. Biomarkers

28. Yang, J., Xu, G., Zheng, Y., Kong, H., Pang, T., Lv, S. and Yang, Q. (2004).

Diagnosis of liver cancer using HPLC-based metabonomics avoiding false-positive result from hepatitis and hepatocirrhosis diseases. J. Chromatogr. B 813, 59-65.

29. Lenz, E. M., Bright, J., Knight, R., Wilson, I. D. and Major, H. (2004).

Cyclosporin A-induced changes in endogenous metabolites in rat urine: a metabonomic investigation using high field 1H NMR spectroscopy, HPLC-TOF/MS and chemometrics. J. Pharm. Biomed. Anal. 35, 599-608.

30. Yang, J., Xu, G., Hong, Q., Liebich, H. M., Lutz, K., Schmülling, R.-M. and

Wahl, H. G. (2004). Discrimination of Type 2 diabetic patients from healthy controls by using metabonomics method based on their serum fatty acid profiles. J. Chromatogr. B 813, 53-58.

31. Bowser, R., Cudkowicz, M. and Kaddurah-Daouk, R. (2006). Biomarkers for

amyotrophic lateral sclerosis. Expert Rev. Mol. Diagn. 6, 387-398. 32. Griffin, J. L., Cemal, C. K. and Pook, M. A. (2004). Defining a metabolic

phenotype in the brain of a transgenic mouse model of spinocerebellar ataxia 3. Physiol. Genomics 16, 334-340.

33. Coen, M., O'Sullivan, M., Bubb, W. A., Kuchet, P. W. and Sorrell, T. (2005).

Proton nuclear magnetic resonance-based metabonomics for rapid diagnosis of meningitis and ventriculitis. Clin. Infect. Dis. 41, 1582-1590.

34. Holmes, E., Tsang, T. M., Huang, J. T.-J., Leweke, F. M., Koethe, D., Gerth,

C. W., Nolden, B. M., Gross, S., Schreiber, D., Nicholson, J. K. and Bahn, S. (2006). Metabolic profiling of CSF: evidence that early intervention may impact on disease progression and outcome in schizophrenia. PLOS Med. 3, e327.

35. Odunsi, K., Wollman, R. M., Ambrosone, C. B., Hutson, A., McCann, S. E.,

Tammela, J., Geisler, J. P., Miller, G., Sellers, T., Cliby, W., Qian, F., Keitz, B., Intengan, M., Lele, S. and Alderfer, J. L. (2005). Detection of epithelial ovarian cancer using 1H-NMR-based metabonomics. Int. J. Cancer 113, 782-788.

36. Lindon, J. C., Nicholson, J. K., Holmes, E., Antti, H., Bollard, M. E., Keun,

H., Beckonert, O., Ebbels, T. M., Reily, M. D., Robertson, D. G., Stevens, G. J., Luke, P., Breau, A. P., Cantor, G. H., Bible, R. H., Niederhauser, U., Senn, H., Schlotterbeck, G., Sidelmann, U. G., Laursen, S. M., Tymiak, A., Car, B. D., Lehman-McKeeman, L., Colet, J.-M., Loukaci, A. and Thomas, C.

48

Page 49: ~Chapter 1~ Introduction 1.1. Biomarkers

(2003). Contemporary issues in toxicology: the role of metabonomics in toxicology and its evaluation by the COMET project. Toxicol. Appl. Pharmacol. 187, 137-146.

37. Lindon, J. C., Holmes, E., Bollard, M. E., Stanley, E. G. and Nicholson, J. K.

(2004). Metabonomics technologies and their applications in physiological monitoring, drug safety assessment and disease diagnosis. Biomarkers 9, 1-31.

38. Nicholson, J. K., Lindon, J. C. and Holmes, E. (1999). 'Metabonomics':

understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data. Xenobiotica 29, 1181-1189.

39. Fiehn, O. (2001). Combining genomics, metabolome analysis, and

biochemical modelling to understand metabolic networks. Comp. Funct. Genomics 2, 155-168.

40. Harrigan, G. G. and Goodacre, R., (eds.). (2003). Metabolic Profiling: Its

Role in Biomarker Discovery and Gene Function Analysis. Kluwer Academic Publishers, Boston.

41. Raamsdonk, L. M., Teusink, B., Broadhurst, D., Zhang, N., Hayes, A., Walsh,

M. C., Berden, J. A., Brindle, K. M., Kell, D. B., Rowland, J. J., Westerhoff, H. V., van Dam, K. and Oliver, S. G. (2001). A functional genomics strategy that uses metabolome data to reveal the phenotype of silent mutations. Nat. Biotech. 19, 45-50.

42. Fiehn, O., Kopka, J., Dormann, P., Altmann, Trethewey, R. N. and

Willmitzer, L. (2000). Metabolite profiling for plant functional genomics. Nat. Biotech. 18, 1157-1161.

43. Lindon, J. C., Holmes, E. and Nicholson, J. K. (2006). Metabonomics

technologies and applications to pharmaceutical research and development. Pharm. Res. 23, 1075-1088.

44. Lindon, J. C., Holmes, E. and Nicholson, J. K. (2004). Metabonomics:

systems biology in pharmaceutical research and development. Curr. Opin. Molec. Ther. 6, 265-272.

45. Griffin, J. L. and Bollard, M. E. (2004). Metabonomics: its potential as a tool

in toxicology for safety assessment and data integration. Curr. Drug Metab. 5, 389-398.

49

Page 50: ~Chapter 1~ Introduction 1.1. Biomarkers

46. Griffin, J. L. (2003). Metabolic profiles to define the genome: can we hear the phenotypes? Phil. Trans. R. Soc. Lond. B 359, 857-871.

47. Tang, H., Wang, Y., Nicholson, J. K. and Lindon, J. C. (2004). Use of

relaxation-edited one-dimensional and two dimensional nuclear magnetic resonance spectroscopy to improve detection of small metabolites in blood plasma. Anal. Biochem. 325, 260-272.

48. Van, Q. N., Chmurny, G. N. and Veenstra, T. D. (2003). The depletion of

protein signals in metabonomics analysis with the WET-CPMG pulse sequence. Biochem. Biophys. Res. Commun. 301, 952-959.

49. de Graaf, R. A. and Behar, K. L. (2003). Quantitative 1H NMR spectroscopy

of blood plasma metabolites. Anal. Chem. 75, 2100-2104. 50. Robertson, D. G., Reily, M. D., Sigler, R. E., Wells, D. F., Paterson, D. A. and

Braden, T. K. (2000). Metabonomics: evaluation fo nuclear magnetic resonance (NMR) and pattern recognition technology for rapid in vivo screening of liver and kidney toxicants. Toxicol. Sci. 57, 320-337.

51. Lindon, J. C., Nicholson, J. K., Holmes, E. and Everett, J. R. (2000).

Metabonomics: metabolic processes studied by NMR spectroscopy of biofluids. Concepts Magn. Reson. 12, 289-320.

52. Lenz, E. M. and Wilson, I. D. (2007). Analytical strategies for

metabonomics. J. Proteom. Res. 6, 443-458. 53. White, L. R., Gårseth, M., Aasly, J. and Sonnewald, U. (2004).

Cerebrospinal fluid from patients with dementia contains increased amounts of an unknown factor. J. Neurosci. Res. 78, 297-301.

54. Dunne, V. G., Bhattachayya, S., Besser, M., Rae, C. and Griffin, J. L. (2005).

Metabolites from cerebrospinal fluid in aneurysmal subarachnoid haemorrhage correlate with vasospasm and clinical outcome: a pattern-recognition 1H NMR study. NMR Biomed. 18, 24-33.

55. Gårset, M., Sonnewald, U., White, L. R., Rød, M., Nygaard, Ø. and Zwart, J.-

A. (2002). Metabolic changes in the cerebrospinal fluid of patients with lumbar disc herniation or spinal stenosis. J. Neurosci. Res. 69, 692-695.

56. Sharma, U., Chaudhury, K., Jagannathan, N. R. and Guha, S. K. (2001). A

proton NMR study of the effect of a new intravasal injectable male contraceptive RISUG on seminal plasma metabolites. Reproduction 122, 431-436.

50

Page 51: ~Chapter 1~ Introduction 1.1. Biomarkers

57. Avena, T. A., Kline, E. E., Smith, A. Y. and Sillerud, L. O. (2005). A

decrease in 1H nuclear magnetic resonance spectroscopically determined citrate in human seminal fluid accompanies the development of prostate adenocarcinoma. J. Urol. 173, 433-438.

58. Silwood, C. J. L., Lynch, E., Claxson, A. W. D. and Grootveld, M. C. (2001).

1H and 13C NMR spectroscopic analysis of human saliva. J. Dent. Res. 81, 422-427.

59. Romero, R., Espinoza, J., Gotsch, F., Kusanovic, J. P., Friel, L. A., Erez, O.,

Mazaki-Tovi, S., Than, N. G., Hassan, S. and Tromp, G. (2006). The use of high-dimensional biology (genomics, transcriptomics, proteomics, and metabolomics) to understand the preterm parturition syndrome. BJOG 113, 118-135.

60. Bock, J. L. (1994). Metabolic profiling of amniotic fluid by proton nuclear

magnetic resonance spectroscopy: correlation with fetal maturation and other clinical variables. Clin. Chem. 40, 56-61.

61. Robertson, D. G., Lindon, J., Nicholson, J. K. and Holmes, E. (2005).

Metabonomics in Toxicity Assessment. Taylor and Francis Group, Inc., Boca Raton, FL.

62. Pears, M. R., Cooper, J. D., Mitchison, H. M., Mortishire-Smith, R. J., Pearce,

D. A. and Griffin, J. L. (2005). High resolution 1H NMR-based metabolomics indicates a neurotransmitter cycling deficit in cerebral tissue from a mouse model of Batten disease. J. Biol. Chem. 280, 42508-42514.

63. Tomlins, A. M., Foxall, P. J. D., Lindon, J. C., Lynch, M. J., Spraul, M.,

Everett, J. R. and Nicholson, J. K. (1998). High resolution magic angle spinning 1H nuclear magnetic resonance analysis of intact prostatic hyperplastic and tumour tissues. Anal. Commun. 35, 113-115.

64. Wang, Y., Bollard, M. E., Keun, H., Antti, H., Beckonert, O., Ebbels, T. M.,

Lindon, J. C., Holmes, E., Tang, H. and Nicholson, J. K. (2003). Spectral editing and pattern recognition methods applied to high-resolution magic-angle spinning 1H nuclear magnetic resonance spectroscopy of liver tissues. Anal. Biochem. 323, 26-32.

65. Garrod, S., Humpher, E., Connor, S. C., Connelly, J. C., Spraul, M.,

Nicholson, J. K. and Holmes, E. (2001). High-resolution 1H NMR and magic angle spinning NMR spectroscopic investigation of the biochemical effects of

51

Page 52: ~Chapter 1~ Introduction 1.1. Biomarkers

2-bromoethanamine in intact renal and hepatic tissue. Magn. Reson. Med. 45, 781-790.

66. Garrison, K. E., Pasas, S. A., Cooper, J. D. and Davies, M. I. (2002). A

review of microdialysis sampling from biological tissues with applications in pharmacokinetics, metabolism and pharmacodynamics. Eur. J. Pharm. Sci. 17, 1-12.

67. Davies, M. I. (1999). A review of microdialysis sampling for

pharmacokinetic applications. Anal. Chim. Acta 379, 227-249. 68. Ungerstedt, U. and Pycock, C. (1974). Functional correlates of dopamine

neurotransmission. Bull. Schweiz Akad. Med. Wiss. 30, 44-55. 69. de la Peña, A., Liu, P. and Derendorf, H. (2000). Microdialysis in peripheral

tissues. Adv. Drug Deliv. Rev. 45, 189-216. 70. Davies, M. I., Cooper, J. D., Desmond, S. S., Lunte, C. E. and Lunte, S. S.

(2000). Analytical considerations for microdialysis sampling. Adv. Drug Deliv. Rev. 45, 169-188.

71. de Lange, E. C. M., de Boer, A. G. and Breimer, D. D. (2000).

Methodological issues in microdialysis sampling for pharmacokinetic studies. Adv. Drug Deliv. Rev. 45, 125-148.

72. Hsiao, J. K., Ball, B. A., Morrison, P. F., Mefford, I. N. and Bungay, P. M.

(1990). Effects of different semipermeable membranes on in vitro and in vivo performance of microdialysis probes. J. Neurochem. 54, 1449-1452.

73. Clough, G. F. (2005). Microdialysis of large molecules. AAPS J. 7, E686-

E692. 74. Rosenbloom, A. J., Sipe, D. M. and Weedn, V. W. (2005). Microdialysis of

proteins: performance of the CMA/20 probe. J. Neurosci. Meth. 148, 147-153.

75. Telting-Diaz, M., Scott, D. O. and Lunte, C. E. (1992). Intravenous

microdialysis sampling in awake, freely-moving rats. Anal. Chem. 64, 806-810.

76. Davies, M. I. and Lunte, C. E. (1995). Microdialysis sampling for hepatic

metabolism studies: impact of microdialysis probe design and implantation technique on liver tissue. Drug Metab. Disp. 23, 1072-1079.

52

Page 53: ~Chapter 1~ Introduction 1.1. Biomarkers

77. Price, K. E., Lunte, C. E. and Larive, C. K. (2008). Development of tissue-targeted metabonomics. Part 1. Analytical considerations. J. Pharm. Biomed. Anal. 46, 737-747.

78. Bergstrom, S. K., Goiny, M., Danielsson, R., Ungerstedt, U., Andersson, M.

and Markides, K. E. (2006). Screening of microdialysates taken before and after induced liver damage; on-line solid phase extraction-electrospray ionization-mass spectrometry. J. Chromatogr. A 1120, 21-26.

79. Palsmeier, R. K. and Lunte, C. E. (1994). Microdialysis sampling in tumor

and muscle: study of the disposition of 3-amino-1,2,4-benzotriazine-1,4-di-N-oxide (SR 4233). Life Sci. 55, 815-825.

80. Lucas, L. H., Wilson, S. F., Lunte, C. E. and Larive, C. K. (2005).

Concentration profiling in rat tissue by high-resolution magic-angle spinning NMR spectroscopy: investigation of a model drug. Anal. Chem. 77, 2978-2984.

81. Tokunaga, N., Yamazaki, T., Akiyama, T., Sano, S. and Mori, H. (2003). In

vivo monitoring of norepinephrine and its metabolites in skeletal muscle. Neurochem. Int. 43, 573-580.

82. Niklasson, M., Holmäng, A., Sjöstrand, M., Strindberg, L. and Lönnroth, P.

(2000). Muscle glucose uptake is effectively activated by ischemia in type 2 diabetic subjects. Diabetes 49, 1178-1185.

83. Woo, K. L. and Lunte, C. E. (2008, Epub ahead of print). The development

of multiple probe microdialysis sampling in the stomach. J. Pharm. Biomed. Anal.

84. Suzuki, H., Moriya, A., Iijima, K., McElroy, K., Fyfe, V. E. and McColl, K.

E. (2003). Validation of microdialysis probes for studying nitrosative chemistry within localized regions of the human upper gastrointestinal tract. Scand. J. Gastroenterol. 38, 856-863.

85. Holovics, H. J., Anderson, C. R., Levine, B. S., Hui, H. W. and Lunte, C. E.

(2007, Epub ahead of print). Investigation of drug delivery by iontophoresis in a surgical wound utilizing microdialysis. Pharm. Res.

86. Ault, J. M., Lunte, C. E., Meltzer, N. M. and Riley, C. M. (1992).

Microdialysis sampling for the investigation of dermal drug transport. Pharm. Res. 9, 1256-1261.

53

Page 54: ~Chapter 1~ Introduction 1.1. Biomarkers

87. Ault, J. M., Riley, C. M., Meltzer, N. M. and Lunte, C. E. (1994). Dermal microdialysis sampling in vivo. Pharm. Res. 11, 1631-1639.

88. Schmidt, S., Banks, R., Kumar, V., Rand, K. H. and Derendorf, H. (2008).

Clinical microdialysis in skin and soft tissues: an update. J. Clin. Pharmacol. 48, 351-364.

89. Gilinksy, M. A., Faibushevish, A. A. and Lunte, C. E. (2001). Determination

of myocardial norepinephrine in freely moving rats using in vivo microdialysis sampling and liquid chromatography with dual-electrode amperometric detection. J. Pharm. Biomed. Anal. 24, 929-935.

90. Tabatabaei, P., Bergström, S. K., Henriksson, R. and Bergenheim, A. T.

(2008). Glucose metabolites, glutamate and glycerol in malignant glioma tumours during radiotherapy. J. Neurooncol. Epub ahead of print.

91. Scott, D. O. and Lunte, C. E. (1993). In vivo microdialysis sampling in the

bile, blood, and liver of rats to study the disposition of phenol. Pharm. Res. 10, 335-342.

92. Zhao, Y., Liang, X. and Lunte, C. E. (1995). Comparison of recovery and

delivery in vitro for calibration of microdialysis probes. Anal. Chim. Acta 316, 403-410.

93. Bungay, P. M., Morrison, P. F. and Dedrick, R. L. (1990). Steady-state

theory for quantitative microdialysis of solutes and water in vivo and in vitro. Life Sci. 46, 105-119.

94. Lönnroth, P., Jansson, P.-A. and Smith, U. (1987). A microdialysis method

allowing characterization of intercellular water space in humans. Am. J. Physiol. 253, E228-E231.

95. Wong, S. L., Wang, Y. and Sawchuck, R. J. (1992). Analysis of zidovudine

distribution to specific regions in rabbit brain using microdialysis. Pharm. Res. 9, 332-338.

96. Wang, C., Wong, S. L. and Sawchuck, R. J. (1993). Microdialysis calibration

using retrodialysis and zero-net flux: application to a study of the distribution of zidovudine to rabbit cerebrospinal fluid and thalamus. Pharm. Res. 10, 1411-1419.

97. Larsson, C. I. (1991). The use of an "internal standard" for control of the

recovery in microdialysis. Life Sci. 49, PL73-PL78.

54

Page 55: ~Chapter 1~ Introduction 1.1. Biomarkers

98. Wisniewski, N., Klitzman, B., Miller, B. and Reichert, W. M. (2001). Decreased analyte transport through implanted membranes: differentiation of biofouling from tissue effects. J. Biomed. Mater. Res. 57, 513-521.

99. Clark, H., Barbari, T. A., Stump, K. and Rao, G. (2000). Histologic

evaluation of the inflammatory response around implanted hollow fiber membranes. J. Biomed. Mater. Res. 52, 183-192.

100. Schiffer, W. K., Mirrione, M. M., Biegon, A., Alexoff, D. L., Patel, V. and

Dewey, S. L. (2006). Serial microPET measures of the metabolic reaction to a microdialysis probe implant. J. Neurosci. Meth. 155, 272-284.

101. Price, K. E., Vandaveer, S. S., Lunte, C. E. and Larive, C. K. (2005). Tissue

targeted metabonomics: metabolic profiling by microdialysis sampling and microcoil NMR. J. Pharm. Biomed. Anal. 38, 904-909.

102. Khandelwal, P., Beyer, C. E., Lin, Q., McGonigle, P., Schechter, L. E. and

Bach, A. C., II. (2004). Nanoprobe NMR spectroscopy and in vivo microdialysis: new analytical methods to study brain neurochemistry. J. Neurosci. Res. 133, 181-189.

103. Khandelwal, P., Beyer, C. E., Lin, Q., Schechter, L. E. and Bach, A. C., II.

(2004). Studying rat brain neurochemistry using nanoprobe NMR spectroscopy: a metabonomics approach. Anal. Chem. 76, 4123-4127.

104. Zhang, S., Yang, X. and Gorenstein, D. G. (2002). Suppression of linearly

responsive residual water. Concepts Magn. Reson. 14, 102-111. 105. Price, W. S. (1999). Water signal suppression in NMR spectroscopy. Ann.

Rep. NMR Spectrosc. 38, 289-354. 106. Claridge, T. D. W. (1999). High-Resolution NMR Techniques in Organic

Chemistry. Elsevier, New York. 107. Griffin, J. L., Nicholls, A. W., Keun, H. C., Mortishire-Smith, R. J.,

Nicholson, J. K. and Kuehn, T. (2002). Metabolic profiling of rodent biological fluids via 1H NMR spectroscopy using a 1 mm microlitre probe. Analyst 127, 582-584.

108. Olson, D. L., Lacey, M. E., Webb, A. G. and Sweedler, J. V. (1999).

Nanoliter-volume 1H NMR detection using periodic stopped-flow capillary electrophoresis. Anal. Chem. 71, 3070-3076.

55

Page 56: ~Chapter 1~ Introduction 1.1. Biomarkers

109. Olson, D. L., Norcross, J. A., O'Neil-Johnson, M., Molitor, P. F., Detlefsen, D. J., Wilson, A. G. and Peck, T. L. (2004). Microflow NMR: concepts and capabilities. Anal. Chem. 76, 2966-2974.

110. Waters, N. J., Garrod, S., Farrant, R. D., Haselden, J. N., Connor, S. C.,

Connelly, J., Lindon, J. C., Holmes, E. and Nicholson, J. K. (2000). High-resolution magic angle spinning 1H NMR spectroscopy of intact liver and kidney: optimization of sample preparation procedures and biochemical stability of tissue during spectral acquisition. Anal. Biochem. 282, 16-23.

111. Wu, C.-L., Taylor, J. L., He, W., Zepeda, A. G., Halpern, E. F., Bielecki, A.,

Gonzalez, R. G. and Cheng, L. L. (2003). Proton high-resolution magic angle spinning NMR analysis of fresh and previously frozen tissue of human prostate. Magn. Reson. Med. 50, 1307-1311.

112. Bollard, M. E., Garrod, S., Holmes, E., Lindon, J. C., Humpfer, E., Spraul, M.

and Nicholson, J. K. (2000). High-resolution 1H and 1H-13C magic angle spinning NMR spectroscopy of rat liver. Magn. Reson. Med. 44, 201-207.

113. Garrod, S., Humpfer, E., Spraul, M., Connor, S. C., Polley, S., Connelly, J.,

Lindon, J. C., Nicholson, J. K. and Holmes, E. (1999). High-resolution magic angle spinning 1H NMR spectroscopic studies on intact rat renal cortex and medulla. Magn. Reson. Med. 41, 1108-1118.

114. Fancy, S.-A., Beckonert, O., Darbon, G., Yabsley, W., Walley, R., Baker, D.,

Perkins, G. L., Pullen, F. S. and Rumpel, K. (2006). Gas chromatography/flame ionisation detection mass spectrometry for the detection of endogenous urine metabolites for metabonomic studies and its use as a complementary tool to nuclear magnetic resonance spectroscopy. Rapid Commun. Mass Spectrom. 20, 2271-2280.

115. Halket, J. M., Przyborowska, A., Stein, S. E., Mallard, W. G., Down, S. and

Chalmers, R. A. (1999). Deconvolution gas chromatography/mass spectrometry of urinary organic acids - potential for pattern recognition and automated identification of metabolic disorders. Rapid Commun. Mass Spectrom. 13, 279-284.

116. Plumb, R. S., Stumpf, C. L., Granger, J. H., Castro-Perez, J., Haselden, J. N.

and Dear, G. J. (2003). Use of liquid chromatography/time-of-flight mass spectrometry and multivariate statistical analysis shows promise for the detection of drug metabolites in biological fluids. Rapid Commun. Mass Spectrom. 17, 2632-2638.

56

Page 57: ~Chapter 1~ Introduction 1.1. Biomarkers

117. Wilson, I. D., Nicholson, J. K., Castro-Perez, J., Granger, J. H., Johnson, K. A., Smith, B. W. and Plumb, R. S. (2005). High resolution "ultra performance" liquid chromatography coupled to oa-TOF mass spectrometry as a tool for differential metabolic pathway profiling in functional genomic studies. J. Proteom. Res. 4, 591-598.

118. Böttcher, C., Roepenack-Lahaye, E. V., Willscher, E., Scheel, D. and

Clemens, S. (2007). Evaluation of matrix effects in metabolite profiling based on capillary liquid chromatography electrospray ionization quadrupole time-of-flight mass spectrometry. Anal. Chem. 79, 1507-1513.

119. Shen, Y., Zhang, R., Moore, R. J., Kim, J., Metz, T. O., Hixson, K. K., Zhao,

R., Livesay, E. A., Udseth, H. R. and Smith, R. D. (2005). Automated 20 kpsi RPLC-MS and MS/MS with chromatographic peak capacities of 1000-1500 and capabilities in proteomics and metabolomics. Anal. Chem. 77, 3090-3100.

120. Tolstikov, V. V., Lommen, A., Nakanishi, K., Tanaka, N. and Fiehn, O.

(2003). Monolithic silica-based capillary reversed-phase liquid chromatography/electrospray mass spectrometry for plant metabolomics. Anal. Chem. 75, 6737-6740.

121. Tolstikov, V. V. and Fiehn, O. (2002). Analysis of highly polar compounds

of plant origin: combination of hydrophilic interaction chromatography and electrospray ion trap mass spectrometry. Anal. Biochem. 301, 298-307.

122. Wilson, I. D., Plumb, R., Granger, J., Major, H., Williams, R. and Lenz, E. M.

(2005). HPLC-MS-based methods for the study of metabonomics. J. Chromatogr. B 817, 67-76.

123. Saito, N., Robert, M., Kitamura, S., Baran, R., Soga, T., Mori, H., Nishioka,

T. and Tomita, M. (2006). Metabolomics for enzyme discovery. J. Proteom. Res. 5, 1979-1987.

124. Soga, T., Ohashi, Y., Ueno, Y., Naraoka, H., Tomita, M. and Nishioka, T.

(2003). Quantitative metabolome analysis using capillary electrophoresis mass spectrometry. J. Proteom. Res. 2, 488-494.

125. Soga, T., Ueno, Y., Naraoka, H., Ohashi, Y., Tomita, M. and Nishioka, T.

(2002). Simultaneous determination of anionic intermediates for Bacillus subtilis metabolic pathways by capillary electrophoresis electrospray ionization mass spectrometry. Anal. Chem. 74, 2233-2239.

57

Page 58: ~Chapter 1~ Introduction 1.1. Biomarkers

126. Ullsten, S., Danielsson, R., Bäckström, D., Sjöberg, P. and Bergquist, J. (2006). Urine profiling using capillary electrophoresis-mass spectrometry and multivariate data analysis. J. Chromatogr. A 1117, 87-93.

127. Lee, R., Ptolemy, A. S., Niewczas, L. and Britz-McKibbin, P. (2007).

Integrative metabolomics for characterizing unknown low-abundance metabolites by capillary electrophoresis-mass spectrometry with computer simulations. Anal. Chem. 79, 403-415.

128. Zomer, S., Guillo, C., Brereton, R. G. and Hanna-Brown, M. (2003).

Toxicological classification of urine samples using pattern recognition techniques and capillary electrophoresis. Anal. Bioanal. Chem. 378, 2008-2020.

129. Pham-Tuan, H., Kaskavelis, L., Daykin, C. A. and Janssen, H.-G. (2003).

Method development in high-performance liquid chromatography for high-throughput profiling and metabonomic studies of biofluid samples. J. Chromatogr. B 789, 283-301.

130. Harrigan, G. G., LaPlante, R. H., Cosma, G. N., Cockerell, G., Goodacre, R.,

Maddox, J. F., Luyendyk, J. P., Ganey, P. E. and Roth, R. A. (2004). Application of high-throughput Fourier-transform infrared spectroscopy in toxicology studies: contribution to a study on the development of an animal model for idiosyncratic toxicity. Toxicol. Lett. 146, 197-205.

131. Vigneau-Callahan, K. E., Shestopalov, A. I., Milbury, P. E., Matson, W. R.

and Kristal, B. S. (2001). Characterization of diet-dependent metabolic serotypes: analytical and biological variability issues in rats. J. Nutr. 131, 924S-932S.

132. Garcia, C. D. and Henry, C. S. (2003). Direct determination of

carbohydrates, amino acids, and antibiotics by microchip electrophoresis with pulsed amperometric detection. Anal. Chem. 75, 4778-4783.

133. Gamache, P. H., Meyer, D. F., Granger, M. C. and Acworth, I. N. (2004).

Metabolomic applications of electrochemistry/mass spectrometry. J. Am. Soc. Mass Spectrom. 15, 1717-1726.

134. Shockcor, J. P., Unger, S. E., Wilson, I. D., Foxall, P. J. D., Nicholson, J. K.

and Lindon, J. C. (1996). Combined HPLC, NMR spectroscopy and ion-trap mass spectrometry with application to the detection and characterization of xenobiotic and endogenous metabolites in human urine. Anal. Chem. 68, 4431-4435.

58

Page 59: ~Chapter 1~ Introduction 1.1. Biomarkers

135. Eriksson, L., Johansson, E., Kettaneh-Wold, N. and Wold, S. (2001). Multi- and Megavariate Data Analysis: Principles and Applications. Umetrics, Sweden.

136. Trygg, J., Holmes, E. and Lundstedt, T. (2007). Chemometrics in

metabonomics. J. Proteom. Res. 6, 469-479. 137. Steuer, R., Morgenthal, K., Weckwerth, W. and Selbig, J. (2007). A gentle

guide to the analysis of metabolomic data in Methods in Molecular Biology. Weckwerth, W. (Eds.). Humana Press, Totowa, NJ,

138. Manly, B. F. J. (2005). Multivariate Statistical Methods: A Primer.

Chapman & Hall/CRC, New York. 139. Lindon, J. C., Holmes, E. and Nicholson, J. K. (2001). Pattern recognition

methods and applications in biomedical magnetic resonance. Prog. Nucl. Magn. Reson. Spectrosc. 39, 1-40.

140. Höskuldsson, A. (1988). PLS regression methods. J. Chemometrics 2, 211-

228. 141. Geladi, P. and Kowalski, B. R. (1986). Partial least-squares regression: a

tutorial. Anal. Chim. Acta 185, 1-17. 142. Abdi, H. (2003). Partial least squares (PLS) regression in Encyclopedia of

Social Sciences Research Methods. Lewis-Beck, M., Bryman, A. and Futing, T. (Eds.). Sage, Thousand Oaks, CA,

143. Holmes, E. and Antti, H. (2002). Chemometric contributions to the evolution

of metabonomics: mathematical solutions to characterising and interpreting complex biological NMR spectra. Analyst 127, 1549-1557.

144. American Heart Association. Heart Disease and Stroke Statistics - 2008

Update. Dallas, Texas: American Heart Association, 2008. © 2008, American Heart Association.

145. Conklin, K. A. (2005). Coenzyme Q10 for prevention of anthracycline-

induced cardiotoxicity. Integr. Cancer Ther. 4, 110-130. 146. Cutler, R. G., Plummer, J., Chowdhury, K. and Heward, C. (2005).

Oxidative stress profiling. Part II. Theory, technology, and practice. Ann. N.Y. Acad. Sci. 1055, 136-158.

59

Page 60: ~Chapter 1~ Introduction 1.1. Biomarkers

147. Cadenas, E. (1989). Biochemistry of oxygen toxicity. Annu. Rev. Biochem. 58, 79-110.

148. Maier, C. M. and Chan, P. H. (2002). Role of superoxide dismutase in

oxidative damage and neurodegenerative disorders. Neuroscientist 8, 323-334.

149. Park, J. L. and Lucchesi, B. R. (1999). Mechanisms of myocardial

reperfusion injury. Ann. Thorac. Surg. 68, 1905-1912. 150. Adibhatla, R. M. and Hatcher, J. F. (2006). Phospholipase A2, reactive

oxygen species, and lipid peroxidation in cerebral ischemia. Free Rad. Biol. Med. 40, 376-387.

151. Scheulen, M. E. and Kappus, H. (1992). Anthracyclines as model

compounds for cardiac toxicity in Tissue-Specific Toxicity: Biochemical Mechanisms. Dekant, W. and Neumann, H.-G. (Eds.). Academic Press, New York, 221-253.

152. Fraga, C. G., Shigenaga, M. K., Park, J.-W., Degan, P. and Ames, B. N.

(1990). Oxidative damage to DNA during aging: 8-hydroxy-2'-deoxyguanosine in rat organ DNA and urine. Proc. Natl. Acad. Sci. USA 87, 4533-4537.

153. Cordis, G. A., Bagchi, D., Maulik, N. and Das, D. K. (1994). High-

performance liquid chromatographic method for the simultaneous detection of malondialdehyde, acetaldehyde, formaldehyde, acetone and propionaldehyde to monitor the oxidative stress in heart. J. Chromatogr. A 661, 181-191.

154. Altschuld, R. A., Gibb, L. and Weissler, A. M. (1970). Metabolism of the

ischemic myocardium. Heart Bull. 19, 109-112. 155. Liedtke, A. J. (1981). Alterations of carbohydrate and lipid metabolism in

the acutely ischemic heart. Prog. Cardiovasc. Dis. 23, 321-336. 156. Calvani, M., Reda, E. and Arrigoni-Martelli, E. (2000). Regulation by

carnitine of myocardial fatty acid and carbohydrate metabolism under normal and pathological conditions. Basic Res. Cardiol. 95, 75-83.

157. Danesi, R., Fogli, S., Gennari, A., Conte, P. and Tacca, M. D. (2002).

Pharmacokinetic-pharmacodynamic relationships of the anthracycline anticancer drugs. Clin. Pharmacokinet. 41, 431-444.

60

Page 61: ~Chapter 1~ Introduction 1.1. Biomarkers

158. Minotti, G., Menna, P., Salvatorelli, E., Cairo, G. and Gianni, L. (2004). Anthracyclines: molecular advances and pharmacologic developments in antitumour activity and cardiotoxicity. Pharmacol. Rev. 56, 185-229.

159. Gewirtz, D. A. (1999). A critical evaluation of the mechanisms of action

proposed for the antitumour effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem. Pharmacol. 57, 727-741.

160. Pedrycz, A., Wieczorski, M. and Czerny, K. (2004). Increased apoptosis in

the adult rat liver after a single dose of adriamycin administration. Ann. Univ. Mariae Curie Sklodowska 59, 313-318.

161. Singal, P. W., Li, T., Kumar, D., Danelisen, I. and Iliskovic, N. (2000).

Adriamycin-induced heart failure: mechanism and modulation. Mol. Cell. Biochem. 207, 77-85.

162. Abdel-Aleem, S., El-Merzabani, M. M., Sayed-Ahmed, M., Taylor, D. A. and

Lowe, J. E. (1997). Acute and chronic effects of adriamycin on fatty acid oxidation in isolated cardiac myocytes. J. Mol. Cell. Cardiol. 29, 789-797.

163. Chaiswing, L., Cole, M. P., St. Clair, D. K., Ittarat, W., Szweda, L. I. and

Oberley, T. D. (2004). Oxidative damage precedes nitrative damage in adriamycin-induced cardiac mitochondrial injury. Toxicol. Pathol. 32, 536-547.

164. El-Gawad, H. M. A. and El-Sawalhi, M. M. (2004). Nitric oxide and

oxidative stress in brain and heart of normal rats treated with doxorubicin: role of aminoguanidine. J. Biochem. Molec. Toxicol. 18, 69-77.

165. Quiles, J. L., Huertas, J. R., Battino, M., Mataix, J. and Ramírez-Tortosa, M.

C. (2002). Antioxidant nutrients and adriamycin toxicity. Toxicol. 180, 79-95.

166. Behnia, K. and Boroujerdi, M. (1999). Inhibition of aldo-keto reductases by

phenobarbital alters metabolism, pharmacokinetics and toxicity of doxorubicin in rats. J. Pharm. Pharmacol. 51, 1275-1282.

167. Akaza, K., Nonami, T., Kurokawa, T., Kobayashi, H., Harada, A., Nakao, A.,

Sugiyama, S., Ozawa, T. and Takagi, H. (1996). Doxorubicin-induced disturbance of energy metabolism after hepatectomy. J. Surg. Res. 61, 454-458.

61

Page 62: ~Chapter 1~ Introduction 1.1. Biomarkers

168. Kalender, Y., Yel, M. and Kalender, S. (2005). Doxorubicin hepatotoxicity and hepatic free radical metabolism in rats: the effects of vitamin E and catechin. Toxicol. 209, 39-45.

169. Öz, E. and İlhan, M. N. (2006). Effects of melatonin in reducing the toxic

effects of doxorubicin. Mol. Cell. Biochem. 286, 11-15. 170. Bagchi, D., Bagchi, M., Hassoun, E. A., Kelly, J. and Stohs, S. J. (1995).

Adriamycin-induced hepatic and myocardial lipid peroxidation and DNA damage, and enhanced excretion of urinary lipid metabolites in rats. Toxicology 95, 1-9.

171. Kwiecień, I., Michalska, M. and Włodek, L. (2006). The selective effect of

cystathionine on doxorubicin hepatotoxicity in tumor-bearing mice. Eur. J. Pharmacol. 550, 39-46.

62

Page 63: ~Chapter 1~ Introduction 1.1. Biomarkers

~Chapter Two~

Tissue-Targeted Metabonomics Development: Analytical Considerations

2.1. Introduction

Tissue-targeted metabonomics provides an alternative to traditional

metabonomics sampling approaches using urine and plasma, which can be obtained

fairly noninvasively and in sufficient quantity and concentration for metabonomics

analytical techniques. Urine and plasma sampling, although popular, is not without

limitations. Time resolution can be limited in these studies, as discussed in Chapter

1. The potential for changes in sample composition due to bacterial growth (as in

urine) or enzymatic degradation (as in plasma) can also cause complications in

sample collection and data interpretation.

Most importantly for this research, sampling biofluids like urine and plasma

does not provide spatial information about metabolic changes. Many metabonomics

studies have made use of tissue biopsies to obtain localized metabolic information.

The tissues studied are diverse and have been derived from both animals and humans

for basic research and clinical applications. They include the liver1-4, kidney4,

brain1,5-7, heart8, prostate9, tumors9-11 and human gut biopsies12. One limitation of this

63

Page 64: ~Chapter 1~ Introduction 1.1. Biomarkers

sampling approach is the termination of the experiment after a biopsy is taken,

utilizing large numbers of animals to characterize metabolic differences and reducing

experimental precision due to animal-to-animal variability. Samples must either be

analyzed using a specialized NMR probe (high resolution magic angle spinning

probe) or the tissue must be homogenized and then undergo an extraction procedure.

A viable alternative to tissue biopsy is microdialysis sampling of extracellular fluid13-

16.

Microdialysis offers an inherent advantage for site-specific metabonomics

sampling. Careful selection of the molecular weight cutoff of the membrane can

selectively exclude macromolecules. Thus, samples are much cleaner and do not

require additional cleanup steps that reduce throughput and increase method error.

Microdialysis samples are typically small in volume (flow rates of ~ 1 μL/min, with

typical sampling times ranging from a few minutes to an hour). While not typically a

problem for metabolic profiling with mass spectrometry, this does require special

consideration when conducting NMR analysis, such as using tube inserts or low

volume probes.

For microdialysis studies, additional sample preparation is required prior to

NMR analysis. Dialysate samples are very low in volume and many components

border on the detection limits of the NMR instrument. Thus, any dilution becomes

problematic. One solution to this problem involves taking dialysate samples to

dryness and reconstituting in a deuterated solution to provide a lock signal and avoid

any dilution effects.

64

Page 65: ~Chapter 1~ Introduction 1.1. Biomarkers

There are two primary options for low volume NMR analysis: capillary tube

inserts and capillary probes or probes with reduced detection volumes. Capillary

tube inserts are made to sit inside a typical 5 mm NMR tube with the aid of a plastic

adapter. The capillary is submerged in sample-matching solvent (usually D2O) and

then the sample is centered in the active region of the magnet for analysis. Although

tube inserts do not provide an efficient filling factor, they allow the analysis of small

volume samples without specialized NMR probes. The low volume detection cells of

specialized capillary probes and nanoprobes can accommodate smaller sample

volumes and typically provide better mass sensitivity than capillary inserts, but they

can be more costly and require flexibility in the NMR setup so that the probe can be

changed.

2.2. Specific Aims

The purpose of this research was to develop and validate analytical

methodology to enable metabolic profiling by 1H-NMR of microdialysis samples.

Because this is a new approach to metabonomics studies, no literature exists detailing

appropriate methodology and characterizing the important analytical considerations

for these experiments. The analytical method involved three primary components

that were characterized in this study: microdialysis sampling, 1H-NMR detection and

multivariate statistical analysis by principal components analysis (PCA).

Microdialysis sampling considerations included probe calibration, as well as sample

preparation procedures that minimized sample composition changes but allowed for

65

Page 66: ~Chapter 1~ Introduction 1.1. Biomarkers

concentration of the sample and reconstitution in a deuterated buffer. 1H-NMR

analysis considerations primarily involved characterization of the small volume

detection scheme. Considerations for the multivariate statistical analysis involved the

integration of the NMR spectra, normalization of the integral data and optimization of

the principal components analysis parameters. Time trend analysis was also

performed on basal dialysate samples by both principal components analysis and

partial least squares regression.

2.3. Materials and Methods

2.3.1. Reagents and Solutions

All deuterated solvents were purchased from Cambridge Isotope Laboratories

(Andover, MA). Salts for Ringer’s solution and other reagents were purchased from

Sigma Chemical Corporation (St. Louis, MO). Ringer’s solution composition was

145 mM NaCl, 2.8 mM KCl, 1.2 mM CaCl2, and 1.2 mM MgCl2. Solutions were

prepared in nanopure water (18.2 MΩ resistivity, Labconco, Kansas City, KS) and

filtered through a 0.22 μm pore filter prior to use.

2.3.2. Microdialysis Methods

2.3.2.1. Microdialysis Probe Preparation and Calibration

All probes were prepared in-house, using polyacrylonitrile (PAN) membrane

(Hospal, Lakewood, CO) of dimensions 350 μm O.D. by 250 μm I.D. and polyimide

tubing (MicroLumen, Tampa, FL) of dimensions 170 μm O.D. by 120 μm I.D. All

probes used Tygon tubing attached at the inlet as a syringe adapter. Vascular probes

66

Page 67: ~Chapter 1~ Introduction 1.1. Biomarkers

67

were prepared with a 10 mm membrane length. Liver probes were linear and also had

a 10 mm membrane length. The outlet of the probe had a 7 cm piece of fused silica

capillary (360 μm O.D. by 200 μm I.D.) attached to the polyimide to use as a needle

guide. Heart probes were linear and were approximately 2 mm in membrane length.

A 1 mm long Tygon tubing “stop” was secured directly behind the active window of

the membrane to aid in implantation of the probe. The outlet of the probe had a 27

gauge stainless steel needle (bent to 45°) attached to aid in implantation. Both the

liver and heart probe are pictured in Figure 2.1. All probes were perfused with

Ringer’s solution at a rate of 1 μL/min and were flushed for 30 minutes prior to the

start of sampling. Samples were collected over 60 minute intervals (producing 60 μL

sample volumes) unless explicitly noted.

Probes were calibrated continuously through the experiment by delivery of 7

μM antipyrine in Ringer’s solution17,18. Antipyrine is an established marker of

microdialysis membrane extraction efficiency18 and will account for global changes

in probe performance. The concentration of antipyrine was chosen by the maximum

concentration that could be used without detection by NMR, to avoid interferences

with endogenous metabolite resonances. Antipyrine was detected using LC/UV at

254 nm (Shimadzu, Columbia, MD). The isocratic LC system was equipped with a

Shimadzu LC-10AD pump and SPD-10AV UV-Vis detector, which was integrated to

EZStart software (version 7.3, Shimadzu Scientific Instruments, Inc., Kyoto, Japan)

with an SCL-10AVP system controller. A Rheodyne 7725i injector fitted with a 25

μL PEEK loop was also used. Samples were injected onto a Phenomenex Synergi

Page 68: ~Chapter 1~ Introduction 1.1. Biomarkers

68

Figure 2.1. Microdialysis probe design. A shows a linear microdialysis probe used for liver microdialysis, with an active membrane length of 10 mm. B shows a linear microdialysis probe used for heart microdialysis, with an active membrane length of 2 mm and a Tygon stop to aid in implantation.

A

B

A

B

Page 69: ~Chapter 1~ Introduction 1.1. Biomarkers

Polar-RP column of 4 μm particle size, of dimensions 2 mm x 150 mm. The mobile

phase was acetonitrile-sodium phosphate (pH 2.5, 25 mM) (17:83, v/v) and was set to

a flow rate of 0.3 mL/min.

2.3.2.2. Animal Setup

All experiments utilized male Sprague-Dawley rats (Sasco, Wilmington, MA),

weight range 350-450 g. Rats were pre-anesthetized by isoflurane inhalation and then

fully anesthetized with a ketamine (67.5 mg/kg)/xylazine (3.4 mg/kg)/acepromazine

(0.67 mg/kg) cocktail, administered subcutaneously. For nonsurvival surgeries, rats

were maintained under anesthesia for the duration of all experiments, using ketamine

booster doses administered as needed intramuscularly at one quarter of the initial

dose. Rats undergoing survival procedures were only given the initial anesthesia

cocktail, which maintained anesthesia for the duration of probe implantation. Rat

body temperature was maintained at 37°C using an electric heating pad.

For survival surgeries, all procedures were performed aseptically. Rats were

given 3 mL saline subcutaneously after the surgery to facilitate recovery and were

housed in a Raturn© system (BASi, West Lafayette, IN). No buprenorphine

analgesic was given to avoid problematic perturbations in basal tissue metabolism.

All animal experiments were performed under approval of the local Institutional

Animal Care and Use (IACUC) committee.

2.3.2.3. Jugular Probe Implantation

The jugular vein was exposed through a small midline incision in the neck

skin tissue. The vein was isolated by separating the vessel from fine connective

69

Page 70: ~Chapter 1~ Introduction 1.1. Biomarkers

70

tissue using blunt dissection and cotton swabs. A flat metal spatula was placed under

the vein to externalize it, and a small incision made in the vessel using a pair of fine

spring scissors. A vascular probe with an introducer was inserted into the vein

toward the heart. Silk sutures were used to secure the probe in the vessel, and the

introducer was then removed. (Surgery was performed by Shannon S. Vandaveer.)

2.3.2.4. Heart Probe Implantation

Pictures to illustrate the surgical procedure are shown in Figure 2.2. A

tracheotomy was performed by externalizing the trachea through an incision on the

midline of the neck. The trachea was isolated using a spatula and any fluid around

the trachea was removed with cotton swabs (Figure 2.2A). A small nick was made

between the rings of the trachea and an intubation tube was immediately inserted and

secured with sutures. The rat was attached to a Digi-Med Sinus Rhythm Analyzer

(Louisville, KY) to monitor heart rate and sinus rhythm. Immediately prior to

opening the chest cavity, the intubation tube was connected to a respirator that

supplied artificial ventilation for the rat. A constant-volume respirator using room air

(Model 683 Rodent Respirator, Harvard Apparatus, Holliston, MA) was used. To

expose the heart, a thoractomy was performed on the left side of the animal by

making an incision between and parallel to the fifth and sixth ribs approximately 1.5

to 2 cm in length (Figure 2.2B), and the pericardium was opened, allowing the

microdialysis probe to be inserted into the myocardium. Specifically, a bent 27 gauge

needle attached to the probe was used to implant the probe into the apex of the

beating heart muscle, near the left descending coronary artery along the longitudinal

Page 71: ~Chapter 1~ Introduction 1.1. Biomarkers

Figure 2.2. Progression of microdialysis probe implantation in the heart. A shows the externalization and ventilation of the trachea. B shows the rat being prepared for the opening of the chest cavity. The rat has been connected to electrodes on three of its legs to monitor the heart rhythm, and an incision has been made in preparation for the thoractomy. C shows the chest cavity open immediately prior to implantation. The lungs have been retracted. The cotton swab in the left hand is used to expose the beating heart for implantation and the forceps in the right hand are holding the bent needle used as an implantation guide for the probe.

A

B

C

A

B

C

71

Page 72: ~Chapter 1~ Introduction 1.1. Biomarkers

72

axis of the heart (Figure 2.2C). The lungs were then fully expanded by blocking the

exhalation tube on the rodent respirator for several seconds. (Some of the surgeries

were performed by Sara L. Thomas.)

2.3.2.5. Brain Probe Implantation

The animal was positioned in a stereotaxic apparatus with the incisor bar set

to level the head between the lambda and bregma fissure lines of the skull. A 1 cm

midline incision was made in the scalp. The incised skin was gently separated from

the skull and then spread open. The exposed scalp was cleaned and dried. A guide

cannula was implanted into the striatum (stereotaxic coordinates from bregma:

anterior 1.0 mm, lateral 2.7 mm, vertical -3.6 mm). Two jeweler’s screws were

partially screwed into the skull on either side of the cannula to act as an anchor for the

dental cement, which was used to fix the cannula to the skull. The incision was

stitched around the dental cement. After implantation, the guide cannula was

replaced with the microdialysis probe, which had a 4 mm active polycarbonate

membrane (CMA Microdialysis AB, Sweden). (Surgery was performed by Shannon

S. Vandaveer.)

2.3.2.6. Liver Probe Implantation

Pictures to illustrate the surgical procedure are shown in Figure 2.3. A 2 cm

incision was made along the midline of the abdomen directly below the diaphragm

and xyphoid process, and the abdominal cavity was opened. The incision was spread

with Bowman retractors and tissue forceps were used to expose the median liver lobe

from beneath the ribs (Figure 2.3B). Two microdialysis probes were implanted using

Page 73: ~Chapter 1~ Introduction 1.1. Biomarkers

Figure 2.3. Progression of microdialysis probe implantation in the liver (survival surgery). A shows the rat prepared prior to surgery. The abdomen has been shaved and cleaned and placed on a heating pad. Immediately after this, the rat was covered with a sterile drape. B shows the midline incision made in the abdomen, with the median liver exposed using tissue forceps. C shows the fused silica in the liver tissue used as the probe implantation guide. D shows the muscle layer being stitched up, with the probe tubing externalized through the incision. E shows the trocar used to tunnel the probe tubing from the abdominal incision out the back of the neck, and F is the closing of the abdominal incision. G shows the rat emerging from anesthesia in the Raturn® bowl. The jacket is utilized to attach the rat to the harness, which signals rat activity and turns the bowl appropriately.

A

B

C

D

E

F

G

A

B

C

D

E

F

G

73

Page 74: ~Chapter 1~ Introduction 1.1. Biomarkers

74

the fused silica capillary as a guide needle (Figure 2.3C). The probes were implanted

1 cm apart, which has been shown by previous studies to be a sufficient distance to

prevent cross-talk between the probes19. The tissue entrance and exit points for each

probe were secured with a small amount of tissue glue applied with a plastic transfer

pipet. After probe implantation, the incision was closed with sutures, with the probe

inlets and outlets externalized through the incision (Figure 2.3D). For survival

surgeries, the probe tubing was tunneled underneath the skin and externalized through

a small incision in the back of the neck (Figure 2.3E). The muscle layer incision was

closed with sutures and the skin was closed with staples (Figure 2.3F).

2.3.2.7. Microdialysis Sample Preparation

All dialysate samples (60 μL original volume) were split into two fractions –

one 50 μL fraction for NMR analysis and one 10 μL fraction for LC/UV analysis of

antipyrine. Fractions for NMR analysis were speedvacuumed to dryness at room

temperature after collection and stored at -20°C in the dark until analysis.

Immediately prior to NMR analysis, the sample was thawed to ambient temperature

and reconstituted in 25 μL of 25 mM deuterated acetate solution in D2O, pD = 7.8,

with 5 mM trimethylsilyl propionic acid (TSP-d4) as an internal standard. Samples

were vortexed for 30 seconds and then transferred to a linear capillary tube insert

(Wilmad-Labglass, Buena, NJ) using a 10 μL syringe with a 10 cm fused silica

needle. The insert was submerged using a PTFE adapter in 600 μL D2O in a 5 mm

NMR tube. A picture of the capillary tube setup is shown in Figure 2.4. The

capillary was filled with 25 μL of a fluorescein solution to aid in visualization.

Page 75: ~Chapter 1~ Introduction 1.1. Biomarkers

75

Figure 2.4. Capillary tube insert in a 5 mm NMR tube.

Page 76: ~Chapter 1~ Introduction 1.1. Biomarkers

76

2.3.3. NMR Spectroscopy

1H-NMR analysis was performed on a Varian INOVA 600 MHz spectrometer

equipped with VnmrJ software (Version 2.1 Revision B, Varian Instruments Inc.,

Palo Alto, CA) at 25ºC. Samples were analyzed on a 5 mm Varian triple-axis

gradient probe. All samples were analyzed with the WET water suppression pulse

sequence. A tip angle of 90º was utilized, with an acquisition time of 1.8 sec and

25200 points. The pulse sequence is shown in Figure 2.5. Spectra were coadded for

3000 transients for a total acquisition time of 95 minutes, zero-filled to 65536 points,

and a line broadening of 1 Hz was applied. All spectra were referenced to TSP-d4 at

0.00 ppm. Resonances were assigned by comparison to standard spectra measured in

the sample solvent solution and literature chemical shift values.

2.3.4. Data Analysis

NMR spectra were exported to ACD/Labs software (Version 10.02, Advanced

Chemistry Development, Inc., Toronto, Ontario) and spectral processing (zero-filling,

line-broadening, baseline correction, and referencing) was performed as described

above. Spectra were manually integrated over 40 defined integral regions between

0.8 and 4.5 ppm, and integral tables were exported as a spreadsheet for data

manipulation. (For sample stability analyses, spectra were manually integrated over

57 defined integral regions.) Integral values were normalized to the TSP-d4 integral

and concentration, as well as the extraction efficiency of the corresponding

microdialysis probe, as measured by the % delivery of antipyrine. Analysis by

principal components, partial least squares regression (PLS) and univariate methods

Page 77: ~Chapter 1~ Introduction 1.1. Biomarkers

77

Figure 2.5. WET pulse sequence parameters.

Tx

Z

1.8 sec

8.1 μsec

wet_1160 wet_1160 wet_1160 wet_1160

32.0 msec 32.0 msec 32.0 msec 32.0 msec

2.0 msec 2.0 msec 2.0 msec 2.0 msec1.0 msecd1 gswet gswet gswet gswet pw

2.0 msec

2.0 msec2.0 msec

2.0 msec

gtw gtw gtw gtw

10 μsecdz

Tx

Z

1.8 sec

8.1 μsec

wet_1160 wet_1160 wet_1160 wet_1160

32.0 msec 32.0 msec 32.0 msec 32.0 msec

2.0 msec 2.0 msec 2.0 msec 2.0 msec1.0 msecd1 gswet gswet gswet gswet pw

2.0 msec

2.0 msec2.0 msec

2.0 msec

gtw gtw gtw gtw

10 μsecdz

Page 78: ~Chapter 1~ Introduction 1.1. Biomarkers

78

were performed with Minitab® Statistical Software (Release 14.20, Minitab, Inc.,

State College, PA). Principal components analysis was performed on a correlation

matrix (data matrix was centered and standardized), with five principal components

calculated. Partial least squares regression between time and metabolic profiles was

also performed on a correlation matrix, with a maximum of five components

calculated. The PLS model was cross-validated through jackknifing.

2.4. Results and Discussion

2.4.1. Microdialysis Sampling

2.4.1.1. Comparison of 1H-NMR Spectra of Plasma and Plasma Dialysate

To illustrate the differences between conventional metabonomics data and

tissue-targeted metabonomics data, a comparison of 1H-NMR spectra of rat plasma

and plasma dialysate taken from the same rat is shown in Figure 2.6. D2O was added

to the plasma to a final concentration of 10% to provide a lock signal, while the

plasma dialysate was prepared as described in the methods (Section 2.3.2.7). The

plasma, as expected, showed underlying broad signals throughout the spectrum due to

proteins and lipids. Specifically, much of the macromolecule signal in plasma comes

from albumin, immunoglobulins, N-acetylated sugars on glycoproteins, lipoproteins,

triglycerides and chylomicrons20. Very few small molecule resonances are detectable

above this background. The major visible sharper resonances in the region from 3.1

to 3.9 ppm correspond to glucose and the sharp singlet at 3.0 ppm is due to creatine.

Page 79: ~Chapter 1~ Introduction 1.1. Biomarkers

79

Figure 2.6. Comparison of the 1H-NMR spectra of (A) plasma and (B) plasma dialysate.

kepplasmapresat072704

8 7 6 5 4 3 2 1 07

8 7 6 5 4 3 2 1 0012345678

B

Chemical Shift (ppm)

A

68 5 4 3 2 1 0

kepplasmapresat072704

8 7 6 5 4 3 2 1 07

8 7 6 5 4 3 2 1 0012345678

B

Chemical Shift (ppm)

A

68 5 4 3 2 1 0

Page 80: ~Chapter 1~ Introduction 1.1. Biomarkers

Other small molecule resonances may be present, but are not sufficiently above the

background for positive identification.

In contrast, the plasma dialysate NMR sample was entirely devoid of any

macromolecule signal. Glucose and creatine resonances are present at the frequencies

indicated above. Lactate, alanine, lysine, valine, leucine, isoleucine, citrate,

tyrosine,and many other small molecules are detectable. The largest differences

between the spectra are noted in the more aliphatic regions (0.5-2.5 ppm). This

region contains resonances from many amino acids, which are key substrates in or

have strong influences on metabolic pathways such as glycolysis, the citric acid cycle,

the urea cycle, glucose-alanine cycle, and many others. Thus, this is a critical area of

the spectrum to characterize in tissue metabolic profiling, which has been achieved

through 1H-NMR analysis of microdialysis samples. In the dialysate NMR spectra,

spectral resolution is high, indicating that the use of inserts for NMR analysis of small

volume dialysate samples does not compromise the quality of the NMR spectrum.

2.4.1.2. Probe Calibration by Delivery of Antipyrine

Quantitation of all the metabolites in any metabolic profile of a dialysate

sample is extremely difficult. To truly quantitate levels of a metabolite in the

extracellular fluid of the tissue surrounding the probe, it is ideal to calibrate the probe

in vivo prior to the start of the experiment with the same compound. Due to the

nature of metabonomics studies, in which analytes are not identified prior to sample

analysis, probe calibration for each individual metabolite is precluded because all the

metabolites cannot be identified prior to analysis. In addition, it would be logistically

80

Page 81: ~Chapter 1~ Introduction 1.1. Biomarkers

81

difficult to calibrate a probe for a large number of metabolites in a single experiment.

However, exact quantitation is not necessarily the goal of these studies – rather, it is

the relative change from baseline levels. This makes probe calibration by delivery a

good option for metabonomics studies using microdialysis sampling. In this method,

the delivery of a standard compound (antipyrine is commonly used) through the

microdialysis probe to the tissue has been shown to be equivalent to the recovery of

the same compound from the tissue to the probe lumen. Thus, the use of a single

marker compound to account for changes in extraction efficiency of many

endogenous small molecules is a simple and efficient way to account for variability in

the data due to changes in probe recovery. Antipyrine was detected in dialysate

samples using the LC/UV method detailed in section 2.3.2.1. Figure 2.7 shows the

separation of antipyrine from endogenous components in basal liver dialysate. In

general, probe extraction efficiency remained constant over the course of the

experiment, but did show variance from rat to rat. Section 2.4.4.1.2 discusses the use

of probe extraction efficiency to remove extraneous variance from the metabonomics

data set.

2.4.2. Sample Preparation Effects and Stability

2.4.2.1. Speedvacuuming Effects

Dialysate samples are primarily collected into an isotonic Ringer’s solution.

Although it is possible to effectively suppress the water resonance in solutions that

are predominately (i.e., 90%) protic water, any analyte resonances within this region

of the spectrum are also suppressed. More significantly, the residual water resonance

Page 82: ~Chapter 1~ Introduction 1.1. Biomarkers

82

Figure 2.7. LC/UV chromatogram of antipyrine in liver dialysate. A shows the chromatogram at full scale and B depicts the same chromatogram at a lower scale to magnify lower intensity peaks. Antipyrine elutes at approximately 7.1 minutes.

1 mV

10 mVA

1 2 3 4 5 6 7 8

Time (minutes)

B 1 mV

10 mV10 mVA

1 mV

1 2 3 4 5 6 7 8

Time (minutes)1 2 3 4 5 6 7 8

Time (minutes)

B

Page 83: ~Chapter 1~ Introduction 1.1. Biomarkers

may distort the spectral baseline, affecting analyte quantitation. For dilute solutions,

the residual water signal may dominate the spectrum, limiting the dynamic range of

the measurement. Therefore, we chose to reduce the water resonance by drying the

dialysate sample and reconstituting in a deuterated solution. This approach is also

advantageous because the sample could be concentrated, improving sample

throughput by reducing the time required for signal-averaging. However, it is

possible that speedvacuuming could alter sample composition.

For this reason, the effect of speedvacuuming on the composition of heart and

plasma dialysate samples was determined. Samples were split into two fractions,

with one frozen at -20ºC and the other speedvacuumed to dryness before storage at -

20ºC. All sample pairs were analyzed by NMR on the same day. Immediately prior

to NMR analysis, frozen samples were diluted to a final TSP-d4 concentration of 5

mM and deuterated acetate concentration of 25 mM. Speedvacuumed samples were

reconstituted in 25 mM deuterated acetate solution in D2O with 5 mM TSP-d4 to the

same final volume as the speedvacuumed samples.

Visual inspection of the spectra revealed few differences between the sample

preparation methods. For heart and plasma dialysate, the carbohydrate regions (< 3

ppm) and aromatic regions (> 5 ppm) showed no detectable differences in both

resonance intensity and resonances present. For both dialysates, small differences

were noted in the 3 – 4.5 ppm region, primarily containing glucose resonances.

The normalized integrals from each sample were averaged for the frozen

samples and for the speedvacuumed samples. A student’s t-test was performed on the

83

Page 84: ~Chapter 1~ Introduction 1.1. Biomarkers

84

two means for each integral region. Only one out of 40 integral regions showed

significant differences between the speedvacuumed and frozen heart dialysate

samples at 95% confidence: 3.22-3.29 ppm. For the plasma dialysate samples, one

region was significant at 95% confidence: 3.75-3.77 ppm. Both of these regions are

dominated by glucose resonances, but do overlap other metabolite resonances.

Principal components analysis (Figure 2.8) showed a clear segregation

between the plasma dialysate and heart dialysate samples (with one set of plasma

dialysate samples appearing as an outlier). No segregation was observed between

dialysate samples that were directly frozen and those that were speedvacuumed

before freezing. Although segregation was observed between heart and plasma

dialysate as expected (with one outlying sample), no segregation between sample

preparation methods was observed within clusters of dialysate types. Therefore, the

scores plot in Figure 2.8 suggests that speedvacuuming did not significantly alter the

sample composition in either plasma or heart dialysate. However, it should be noted

that sample composition is not exactly the same, as corresponding frozen and

speedvacuumed sample points do not overlap in the score plot. Measurement error is

also reflected in the scores plot in Figure 2.8, accounting for some of the lack of

overlap of sample pairs. Thus, speedvacuuming alters dialysate composition slightly,

but not in a large and systematic manner as would be indicated by clustering in the

scores plot. The univariate analysis and spectral examination detailed previously

confirmed these conclusions. Although it is apparent that speedvacuuming does alter

dialysate composition slightly due to loss of volatile components (primarily acetate)

Page 85: ~Chapter 1~ Introduction 1.1. Biomarkers

85

Figure 2.8. Score plot comparing sample preparation methods for dialysate samples. ♦ represents samples speedvacuumed to dryness and reconstituted in deuterated solution immediately prior to NMR analysis. ∇ represents samples frozen prior to NMR analysis and then diluted to 10% D2O. Each point is labeled with its individual sample number, and lines are drawn to connect sample pairs visually.

-6 -4 -2 0 2 4 6 8 10

-6

-4

-2

0

2

4

6

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Heart Dialysate

Plasma Dialysate2

1

2

3

1

3

12

4

3

41

2

3

-6 -4 -2 0 2 4 6 8 10

-6

-4

-2

0

2

4

6

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Heart Dialysate

Plasma Dialysate2

1

2

3

1

3

12

4

3

41

2

3

-6 -4 -2 0 2 4 6 8 10

-6

-4

-2

0

2

4

6

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Heart Dialysate

Plasma Dialysate2

1

2

3

1

3

12

4

3

41

2

3

Page 86: ~Chapter 1~ Introduction 1.1. Biomarkers

or possible degradation of unidentified unstable compounds, the changes it causes are

confined to a small region of the NMR spectrum. Speedvacuuming dialysate samples

also improves sample stability and spectral quality by improving water suppression.

Although reconstitution in D2O can cause loss of resonances from exchangeable

protons, in general for small organic molecules there is less information lost by this

route than by suppression of the solvent resonance in 90% H2O.

2.4.2.2. Recovery by Dissolution

Biofluid composition is complex, especially plasma, whose components have

varying degrees of solubility in aqueous solution. Additionally, many of the dialysate

samples collected for this project were speedvacuumed to dryness and reconstituted

in half the original volume, potentially exacerbating any solubility issues.

The recovery of plasma dialysate components by reconstitution in Ringer’s

solution was examined. Plasma dialysate was collected and speedvacuumed to

dryness. The plasma was reconstituted in deuterated acetate solution in D2O

(pD=7.8) and analyzed by NMR. Any solution remaining in the sample vial was

removed and the same volume of methanol-d4 was added to solubilize any residual

metabolites that were not dissolved when the plasma dialysate was initially

reconstituted. The NMR spectrum measured for this residual metabolite solution was

compared to a control methanol-d4 spectrum and to the original plasma dialysate

spectrum. Only solvent resonances were detected in the residual metabolite solution,

indicating that no detectable levels of metabolites were left undissolved by the

86

Page 87: ~Chapter 1~ Introduction 1.1. Biomarkers

reconstitution buffer. No solubility concerns were noted with the dialysate sample

preparation procedure.

2.4.2.3. pH Effects on Dissolution

In NMR experiments, solution pH is an important concern, as many

resonances have a pH-dependent chemical shift. In metabonomics NMR experiments

in particular, the choice of an appropriate pH for the sample as well as control of the

pH with a buffer is important. Metabonomics NMR spectra tend to be crowded with

hundreds of resonances, so even small changes in the chemical shift can change the

appearance of the spectrum. When applying bucketing procedures for statistical

analysis, the width of the buckets is typically so small (0.04 ppm) even a small

change in pH can easily shift resonances between buckets. Therefore, the

reproducibility of NMR spectra generated from the same sample at a normal in vivo

pH and low pH was examined.

The purpose of this experiment was to examine the effect of pH on the NMR

spectra of plasma dialysate. Two separate plasma dialysate samples (speedvacuumed

to dryness after collection and stored at -20ºC until analysis) were reconstituted in 25

mM deuterated acetate in D2O, one with a pD of 7.8 and the other with a pD of 3.4.

Spectra were examined for the shifting or disappearance of resonances. No major

changes were noted when comparing the spectra. Most changes occurred in

glutamate resonances, with shifts of 0.02 ppm and 0.04 ppm occurring at the

resonances at 2.43 ppm and 2.09 ppm, respectively. Overall, the pD had a minimal

effect on the NMR spectrum, with an average change in ppm of 0.009 over

87

Page 88: ~Chapter 1~ Introduction 1.1. Biomarkers

88

approximately 4 pH units. Therefore, for dialysate samples reconstituted in buffer,

small changes in pH should minimally change the NMR spectrum and therefore

integration parameters.

2.4.2.4. Speedvacuumed Sample Stability

The storage conditions of the speedvacuumed samples were important in that

the need for long NMR acquisition times to allow for signal averaging combined with

the limitations on instrument access limit the number of samples that can be analyzed

in a single day, requiring that samples from the same set be stored and analyzed on

different days. It was therefore necessary to establish the stability window of the

dialysate samples.

The appropriate storage conditions of the speedvacuumed samples were

assessed. Plasma dialysate samples were split into equal volume fractions and all

samples were speedvacuumed to dryness. Half of the speedvacuumed samples were

stored at -20ºC and the other half were stored at 4ºC. All samples were reconstituted

in equal volumes of 25 mM deuterated acetate solution in D2O with 5 mM TSP-d4

immediately prior to NMR analysis. Samples from each storage condition were

analyzed on days 0 (immediately after speedvacuuming to determine the initial

sample composition), 1, 2 and 7.

Results showed a clear demonstration of sample instability for some

components at 4ºC and improved sample stability at -20ºC. Figure 2.9 shows

resonances from 3.0-4.5 ppm, where resonances from glucose tend to dominate the

spectrum. Figure 2.10 shows resonances from 0.5-3.0 ppm, containing the aliphatic

Page 89: ~Chapter 1~ Introduction 1.1. Biomarkers

89

Figure 2.9. Comparison of NMR spectra acquired from speedvacuumed and reconstituted plasma dialysate samples stored at two different temperatures. A shows the spectrum of the original plasma dialysate acquired directly after sampling and speedvacuuming. B shows the spectrum acquired from the plasma dialysate sample stored in the refrigerator (4°C) for 7 days prior to analysis. C shows the spectrum acquired from the plasma dialysate sample stored in the freezer (-20°C) for 7 days prior to analysis.

A

B

C

3.03.33.63.94.24.5

1H-NMR Chemical Shift (ppm)

A

B

C

3.03.33.63.94.24.5

1H-NMR Chemical Shift (ppm)

Page 90: ~Chapter 1~ Introduction 1.1. Biomarkers

90

Figure 2.10. Comparison of NMR spectra acquired from speedvacuumed and reconstituted plasma dialysate samples stored at two different temperatures. A shows the spectrum of the original plasma dialysate acquired directly after sampling and speedvacuuming. B shows the spectrum acquired from the plasma dialysate sample stored in the refrigerator (4°C) for 7 days prior to analysis. C shows the spectrum acquired from the plasma dialysate sample stored in the freezer (-20°C) for 7 days prior to analysis.

1.0

1.52.0

A

B

C

A

B

C

2.5

1H-NMR Chemical Sh

1.52.0

ift (ppm)

1.02.5

1H-NMR Chemical Shift (ppm)

Page 91: ~Chapter 1~ Introduction 1.1. Biomarkers

resonances of many amino acids and other metabolites. Aliphatic resonances showed

substantial changes seven days after storage at 4ºC. Although some resonances (such

as alanine) showed a decline in relative intensity, many new resonances appeared.

Minor changes in the frozen samples were noted, although these were smaller relative

to changes observed in the refrigerated samples. Specifically, six out of 57 integral

regions were considered significantly different at 95% confidence when comparing

the frozen samples analyzed on the 7th day to the original sample integral values. In

contrast, 18 of 57 integral regions were considered significantly different at 95%

confidence when comparing the refrigerated samples analyzed on the 7th day to the

original sample integral values. Most of the changes in sample composition for both

frozen and refrigerated samples were observed in the 3.0 – 4.0 ppm region, which

primarily contains glucose resonances. Upon inspection of the NMR spectra, the

levels of glucose stayed constant. The differences observed are due to unidentified

resonances growing into the spectra as time elapsed. Also, when comparing the

integral values for frozen and refrigerated samples analyzed on the 7th day, 23 integral

regions were considered significantly different at 95% confidence. The significant

integral regions were also primarily found in the 3.0 – 4.0 ppm region. The exact

integral regions showing significant differences are listed in Table 2.1.

In summary, these comparative studies showed instability in sample

composition at 4°C, but improved stability at -20°C. It should be noted that changes

in sample composition were observed in the frozen samples. Although minor, they

could still alter the integral values obtained in subsequent experiments. In this study,

91

Page 92: ~Chapter 1~ Introduction 1.1. Biomarkers

Original/Frozen Original/Refrigerated Frozen/Refrigerated 1.06-1.09 1.06-1.09 1.09-1.18 1.09-1.18 1.18-1.23 1.18-1.23 1.91-1.93 2.39-2.47 2.74-3.00 2.74-3.00 3.06-3.17 3.06-3.17 3.17-3.22 3.17-3.22 3.22-3.28 3.36-3.39 3.36-3.39

3.39-3.41 3.39-3.41 3.39-3.41 3.41-3.43 3.41-3.43 3.41-3.43

3.44-3.46 3.44-3.46 3.50-3.52 3.50-3.52 3.52-3.54 3.54-3.56 3.54-3.56

3.56-3.58 3.56-3.58 3.70-3.73 3.70-3.73 3.72-3.75 3.77-3.80 3.80-3.85 3.85-3.87

3.87-3.90 3.93-3.94 3.93-3.94 3.94-4.09 3.94-4.09

4.09-4.15 4.09-4.15 4.09-4.15 6.80-8.00

Table 2.1. Integral regions significantly different between the original, frozen and refrigerated sample groups, as determined by the student’s t test at a 95% confidence level. Not all the possible integral regions are shown in the table – only those shown to cause significant differences between the sample groups.

92

Page 93: ~Chapter 1~ Introduction 1.1. Biomarkers

both frozen and refrigerated samples were analyzed 1, 2 and 7 days after analysis of

the original sample. The day 1 and 2 spectra acquired from frozen samples were very

similar in composition to the original sample, suggesting that it is only after a period

of a few days that sample composition changes significantly in the frozen samples.

Thus, speedvacuumed samples were frozen until NMR analysis, and samples from

the same experiment were analyzed within one day of each other to avoid introducing

variance from sample stability.

2.4.3. NMR Analysis

The analysis of small volume samples can be challenging for NMR

experiments. Conventional NMR analysis uses samples of about 600 μL for normal 5

mm NMR tubes and about 220 μL for specialized 5 mm Shigemi tubes. (Shigemi

NMR tubes reduce the required sample volume by utilizing glass plugs in the tube

whose magnetic susceptibility is matched to the sample solvent. The sample volume

needs only to fill the active window of the probe.) Smaller diameter tubes, typically 3

mm in diameter, can also be used. Conventional 3 mm tubes require about 230 μL

for analysis and 80 μL for specialized Shigemi tubes. Even the smaller diameter

tubes require sample volumes that would yield poor time resolution (a few hours) for

microdialysis sampling (using typical μL/min flow rates). One final alternative

utilizes microcell assemblies that insert smaller tubes inside conventional 5 mm NMR

tubes, such as those sold by Wilmad-Labglass, Inc (Figure 2.4). These inserts come

in a variety of geometries, including cylindrical (8 μL/cm volume), spherical bulb (18

μL total volume) and cylindrical bulb (110 μL total volume). Cylindrical inserts were

93

Page 94: ~Chapter 1~ Introduction 1.1. Biomarkers

chosen because sample insertion and shimming were easier with this insert probe

geometry.

Specialized NMR probes are an alternative to reduce the sample volume

requirements of the NMR experiment. A microcoil probe (CapNMR probe produced

by Protasis/MRM) used in these experiments was composed of a length of capillary

with a bubble cell. The observe volume of this bubble cell was 1.5 μL and total probe

volume was approximately 10 μL. Thus, sample volume requirements are reduced

significantly when compared to traditional NMR analysis. A further advantage of

microcoil NMR analysis is increased mass sensitivity, meaning that given the same

mass of analyte detected, the microcoil probe will give a larger response than a

traditional 5 mm NMR probe21. Another study of microdialysis sampling for

metabonomics studies used a Varian nanoprobe cell15, which required 20 μL sample

volumes.

A comparative experiment was conducted to characterize the analysis of

microdialysis samples by NMR using both specialized tube inserts and a specialized

microcell probe. Plasma dialysate samples were collected and split into two equal

volume fractions. The samples were speedvacuumed to dryness and reconstituted in

the same volume of deuterated acetate solution. For each sample pair, one sample

was placed in a cylindrical tube insert in a conventional 5 mm NMR tube (Figure

2.4). The sample was analyzed as usual on a triple-axis gradient probe. The other

sample was injected into the capillary microcell probe until the flow cell was

completely filled with sample. Samples were acquired using WET water suppression

94

Page 95: ~Chapter 1~ Introduction 1.1. Biomarkers

95

with a 90° tip angle calibrated for each probe. Spectra were processed as described in

the methods and visually compared.

The results of the comparative study are shown in Figure 2.11. No significant

differences between sample analysis methods were noted from 3.0-4.5 ppm.

However, differences were noted in other regions of the spectra. First, in Figure 2.11,

resonances from the capillary microcell probe are broader than those analyzed using

tube inserts. This is not necessarily observed in all resonances – for example, 7.75

ppm shows sharp resonances for both analytical approaches. The shims were similar

between the two analytical approaches, as judged by the linewidth of the internal

standard, TSP-d4. So, the broad resonances are likely not due to poorer shimming of

the probe, but perhaps solute interactions with the surface of the capillary. Both A

and B in Figure 2.11 also show differences in resonances detected, both in the

appearance of new resonances in the samples analyzed by the microprobe and in

differences in relative intensity of various resonances. New resonances are noted at

2.20 and 7.75 ppm. Changes in relative resonance intensity were noticed in the

region of 6.75-7.35 ppm, as well as the doublet next to lactate (1.30 ppm), alanine

(1.45 ppm), and the mixture of resonances at 2.10 ppm.

The results from this comparative study indicate that the use of tube inserts for

NMR analysis of microdialysis samples is a superior alternative to the use of capillary

microprobes for this specific application. Interactions of the sample with the

capillary wall resulted in broadening of some of the resonances, therefore poorer

spectral quality was obtained. Although the capillary microprobes offer superior

Page 96: ~Chapter 1~ Introduction 1.1. Biomarkers

96

Figure 2.11. Comparison of plasma dialysate analysis by (1) a cylindrical tube insert with a conventional triple-axis gradient probe and (2) a capillary microprobe. Results are representative of the experimental results. A shows resonances from 0.75-3.0 ppm and B shows resonances from 6.0-9.0 ppm.

2.5 2.0 1.5 1.0

8.5 8.0 7.5 7.0 6.5 6.0

1H Chemical Shift (ppm)

A

B

1

2

1

2

2.5 2.0 1.5 1.02.5 2.0 1.5 1.0

8.5 8.0 7.5 7.0 6.5 6.08.5 8.0 7.5 7.0 6.5 6.0

1H Chemical Shift (ppm)

A

B

1

2

1

2

Page 97: ~Chapter 1~ Introduction 1.1. Biomarkers

detection volumes relative to the NMR tube inserts, the disadvantages preclude their

use in these studies. Not only is the spectral quality problematic, the flow probe

format of the microprobe also reduces the flexibility in the NMR setup. In contrast,

samples in tube inserts can be analyzed using nearly any probe, including cryoprobes.

Although tube inserts do not provide as efficient a filling factor as the capillary

probes, they allow the analysis of small volume samples without specialized NMR

probes and generate spectral data of superior quality in this application.

2.4.4. Data Analysis

2.4.4.1. Data Pretreatment

Once the NMR spectrum is acquired, steps must be taken to translate the

spectral domain data into a form more appropriate for statistical analysis.

Interpretation of large metabonomics data sets can be cumbersome when considered

in a univariate manner. Multivariate statistical methods such as principal components

analysis (PCA) have been a tremendous tool for metabonomics researchers for data

evaluation. However, the quality of data input for PCA determines the quality of the

results obtained. The purpose of this study was to evaluate the effects of data

pretreatment on PCA of tissue-targeted metabonomics data, including integration

methodology, data normalization and data standardization. This pretreatment can

strongly affect the statistical analysis and the interpretation of the results. Thus, data

pretreatment steps must be chosen that will give results that accurately reflect trends

in the data, rather than bringing irrelevant artifacts to light.

97

Page 98: ~Chapter 1~ Introduction 1.1. Biomarkers

98

2.4.4.1.1. Data Integration

First, the method of integration was considered. Many metabonomics studies

use bucketing, an automated approach to integration in which the NMR spectrum is

divided somewhat arbitrarily into integral regions of equal ppm width22. This is by

far the most popular approach to metabonomics integration, as the automation is

attractive to many researchers. Another approach is to use manual integration,

whereby the researcher examines the spectra and determines the divisions between

integral regions. The regions will likely not be of equal width, but the end points of

the regions will occur at natural breaking points in the spectra. The manual

integration divisions are kept consistent between spectra.

In this comparison, a sample set of basal heart and liver dialysate was

integrated in two ways. First, the spectra were bucketed from 4.5-0.8 ppm after

spectral processing in ACD/Labs software with settings of 0.04 ppm bucket width

and 10% looseness. (The % looseness setting determines the flexibility that the

program is allowed in dividing spectra, so as to avoid splitting a resonance into two

integral regions as much as possible.) Second, the same set of spectra was manually

integrated from 4.5-0.8 ppm. Both sets of integrals were normalized to TSP-d4

intensity and to the average % delivery of the microdialysis probe. PCA was

performed on a correlation matrix, and the results are shown in Figure 2.12. The

resulting scores plots were similar, with clusters for the liver dialysate samples

observed in roughly the same coordinates in each plot. The heart dialysate shows

about the same amount of spread with each integration mode. Although slight

Page 99: ~Chapter 1~ Introduction 1.1. Biomarkers

99

on basal heart and liver dialysate data. A shows spectral data integrated by bucketing and B shows spectral data integrated manually. represents basal liver dialysate data and represents basal heart dialysate data.

15

Figure 2.12. Comparison of integration methods

-10 -5 0 5 10-10

-8

-6

-4

-2

0

2

4

6

8

Prin

cipa

l Com

pone

nt 2

Principal Component 1

A

B

-10 -5 0 5 10-10

-8

-6

-4

-2

0

2

4

6

8

15

Prin

cipa

l Com

pone

nt 2

Principal Component 1

-10 -5 0 5 10-10

-8

-6

-4

-2

0

2

4

6

8

15

Prin

cipa

l Com

pone

nt 2

Principal Component 1

A

B

-10 -5 0 5 10-10

-8

-6

-4

-2

0

2

4

6

8

15

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Page 100: ~Chapter 1~ Introduction 1.1. Biomarkers

differences in data coordinates were noted, none of the changes would alter the data

interpretation. Thus, no significant differences were noted between integration

modes.

The occupation of many integral regions is one means by which a molecule

may dominate the PCA (in this case, glucose). Thus, the decisions that the researcher

makes when dividing the NMR spectrum into integral regions may determine which

molecules become important in the PCA. Most metabonomics experiments utilize

bucketing, perhaps the most impartial method of dividing the NMR spectrum into

integral regions. However, divisions can occur across resonances and it may not be

advantageous for all data sets, particularly for tissue-targeted metabonomics data,

where glucose is high in intensity and occupies a large space in the NMR spectrum.

Our studies have shown that the integration method does not significantly alter the

interpretation of the data set. Subsequent analyses will be performed with manual

integration, allowing for more control over how the integrals represent the data set

and decreasing complications from shifting of resonances between integral regions.

2.4.4.1.2. Data Normalization

Data normalization was also examined. Metabonomics data analysis by PCA

is based on variance in the data set. The assumption is that most of the variance in

the data is from metabolic changes induced by the metabolic stressor. However, this

is not necessarily the case. Variance can be introduced into these experiments by a

variety of routes, including error in the actual analysis. In tissue-targeted

metabonomics studies, variations in the probe extraction efficiency can also cause

100

Page 101: ~Chapter 1~ Introduction 1.1. Biomarkers

101

changes in metabolite concentration unrelated to the metabolic stressor. With this in

mind, it is important to identify and account for sources of variance unrelated to the

metabolic stressor so that changes related to the metabolic stressor can be more easily

and confidently identified.

Three principal component analyses were performed: on the raw data set, the

data set normalized to TSP-d4, and the data set normalized to both TSP-d4 and the

average extraction efficiency for the microdialysis probe (as gauged by the %

delivery of antipyrine). Manual integrals from the basal heart and liver dialysate data

set were again utilized. Figure 2.13 shows the three scores plots generated. Without

normalization the data shows scatter, with no clustering or segregation between heart

and liver dialysate. Data normalized to TSP-d4 intensity showed segregation between

dialysate types, but with no real clustering. Finally, data normalized to both TSP-d4

and extraction efficiency of the microdialysis probes showed a tight cluster of liver

dialysate samples that were segregated from the more scattered heart dialysate

samples. Although the liver dialysate data clustered tightly when normalized to both

TSP-d4 and % delivery, the heart dialysate data showed less variance when

normalized only to TSP-d4.

From the progression of effects of normalization observed in Figure 2.13, it is

clear that the variation in the data due to these two factors is greater than the variation

between liver and heart dialysate composition. Therefore, the initial PCA was

dominated by experimental (not metabolic) variance and underlying patterns could

not be resolved. For the liver dialysate data, the more variation corrected for prior to

Page 102: ~Chapter 1~ Introduction 1.1. Biomarkers

102

Figure 2.13. Comparison of data normalization methods on basal heart and liver dialysate data. A shows unnormalized spectral data, B shows spectral data normalized to the internal standard, TSP, and C shows spectra data normalized to both the internal standard and to the average extraction efficiency of the microdialysis probe. represents basal liver dialysate data and represents basal heart dialysate data.

10 15

-5 0 5

-8

-6

-4

-2

0

2

4

6

8

Prin

cipa

l Com

pone

nt 2

Principal Co

-5 0 5

-8

-6

-4

-2

0

2

4

6

8mponent 1

10 15

Prin

cipa

l Com

pone

nt 2

Principal Comp

-5 0 5

-8

-6

-4

-2

0

2

4

6

8

onent 1

10 15

Prin

cipa

l Com

pone

nt 2

Principal Comp

A

B

C

-5 0 5

-8

-6

-4

-2

0

2

4

6

8

onent 1

10 15Pr

inci

pal C

ompo

nent

2

Principal Co

-5 0 5

-8

-6

-4

-2

0

2

4

6

8mponent 1

10 15

Prin

cipa

l Com

pone

nt 2

Principal Comp

-5 0 5

-8

-6

-4

-2

0

2

4

6

8

onent 1

10 15

Prin

cipa

l Com

pone

nt 2

Principal Comp

A

B

C

onent 1

Page 103: ~Chapter 1~ Introduction 1.1. Biomarkers

103

the PCA, the tighter the clustering. This was not observed with the heart dialysate

data, as more scatter was observed after normalization to probe delivery. This is

likely due to two key differences in heart and liver microdialysis probe performance,

as shown in Figure 2.14. First, due to spatial constraints in probe implantation in the

heart apex, the membrane length is 8 mm shorter than the liver microdialysis probe.

Thus, the extraction efficiency is approximately 30% less in the heart than in the

liver. Second, the heart is a continuously moving organ, unlike the liver. This leads

to greater variation in probe extraction efficiency. When this is considered with the

smaller overall extraction efficiency of the heart microdialysis probes, it is likely that

corrections for probe performance does not effectively remove variation in the heart

dialysate data set. These results are consistent with what would be expected both

experimentally and biologically. Subsequent analyses of liver dialysate data will

include normalization to TSP-d4 and the probe extraction efficiency, while heart

dialysate will only include normalization to TSP-d4.

2.4.4.1.3. Data Standardization

The disparity between metabolite levels in an NMR spectrum from tissue-

targeted metabonomics data seems to be exacerbated relative to typical metabonomics

data because most of the molecules of interest in the NMR spectrum are present at

concentrations at least ten times lower than the most abundant molecule, glucose.

However, the patterns in their changes are as important, if likely not more important,

than that of glucose. This is an important consideration for data pretreatment,

particularly in the choice of a correlation or covariance matrix for principal

Page 104: ~Chapter 1~ Introduction 1.1. Biomarkers

104

Figure 2.14. The % delivery of antipyrine in heart (n=4) and liver (n=5) microdialysis experiments.

0 2 4 6 8 10 12

0

20

40

60

% D

eliv

ery

of A

ntip

yrin

e

Time (hour)

Liver

Heart

0 2 4 6 8 10 12

0

20

40

60

% D

eliv

ery

of A

ntip

yrin

e

Time (hour)

Liver

Heart

Page 105: ~Chapter 1~ Introduction 1.1. Biomarkers

105

components analysis. PCA can be performed using either a correlation matrix or a

covariance matrix. Covariance matrix data has been unstandardized, while

correlation matrix data has been standardized to a zero mean and unit variance.

Higher intensity resonances would have a greater influence on the principal

components analysis using a covariance matrix, while use of a correlation matrix

would place equal emphasis on all variables, regardless of intensity. Thus, the effect

of data standardization was examined.

Figure 2.15 is a comparison of loadings values from a PCA performed on the

same data set using a correlation and covariance matrix. The loadings values from

the PCA on a covariance matrix show a greater range of values, with most variables

contributing to the analysis minimally. When these loadings values are aligned with

the corresponding resonances in the NMR spectrum as in Figure 2.16, a correlation

can be noted between the highest intensity resonances and the largest loading values.

In contrast, the loadings values for PCA performed on a correlation matrix show

much less of a range than the covariance matrix loadings, but now each variable

shows roughly equal importance to the PCA. Note from the corresponding NMR

resonances that no correlation can be found between loadings values and resonance

intensity as in the covariance matrix PCA.

The loadings plot in Figure 2.15 demonstrates clearly the importance of

standardization in the data set when the dynamic range of the data is high. If no

standardization occurs, the high intensity metabolites dominate the PCA. However,

contributions from individual variables become more equal when standardization is

Page 106: ~Chapter 1~ Introduction 1.1. Biomarkers

106

Figure 2.15. Comparison of loadings values for the same data set with a principal components analysis performed using a correlation matrix and covariance matrix. NMR spectra positioned above the graph approximate the resonances that correspond to each integral region. represents loadings calculated from a correlation matrix. represents loadings from a covariance matrix.

4.0 3.5 3.04.04.1 3.9 3.5 3.0 .5 1.01.5 1.0

010203040-0.050.000.050.100.150.200.250.300.350.400.450.50

Load

ings

Val

ue

Integral Region

4.0 3.5 3.04.04.1 3.9 3.5 3.0 .5 1.01.5 1.0.5 1.01.5 1.0

010203040-0.050.000.050.100.150.200.250.300.350.400.450.50

Load

ings

Val

ue

Integral Region

Page 107: ~Chapter 1~ Introduction 1.1. Biomarkers

applied (that is, when a correlation matrix is used). There is still some variation in

importance to the PCA, but overall, the concentration of the compound does not

determine its importance in the analysis. A potential problem with the use of a

correlation matrix is the lack of discrimination between variables. When everything

becomes important, in essence, everything loses its importance. It becomes difficult

to identify just a few resonances that might be important biomarkers of the conditions

being studied. However, this concern is not enough to prevent the use of correlation

matrices in these studies. Thus, all PCA analyses were performed using correlation

matrices to equalize variable contribution to the statistical analysis23.

2.4.5. Analysis of Tissue-Targeted Metabonomics Data: Perspectives and Time

Trends

Metabonomics data sets can be analyzed on three primary levels: multivariate,

univariate and by spectral examination. Most metabonomics studies solely use

multivariate statistics to interpret trends in the data, since it is an efficient way to

examine the data. However, this may provide an incomplete picture of trends in the

data. It is also important to characterize any trends in basal metabolism or from

experimental parameters, such as from anesthesia, diurnal rhythms, food/fasting

cycles and animal activity, which will be more fully discussed in Chapter 3. For

tissue-targeted metabonomics in particular, the description of time trends is key to

data interpretation.

These data analysis strategies were applied to a data set comparing basal liver

dialysate from both anesthetized and awake rats. For the anesthetized rats, sampling

107

Page 108: ~Chapter 1~ Introduction 1.1. Biomarkers

108

began 30 minutes after probe implantation and continued every hour for 12 hours.

For the awake rats, sampling began 24 hours after probe implantation to allow the

animals sufficient time to recover from the anesthesia and continued every hour for

96 hours. Comparisons were made between tissue metabolic states (awake,

anesthetized, and an awake rat that developed an infection post-surgery).

Additionally, time trends were analyzed by both PCA and PLS regression.

First, general groupings in the data were examined. Principal components

analysis of the data, shown in Figure 2.16, shows three distinct groupings of data

points along principal components 1 and 3. These three groupings correspond to

previously noted metabolic states of the animal: awake healthy, anesthetized and

awake infected. Principal component 1 separates the healthy awake samples from the

anesthetized and awake infected samples. Principal component 3 separates the awake

infected samples from the anesthetized samples. The loadings indicate that principal

component 1 is a contrast axis between time after probe implantation and all integral

regions except those corresponding to lactate, meaning that the time variable has a

negative loading while all loadings for the integral regions are positive in magnitude.

Loadings for principal component 3 show lactate, valine, and resonances in the 3.51-

3.63 ppm region (including some glucose resonances) and the 2.46-2.60 ppm region

as important in segregating the samples from anesthetized and awake infected

animals.

The data was also analyzed in a univariate manner by plotting time after probe

implantation against the normalized integrals from each integral region of the NMR

Page 109: ~Chapter 1~ Introduction 1.1. Biomarkers

109

Figure 2.16. Comparison of liver metabolic states by principal components analysis. represents samples of basal liver dialysate from an awake rat with sepsis (n=1). represents samples of basal liver dialysate taken from healthy awake rats (n=3). ♦ represents samples of basal liver dialysate taken from anesthetized healthy rats (n=4).

-8 -6 -4 -2 0 2 4 6 8 10 12-4

-3

-2

-1

0

1

2

3

4

5

6

7

Prin

cipa

l Com

pone

nt 3

Principal Component 1-8 -6 -4 -2 0 2 4 6 8 10 12

-4

-3

-2

-1

0

1

2

3

4

5

6

7

Prin

cipa

l Com

pone

nt 3

Principal Component 1

Page 110: ~Chapter 1~ Introduction 1.1. Biomarkers

110

spectrum (40 in total). This allowed both time trends and segregation of groups

within a single variable to be visualized. Examination of the plots showed

discrepancies in regions containing glucose resonances. As shown in Figure 2.17,

although A and B both contain glucose resonances, the trends observed in the samples

from awake infected rats differ remarkably between these regions, while the trends

observed in the samples from awake healthy and anesthetized rats remain the same.

The 3.51-3.63 ppm region was also shown to be important in principal component 3

for segregating the samples from anesthetized rats from the samples from awake

infected rats. Spectral examination of the data confirmed the findings from the PCA

and univariate data analysis.

PCA (described above) showed a time trend along principal component 2, as

illustrated in Figure 2.18. This time trend was unidirectional and confined to the

samples from anesthetized animals. The samples from awake (both healthy and

infected) animals did not show a definitive time trend although a trend was observed

in the univariate time plots in Figure 2.17. Loadings for principal component 2 (not

shown) indicate glucose, creatine and lysine as being most important in defining the

time trends for samples from anesthetized animals.

PLS regression showed a slightly different time trend. PLS was performed

separately on correlation matrices for samples from anesthetized and awake animals,

correlating the NMR integral data with sampling time. The anesthetized sample set

showed a good correlation between the predictors (NMR integrals) and responses

(sampling time), with an R2 of 0.916 for 2 components and an R2 predicted of 0.874

Page 111: ~Chapter 1~ Introduction 1.1. Biomarkers

111

Figure 2.17. Comparison of liver glucose levels over time. All rat samples are plotted. represents samples of basal liver dialysate from an awake rat with sepsis. represents samples of basal liver dialysate taken from healthy awake rats (n=3). ♦ represents samples of basal liver dialysate taken from anesthetized healthy rats (n=4). A shows changes in the 3.51-3.52 ppm region and B shows changes in the 3.36-3.46 ppm region.

100 110 120

0 10 20 30 40 50 60 70 80 90

0

5

10

15

20

25

30

35

40

45

Nor

mal

ized

Pea

k In

tens

ity

Time Past Probe Implantation (

0 10 20 30 40 50 60 70 80 90

0

50

100

150

200

250

300

350 Hr)

100 110 120

Nor

mal

ized

Pea

k In

tens

ity

Time Past Probe Implantation (

0 10 20 30 40 50 60 70 80 90

0

5

10

15

20

25

30

35

40

45

Hr)

A

B

100 110 120

Nor

mal

ized

Pea

k In

tens

ity

Time Past Probe Implantation (

0 10 20 30 40 50 60 70 80 90

0

50

100

150

200

250

300

350 Hr)

100 110 120

Hr)

A

B

Nor

mal

ized

Pea

k In

tens

ity

Time Past Probe Implantation (

Page 112: ~Chapter 1~ Introduction 1.1. Biomarkers

112

Figure 2.18. Time trends observed in basal liver dialysate using principal components analysis. represents samples of basal liver dialysate from an awake rat with sepsis. represents samples of basal liver dialysate taken from healthy awake rats. ♦ represents samples of basal liver dialysate taken from anesthetized healthy rats. The arrow shows the direction of the time trend observed in the anesthetized rats.

-8 -6 -4 -2 0 2 4 6 8 10 12

-6

-4

-2

0

2

4

6

8

Prin

cipa

l Com

pone

nt 2

Principal Component 1-8 -6 -4 -2 0 2 4 6 8 10 12

-6

-4

-2

0

2

4

6

8

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Page 113: ~Chapter 1~ Introduction 1.1. Biomarkers

113

for 2 components, as determined through cross-validation. The standardized

coefficients generated, which are similar to PCA loadings, are shown in Figure

2.19A. Again, there is a contrast between glucose and creatine, but glucose is now

also contrasted with lactate and citrate. These resonances were not significant in the

PCA loadings, likely because the samples from awake animals were also included in

the analysis. Figure 2.19B shows the coefficients for the PLS regression correlation

between the NMR integral data for the awake samples and the sampling time. Here,

the correlation is not as strong as observed for the samples from anesthetized rats,

with an R2 of 0.488 and an R2 predicted of 0.152, as determined by cross-validation.

A similar trend was also observed in the PCA analysis. However, a loose correlation

was still defined by the analysis largely by a contrast between the lysine, isoleucine,

and valine and citrate, creatine, alanine and β-hydroxybutyrate resonances.

This research examines a three-tiered approach for the analysis of a

metabonomics data set. Principal components analysis provided a good overall

description of the trends in the data set. Groupings in the data were identified on the

scores plot with principal components 1 and 3, while time trends were identified on

the scores plot with principal components 1 and 2. While these trends were

confirmed by examining the data from a univariate and spectral perspective, no

significant trends were overlooked by the PCA. Analysis on the univariate and

spectral levels, however, is still important to more fully describe trends identified in

the multivariate analysis. The multivariate analysis can streamline the more time

consuming univariate and spectral analysis steps by focusing attention on the most

Page 114: ~Chapter 1~ Introduction 1.1. Biomarkers

114

Figure 2.19. Standardized coefficients for PLS regression analysis. A shows the coefficients from PLS analysis of basal liver dialysate from anesthetized rats. Model statistics showed an R2 of 0.916 and an R2 predicted of 0.874. B shows the coefficients from PLS analysis of basal liver dialysate from awake rats. Model statistics showed an R2 of 0.488 and an R2 predicted of 0.152. The approximate correspondence of the integral regions to chemical shift is shown by the chemical shift scale above each graph.

40 35 30 25 20

-0.10

-0.05

0.00

0.05

15 10 5

Stan

dard

ized

Coe

ffici

ent

Integral Region

510152025303540

-0.4

-0.3

-0.2

-0.1

0.0

0.1

0.2

0.3

0.4

Sta

ndar

dize

d C

oeffi

cien

t

Integral Region

A

B1H Chemical Shi

4.5

ft (ppm)

0.82.65

1H Chemical Shi

4.5

ft (ppm)

0.82.65

40 35 30 25 20

-0.10

-0.05

0.00

0.05

15 10 5

Stan

dard

ized

Coe

ffici

ent

Integral Region40 35 30 25 20

-0.10

-0.05

0.00

0.05

15 10 5

Stan

dard

ized

Coe

ffici

ent

Integral Region

510152025303540

-0.4

-0.3

-0.2

-0.1

0.0

0.1

0.2

0.3

0.4

Sta

ndar

dize

d C

oeffi

cien

t

Integral Region510152025303540

-0.4

-0.3

-0.2

-0.1

0.0

0.1

0.2

0.3

0.4

Sta

ndar

dize

d C

oeffi

cien

t

Integral Region

A

B1H Chemical Shi

4.5

ft (ppm)

0.82.65

1H Chemical Shi

4.5

ft (ppm)

0.82.65

1H Chemical Shi

4.5

ft (ppm)

0.82.65

1H Chemical Shi

4.5

ft (ppm)

0.82.65

Page 115: ~Chapter 1~ Introduction 1.1. Biomarkers

relevant integral regions, both increasing throughput and ensuring an in-depth

analysis of the data set.

Because a time course of microdialysis samples are collected from a single

animal, the data generated by tissue-targeted metabonomics naturally lends itself to

analysis of time trends as opposed to the typical group classification. Time trends can

be observed with PCA and then more accurately described using PLS regression.

PCA was useful in identifying data sets with strong time trends, but was unable to

identify weaker time trends. The principal components loadings correctly identified

most of the key resonances responsible for defining strong time trends, as they

corresponded to the coefficients calculated from PLS regression. However, compared

with PCA, PLS identified more resonances as significant. Additionally, PLS

regression could identify and describe both strong and weak time trends, as illustrated

by the samples from anesthetized and awake animals.

2.5. Conclusions

Tissue-targeted metabonomics shares many of the same goals as “traditional”

metabonomics studies, the primary goal being the identification of new biomarkers

without the prejudice of the more common single biomarker assays. Theoretically, by

expanding the scope of metabolites studied, the probability of uncovering new

biomarkers increases. However, tissue-targeted metabonomics differs from

traditional metabonomics in several ways. First, of course, is the site specificity of

the sampling approach. Second, microdialysis sampling does not require biopsy of

115

Page 116: ~Chapter 1~ Introduction 1.1. Biomarkers

the tissue of interest and the tissue remains intact. Third, because many samples can

be obtained from the same animal over the time course of the experiment, tissue-

targeted metabonomics better lends itself to the examination of time trends in

metabolic data, rather than simpler group classification experiments. This is not to

imply that traditional metabonomics cannot study time trends (or that tissue-targeted

metabonomics cannot study group classification experiments) – in fact, many do24,25.

However, the vast majority of metabonomics studies using urine or plasma sampling

report group classifications as the end result of data interpretation. Finally, an

additional benefit of the time trend approach of tissue-targeted metabonomics is the

use of an animal as its own control, as is also done in some conventional

metabonomics studies. By characterizing the basal metabolic patterns of an animal

prior to perturbing its metabolism, corrections for animal-to-animal variation can be

applied, potentially improving data interpretation.

This tension between high and low abundance metabolites is a significant

barrier to metabolic profiling experiments in general, both for detection schemes and

data analysis. The instrumental dynamic range required to effectively characterize

the complete metabolome is immense. Thus, it should be considered that while the

data generated by metabonomics analysis is extensive, it is not comprehensive. Many

metabolites of key significance to the metabolic perturbation being studied but lower

than the detections limits of the instrument may be excluded. A related problem is

the difficulty of detecting metabolites present in low abundance due to overlap with

the more intense resonances of the major components, an inherent consequence of

116

Page 117: ~Chapter 1~ Introduction 1.1. Biomarkers

performing the analysis on the intact mixture without a separation. This problem can

be partially alleviated by using a higher field magnet since dispersion increases

linearly with magnetic field. Still, there are practical limits such as the

instrumentation available to the investigator, and even at the highest field available

many important low abundance metabolites will still be missed due to resonance

overlap. Dynamic range is also a problem in metabonomics data analysis. A balance

must be established between the dominance of high abundance metabolites in the

multivariate analysis and the inflation of regions of noise when trying to increase the

contribution of low abundance metabolites in the multivariate analysis.

In addition to dynamic range, the complexity of metabonomics data analyses

can create challenges for the technique. Balancing a thorough investigation of the

data with time constraints can be difficult. Using PCA to identify the pertinent trends

in the data, while efficient, can easily overlook important metabolic trends,

particularly from lower abundance metabolites, which tend to become overshadowed

in principal components analysis even with unit variance scaling. The studies

presented in this research suggest that principal components analysis gives an

accurate overview of the data and can be used to focus deeper data analysis. Partial

least squares regression is also a powerful tool, particularly in describing time trends

in the data. For both of these multivariate statistical techniques, it is important to

probe the data further using univariate analysis and spectral inspection.

In summary, tissue-targeted metabonomics focuses on biomarker discovery

for localized metabolic perturbations, as observed through time trends in the

117

Page 118: ~Chapter 1~ Introduction 1.1. Biomarkers

metabolic profiles. These time trends point to potential biomarkers for the condition,

leading to more targeted single biomarker diagnostic assays. Perhaps more

importantly, the improved time resolution with reduced animal-to-animal variability

enables the researcher to observe the development or recession of a physiological

condition, as well as to identify the key metabolic pathways involved throughout the

onset or retraction of a metabolic perturbation. These perturbations may be

identifiable well before any changes are observed in urine or plasma. Thus, the use of

tissue-targeted metabonomics may lead to tests that enable the detection of site-

specific diseases or drug toxicity much faster than conventional assays.

2.6. References 1. Nicholas, P. C., Kim, D., Crews, F. T. and MacDonald, J. M. (2008). 1H

NMR-based metabolomic analysis of liver, serum, and brain following ethanol administration in rats. Chem. Res. Toxicol. 21, 408-420.

2. Soga, T., Baran, R., Suematsu, M., Ueno, Y., Ikeda, S., Sakurakawa, T.,

Kakazu, Y., Ishikawa, T., Robert, M., Nishioka, T. and Tomita, M. (2006). Differential metabolomics reveals ophthalmic acid as an oxidative stress biomarker indicating hepatic glutathione consumption. J. Biol. Chem. 281, 16768-16776.

3. Wang, Y., Bollard, M. E., Keun, H., Antti, H., Beckonert, O., Ebbels, T. M.,

Lindon, J. C., Holmes, E., Tang, H. and Nicholson, J. K. (2003). Spectral editing and pattern recognition methods applied to high-resolution magic-angle spinning 1H nuclear magnetic resonance spectroscopy of liver tissues. Anal. Biochem. 323, 26-32.

4. Garrod, S., Humpher, E., Connor, S. C., Connelly, J. C., Spraul, M.,

Nicholson, J. K. and Holmes, E. (2001). High-resolution 1H NMR and magic angle spinning NMR spectroscopic investigation of the biochemical effects of 2-bromoethanamine in intact renal and hepatic tissue. Magn. Reson. Med. 45, 781-790.

118

Page 119: ~Chapter 1~ Introduction 1.1. Biomarkers

5. Griffin, J. L., Cemal, C. K. and Pook, M. A. (2004). Defining a metabolic phenotype in the brain of a transgenic mouse model of spinocerebellar ataxia 3. Physiol. Genomics 16, 334-340.

6. Pears, M. R., Cooper, J. D., Mitchison, H. M., Mortishire-Smith, R. J., Pearce,

D. A. and Griffin, J. L. (2005). High resolution 1H NMR-based metabolomics indicates a neurotransmitter cycling deficit in cerebral tissue from a mouse model of Batten Disease. J. Biol. Chem. 280, 42508-42514.

7. Viant, M. R., Lyeth, B. G., Miller, M. G. and Berman, R. F. (2005). An

NMR metabolomic investigation of early metabolic disturbances following traumatic brain injury in a mammalian model. NMR Biomed. 18, 507-516.

8. Barba, I., Jaimez-Auguets, E., Rodriguez-Sinovas, A. and Garcia-Dorado, D.

(2007). 1H NMR-based metabolomic identification of at-risk areas after myocardial infarction in swine. Magn. Reson. Mater. Phy. 20, 265-271.

9. Tomlins, A. M., Foxall, P. J. D., Lindon, J. C., Lynch, M. J., Spraul, M.,

Everett, J. R. and Nicholson, J. K. (1998). High resolution magic angle spinning 1H nuclear magnetic resonance analysis of intact prostatic hyperplastic and tumour tissues. Anal. Commun. 35, 113-115.

10. Griffin, J. L. and Kauppinen, R. A. (2007). Tumour metabolomics in animal

models of human cancer. J. Proteome Res. 6, 498-505. 11. Yang, Y., Li, C., Nie, X., Feng, X., Chen, W., Yue, Y., Tang, H. and Deng, F.

(2007). Metabonomic studies of human hepatocellular carcinoma using high-resolution magic angle spinning 1H NMR spectroscopy in conjunction with multivariate data analysis. J. Proteome Res. 6, 2605-2614.

12. Wang, Y., Cloarec, O., Tang, H., Lindon, J. C., Holmes, E., Kochhar, S. and

Nicholson, J. K. (2008). Magic angle spinning NMR and 1H-31P heteronuclear statistical total correlation spectroscopy of intact human gut biopsies. Anal. Chem. 80, 1058-1066.

13. Bergström, S. K., Goiny, M., Danielsson, R., Ungerstedt, U., Andersson, M.

and Markides, K. E. (2006). Screening of microdialysates taken before and after induced liver damage; on-line solid phase extraction-electrospray ionization-mass spectrometry. J. Chromatogr. A 1120, 21-26.

14. Khandelwal, P., Beyer, C. E., Lin, Q., McGonigle, P., Schechter, L. E. and

Bach, A. C., II. (2004). Nanoprobe NMR spectroscopy and in vivo microdialysis: new analytical methods to study brain neurochemistry. J. Neurosci. Methods 133, 181-189.

119

Page 120: ~Chapter 1~ Introduction 1.1. Biomarkers

15. Khandelwal, P., Beyer, C. E., Lin, Q., Schechter, L. E. and Bach, A. C., II.

(2004). Studying rat brain neurochemistry using nanoprobe NMR spectroscopy: a metabonomics approach. Anal. Chem. 76, 4123-4127.

16. Price, K. E., Vandaveer, S. S., Lunte, C. E. and Larive, C. K. (2005). Tissue

targeted metabonomics: metabolic profiling by microdialysis sampling and microcoil NMR. J. Pharm. Biomed. Anal. 38, 904-909.

17. Wang, Y., Wong, S. L. and Sawchuk, R. J. (1993). Microdialysis calibration

using retrodialysis and zero-net flux: application to a study of the distribution of zidovudine to rabbit cerebrospinal fluid and thalamus. Pharm. Res. 10, 1411-1419.

18. Yokel, R. A., Allen, D. D., Burgio, D. E. and McNamara, P. J. (1992).

Antipyrine as a dialyzable reference to correct differences in efficiency among and within sampling devices during in vivo microdialysis. J. Pharmacol. Toxicol. Methods 27, 135-142.

19. Davies, M. I. and Lunte, C. E. (1995). Microdialysis sampling for hepatic

metabolism studies. Impact of microdialysis probe design and implantation technique on liver tissue. Drug Metab. Disp. 23, 1072-1079.

20. Foxall, P. J. D., Spraul, M., Farrant, R. D., Lindon, J. C., Neild, G. H. and

Nicholson, J. K. (1993). 750 MHz 1H-NMR spectroscopy of human blood plasma. J. Pharm. Biomed. Anal. 11, 267-276.

21. Olson, D. L., Norcross, J. A., O'Neil-Johnson, M., Molitor, P. F., Detlefsen,

D. J., Wilson, A. G. and Peck, T. L. (2004). Microflow NMR: concepts and capabilities. Anal. Chem. 76, 2966-2974.

22. Farrant, R. D., Lindon, J. C., Rahr, E. and Sweatman, B. C. (1992). An

automatic data reduction and transfer method to aid pattern recognition analysis and classification of NMR spectra. J. Pharm. Biomed. Anal. 10, 141-144.

23. Craig, A., Cloarec, O., Holmes, E., Nicholson, J. K. and Lindon, J. C. (2006).

Scaling and normalization effects in NMR spectroscopic metabonomics data sets. Anal. Chem. 78, 2262-2267.

24. Waters, N. J., Holmes, E., Williams, A., Waterfield, C. J., Farrant, R. D. and

Nicholson, J. K. (2001). Time-related metabolic effects of α-naphthylisothiocyanate on liver, urine, and plasma in the rat: an integrative metabonomic approach. Chem. Res. Toxicol. 14, 1401-1421.

120

Page 121: ~Chapter 1~ Introduction 1.1. Biomarkers

25. Yap, I. K. S., Clayton, T. A., Tang, H., Everett, J. R., Hanton, G., Provost, J.-

P., Le Net, J.-L., Charuel, C., Lindon, J. C. and Nicholson, J. K. (2006). An integrated metabonomics approach to describe temporal metabolic disregulation induced in the rat by the model hepatotoxin allyl formate. J. Proteom. Res. 5, 2675-2684.

121

Page 122: ~Chapter 1~ Introduction 1.1. Biomarkers

~Chapter Three~

Tissue-Targeted Metabonomics Development: Biological Considerations

3.1. Introduction

Tissues have differing energy requirements and utilize different pathways for

energy generation, according to their specific function and the levels of enzymes

present in the tissue1. Thus, the differing functions an organism’s tissues serve are

reflected in their metabolic profiles. The brain requires a steady supply of glucose

(15% of the body’s total energy consumption) and oxygen (20% of the body’s total

oxygen consumption) to generate enough energy to maintain membrane potentials

necessary for transmitting nerve impulses1-3. Even within the brain, a notably

heterogeneous organ, the different regions have different metabolic needs1. Although

glucose is the primary fuel, the brain can utilize ketone bodies for energy generation

under fasting conditions. The muscle requires energy for contraction of the tissue,

allowing it to do work. It can utilize a variety of energy substrates, including fatty

acids (used while resting), glucose (used during exertion) and ketone bodies. During

exertion, energy is produced by the dephosphorylation of muscle creatine phosphate

stores and glucose is generated from the muscle’s glycogen stores. The glucose is

122

Page 123: ~Chapter 1~ Introduction 1.1. Biomarkers

then metabolized. Glycolytic flux is greater than flux through the citric acid cycle,

which causes accumulation of lactate and alanine in the tissue1-3. Fatty acids are used

during resting while the muscle’s glycogen stores are depleted. Muscle tissue will

also breakdown the protein in the tissue for energy under starvation conditions. The

heart, with its constant pumping and activity, maintains a more constant energy usage

than skeletal muscle and is strictly aerobic in its metabolism. Like the brain, it has

few energy stores and requires a steady supply of oxygen and energy substrates,

primarily fatty acids but also glucose, lactate and ketone bodies1-3. The liver is

perhaps the most metabolically diverse organ in the body. It uses glucose, fatty acids

and amino acids to supply its own energy demands. What makes it distinct is that it

also assists other organs and tissues in their metabolic needs through the synthesis of

fatty acids, glucose, glycerol, amino acids and ketone bodies, the

metabolism/detoxification of exogenous compounds, the regulation of blood glucose

levels and the elimination of nitrogenous compounds through the urea cycle1,2.

Not only does metabolism vary between tissues, but it also varies over time

within the same tissue. It has been well established that tissue metabolism can

change over time due to a variety of factors. Biological rhythms are cycles that are

influenced by environmental cues, primarily light/dark cycles4. Circadian rhythms

are biological rhythms on a roughly 24 hour period4,5. These particular rhythms occur

without entrainment by any external cues, suggesting that there is some internal

pacemaker responsible for establishing these rhythms6. (Diurnal rhythms are those

cycles that are controlled by external stimuli6.) Rhythms or cycles in metabolism are

123

Page 124: ~Chapter 1~ Introduction 1.1. Biomarkers

under the control of the suprachiasmatic nucleus (SCN), the “pacemaker” of the

body7. The SCN has been shown to regulate melatonin production by the pineal

gland7, but the exact mechanism of control is poorly understood8. Rhythms are

observed in a variety of processes and metabolite levels that all relate directly or

indirectly to tissue metabolism (and possibly directly or indirectly to each other) –

glucose uptake, plasma glucose levels, blood pressure, heart rate, hormone secretion,

activity levels, sleep/wake cycles, body temperature, melatonin levels, cortisol levels

and eating4,7,9.

In the liver, circadian rhythms in carbohydrate metabolism are well

established6,10. But, these have not been studied in a comprehensive manner – rather,

the information is pieced together from various studies. Enzymes important to

metabolic pathways such as the citric acid cycle have been shown to exhibit circadian

rhythmicity in their levels8,11. In addition, “clock genes” have been identified in

nonneural tissue, including the liver, suggesting that at least some metabolic rhythms

are intrinsic to the liver tissue itself, outside of the influence of the SCN6,12.

Rhythmicity in liver metabolism has been studied for cholesterol metabolism, amino

acid metabolism and glucose metabolism6. Both hydroxymethylglutaryl-coenzyme A

(HMG-CoA) reductase, a participant in cholesterol synthesis, and cholesterol 7α-

monooxygenase, a participant in bile acid synthesis from cholesterol have an

established circadian rhythm6. Amino acid metabolism circadian rhythms are less

clear – rhythms in actual metabolites appear to be secondary to eating cycles, but

some enzyme cycles have been shown to be independent of eating cycles. These

124

Page 125: ~Chapter 1~ Introduction 1.1. Biomarkers

125

enzymes include tyrosine transaminase, alanine transaminase, tryptophan 2,3-

dioxygenase, lactate dehydrogenase, L-serine dehydratase and arginase6. There are

strong correlations between light/dark cycles and many key enzymes involved in

glycolysis11. Some glucose metabolism cycles, specifically the citric acid cycle, are

less established. Isocitrate dehydrogenase and malate dehydrogenase (malic enzyme)

have been studied, but it is unclear if the rhythms observed are circadian or secondary

to feeding cycles6. Additionally, it has been shown that glycogen levels (as well as

gluconeogenic and glycogen metabolic enzymes) in the liver vary over the course of

the day, with maximum levels occurring during the active/dark period11.

Most of the studies on basal changes in liver metabolism have been performed

examining enzyme levels and individual metabolite levels. This is true of metabolic

studies in a variety of tissues, not just the liver. Very few, if any, studies have been

performed with metabolic profiling experiments. To date, many metabonomics

studies have been published that characterize basal metabolic trends in animals and

humans, as well as the factors contributing to these changes13. Most of these studies

have focused on whole body metabolic trends, with urine the predominant sample

analyzed. Basal metabolism is affected by a myriad of factors, some genetic, some

environmental. Not surprisingly, distinct metabolic differences occur between

species, as illustrated in Figure 3.1, which compares urine samples from rat, mouse

and human subjects. Differences in the metabolic profiles of different genetic strains

of rats13,14 and mice15,16 have been clearly demonstrated, as have differences between

genders of the same strain14,16,17. The hormonal fluctuations experienced by female

Page 126: ~Chapter 1~ Introduction 1.1. Biomarkers

-10 -8 -6 -4 -2 0 2 4 6 8

-4

-3

-2

-1

0

1

2

3

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Figure 3.1. Analysis of urine from three different species. The samples analyzed are aliquots from pooled urine samples. represents human urine (n=7), represents rat urine (n=9) and Δ represents mouse urine (n=10).

126

Page 127: ~Chapter 1~ Introduction 1.1. Biomarkers

rats causes metabolic pertubations over the course of the estrus cycle17, which is why

most metabonomics studies are limited to male subjects. The age of the animal is

also an important factor, particularly in the early development stage18. The animal

shows large changes in body weight over the first few months of life and its gut

microflora is still developing. Animal diet, including the food supplied, as well as the

time of food consumption and amount of water consumption can all dramatically

affect urinary metabolic profiles18-20. Related to this is the composition of the gut

microflora in the intestines, which can vary due to a variety of genetic and

environmental factors21. Environmental factors such as room temperature (and

fluctuations in) and noise (affecting the animal’s sleep quality and stress levels) also

change animal metabolism. Finally, general diurnal rhythms in urinary metabolic

profiles have been observed, with distinctions observed between samples collected in

the morning and at night14,16,17.

3.2. Specific Aims

Nearly all of the metabonomic studies of basal metabolism have been

performed in biofluids, and these studies are almost exclusively in urine. Urine can

give some information on liver function, but its composition is influenced by a variety

of organs, as reviewed in Section 1.2.2.2. Information is lacking on basal trends in

tissue metabolism, as observed through the metabolic profiling of tissue biopsies or

dialysate samples of the tissue extracellular fluid.

127

Page 128: ~Chapter 1~ Introduction 1.1. Biomarkers

Thus, the focus of the study was to apply tissue-targeted metabonomics to the

study of basal metabolism in tissue. These studies will lend new insight to both the

metabonomics community and the biological community interested in circadian

rhythms in tissue metabolism. Experiments were performed to examine and describe

circadian rhythms in tissue metabolism, as well as to establish the expected animal-

to-animal variability in metabolism. Additionally, the effects of anesthesia on basal

tissue metabolism were studied, as this is a component of the tissue-targeted

metabonomics experiment. The focus of this study will be liver metabolism, although

some perspective will also be given on myocardial tissue metabolism.

3.3. Materials and Methods

All materials and methods are described in Section 2.3. Additions to the

methods are described below.

3.3.2.2. Animal Setup

Complete consistency between experiments was attempted to avoid

introducing variance from circadian rhythms and environmental factors. All surgeries

were performed at the same time of day and sampling commenced at the same time as

well. Rats were fed the same rodent chow (LabDiet 5001 Rodent Diet) and

maintained on a 12 hour light/dark cycle (6:00 AM/PM).

128

Page 129: ~Chapter 1~ Introduction 1.1. Biomarkers

129

3.4. Results and Discussion

3.4.1. Comparison of Dialysate Metabolic Profiles from Various Tissues

The primary purpose of this study was to characterize differences in basal

tissue metabolism. Each tissue will have its own distinctive metabolic profile under

basal conditions, due to variance in metabolism. Tissues preferentially use different

substrates for energy generation and other major metabolic processes21, as reviewed

in the chapter introduction (Section 3.1). In the heart, energy is generated through β-

oxidation of fatty acids and glucose metabolism via the citric acid cycle. Brain

metabolism is dominated by glucose metabolism through glycolysis and the citric

acid cycle. Finally, liver metabolism is dominated by glycolysis, gluconeogenesis, β-

oxidation of fatty acids and fatty acid synthesis. The levels of extracellular

metabolites will reflect these differences in dominant metabolic pathways.

Figures 3.2, 3.3 and 3.4 show differences in the NMR spectra reflecting the

metabolic profiles of basal dialysate sampled from heart, brain and liver tissues in

anesthetized rats. In general, liver dialysate tended to be more concentrated than

brain and heart dialysate. This is partially due to the fact that the liver can

accommodate a larger active probe membrane length (10 mm) than brain or heart

tissues (2-4 mm), increasing the extraction efficiency of the membrane by a factor of

3-5. This can be accounted for in data analysis by normalizing to probe delivery, as

described previously. Although the brain is as large as or larger than the liver, the

heterogeneity of the tissue makes it necessary to reduce the probe membrane size to

sample from a single region (in this case, the striatum). The heart is smaller in size

Page 130: ~Chapter 1~ Introduction 1.1. Biomarkers

130

Figure 3.2. Comparison of 1H-NMR spectra (0.5-3.0 ppm) reflecting the metabolic profiles of different tissue dialysates. A: heart dialysate; B: brain dialysate; C: liver dialysate.

2.5 2.0 1.5 1.0

1H-NMR Chemical Shift (ppm)

A

B

C

2.5 2.0 1.5 1.0

1H-NMR Chemical Shift (ppm)

A

B

C

Page 131: ~Chapter 1~ Introduction 1.1. Biomarkers

131

Figure 3.3. Comparison of 1H-NMR spectra (3.0-4.5 ppm) reflecting the metabolic profiles of different tissue dialysates. A: heart dialysate; B: brain dialysate; C: liver dialysate.

4.4 4.3 4.2 4.1 4.0 3.9 3.8 3.7 3.6 3.5 3.4 3.3 3.2 3.1 3.0

A

B

C

1H-NMR Chemical Shift (ppm)4.4 4.3 4.2 4.1 4.0 3.9 3.8 3.7 3.6 3.5 3.4 3.3 3.2 3.1 3.0

A

B

C4.4 4.3 4.2 4.1 4.0 3.9 3.8 3.7 3.6 3.5 3.4 3.3 3.2 3.1 3.04.4 4.3 4.2 4.1 4.0 3.9 3.8 3.7 3.6 3.5 3.4 3.3 3.2 3.1 3.0

A

B

C

1H-NMR Chemical Shift (ppm)

Page 132: ~Chapter 1~ Introduction 1.1. Biomarkers

132

Figure 3.4. Comparison of 1H-NMR spectra (6.0-9.0 ppm) reflecting the metabolic profiles of different tissue dialysates. A: heart dialysate; B: brain dialysate; C: liver dialysate.

8.5 8.0 7.5 7.0 6.5 6.0

A

B

C

1H-NMR Chemical Shift (ppm)

8.5 8.0 7.5 7.0 6.5 6.0

A

B

C

1H-NMR Chemical Shift (ppm)

Page 133: ~Chapter 1~ Introduction 1.1. Biomarkers

than both the brain and liver, and implantation is complicated both by the movement

of the tissue during implantation and the need to implant in the muscle wall of the

heart and not in the atrial or ventricular chambers, thus necessitating a smaller

membrane length. It is also possible that the liver extracellular fluid is simply more

concentrated than the brain or heart extracellular fluid. Finally, the liver and heart are

both highly perfused organs, increasing the extraction efficiency of the membrane in

these tissues.

In the aliphatic region of the NMR spectra (Figure 3.2), the heart and liver

dialysate show some differences but are generally more similar to each other than

each individually are to brain dialysate. Brain dialysate shows a noticeable absence

or depletion of alanine, β-hydroxybutyrate, valine, leucine, isoleucine and citrate.

Heart and liver dialysate show more similarities, probably due to the fact that both are

so well perfused and therefore reflect to a certain extent metabolite levels in the

plasma. The region of 2.2-2.5 ppm seems to most clearly show differences in tissue

metabolism in this region of the spectrum. For example, glutamine is observed in

heart dialysate, but is not identifiable in brain and liver dialysate. In the glucose

region of the NMR spectra (Figure 3.3), less differences are noted. There were

relative differences in the levels of lactate and glucose, but both were present in

significant concentrations. Creatine levels were fairly constant across the dialysate

types. Because glucose is such a dominant metabolite in all three dialysate types and

has multiple resonances, any resonances overlapping glucose will not be detected.

Thus, information is lost about lower level metabolites occurring in this region of the

133

Page 134: ~Chapter 1~ Introduction 1.1. Biomarkers

134

spectrum, which could potentially include metabolites like taurine and choline. In

this region of the spectra, key differences were noted in the region of 3.55-3.7 ppm.

The brain dialysate shows a significant level of myo-inositol and glycine that is not

seen in the liver and heart dialysate. The aromatic region of the NMR spectra (6.0-

9.0 ppm), shown in Figure 3.4, contains resonances from low abundance metabolites.

Heart and liver dialysate shows a signal from tyrosine that is not seen in brain

dialysate. In general, liver dialysate has more abundant aromatic metabolites than

brain or heart dialysate.

Principal components analysis (PCA) was performed to compare the

metabolic profiles obtained under basal metabolic conditions from heart, brain and

liver in anesthetized rats. The resulting scores plot is shown in Figure 3.5. In this

analysis, the integral data was only normalized to the internal standard, TSP-d4. In

chapter 2, it was demonstrated that normalizing to the average % delivery of the

probe adds variance to the heart dialysate metabolic profiles, rather than removing it.

Additionally, no % delivery information was available for the brain dialysate

samples. Therefore, there is some variance in the data set from probe delivery that

could not be accounted for in this analysis. In the scores plot, groupings are observed

between dialysate types, although they do not show tight clustering. Principal

component 1 distinguishes clearly between brain and heart dialysate, with some

delineation of heart and liver dialysate present. However, it is only in combination

with principal component 3 that heart and liver dialysate are able to be distinguished.

The loadings for principal component 1 indicate that nearly every integral region is

Page 135: ~Chapter 1~ Introduction 1.1. Biomarkers

135

Figure 3.5. Comparative PCA study of metabolic profiles representing basal metabolism in liver () (n=4), heart () (n=3) and brain () (n=1) tissues of anesthetized rats. * indicates a sample of basal heart dialysate that is an outlier.

-8 -6 -4 -2 0 2 4 6 8 10 12-5.0-4.5-4.0-3.5-3.0-2.5-2.0-1.5-1.0-0.50.00.51.01.52.02.53.0

Prin

cipa

l Com

pone

nt 3

Principal Component 1

*

-8 -6 -4 -2 0 2 4 6 8 10 12-5.0-4.5-4.0-3.5-3.0-2.5-2.0-1.5-1.0-0.50.00.51.01.52.02.53.0

Prin

cipa

l Com

pone

nt 3

Principal Component 1

*

Page 136: ~Chapter 1~ Introduction 1.1. Biomarkers

important in defining the separation for this axis, with the exception of creatine and

empty regions of the spectrum at 2.17-2.36 ppm and 1.09-1.18 ppm. The loadings for

principal component 3 are more defined/focused, with glycine, β-hydroxybutyrate,

valine, glutamine, alanine, lysine and citrate resonances most responsible for the

trends observed along this axis.

The data presented demonstrates that tissues exhibit unique metabolic profiles

that typically correspond with the known preferred energy generation pathways for

the tissue, with some exceptions. For example, in the brain, no ketone bodies were

present in the extracellular fluid (ECF), even though the rat was anesthetized and

therefore fasting during sampling. Also, no citrate was present in the ECF, even

through the brain utilizes the citric acid cycle as a primary means of energy

generation. Very few studies have been performed correlating the ECF composition

with energy state of the cell and with its internal composition of energy metabolites.

One study has demonstrated that lactate, adenosine, inosine and hypoxanthine levels

in dialysate samples from porcine hearts correlate fairly well to levels of the

compounds extracted from biopsy samples taken from the same tissue, both under

normal and ischemic conditions25. However, literature characterizing other

endogenous metabolites or tissues remains scarce. These results give a new

perspective to basal tissue metabolism studies.

3.4.2. Time Trends in Metabolism: Heart and Liver Tissues

There are differences in metabolism between tissues, but perhaps more

importantly to this research, basal tissue metabolism in each tissue changes over time

136

Page 137: ~Chapter 1~ Introduction 1.1. Biomarkers

137

for a variety of reasons, including experimental factors such as anesthesia and

circadian rhythms. These individual issues will be explored in detail in subsequent

sections. For these experiments, basal tissue metabolism was characterized for heart

and liver tissues.

Heart dialysate was sampled from anesthetized rats. Samples were collected

every 60 minutes for up to 10 hours. PCA and partial least squares regression (PLS)

was performed on NMR integrals normalized to TSP-d4 (the NMR internal standard)

only. (Prior studies, detailed in Chapter 2, have shown that normalization to the %

delivery of the microdialysis probe adds variance to data sets obtained from heart

dialysate.) The scores plot from the PCA is shown in Figure 3.6. A time trend is

observed along the principal component 2 axis, with early samples beginning at lower

scores on the axis and increasing (to varying degrees) over time. PCA identified

creatine, alanine, isoleucine and glucose as the most important in defining the time

trend in the second component. Specifically, alanine, creatine and isoleucine increase

over time as glucose decreases. Creatine time trends are shown in Figure 3.7, and are

representative of the time trends observed in alanine and isoleucine. Glucose time

trends are shown in Figure 3.8. Increases in creatine are likely due to

dephosphorylation of the store of creatine phosphate in cardiac tissue. With the rat in

a fasting state due to anesthesia and glucose levels falling, the cardiac tissue must turn

to other means to supply energy. Creatine phosphate is stored in tissue for this very

purpose. Increases in alanine are likely due to perturbations in the glucose-alanine

cycle26, in which pyruvate is transaminated with glutamate to yield alanine and α-

Page 138: ~Chapter 1~ Introduction 1.1. Biomarkers

138

-8 -6 -4 -2 0 2 4 6 8 10 12 14-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

5

6

7

8

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Figure 3.6. PCA of basal heart dialysate metabolic profiles (n=8). The arrow indicates the direction of the time trend. Data points are labeled by hour of sampling. Symbols are as follows: , hour 1; , hour 2; , hour 3; ◊, hour 4; , hour 5; , hour 6; , hour 7; Δ, hour 8; ♦, hour 9; , hour 10.

Page 139: ~Chapter 1~ Introduction 1.1. Biomarkers

0 2 4 6 8 10468

101214161820222426283032343638

Nor

mal

ized

Inte

gral

Time (Hour)

Figure 3.7. Creatine levels in basal heart dialysate over time (n=8). Individual rats are differentiated by different symbols to show the time trend for each rat.

139

Page 140: ~Chapter 1~ Introduction 1.1. Biomarkers

0 2 4 6 8 10

0

50

100

150

200

250

Nor

mal

ized

Inte

gral

Time (Hour)

Figure 3.8. Glucose levels in basal heart dialysate over time (n=8). Individual rats are differentiated by different symbols to show the time trend for each rat.

140

Page 141: ~Chapter 1~ Introduction 1.1. Biomarkers

ketoglutarate. The large amount of glucose near the beginning of the experiment

likely causes a flux in the glycolytic pathway, resulting in a buildup of pyruvate that

is available for transamination and subsequent transport to the liver. Isoleucine was

an unexpected significant metabolite. Although it can participate in metabolism in

the heart tissue, it is typically not thought of as a key metabolite in heart metabolism.

Isoleucine levels may also be affected by transamination pathways, which are also

affected by pyruvate levels as well as α-ketobutyrate levels. Partial least squares

analysis was also performed on this data set, correlating the NMR integrals

(normalized only to TSP) to time. A 5 component model was calculated, with an R2

of 0.86 and R2 predicted of 0.76 (R2 predicted values are calculated by cross

validating the model and are an indication of the model robustness). Interestingly, the

PLS model identified a few metabolites as important in defining the time trends in the

data set that PCA did not identify. Glucose was identified by both analyses as key to

the time trend in the data, but PLS identified myo-inositol, leucine and valine as

important as well, although not of the same importance as glucose. Leucine and

valine are likely changing in a similar transamination reaction like isoleucine, as they

are all branched-chain amino acids and participate in similar metabolic pathways.

Myo-inositol is produced from glucose-6-phosphate, indicating that this may also be

indicative of glycolytic flux in the cardiac tissue.

The basal metabolism of liver tissue in anesthetized animals was also studied.

Samples were collected every 60 minutes for up to 12 hours. PCA and PLS were

performed on NMR integrals normalized to TSP-d4, the average % delivery of the

141

Page 142: ~Chapter 1~ Introduction 1.1. Biomarkers

142

microdialysis probe and the integral value for the initial time point prior to PCA and

PLS analysis. The PCA scores plot is shown in Figure 3.9. It is shown on the same

scale as Figure 3.6 to allow for comparison between the time trends in heart and liver

tissue. The scores plot shows that the first principal component is responsible for

describing the time trends in liver dialysate from anesthetized rats. The loadings for

the first principal component indicate that lysine, alanine, valine, creatine, glucose

and myo-inositol are significant. Figure 3.10 shows the changes in lysine, which is

representative of the changes in alanine, valine and creatine. These metabolites

increase over time. Figure 3.11 shows changes in glucose, which are similar to

changes in myo-inositol. Both decrease over time. Note that many of the same

metabolites are affected in both the heart and liver tissue. Creatine increases,

indicating the liver uses its stores of creatine phosphate as the animal fasts and

glucose levels drop. Alanine increases as glucose decreases, again indicating flux

through glycolysis, resulting in transamination of pyruvate. Valine and myo-inositol,

identified as key in the PLS model for heart metabolism, are also important in liver

metabolism, according to the PCA. Lysine appears to be unique to basal liver

metabolism time trends in anesthetized rats. It reacts with α-ketoglutarate and a

reductase enzyme to form saccharopine. A PLS model was also calculated to

correlate basal liver metabolism in anesthetized rats to time. The model was quite

strong, with an R2 value of 0.99 and and R2 predicted value of 0.98. In this case, PLS

identified the same metabolites as significant as the PCA model did.

Page 143: ~Chapter 1~ Introduction 1.1. Biomarkers

-8 -6 -4 -2 0 2 4 6 8 10 12 14-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

5

6

7

8

Prin

cipa

l Com

pone

nt 4

Principal Component 1

Figure 3.9. PCA of basal liver dialysate metabolic profiles (n=5). Data points are labeled to show time trends. Symbols are as follows: , hour 1; , hour 3; , hour 5; ◊, hour 7; , hour 7; , hour 11.

143

Page 144: ~Chapter 1~ Introduction 1.1. Biomarkers

0 2 4 6 8 10 12

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

Nor

mal

ized

Inte

gral

Time (Hour)

Figure 3.10. Lysine levels in basal liver dialysate from anesthetized rats over time (n=5). Individual rats are differentiated by different symbols to show the time trend for each rat.

144

Page 145: ~Chapter 1~ Introduction 1.1. Biomarkers

0 2 4 6 8 10 120.0

0.2

0.4

0.6

0.8

1.0

Nor

mal

ized

Inte

gral

Time (Hour)

Figure 3.11. Glucose levels in basal liver dialysate from anesthetized rats over time (n=5). Individual rat experiments are differentiated by different symbols to show the time trend for each rat.

145

Page 146: ~Chapter 1~ Introduction 1.1. Biomarkers

Changes in basal liver metabolism in awake rats over time were also

characterized. Samples were collected from awake rats every 60 minutes up to 72

hours. PCA and PLS models were calculated from NMR integrals normalized to

TSP-d4, the average % delivery of the microdialysis probe and the integral value for

the initial time point. For the PCA model, no correlations were noted to time in the

scores plots (5 principal component axes were examined). The PLS model showed a

weak correlation to time (R2 = 0.74), but the model itself was poor (R2 predicted =

0.27). No significant time trends were identified in this data set.

Overall, it appears that changes in basal metabolism in both the heart and liver

are highly linked to changes in carbohydrate metabolism - specifically, glucose. Both

tissues show evidence of flux through glycolysis, as many of the amino acids that rise

in level are typically produced as a result of transamination with pyruvate or other

glycolytic intermediates. The citric acid cycle is the rate-limiting step in

carbohydrate metabolism, so it stands to reason that an abundance of glucose, as

observed at the beginning of the sampling period, would rapidly progress through

glycolysis and accumulate while waiting to enter the citric acid cycle. In addition to

glucose metabolism, creatine phosphate stores in both tissues are depleted over the

course of the experiment, which is expected when rats are under low energy

conditions like fasting, such as in these experiments.

3.4.3. Anesthesia Effects

Microdialysis experiments require the use of anesthesia during probe

implantation. This can cause significant perturbations to the animal’s local and

146

Page 147: ~Chapter 1~ Introduction 1.1. Biomarkers

general metabolism. Anesthesia affected liver metabolism to a much greater extent

than circadian rhythms or changes in animal activity. The anesthetics used in these

experiments were isoflurane, ketamine, xylazine and acepromazine. Isoflurane is an

inhalation anesthetic used to sedate the rat briefly during administration of the

anesthesia cocktail, which included ketamine, xylazine and acepromazine. Ketamine

is a dissociative injectable anesthetic, that acts to change the reaction of the central

nervous system to sensory stimuli27. Ketamine is known to cause tremoring and

muscle rigidity, so it is commonly administered in a cocktail with one or two other

agents28, typically xylazine and acepromazine (also known as acetylpromazine

maleate). Xylazine is both a sedative and an analgesic that causes muscle relaxation,

and acepromazine is primarily used for its sedative properties29.

Comparison experiments to determine the effects of anesthesia on basal liver

metabolism were performed by sampling liver dialysate from anesthetized and awake

rats. The liver was chosen as a model tissue for two primary reasons. First, the

surgery was simpler and more amenable to awake studies than heart microdialysis.

Second, the liver was planned for use in subsequent experiments detailed in Chapter

4, so it was important to characterize these perturbations to interpret the data in the

subsequent studies.

Probe implantation was performed in the same manner for both awake and

anesthetized animals. For the anesthetized animal experiments, sampling began about

one hour after probe implantation and continued for approximately 12 hours

thereafter. Anesthetized animal experiments are limited to about 12 hours, primarily

147

Page 148: ~Chapter 1~ Introduction 1.1. Biomarkers

148

due to limitations in the time the rat can be maintained under anesthesia and to avoid

the onset of infection. It should be noted that anesthetized animal experiments can be

conducted for longer than 12 hours if the probe implantation is under sterile

conditions and if the rat can be maintained under anesthesia. However, for these

experiments, 12 hours of sampling was sufficient. For the awake animal experiments,

sampling began about 24 hours after probe implantation, when the rat was fully

recovered from the implantation and the anesthesia had left its system. Sampling

continued for 72 hours. Experiments were also performed during which sampling

commenced immediately after probe implantation to study the metabolic effects of

anesthesia leaving the rat.

A comparison of the metabolic profiles in awake and anesthetized rats is

shown in the scores plot in Figure 3.12. A separation between metabolic profiles of

awake and anesthetized animals is observed in the plot, demonstrating that there are

metabolic differences caused by the anesthesia. This separation is defined solely by

principal component 1. Also, the size of the clusters shows a marked difference

between awake and anesthetized samples. It is evident that anesthesia introduces

large amounts of variance into the data set and that awake animals experience

relatively much less metabolic variance over time. This additional variance in the

anesthetized animals is defined by both the first and second principal component

axes.

Figure 3.13 shows the time trends observed in the scores plot in Figure 3.12.

Figure 3.12 shows data from multiple rats for each group. Figure 3.13 is the same

Page 149: ~Chapter 1~ Introduction 1.1. Biomarkers

149

rats (n=4). represents samples from awake animals and represents samples from anesthetized animals.

Figure 3.12. Comparison of basal liver metabolites in awake (n=3) and anesthetized

-6 -4 -2 0 2 4 6 8 10 12 14

-8

-6

-4

-2

0

2

4

6

Prin

cipa

l Com

pone

nt 2

Principal Component 1-6 -4 -2 0 2 4 6 8 10 12 14

-8

-6

-4

-2

0

2

4

6

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Page 150: ~Chapter 1~ Introduction 1.1. Biomarkers

150

Figure 3.13. Time trajectory plots comparing the basal liver metabolism of awake and anesthetized rats. represents averaged samples from awake animals (n=3) and represents averaged samples from anesthetized animals (n=4). A shows a comparison of time trajectories from awake and anesthetized animals and B shows an expanded view of the time trajectory in awake animals shown in A.

8

-6 -4 -2 0 2 4 6

-4

-2

0

2

4

6

Prin

cipa

l Com

pone

nt 2

Principal Component 1

-6 -5 -4 -3-1.0

-0.5

0.0

0.5

1.0

-2

Prin

cipa

l Com

pone

nt 2

Principal Component 1

A

B-6 -4 -2 0 2 4

-4

-2

0

2

4

6

6 8

Prin

cipa

l Com

pone

nt 2

Principal Component 1

-6 -5 -4 -3-1.0

-0.5

0.0

0.5

1.0

-2

Prin

cipa

l Com

pone

nt 2

Principal Component 1

A

B

Page 151: ~Chapter 1~ Introduction 1.1. Biomarkers

data set, but data points from individual rats were averaged at each time point to more

easily track time trends in the data. Arrows in the plot show the directions of the

metabolic time trends. In Figure 3.13A, note that anesthetized animals experience a

large unidirectional time trend that is not observed in the awake animals. This is

indicative of a large shift in metabolism that does not return to the original levels of

metabolites over the time of the experiment. Figure 3.13B shows an expanded view

of the metabolic time trends for the awake animals. No patterns are observed,

whether unidirectional or cyclical. The PCA loadings for principal component 1

identify all integral regions as significant in defining the separation between awake

and anesthetized animals, except for both regions containing lactate. The time trend

along principal component 2 for the anesthetized rats is defined by glucose and

creatine.

These experiments illustrate an important point for these experiments: the

variance introduced by anesthesia is much greater than the variance introduced by

animal activity, feeding/fasting cycles and diurnal variations in metabolism for tissue-

targeted metabonomics experiments, as all of these potential sources of variance are

included in the awake animal data set. This introduction of large amounts of variance

unrelated to the metabolic stressor of the metabonomics experiment can be

detrimental to the success of the experiment and data interpretation. Thus, although

some microdialysis experiments may be better suited to the use of anesthetized

animals, tissue-targeted metabonomics studies in liver tissue should avoid the use of

anesthesia during sampling from the animal.

151

Page 152: ~Chapter 1~ Introduction 1.1. Biomarkers

One of the main reasons for the dramatic time trends observed in the

anesthetized animal samples is the effect of anesthesia on glucose levels. Glucose

was identified in the principal component loadings for the analysis shown in Figures

3.12 and 3.13 as important both in distinguishing between awake and anesthetized

animals and in establishing the time trend observed in anesthetized animals. Both

ketamine/xylazine and isoflurane anesthetics have been shown to cause

hyperglycemia in rodents. Xylazine in particular is an α2-adrenergic agonist and

causes a massive release of glucose stores from the liver upon initial administration28.

Isoflurane also causes surges in glucose levels independently, but the mechanism for

this effect is less clear28. Figure 3.14 shows the dramatic trends in glucose observed

in rats under anesthesia, those emerging from anesthesia and those completely

removed from anesthesia. Rats under anesthesia initially have large amounts of

glucose in the liver extracellular fluid, which fall dramatically over the course of

several hours, almost by a factor of ten. Glucose levels begin to level around 20

hours as it clears from the extracellular fluid and as the anesthesia wears off. To

avoid introducing glucose as a large source of variance unrelated to the experimental

metabolic stressor, Figure 3.14 suggests that sampling should begin at least 20 hours

post-probe anesthesia. It should be noted that in experiments with anesthesia, rats are

fasting, as they are unable to eat. It is difficult to determine what, if any, effect

fasting has on the levels of glucose in the experiment.

152

Page 153: ~Chapter 1~ Introduction 1.1. Biomarkers

153

0 20 40 60 80 100 120

0

20

40

60

80

100

120

140

160

180

200

220

Nor

mal

ized

inte

gral

Time (hour)

Figure 3.14. Timecourse of glucose in the liver extracellular fluid, as affected by the anesthetic agents ketamine, xylazine and isoflurane. represents samples from anesthetized animals, represents samples from animals emerging from anesthesia and represents samples from awake animals.

Page 154: ~Chapter 1~ Introduction 1.1. Biomarkers

154

3.4.4. Animal-to-Animal Variability

In any in vivo experiment, some variance in the data set will originate from

animal-to-animal variability in metabolism. Each animal will exhibit its own unique

metabolic profile, due to small variations in metabolic rate (related to age and body

weight) and other undetermined innate factors. In all experiments, every effort was

taken to maintain consistency in the animals studied, through the use of male

Sprague-Dawley rats from the same commercial source, within the weight range 350-

450 g. Because rat weight tracks well with age, the rats used should be similar in age.

The data analyses shown in this section is the same data as shown in Section 3.4.2,

but relabeled to look at the data from a different perspective.

Animal-to-animal variability in heart metabolism is illustrated in the PCA

scores plot in Figure 3.15. The first principal component separates the different

animals, giving information on what causes the animal-to-animal variability in heart

microdialysis experiments. The loadings for the first principal component show

valine, leucine and glutamine as most important in differentiating between individual

rats. A univariate plot of valine time trends in each rat is shown in Figure 3.16.

There are no definable time trends for valine in these experiments – rather, it is the

differences in levels of valine between rats that makes it so significant. The other

significant integral regions, including leucine and glutamine, show similar trends. In

this data set, some variance may still be present from the % delivery of the

microdialysis probe, as the data was not normalized to this. (This was determined to

be inappropriate for heart dialysate data, as shown in Section 2.4.4.1.2.) This could

Page 155: ~Chapter 1~ Introduction 1.1. Biomarkers

-6 -4 -2 0 2 4 6 8 10 12-8

-6

-4

-2

0

2

4

6

8

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Figure 3.15. PCA of basal heart dialysate metabolic profiles (n=8). Individual rats are differentiated by different symbols to show the time trend for each rat.

155

Page 156: ~Chapter 1~ Introduction 1.1. Biomarkers

0 2 4 6 8 100

1

2

3

4

5

6

7

8

Nor

mal

ized

Inte

gral

Time (Hour)

Figure 3.16. Valine levels in basal heart dialysate from anesthetized rats over time (n=8). Individual rats are differentiated by different symbols to show the time trend for each rat.

156

Page 157: ~Chapter 1~ Introduction 1.1. Biomarkers

157

appear as animal-to-animal variability in the scores plot. It should also be noted that

while there is variance between animal metabolism overall along the first principal

component axis, there is also variance among the animals with regard to the way the

metabolism changes over time. Along the second principal component axis in Figure

3.15, there is a large variation between the “length” of the time trends. This indicates

that some animals experience a substantial change in basal metabolism over the

course of sampling, while others experience much less change. In fact, the first

principal component axis sorts rats based on the magnitude of the metabolic change

experienced over time, with rats experiencing greater change at higher scores values.

Figure 3.17 shows the scores plot illustrating animal-to-animal variability in

liver metabolism in anesthetized rats. All variance between rats should be primarily

due to differences between animals, as other identified sources of variance have been

corrected for in the raw data set prior to analysis. The fourth principal component

axis best defines metabolic differences between the rats. In general, the anesthetized

liver experiments showed much less animal-to-animal variation in metabolism than

the heart microdialysis experiments, as observed by both multivariate and univariate

analysis of the data. β-hydroxybutyrate and lactate were most significant in

differentiating between anesthetized animals, as they show the most variation from

animal to animal. Liver metabolism in awake rats contains animal-to-animal

variation, but it is more difficult to define. Figure 3.18 shows the scores plot for the

data set. Unlike the previous scores plot, there is no clearly defined axis describing

animal-to-animal variation in the data set, although it is present.

Page 158: ~Chapter 1~ Introduction 1.1. Biomarkers

-6 -4 -2 0 2 4 6 8 10 12-8

-6

-4

-2

0

2

4

6

8

Prin

cipa

l Com

pone

nt 4

Principal Component 1

Figure 3.17. PCA of basal liver dialysate metabolic profiles in anesthetized rats (n=5). Individual rats are differentiated by different symbols to show the time trend for each rat.

158

Page 159: ~Chapter 1~ Introduction 1.1. Biomarkers

-6 -4 -2 0 2 4 6 8 10 12-8

-6

-4

-2

0

2

4

6

8

Prin

cipa

l Com

pone

nt 3

Principal Component 1

Figure 3.18. PCA of basal liver dialysate metabolic profiles in awake rats (n=4). Individual rats are differentiated by different symbols to show the time trend for each rat.

159

Page 160: ~Chapter 1~ Introduction 1.1. Biomarkers

160

Animal-to-animal variation is an important consideration for metabonomics

experiments. Studies are needed to clearly establish expected variance from innate

differences in metabolism between animals, so that metabolic changes induced by a

metabolic stressor can be confidently identified. In tissue targeted metabonomics,

which is primarily focused on the identification and analysis of time trends in tissue

tabolism, it is important to establish both differences between animal metabolism

t single time points and differences between basal tissue metabolism over the time

ourse of the experiment. In these studies, liver and heart tissues showed different

etabolites as significant in varying between animals. Heart tissue showed the most

ariance from amino acids (leucine, valine and glutamine), while liver tissue showed

e most variance from energy substrates or intermediates (lactate and β-

ydroxybutyrate).

.4.5. Circadian Rhythms

The presence of circadian rhythms was investigated in samples taken from the

animal experiments, due to their sampling period being 12 hours or less. PCA and

PLS have already shown a poor correlation of liver metabolism in awake rats to time,

as detailed in Section 3.4.2. However, it is possible that cyclical trends in the data

could be missed, so the data was examined in a univariate manner. Only one rat

exhibited cyclical trends in some metabolites, including leucine, isoleucine, valine,

alanine and citrulline. Figure 3.19 shows levels of valine in the liver of three

different awake rats over the course of three days of sampling. Roughly 24 hour

me

a

c

m

v

th

h

3

liver of awake rats. Circadian rhythms should not be significant in anesthetized

Page 161: ~Chapter 1~ Introduction 1.1. Biomarkers

161

Figure 3.19. Timecourse of valine in the extracellular fluid of liver tissue of awake rats (n=3). The gray bars represent the dark period of the day, 6:00 PM – 6:00 AM.

0 10 20 30 40 50 60 70 80 90 1000.6

0.7

0.8

0.9

1.0

1.1

1.2

1.3

1.4

1.5

1.6

1.7

1.8

1.9

2.0

Nor

mal

ized

Inte

gral

Time (hour)0 10 20 30 40 50 60 70 80 90 100

0.6

0.7

0.8

0.9

1.0

1.1

1.2

1.3

1.4

1.5

1.6

1.7

1.8

1.9

2.0

Nor

mal

ized

Inte

gral

Time (hour)

Page 162: ~Chapter 1~ Introduction 1.1. Biomarkers

162

cycles of valine levels are observed in one rat. The other rats show little to no cycling

in metabolite levels. This plot is representative of trends observed in leucine,

isoleucine, alanine and citrulline. All other metabolites showed very little cycling in

levels over a 24 hour period. In addition to metabolite levels, cycling in animal

activity was noted in all three rats, as expected. An example of animal activity is

given in Figure 3.20. The rat with the greatest difference between activity in the

light and dark period was the one with the cycling observed in metabolite levels. The

other two rats showed activity cycling consistent with the time of day, but the

differences in activity levels between the light and dark periods were less distinct.

These experiments have demonstrated that circadian rhythms do not

significantly impact the composition of the liver extracellular fluid, at least in a large

and consistent manner. While one rat showed cycling in a few metabolite levels, this

was not observed in the other two rats studied. The fact that these effects are limited

to a few metabolites and only in a small number of rats is positive for tissue-targeted

metabonomics data interpretation. Little variance is added to the data set by circadian

rhythms, making interpretation of other more interesting metabolic effects easier.

The relatively small magnitude of circadian rhythms on liver extracellular fluid

composition is confirmed in Figure 3.12, showing that metabolic changes from

anesthesia are much greater than any circadian rhythms observed. This is the first

known study of circadian rhythms in the composition of liver extracellular fluid.

Numerous microdialysis studies of circadian rhythms in neurotransmitters have been

Page 163: ~Chapter 1~ Introduction 1.1. Biomarkers

163

Figure 3.20. Animal activity histogram for one rat. The y axis represents the number of times that the optical sensor in the Raturn® system was activated over an hour period. The gray bars represent the dark period of the day, 6:00 PM – 6:00 AM.

20 40 60 800

20

40

60

80

100

Num

ber o

f Sen

sor E

vent

s

Time (Hour)20 40 60 80

0

20

40

60

80

100

Num

ber o

f Sen

sor E

vent

s

Time (Hour)

Page 164: ~Chapter 1~ Introduction 1.1. Biomarkers

performed in the brain tissue. This is an important study for tissue-targeted

metabonomics experiments, helping in interpretation of the data acquired.

3.4.6. Tissue Immunological Response

Histology studies were performed on all tissues. Hearts and livers were

harvested immediately after the end of the experiment, and the tissue was fixed in

10% buffered formalin and stored at 4°C for at least a week. The tissue was

embedded in paraffin, sliced and stained in hematoxylin and eosin. Tissue responded

as described previously30. Probes implanted in anesthetized animals were in the

tissue for 12-13 hours. Small amounts of damage from implantation were noted, but

the primary tissue immune reaction was the beginning of neutrophil implantation into

the implantation site. Awake animals had the probe implanted for much longer

periods of time, typically around 96 hours. Consequently, the immune reaction was

much greater. Neutrophil infiltration was much greater, some macrophage

recruitment was present and a fibrotic capsule had begun to form around the probe.

Unfortunately, the effects of probe implantation on metabolic profiles were unable to

be determined. This is a common quandary in microdialysis experiments, as samples

of extracellular fluid cannot be taken without the microdialysis probe.

Histology did show that one awake rat developed an infection over the course

of the experiments. (Anesthetized rat experiments are not concerned with infection,

as sampling is completed before infection begins to develop.) These results were

reported in Chapter 2, Section 2.4.5 and Figures 2.16 and 2.17. It is important to

comment here that infection causes changes in the metabolic profiles of liver tissue

164

Page 165: ~Chapter 1~ Introduction 1.1. Biomarkers

(and presumably any other tissue). Thus, it is important to maintain sterile field

during probe implantation, closely monitor rats for signs of infection during sampling

and perform tissue histological analysis after sampling to determine if an infection

developed. Conversely, this finding could provide an interesting future application

for tissue-targeted metabonomics studies as a potentially more sensitive detector of

sepsis.

3.5. Conclusions

These results demonstrate the potential of tissue-targeted metabonomics for

studies requiring metabolic information from specific tissues. The ability to collect

multiple samples over time from the same animal offers a unique advantage for the

use of microdialysis sampling in metabonomics studies. As with all in vivo studies,

metabolite levels are prone to change from factors unrelated to the metabolic stressor

of the experiment, due to metabolite level variability between animals, circadian

rhythms and animal activity. It is particularly important to characterize these trends

in metabonomics studies because of the large number of interconnected endogenous

metabolites monitored in a single analysis. The union of microdialysis sampling with

metabonomics studies also presents anesthesia as an additional factor to be

considered in characterizing metabolic perturbations unrelated to the metabolic

stressor.

Tissue-targeted metabonomics data sets experience variance from a variety of

sources. Tissues have different metabolic profiles, according to their specific

165

Page 166: ~Chapter 1~ Introduction 1.1. Biomarkers

functions and energy needs. Within the same tissue, variance occurs primarily from

anesthesia (if sampling from anesthetized animals) and animal-to-animal variability.

Circadian rhythms do not appear to contribute significantly to variance in basal

metabolism.

3.6. References

1. Frayn, K. A. (1996). Metabolic Regulation: A Human Perspective. Portland Press, London.

2. Mathews, C. K., van Holde, K. E. and Ahern, K. G. (2000). Biochemistry.

Addison Wesley Longman, Inc., New York. 3. Gibson, D. M. and Harris, R. A. (2002). Metabolic Regulation in Mammals.

Taylor and Francis, New York. 4. Benstaali, C., Mailloux, A., Bogdan, A., Auzeby, A. and Touitou, Y. (2001).

Circadian rhythms of body temperature and motor activity in rodents: their relationships with the light-dark cycle. Life Sci. 68, 2645-2656.

5. Holzberg, D. and Albrecht, U. (2003). The circadian clock: a manager of

biochemical processes within the organism. J. Neuroendocrin. 15, 339-343. 6. Davidson, A. J., Castanon-Cervantes, O. and Stephan, F. K. (2004). Daily

oscillations in liver function: diurnal vs circadian rhythmicity. Liver Internat. 24, 179-186.

7. Borjigin, J. and Liu, T. (2007). Application of long-term microdialysis in

circadian rhythm research. Pharmacol. Biochem. Behav. Epub ahead of print. 8. Froy, O. (2007). The relationship between nutrition and circadian rhythms in

mammals. Frontiers Neuroendocrin. 28, 61-71. 9. la Fleur, S. E. (2003). Daily rhythms in glucose metabolism: suprachiasmatic

nucleus output to peripheral tissue. J. Neuroendocrin. 15, 10. Walker, P. R., Bonney, R. J. and Potter, V. R. (1974). Diurnal rhythms of

hepatic carbohydrate metabolism during development of the rat. Biochem. J. 140, 523-529.

166

Page 167: ~Chapter 1~ Introduction 1.1. Biomarkers

11. Kaminsky, Y. G. and Kosenko, E. A. (1987). Diurnal rhythms in liver

carbohydrate metabolism: comparative aspects and critical review. Comp. Biochem. Physiol. 86B, 763-784.

12. Shibata, S. (2004). Neural regulation of the hepatic circadian rhythm.

Anatom. Rec. A 280A, 901-909. 13. Robosky, L. C., Wells, D. F., Egnash, L. A., Manning, M. L., Reily, M. D.

and Robertson, D. G. (2005). Metabonomic identification of two distinct phenotypes in Sprague-Dawley (Crl:CD(SD)) rats. Toxicol. Sci. 87, 277-284.

14. Williams, R. E., Lenz, E. M., Evans, J. A., et al. (2005). A combined 1H

NMR and HPLC-MS-based metabonomic study of urine from obese (fa/fa) Zucker and normal Wistar-derived rats. J. Pharm. Biomed. Anal. 38, 465-471.

15. Gavaghan, C. L., Holmes, E., Lenz, E., Wilson, I. D. and Nicholson, J. K.

(2000). An NMR-based metabonomic approach to investigate the biochemical consequences of genetic strain differences: application to C57BL10J and Alpk:ApfCD mouse. FEBS Lett. 484, 169-174.

16. Plumb, R., Granger, J., Stumpf, C., Wilson, I. D., Evans, J. A. and Lenz, E.

(2003). Metabonomic analysis of mouse urine by liquid-chromatography-time of flight mass spectrometry (LC-TOFMS): detection of strain, diurnal and gender differences. Analyst 128, 819-823.

17. Bollard, M. E., Holmes, E., Lindon, J. C., et al. (2001). Investigations into

biochemical changes due to diurnal variation and estrus cycle in female rats using high-resolution 1H NMR spectroscopy of urine and pattern recognition. Anal. Biochem. 295, 194-202.

18. Bell, J. D., Sadler, P. J., Morris, V. C. and Levander, O. A. (1991). Effect of

aging and diet on proton NMR spectra of rat urine. Magn. Reson. Med. 17, 414-422.

19. Phipps, A. N., Wright, B., Stewart, J. and Wilson, I. D. (1997). Use of proton

NMR for determining changes in metabolite excretion profiles induced by dietary changes in the rat. Pharm. Sci. 3, 143-146.

20. Gu, H., Chen, H., Pan, Z., et al. (2007). Monitoring diet effects via biofluids

and their implications for metabolomics studies. Anal. Chem. 79, 89-97.

167

Page 168: ~Chapter 1~ Introduction 1.1. Biomarkers

21. Nicholls, A. W., Mortishire-Smith, R. J. and Nicholson, J. K. (2003). NMR spectroscopic-based metabonomic studies of urinary metabolic variation in acclimatizing germ-free rats. Chem. Res. Toxicol. 16, 1395-1404.

22. Wang, Y., Wong, S. L. and Sawchuk, R. J. (1993). Microdialysis calibration

using retrodialysis and zero-net flux: application to a study of the distribution of zidovudine to rabbit cerebrospinal fluid and thalamus. Pharm. Res. 10, 1411-1419.

23. Yokel, R. A., Allen, D. D., Burgio, D. E. and McNamara, P. J. (1992).

Antipyrine as a dialyzable reference to correct differences in efficiency among and within sampling devices during in vivo microdialysis. J. Pharmacol. Toxicol. Methods 27, 135-142.

24. Davies, M. I. and Lunte, C. E. (1995). Microdialysis sampling for hepatic

metabolism studies. Impact of microdialysis probe design and implantation technique on liver tissue. Drug Metab. Disp. 23, 1072-1079.

25. Kavianipour, M., Wikstrom, G., Ronquist, G. and Waldenstrom, A. (2003).

Validity of the microdialysis technique for experimental in vivo studies of myocardial energy metabolism. Acta Physiol. Scand. 179, 61-65.

26. Thomassen, A., Nielsen, T. T., Bagger, J. P. and Thuesen, L. (1984).

Myocardial glutamate and alanine exchanges related to carbohydrate metabolism in patients with normal and stenotic coronary arteries. Clin. Physiol. 4, 425-434.

27. Wright, M. (1982). Pharmacologic effects of ketamine and its use in

veterinary medicine. J. Am. Vet. Med. Assoc. 180, 1462-1471. 28. Saha, J. K., Xia, J., Grondin, J. M., Engle, S. K. and Jakubowski, J. A.

(2005). Acute hyperglycemia induced by ketamine/xylazine anesthesia in rats: mechanisms and implications for preclinical models. Exp. Biol. Med. 230, 777-784.

29. Jones, L. M., Booth, N. H. and McDonald, L. E. (Eds.). (1977). Veterinary

Pharmacology and Therapeutics. Iowa State University Press, Ames. 30. Davies, M. I. and Lunte, C. E. (1995). Microdialysis sampling for hepatic

metabolism studies: impact of microdialysis probe design and implantation technique on liver tissue. Drug Metab. Disp. 23, 1072-1079.

168

Page 169: ~Chapter 1~ Introduction 1.1. Biomarkers

~Chapter Four~

Tissue-Targeted Metabonomics: Application to the Study of Oxidative Stress

4.1. Introduction

Tissue-targeted metabonomics has the potential to give a new perspective on

metabolic changes in specific tissues that experience oxidative stress. Oxidative

stress was reviewed in the Introduction, Section 1.5. Some metabonomics studies

have been previously conducted to examine the effects of oxidative stress on brain1,2,

liver3-6, kidney7,8, vascular smooth muscle9,10 and neonate urine11. These studies have

examined oxidative stress in tissues originating from various sources, including

traumatic brain injury, hepatotoxicity, kidney transplantation, schizophrenia, diet and

fatty liver disease. The studies use a mixture of biofluid and tissue extract analysis,

often comparing the metabolic profiles to concurrent proteomic or genomic data. In

these studies, no clear metabolic patterns resulting from oxidative stress were

identified. A few endogenous metabolites changed with the timecourse of oxidative

damage, including ophthalmic acid, allantoin, taurine, polyunsaturated fatty acids and

ascorbate, although no consistent metabolic changes were noted among the studies.

To date, no metabonomics studies have been performed examining two common

169

Page 170: ~Chapter 1~ Introduction 1.1. Biomarkers

causes of oxidative stress, ischemia/reperfusion and chemically induced oxidative

stress by doxorubicin. Additionally, no metabonomics studies have been performed

characterizing oxidative stress in specific tissues with microdialysis sampling – all

tissue-specific work has been done through biopsies, so animal-to-animal variability

is high in these experiments.

4.2. Specific Aims

With this in mind, the purpose of this research was to apply tissue-targeted

metabonomics to the study of oxidative stress, as induced locally by doxorubicin.

Prior to in vivo studies, in vitro studies were performed to determine the solution

stability of doxorubicin and its behavior in a microdialysis system. Myocardial

oxidative stress, as induced by doxorubicin and ischemia/reperfusion, was

characterized in the anesthetized rat. However, the research primarily focused on

hepatic oxidative stress, as induced by doxorubicin. This was examined in both

awake and anesthetized animals. Additionally, a marker of oxidative stress, the

antioxidant glutathione, was monitored in liver dialysate samples and correlated to

changes in metabolic profiles. The purpose behind the glutathione analyses was to

better identify metabolic changes specific to the oxidative stress induced by

doxorubicin by providing some metric of oxidative stress to which to correlate the

metabolic profiles.

170

Page 171: ~Chapter 1~ Introduction 1.1. Biomarkers

4.3. Materials and Methods

See Section 2.3. Deviations or additions to the methods described are noted in

the following sections.

4.3.1. Reagents and Solutions

Doxorubicin hydrochloride, ≥ 98% purity, was purchased from Sigma

Chemical Corporation (St. Louis, MO), as were the glutathione and glutathione

disulfide standards.

4.3.2. Microdialysis Methods

4.3.2.1. In Vitro Experiments

Doxorubicin in vitro experiments were performed to characterize the behavior

of doxorubicin in the microdialysis setup. A 37°C bath was prepared using a 118 mL

polypropylene specimen jar filled with 80 mL of Ringer’s solution. (A polypropylene

container was used in place of the typical glass container, due to doxorubicin’s

propensity to stick to glass surfaces.) The specimen jar was kept at constant

temperature using a Thermolyne Dri-Bath and was stirred slowly throughout the

experiment by placing the bath on a magnetic stirrer plate. The specimen jar was

isolated using a lid, with holes drilled in the lid for the microdialysis probes. The

probe membrane was submerged in the middle of the bath, and the probe was secured

in position. The bath was allowed to equilibrate to temperature for 30 minutes and

the probes were allowed to equilibrate while being perfused at 1 μL/min for an

additional 30 minutes. Delivery experiments were typically performed first. In these

experiments, the bath contained Ringer’s solution and the probe was perfused with 10

171

Page 172: ~Chapter 1~ Introduction 1.1. Biomarkers

172

μM doxorubicin and 10 μM antipyrine in Ringer’s solution. (Concentrations were

varied in some experiments.) In the recovery experiments, the bath contained 10 μM

doxorubicin and 10 μM antipyrine in Ringer’s solution and the microdialysis probe

was perfused with Ringer’s solution. Samples were collected every 15 minutes for

LC/UV analysis, as described in Section 2.3.2.1 and 4.3.3.

4.3.2.2. Isoflurane Anesthesia

A subset of rats was anesthetized using isoflurane inhalation anesthesia to

determine the effects of anesthesia on the timecourse of glutathione and glutathione

disulfide in liver dialysate. The setup is shown in Figure 4.1. The isoflurane was

delivered to the rat using an isoflurane vaporizer (VetEquip, Pleasanton, CA). The

vaporizer mixed isoflurane vapor with an aerobic gas mixture (95% oxygen/5%

carbon dioxide). The airflow was maintained at 1.5 L/min. Rats were initially

anesthetized at 5% isoflurane. The level was lowered to approximately 2% to

maintain anesthesia, but was adjusted as necessary to maintain appropriate levels of

sedation. Exhaled isoflurane was trapped using a VaporGuard activated charcoal

absorption filter.

4.3.2.3. Doxorubicin Dosage

Doxorubicin was dosed in two different ways. First, preliminary studies for

doxorubicin were performed in heart microdialysis experiments with anesthetized

rats. In these studies, doxorubicin was dosed at 15 mg/kg intraperitoneally. Second,

doxorubicin was dosed locally through the microdialysis probe in the liver

microdialysis experiments. Two probes were implanted in the median lobe of the

Page 173: ~Chapter 1~ Introduction 1.1. Biomarkers

173

Figure 4.1. Isoflurane inhalation anesthesia setup, as utilized during a liver microdialysis experiment. A labels the vaporizer and B designates the charcoal filter. The syringe pump and fraction collector on the right show the microdialysis sampling equipment.

A

B

A

B

Page 174: ~Chapter 1~ Introduction 1.1. Biomarkers

174

liver (Figure 4.2). One probe (posterior, Probe A) was used to monitor basal changes

in the liver extracellular fluid composition and the second probe (anterior, Probe B)

was used to both dose doxorubicin and monitor changes in liver metabolism due to

the doxorubicin dose. The probes were spaced 1 cm apart, a distance previously

established to prevent crosstalk between the probes12. Probes were perfused at 1

μL/min with 7 μM antipyrine in Ringer’s solution. After collection of basal dialysate

from both probes, the perfusate for Probe B was switched to 2 mg/mL doxorubicin

dissolved in the original perfusate solution. Probe B was perfused for 1 hour with the

doxorubicin solution, after which the perfusate was returned to the original

composition. In anesthetized rat experiments, basal dialysate was collected for three

hours, with samples collected every 60 minutes. After the doxorubicin dose, samples

were collected every 60 minutes for eight hours. In awake rat experiments, basal

dialysate samples were collected every 60 minutes for 24 hours. After the

doxorubicin dose, sampling continued for 47 hours.

4.3.3. LC/UV Doxorubicin Analysis

Doxorubicin was detected in liver dialysate samples using LC/UV, with the

system described in Section 2.3.2.1. The isocratic method utilized the mobile phase

25% acetonitrile/75% 25 mM sodium phosphate buffer, pH = 2.5. The flow rate was

0.3 mL/min, and the detection wavelength was 254 nm.

4.3.4. CE/UV Glutathione Analysis

Glutathione (GSH) and glutathione disulfide (GSSG) were quantified in liver

dialysate using a CE/UV method similar to that previously described13. A homebuilt

Page 175: ~Chapter 1~ Introduction 1.1. Biomarkers

175

Figure 4.2. Dual probe approach to doxorubicin studies in the liver. A shows the basic implantation geometry, with B showing the localized dosing of doxorubicin through Probe B.

1 cm

Probe A

Probe B

Blood flow

Median liver lobe

Probe A

Probe B

Blood flow

A

B

1 cm

Probe A

Probe B

Blood flow

Median liver lobe

Probe A

Probe B

Blood flow

A

B

Page 176: ~Chapter 1~ Introduction 1.1. Biomarkers

176

CE/UV system was used, with a Spellman CZE1000R high voltage power supply and

SpectraPhysics UV1000 detector. A 50 μm inner diameter (360 μm outer diameter)

fused silica capillary was utilized, cut to 60 cm in total length. A 1 cm detection

window was burned in the capillary coating, making the capillary 45 cm in effective

length. The background electrolyte (BGE) was 0.1 M ammonium chloride with 0.5

mM tetradecyltrimethylammonium bromide (TTAB), pH = 8.4. TTAB, a cationic

surfactant, was used to reverse the electroosmotic flow of the separation. Samples

were injected onto the capillary electrokinetically by applying a potential of -10 kV

for 30 seconds, followed by an electrokinetic injection of 0.1 M NaOH for 60 seconds

at -10 kV for pH-mediated stacking. The separation potential was -10 kV, and the

detection wavelength was 214 nm. Data was acquired using Chrom&Spec software

(version 1.52L, Ampersand International, Beachwood, OH). A sample

electropherogram is shown in Figure 4.3. At the beginning of each day, the capillary

was fully conditioned using the following rinsing procedure (times represent the

length of time a pressure of 10 psi was applied): 10 minutes, methanol; 2 minutes,

water; 10 minutes, 1 M HCl; 2 minutes, water; 20 minutes, 0.1 M NaOH; 2 minutes,

water; 10 minutes, BGE. In between injections, the capillary was rinsed with

hydrodynamic injections of 0.1 M NaOH and BGE, 4 minutes each.

4.3.5. LC/EC Glutathione Analysis

Glutathione (GSH) and glutathione disulfide (GSSG) were quantified in liver

dialysate using a dual electrode LC/EC method similar to those previously described

in literature14-17. The LC/EC system was composed of a single Shimadzu LC-10AD

Page 177: ~Chapter 1~ Introduction 1.1. Biomarkers

177

Figure 4.3. CE/UV electropherogram of basal liver dialysate. The arrow indicates the peak representing GSSG.

1 μA

1 2 6543 110987 1

Time (min)

ch1

1 μA

1 2 6543 110987 1

Time (min)

ch1

Page 178: ~Chapter 1~ Introduction 1.1. Biomarkers

178

pump and a Rheodyne 7725i injector, fitted with a 25 μL PEEK injection loop (10 μL

injection volume). The inlet tubing for the pump was composed of PEEK, to prevent

the reoxygenation of the mobile phase after sparging. A Phenomenex Synergi Hydro-

RP column, 2 mm x 150 mm, 4 μm particle size was used. The mobile phase

consisted of 98% 0.12 M monochloroacetic acid 7 mM sodium octylsulfonic acid, pH

= 3.0/2% methanol, at a flow rate of 0.35 mL/min. A dual gold-mercury electrode

system was used as the detection scheme. The electrode consisted of two 3 mm gold

electrodes in a series configuration. The gold electrodes were amalgamated and

placed in the electrode block with a thin-layer gasket 0.005” thick. A Ag/AgCl

reference electrode was utilized. Two BAS LC-4C amperometric detectors were

utilized and interfaced to Chrom&Spec software (v. 1.52L, Ampersand International,

Inc., Beechwood, OH). The upstream electrode was set to -1.0 V (range = 1 μA) and

the downstream electrode was set to 0.15 V (range = 50 nA). The upstream electrode

was used for disulfide reduction, and the downstream electrode was used for thiol

detection. In all microdialysis experiments for the detection of glutathione, samples

were collected into an equal volume of acid (0.12 M monochloroacetic acid pH = 3.0)

to stabilize the thiol/disulfide ratio. A representative chromatogram of a liver

dialysate sample is shown in Figure 4.4.

4.3.6. NMR Spectroscopy

See Section 2.3.3.

4.3.7. Data Analysis

See Section 2.3.4.

Page 179: ~Chapter 1~ Introduction 1.1. Biomarkers

179

Figure 4.4. LC/EC chromatogram of liver dialysate. 1 corresponds to GSH and 2 corresponds to GSSG.

ch1

2 4 1686 10 12 14

1 nA1

2

Time (min)

ch1

2 4 1686 10 12 142 4 1686 10 12 14

1 nA1

2

Time (min)

Page 180: ~Chapter 1~ Introduction 1.1. Biomarkers

4.4. Results and Discussion

Tissue-targeted metabonomics lends itself to the study of conditions that

induce spatially-specific metabolic changes that would otherwise go undetected by

conventional metabonomics sampling approaches. One such condition is oxidative

stress, which can be induced by a variety of mechanisms. Here, metabolic changes

induced by ischemia/reperfusion or doxorubicin in heart and liver tissue are

presented, along with concurrent studies of oxidative stress using established

biomarkers, glutathione and its disulfide. However, initially, the behavior of

doxorubicin in the analytical setup was characterized.

4.4.1. Doxorubicin Method Development

Doxorubicin presented some significant analytical challenges for both

microdialysis and LC experiments, due to its hydrophobicity and propensity to

photodegradation. Doxorubicin was detected by LC/UV in liver dialysate, as detailed

in Section 4.3.3. Carryover for the method was about 1%. Because doxorubicin is

fairly hydrophobic, sticking to the glass syringe or tubing components created a

significant carryover problem. This was corrected by rinsing the syringe with three

volumes of isopropanol followed by three volumes of Ringer’s solution (the sample

matrix). The injection loop (25 μL volume) was also rinsed with four volumes of

isopropanol followed by four volumes of Ringer’s solution. The linear range for

doxorubicin was 1-75 μM, and precision was 2-3%. A series of in vitro experiments

were performed to characterize doxorubicin behavior with probe components (tubing

and membrane), as well as with the glass syringe used for perfusion. Additionally,

180

Page 181: ~Chapter 1~ Introduction 1.1. Biomarkers

181

the degradation of doxorubicin over time and by light was studied. The setup for

these experiments is described in Section 4.3.2.1.

4.4.1.1. Doxorubicin Adsorption in Microdialysis Experiments

The adsorption of doxorubicin to microdialysis probe components was

examined. Microdialysis probes are composed of Tygon tubing (used as an adapter

between the probe and syringe), polyimide tubing and the membrane, which can be

composed of many different polymers. (Membrane selection will be examined in

Section 4.4.1.3.) For linear microdialysis probes, either polyimide tubing or fused

silica is used. In this laboratory, polyimide is the tubing of choice, due to its

improved flexibility, making surgical probe implantation and tubing tunneling under

the skin easier. However, polyimide may be more prone to interactions with

hydrophobic compounds. Thus, it is important to characterize in vitro the effects of

the equipment setup and microdialysis probe composition on doxorubicin levels in

the perfusate.

Figure 4.5 shows the adsorption of doxorubicin to the microdialysis probe

setup. In this experiment, the doxorubicin solution was analyzed in three ways: the

perfusate solution prior to perfusing through the microdialysis probe, the solution

perfused through a length of polyimide tubing and the solution perfused through a

microdialysis probe (1 cm PAN membrane active window). The level of doxorubicin

was constant for both the solution from the polyimide tubing and from the

microdialysis probe, showing a constant doxorubicin delivery over time. A small but

significant loss of doxorubicin was observed after perfusing the solution through the

Page 182: ~Chapter 1~ Introduction 1.1. Biomarkers

0 50 100 150 200 250 300 350 4000

2000

4000

6000

8000

10000

12000

14000

16000

18000

20000

Pea

k H

eigh

t

Time (min)

Figure 4.5. Examination of doxorubicin adsorbance to the probe components. The dashed line indicates the initial level of doxorubicin in the perfusate solution (10 μM doxorubicin in Ringer’s solution). indicates the level of doxorubicin after perfusing the solution through a length of polyimide tubing. ◊ indicates the level of doxorubicin after perfusing the solution through a complete microdialysis probe with a 10 mm PAN membrane.

182

Page 183: ~Chapter 1~ Introduction 1.1. Biomarkers

183

polyimide tubing alone, indicating adsorption in some component of the in vitro

experiment setup. This could be due to one of three components that the perfusate

solution is exposed to: the glass syringe, Tygon tubing or polyimide tubing. Figure

4.6 shows a repeat of the experiment performed in Figure 4.5, but each of the probe

components were examined individually. The loss of doxorubicin from the syringe,

Tygon tubing and polyimide tubing was the same. Therefore, it can be concluded that

the primary source of adsorption of doxorubicin in its perfusion through a

microdialysis probe originates with the glass syringe. This is the first component of

the perfusion setup that the doxorubicin solution contacts. While it is important to

note these sources of adsorption, this should not pose a problem for in vivo

microdialysis experiments. The levels of doxorubicin in the solution from the syringe

are constant over time, indicating that adsorption will not cause unwanted fluctuations

in doxorubicin levels, which could potentially introduce variance into the metabolic

profile data.

4.4.1.2. Doxorubicin Solution Stability

The stability of doxorubicin solutions was a concern, as it is known to

undergo degradation, particularly at lower concentrations. It has been demonstrated

that doxorubicin is most stable at 2 mg/mL (~3 mM) concentration for storage

purposes18. However, the doxorubicin concentration in at least some of the dialysate

samples was expected to be in the low micromolar range. Therefore, it was important

to establish the window of stability during which the dialysate samples could be

analyzed for doxorubicin levels by LC/UV. A set of doxorubicin standards (10, 5, 1

Page 184: ~Chapter 1~ Introduction 1.1. Biomarkers

0 50 100 150 200 2500

5000

10000

15000

20000

25000

30000

Pea

k H

eigh

t

Time (min)

Figure 4.6. Examination of doxorubicin adsorbance to individual microdialysis probe components. The perfusate solution was 10 μM doxorubicin in Ringer’s solution. indicates the level of doxorubicin in the perfusate, which was stored in the dark in a polypropylene vial through the experiment. Δ indicates the level of doxorubicin after perfusing the solution through a length of Tygon tubing. ◊ indicates the level of doxorubicin after perfusing the solution through a length of polyimide tubing. indicates the level of doxorubicin sampled directly from the glass syringe used to perfuse solution through the tubing.

184

Page 185: ~Chapter 1~ Introduction 1.1. Biomarkers

185

and 0.5 μM) were prepared in Ringer’s solution and analyzed by LC/UV in triplicate.

(Standards were stored in the dark at room temperature between injections on the

same day.) The remaining standard solutions were then aliquoted into 10 μL portions

in polypropylene vials, protected from light and stored at 4°C. The doxorubicin stock

solution (2 mg/mL in Ringer’s solution) was also stored with the standard solutions.

After 2 days, fresh standards were prepared from the stock solution and analyzed,

along with the two-day-old standards. Figure 4.7 shows the results of the study. The

standards prepared from the stock solution on the second day match the calibration

curve generated on the initial day of analysis, confirming that the doxorubicin stock

solution is stable at the high concentration and under the storage conditions described.

A large drop in response was observed for the older standard solutions. The lower

doxorubicin standard concentrations (1 and 0.5 μM) did not give a detectable signal.

The results of this study suggest that dialysate samples should be analyzed without

delay after collection to avoid any quantitation problems due to degradation.

Another concern with the microdialysis setup is the stability of the

doxorubicin solution in the glass syringe over the course of the experiment.

Depending on the doxorubicin dosing scheme, doxorubicin could remain in the

syringe for several hours at room temperature while being perfused through the

microdialysis probe. Photodegradation effects could be avoided by covering the

syringe with foil, but other degradation mechanisms could cause significant changes

in doxorubicin levels over time in the syringe. To characterize this potential problem,

a solution of 10 μM doxorubicin and 10 μM antipyrine in Ringer’s solution was

Page 186: ~Chapter 1~ Introduction 1.1. Biomarkers

0 2 4 6 8 10

0

5000

10000

15000

20000

25000

Peak

Hei

ght

Concentration (μM)

Figure 4.7. Stability of doxorubicin standards in Ringer’s solution. indicates standard solutions prepared on day 0, the first day of analysis. indicates standard solutions prepared two days after the initial analysis from the original stock solution. indicates standard solutions prepared on day 0 and analyzed two days later.

186

Page 187: ~Chapter 1~ Introduction 1.1. Biomarkers

187

prepared, and two glass 1 mL syringes (Hamilton Company, Reno, NV) were filled

with the solution. The syringes were covered in foil and placed in the syringe pump.

The flow was set to 1 μL/min. Samples were collected hourly over the course of the

day and analyzed by LC/UV immediately. The perfusate solution (stored in the dark

in a polypropylene vial) was also analyzed over the course of the day. The syringes

(and perfusate solution) were then allowed to sit overnight at room temperature, still

wrapped in foil. The following day, the delivery experiment was repeated, and

injections were made of solution from the syringe to show the long-term stability of

the solution in the syringe. Figure 4.8 shows the stability of both antipyrine and

doxorubicin over time. Antipyrine showed good stability with no adsorption of the

analyte to the syringe, which was expected. Doxorubicin levels in the solution from

the syringes were significantly lower at 95% confidence than levels in the initial

solution due to adsorption of the analyte to the glass syringe. However, doxorubicin

in all experiment groups showed fairly good stability, particularly over the course of

the initial 10 hours of analysis. The RSD of doxorubicin levels in all experiment

groups was approximately 5%. Therefore, although doxorubicin showed adsorption

to the walls of the glass syringe, this did not impact the stability of doxorubicin levels

coming from the syringe. This is an important point for the dosage of doxorubicin by

delivery through the microdialysis probe.

Doxorubicin has been shown to degrade in the presence of light18,19.

Photodegradation of a solution of 10 μM doxorubicin was examined. The solution

was split into two polypropylene vials. One vial was stored at room temperature in

Page 188: ~Chapter 1~ Introduction 1.1. Biomarkers

188

Figure 4.8. Stability of doxorubicin (A) and antipyrine (B) in a glass syringe over time. indicates solute levels in the solution stored in a polypropylene vial in the dark, and indicates solute levels in the solution stored in a glass syringe in the dark.

1200 1400 1600

-200 0 200 400 600 800 10006000

8000

10000

12000

14000

Pea

k H

eigh

t

Time (min)

-200 0 200 400 600 800 1000

12000

14000

16000

18000

20000

1200 1400 1600

Pea

k H

eigh

t

Time (min)

A

B

-200 0 200 400 600 800 10006000

8000

10000

12000

14000

1200 1400 1600

Pea

k H

eigh

t

Time (min)

-200 0 200 400 600 800 1000

12000

14000

16000

18000

20000

1200 1400 1600

Pea

k H

eigh

t

Time (min)

A

B

Page 189: ~Chapter 1~ Introduction 1.1. Biomarkers

189

the dark, while the other vial was stored at room temperature at normal indoor

lighting conditions on the laboratory bench. Samples were analyzed periodically

from both solutions. The results of the experiment are shown in Figure 4.9. The

results from this experiment are difficult to interpret. The solution stored in the dark

showed a low response from doxorubicin at three consecutive timepoints (150-250

minutes) that returned to initial levels for the last two time points. The origin of these

low data points is unknown. Samples taken from the solution stored in light showed a

steady decline in doxorubicin levels over the course of the sampling period (~6

hours). It is unclear whether this is a significant decline, due to the variation

observed from samples analyzed from the dark solution. Regardless, some

photodegradation was expected, and vials were covered in foil to lessen its effects in

doxorubicin quantitation.

4.4.1.3. Doxorubicin In Vitro Behavior

Microdialysis probes were calibrated with doxorubicin in vitro to characterize

the behavior of the probe and approximate its behavior in vivo. Each probe was

characterized by both a delivery and a recovery experiment, as described in Section

4.3.2.1. Probe extraction efficiency can be calculated by characterizing either probe

delivery or recovery – theoretically, either technique should yield the same probe

extraction efficiency. This has been demonstrated for linear probes previously by this

laboratory20. In general, delivery experiments were performed first, followed by the

recovery experiment. Figure 4.10 shows the result of microdialysis probe calibrations

by doxorubicin, with the recovery and delivery experiments performed in different

Page 190: ~Chapter 1~ Introduction 1.1. Biomarkers

-50 0 50 100 150 200 250 300 350

6000

8000

10000

12000

14000

16000

18000

20000

22000

Pea

k H

eigh

t

Time (min)

Figure 4.9. Photostability of 10 μM doxorubicin in Ringer’s solution. The dashed line indicates the initial level of doxorubicin in the perfusate solution (10 μM doxorubicin in Ringer’s solution). indicates the solution stored in the dark, and indicates solution stored on the benchtop, exposed to normal indoor lighting conditions.

190

Page 191: ~Chapter 1~ Introduction 1.1. Biomarkers

191

Figure 4.10. Method for calibration of microdialysis probes (1 cm PAN membrane) with doxorubicin. indicates the delivery of doxorubicin, as performed first in the calibration experiment. indicates the recovery of doxorubicin, as performed second in the calibration experiment. indicates the delivery of doxorubicin, as performed second in the calibration experiment. indicates the recovery of doxorubicin, as performed first in the calibration experiment.

0 20 40 60 80 100 120 1400

10

20

30

40

50

60

70

80

90

100

Prob

e E

xtra

ctio

n E

ffici

ency

Time (min)0 20 40 60 80 100 120 140

0

10

20

30

40

50

60

70

80

90

100

Prob

e E

xtra

ctio

n E

ffici

ency

Time (min)

Page 192: ~Chapter 1~ Introduction 1.1. Biomarkers

192

orders. For both experiments, the delivery of doxorubicin was level and consistent

throughout the experiment. The delivery was unaffected by the order of the

calibration experiment. The recovery of doxorubicin showed significant differences

between experiments. Additionally, the observed probe extraction efficiency began

low for each recovery experiment and took about 30 minutes for extraction efficiency

values to stabilize. The order of the experiment significantly changed the extraction

efficiency values observed for the recovery portion of the experiment, by 20-25%.

The reason for this large discrepancy is unclear, but is likely connected to

doxorubicin’s hydrophobicity. Doxorubicin readily adsorbs to PAN membrane. The

membrane is a deep red color after perfusion with doxorubicin. Immersion of the

probe in a doxorubicin solution rapidly increases the amount of doxorubicin adsorbed

to the membrane, while this seems to be a slower process in the delivery experiments.

In these experiments, the doxorubicin solution is only perfused through the probe,

while the probe itself is submerged in Ringer’s solution.

When such a large (and reproducible) discrepancy is observed in data, it is

advisable to verify the analytical methodology and technique of the scientist

performing the experiment. With this in mind, the in vitro experiments described

above were repeated by two scientists. Probes were constructed by each scientist, and

each scientist independently calibrated one probe fabricated by the other (two probes

total). All experiments were performed on the same day. The probes were perfused

with 10 μM doxorubicin and 10 μM antipyrine in Ringer’s solution. Delivery

experiments were performed first, followed by recovery experiments. Figure 4.11

Page 193: ~Chapter 1~ Introduction 1.1. Biomarkers

193

Figure 4.11. Comparison of microdialysis probe (1 cm PAN membrane) calibration technique, as performed by two different scientists. Calibrated for doxorubicin (A) and antipyrine (B) with two different probes – one made by each scientist. indicates delivery values as determined by scientist #1. indicates recovery values as determined by scientist #1. indicates delivery values as determined by scientist #2. indicates recovery values as determined by scientist #2.

120

0 20 40 60 80 1000

10

20

30

40

50

60

70

80

90

100

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

0 20 40 60 80 1000

10

20

30

40

50

60

70

80

90

100

120

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

A

B

0 20 40 60 80 1000

10

20

30

40

50

60

70

80

90

100

120

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

0 20 40 60 80 1000

10

20

30

40

50

60

70

80

90

100

120

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

A

B

Page 194: ~Chapter 1~ Introduction 1.1. Biomarkers

shows the results of the experiment. Probe calibration by antipyrine shows consistent

results, with matching values for probe delivery and recovery over time. The

antipyrine results indicate the precision of both the probe fabrication and analytical

methodology, so any discrepancies observed in the doxorubicin data were due to the

behavior of doxorubicin itself. The doxorubicin results show good reproducibility for

the delivery of doxorubicin, reaching a similar steady-state level, both between

scientists and probes. These values were also close to those obtained to antipyrine.

However, there is a large difference between the delivery and recovery values for

both probes, as determined by both scientists. The recovery shows variation at the

initial time points, with some convergence in recovery values observed at later time

points. Although the recovery of doxorubicin in vitro was problematic, the delivery

was really of primary concern. This is the mechanism by which the drug will be

delivered to the tissue. It is consistent and reproducible, indicating that the drug dose

will be consistent as well.

The effect of the probe membrane material on the in vitro behavior of

doxorubicin was examined. Many membrane polymers are available, but

polyacrylonitrile (PAN) and cuprophan membranes were characterized in this

investigation because of their accessibility and successful use in previous

microdialysis experiments. PAN membranes are advantageous for microdialysis

experiments because the membrane is robust, i.e., does not kink or break easily. This

simplifies both fabrication and implantation. PAN is a hydrophilic membrane with a

20 kDa molecular weight cutoff (MWCO) and is highly negatively charged on the

194

Page 195: ~Chapter 1~ Introduction 1.1. Biomarkers

195

membrane surface20. Cuprophan (regenerated cellulose) is also a hydrophilic

membrane, but the membrane surface is neutral20. It has a 12 kDa MWCO and is

much more brittle than PAN membrane. The delivery and recovery for doxorubicin

and antipyrine (as described for previous experiments) was determined for

microdialysis probes made of 1 cm PAN or 1 cm cuprophan membrane, both

prepared in-house. Figure 4.12 shows the results of the experiment. Antipyrine

behaved similarly with both membrane types. (One probe with a PAN membrane

showed lowered recovery values, but this was the only probe out of numerous in vitro

experiments that exhibited this behavior for antipyrine.) Doxorubicin showed

significant differences between the delivery values obtained from the PAN and

cuprophan membranes. Both cuprophan and PAN showed much lower recovery

values for doxorubicin. Thus, this discrepancy between delivery and recovery values

of doxorubicin was not remedied using the cuprophan membrane.

Recovery and delivery values can change with analyte concentration. The

effects of doxorubicin concentration on probe delivery and recovery were

characterized (antipyrine was used as a control). Probes were calibrated by both

delivery and recovery of antipyrine and doxorubicin at 10, 50 and 100 μM

concentrations for both analytes in Ringer’s solution. Both PAN and cuprophan

membranes were compared. The results are shown in Figure 4.13. (In the 100 μM

experiment, the cuprophan membrane was torn, and recovery results could not be

obtained.) Overall, no concentration dependence was noted for the recovery or

Page 196: ~Chapter 1~ Introduction 1.1. Biomarkers

196

Figure 4.12. The effects of membrane composition on the probe extraction efficiency for doxorubicin (A) and antipyrine (B). indicates delivery values for the probe made with PAN membrane. indicates recovery values for the probe made with PAN membrane. indicates delivery values for the probe made with cuprophan membrane. indicates recovery values for the probe made with cuprophan membrane.

160

0 20 40 60 80 100 120 1400

10

20

30

40

50

60

70

80

90

100

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

0 20 40 60 80 100 120 1400

10

20

30

40

50

60

70

80

90

100

160

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

A

B

0 20 40 60 80 100 120 1400

10

20

30

40

50

60

70

80

90

100

160

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

0 20 40 60 80 100 120 1400

10

20

30

40

50

60

70

80

90

100

160

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

A

B

Page 197: ~Chapter 1~ Introduction 1.1. Biomarkers

197

Figure 4.13. The effect of solute concentration on probe extraction efficiency for both PAN (A, C) and cuprophan (B, D) membranes. Doxorubicin is measured in A and B, and antipyrine is measured in C and D. Square symbols show the probe extraction efficiency for a 10 μM solution, circle symbols show the probe extraction efficiency for a 50 μM solution, and triangle symbols show the probe extraction efficiency for a 100 μM solution. Closed symbols show probe delivery and open symbols show probe recovery.

0 20 40 60 80 100 1200

10

20

30

40

50

60

70

80

90

100P

robe

Ext

ract

ion

Effi

cien

cy

Time (min)0 20 40 60 80 100 120

0

10

20

30

40

50

60

70

80

90

100

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

0 20 40 60 80 100 1200

10

20

30

40

50

60

70

80

90

100

Prob

e E

xtra

ctio

n Ef

ficie

ncy

Time (min)0 20 40 60 80 100 120

0

10

20

30

40

50

60

70

80

90

100

Prob

e E

xtra

ctio

n Ef

ficie

ncy

Time (min)

A B

C D

0 20 40 60 80 100 1200

10

20

30

40

50

60

70

80

90

100P

robe

Ext

ract

ion

Effi

cien

cy

Time (min)0 20 40 60 80 100 120

0

10

20

30

40

50

60

70

80

90

100

Pro

be E

xtra

ctio

n E

ffici

ency

Time (min)

0 20 40 60 80 100 1200

10

20

30

40

50

60

70

80

90

100

Prob

e E

xtra

ctio

n Ef

ficie

ncy

Time (min)0 20 40 60 80 100 120

0

10

20

30

40

50

60

70

80

90

100

Prob

e E

xtra

ctio

n Ef

ficie

ncy

Time (min)

A B

C D

Page 198: ~Chapter 1~ Introduction 1.1. Biomarkers

delivery of either antipyrine or doxorubicin. Probes behaved as expected for each

membrane type and analyte, as determined by previous experiments.

In summary, doxorubicin shows deviations from “ideal” behavior for

microdialysis experiments, including adsorption to glass and discrepancies between

the delivery and recovery of the analyte by the microdialysis probe. In these studies,

these discrepancies were characterized to aid in the interpretation of data in the in

vivo experiments. These experiments demonstrated an important property of

doxorubicin that was key in data interpretation for metabonomics experiments. First,

the delivery of doxorubicin was very consistent over time, matching the delivery

observed for antipyrine (the internal standard). In most of the experiments,

doxorubicin was dosed through the microdialysis probe. These results show that the

doxorubicin dose will be consistent between animals and likely will not add to

variation in the experiments.

4.4.2. Myocardial Oxidative Stress

One of the most susceptible tissue targets of oxidative stress is the heart.

Whether the oxidative stress is physiologically induced by ischemia/reperfusion or is

chemically induced by the administration of a drug such as doxorubicin, metabolism

in the tissue is disrupted, resulting in detrimental and perhaps permanent effects to the

tissue, including contractile dysfunction and congestive heart failure.

Preliminary studies were performed to characterize metabolic changes

induced in heart tissue by both physiologically induced and chemically induced

oxidative stress. All experiments were performed on anesthetized animals, as the

198

Page 199: ~Chapter 1~ Introduction 1.1. Biomarkers

199

induction of myocardial ischemia is nearly impossible to perform in awake animals,

for both surgical and ethical reasons. In these experiments, basal heart dialysate was

collected from rats for two hours (samples collected every 30 minutes) prior to the

induction of oxidative stress. Oxidative stress was induced physiologically by

temporary ligation of the left anterior descending coronary artery or induced

chemically by the administration of a systemic dose of doxorubicin (15 mg/kg

intraperitoneally).

The metabolic consequences of both methods of inducing oxidative stress

were compared in the principal components analysis (PCA) scores plot in Figure

4.14. The analysis indicates that physiological and chemical oxidative stress may

have different metabolic signatures in heart tissue. The right-facing arrow in the

scores plot indicates the metabolic trend observed as a result of ischemia/reperfusion,

while the downward-facing arrow shows the direction of the metabolic trend induced

by doxorubicin administration. In this scores plot, the first principal component

distinguishes between control experiment samples and ischemia/reperfusion

experiment samples. The second principal component distinguishes between control

experiment samples and doxorubicin experiment samples. The loadings for the first

principal component show lysine, alanine, glutamine, lactate, citrate, β-

hydroxybutyrate and glucose as important. Interestingly, creatine is not identified in

this analysis as important, even though ischemia/reperfusion is known to significantly

alter the levels of creatine through dephosphorylation of creatine phosphate. In

Figure 4.15, the simultaneous metabolic changes induced by ischemia/reperfusion in

Page 200: ~Chapter 1~ Introduction 1.1. Biomarkers

200

myocardial ischemia/reperfusion and indicates samples from rats dosed with doxorubicin. The arrows designate the direction of metabolic trends resulting from ischemia/reperfusion or doxorubicin dose. Integral data was normalized to the initial time point prior to PCA.

Figure 4.14. Principal components analysis of heart dialysate metabolic profiles. Δ indicates samples from control rats, indicates samples from rats undergoing

-10 -5 0 5 10 15 20

-9

-8

-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

Prin

cipa

l Com

pone

nt 2

Principal Component 1-10 -5 0 5 10 15 20

-9

-8

-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

Prin

cipa

l Com

pone

nt 2

Principal Component 1

Page 201: ~Chapter 1~ Introduction 1.1. Biomarkers

201

Figure 4.15. Comparison of NMR spectra of heart dialysate from a rat experiencing myocardial ischemia/reperfusion. A depicts basal heart dialysate, B depicts ischemic heart dialysate and C depicts reperfused heart dialysate. Three key metabolites in the spectra are labeled: 1, lactate; 2, creatine; 3, glucose.

4.10 4.00 3.90 3.80

1H Chemical Shift (ppm)

A

B

C

2

3

1

4.10 4.00 3.90 3.80

1H Chemical Shift (ppm)

A

B

C

4.10 4.00 3.90 3.804.10 4.00 3.90 3.804.10 4.00 3.90 3.80

1H Chemical Shift (ppm)

A

B

C

2

3

1

Page 202: ~Chapter 1~ Introduction 1.1. Biomarkers

three key energy metabolites are depicted. Glucose falls rapidly and lactate increases

after the induction of ischemia. Creatine remains level, with a small increase during

the reperfusion period. These results are in agreement with the PCA loadings.

Doxorubicin affects different metabolites; the loadings for the second principal

component indicate that glucose, valine, citrulline and creatine levels are altered in

response to doxorubicin administration.

The results described above are preliminary, with the doxorubicin-dosed

group only containing n=1. However, early indications suggest a differential

metabolic response. Some of the metabolites changing in response to myocardial

ischemia/reperfusion are in accordance with previously established trends in

literature, with glucose and lactate changing as a result of the heart muscle’s switch to

anaerobic metabolism under restricted blood flow21. The other metabolites (lysine,

alanine, glutamine, citrate and β-hydroxybutyrate) are involved in energy

metabolism, but through many different metabolic pathways. Doxorubicin may tap

into different pathways than ischemia/reperfusion, with different metabolites

earmarked as important. No anaerobic metabolism was induced, but alterations in

energy stores were noted, as were changes in nitrogen metabolism. This is the first

time that myocardial oxidative stress has been examined using metabonomics,

particularly using a sampling technique to focus on the actual tissue metabolism.

Additionally, this is the first metabonomics comparison of oxidative stress events

induced by different mechanisms.

202

Page 203: ~Chapter 1~ Introduction 1.1. Biomarkers

In spite of these preliminary results, it was decided to move the oxidative

stress investigations to the liver. Experimentally, microdialysis sampling of heart

tissues imposes restrictions on the metabonomics studies that can be overcome by

moving to liver studies. Heart microdialysis probes are limited in the active

membrane length, thus limiting probe extraction efficiency. By lowering the

concentration of metabolites in the dialysate sample, several possible negative effects

on the method could result. First, the number of detected metabolites will likely

decrease because the concentration in the original dialysate sample will be lowered

due to poorer extraction efficiency. Second, the throughput of the method could be

lowered due to increased NMR experiment time to compensate for the more dilute

samples by signal averaging longer. Third, the time resolution of the method could

be worsened by sampling for longer periods of time to concentrate the sample to

compensate for the lower extraction efficiency of the heart microdialysis probes.

Thus, the decision was made to move to liver microdialysis experiments. For these

experiments, the active membrane length can be longer, thus increasing probe

extraction efficiency. Additionally, the microdialysis experiments are more amenable

to awake animal studies.

4.4.3. Hepatic Oxidative Stress

In these experiments, doxorubicin was used to chemically induce oxidative

stress only in the liver tissue immediately surrounding one of the implanted

microdialysis probes. One of the advantages of microdialysis sampling is that drugs

can be delivered to the surrounding tissue while the endogenous metabolites are

203

Page 204: ~Chapter 1~ Introduction 1.1. Biomarkers

collected from the same tissue being dosed. By adding a second probe in the liver

lobe unaffected by the doxorubicin dose as a control (referred to as Probe A), natural

changes in liver metabolism could be identified and separated from metabolic

changes induced by doxorubicin (Figure 4.2).

Doxorubicin can be dosed to examine toxicity from two perspectives – acute

and chronic. The cardiotoxic side effects of doxorubicin are typically the result of

chronic toxicity, induced by a constant, lower dose of the drug. However, in these

studies, the acute toxicity of doxorubicin was probed for several reasons. First, the

liver was studied rather than the heart, making induction of cardiotoxicity less

interesting. Second, acute toxicity studies are more efficient and easier to study by

microdialysis because the timescale is hours to days rather than weeks to induce the

metabolic change. Finally and most importantly, it would be difficult to separate the

initial toxic effects of doxorubicin from changes caused by tissue repair and

adaptation in chronic toxicity studies22. It should be acknowledged that the toxic side

effects of doxorubicin may differ depending on the dose or time given23-25. For

example, apoptosis may be observed at lower concentrations of doxorubicin, while

free radical-mediated toxicity is dominant at higher concentrations23. Doxorubicin

has been shown to have varying degrees of toxicity when it is administered at

different times of day26. In these studies, the doxorubicin dose was given at the same

time of day for each rat.

204

Page 205: ~Chapter 1~ Introduction 1.1. Biomarkers

205

4.4.3.1. Doxorubicin Dosage Studies

The doxorubicin dosing scheme in liver microdialysis experiments was

described in Section 4.3.2.3. Doxorubicin was analyzed in dialysate from both probes

using LC/UV, in order to both monitor the time course of doxorubicin and confirm

that no doxorubicin was detected in the control probe. Figure 4.16 shows

doxorubicin levels over time. There is an initial spike of the drug when it is perfused

through the probe, and then it gradually decays over the next few hours. Thus, the

dose of doxorubicin provides a single, intense metabolic insult to the tissue. Figure

4.16 also confirms that this metabolic insult is localized, as no doxorubicin was

detected in the dialysate from the control probe A. This is further confirmed by

histological analysis of the liver tissue, as shown by the representative picture in

Figure 4.17. The tissue around probe B shows a large necrotic zone, which is lighter

in color than the surrounding healthy tissue. This zone does not extend to the tissue

around probe A, confirming that the dialysate sampled at probe A should be

representative of the natural metabolic profile of healthy liver tissue.

The time course of damage from the doxorubicin dose was examined through

a histology study. In this study, probes were implanted as described previously. Rats

were maintained under anesthesia. Thirty minutes after probe implantation, probes

were perfused with Ringer’s solution for three hours to mimic the basal dialysate

sampling period. The perfusate was then switched to 2 mg/mL doxorubicin in

Ringer’s solution for one hour. At the end of the dosing period, the rats were put

down at various time points (0, 2, 5, 6 and 8 hours post-dose). Livers were harvested

Page 206: ~Chapter 1~ Introduction 1.1. Biomarkers

-4 -2 0 2 4 6 8-100

0

100

200

300

400

500

600

700

Dox

orub

icin

Dia

lysa

te C

once

ntra

tion

(uM

)

Time (hour)

Figure 4.16. Timecourse of doxorubicin dosage and clearance through the microdialysis probe, as measured by the dialysate concentration. represents samples from Probe A, the undosed/control probe and Δ represents samples from Probe B, through which doxorubicin was dosed.

206

Page 207: ~Chapter 1~ Introduction 1.1. Biomarkers

207

Figure 4.17. Histology of liver with two microdialysis probes implanted (A and B). Doxorubicin was perfused through probe B. The light area around Probe B is a zone of necrotic tissue.

A

B

A

B

Page 208: ~Chapter 1~ Introduction 1.1. Biomarkers

208

and prepared for histological analysis. Dr. Mike Thompson, pathologist at Lawrence

Memorial Hospital, helped in the interpretation of the slides. The results of the

histological study are shown in Table 4.1. The control probe (Probe A) showed a

normal immune response to the probe, with small amounts of damage from probe

implantation and some neutrophil recruitment at later time points. Very little tissue

damage occurred around Probe B immediately after dosing – it became more evident

2 hours post-dose. Maximum damage appeared to occur 5 hours post-dose.

Consistently, the amount of damage around Probe A from implantation and immune

response was substantially lower than the damage induced by doxorubicin. The data

from this study was also used to approximate a doxorubicin “diffusion coefficient”

from the probe through the tissue. The maximum distance of damage from the probe

was measured and divided by the time post-dose. The results are shown in Table 4.2.

Diffusion coefficient approximations were fairly consistent among the time points,

with an average coefficient of 11 nm/min through the liver tissue for doxorubicin.

Additionally, the pharmacokinetics of the doxorubicin dose was explored, as

no literature has been published on the dosing of doxorubicin through a microdialysis

probe. The doxorubicin dose (perfusate concentration of 2 mg/mL perfused through

the microdialysis probe at 1 μL/min for 1 hour) was given in three experimental

settings: in vitro, in the liver of anesthetized animals and in the liver of awake

animals. All microdialysis probes utilized were of the same membrane length. The

overall time course for each set of experiments is shown in Figure 4.18. The

pharmacokinetics of doxorubicin in awake and anesthetized animals does not differ

Page 209: ~Chapter 1~ Introduction 1.1. Biomarkers

Time Post-Dose (Hour) Necrotic Zone Area Observations Probe A: 0 μm2 Probe A: Minimal change 0 Probe B: 0 μm2 Probe B: Minimal change

Probe A: 5.4 x 103 μm2 Probe A: Some cell death from probe implantation

2 Probe B: 1.3 x 107 μm2

Probe B: Liver toxicity developing; geographic necrosis evident

Probe A: 7.4 x 105 μm2 Probe A: Some cell death from probe implantation 5

Probe B: 1.9 x 107 μm2 Probe B: Zonal necrosis, but peripheral region still viable

Probe A: 1.3 x 104 μm2 Probe A: Some cell death from probe implantation 6

Probe B: 2.8 x 107 μm2 Probe B: Zonal necrosis, but peripheral region still viable

Probe A: 2.2 x 104 μm2 Probe A: Some cell death from probe implantation 8

Probe B: 1.1 x 107 μm2 Probe B: Zonal necrosis, but peripheral region still viable

Table 4.1. Progression of damage from doxorubicin, as gauged by histological analysis of liver tissue harvested at various time points post-dose.

209

Page 210: ~Chapter 1~ Introduction 1.1. Biomarkers

Time Post-Dose (Hour)

Maximum Distance (mm)

Diffusion Coefficient (nm/min)

2 1.2 10 5 3.1 10 6 5.0 14 Average: 11

Table 4.2. Doxorubicin diffusion coefficient approximation, as estimated from the histology study of the time course of doxorubicin damage.

210

Page 211: ~Chapter 1~ Introduction 1.1. Biomarkers

-1 0 1 2 3 4 5 6 7 8 9 10

10

100

1000

Dox

orub

icin

Con

cent

ratio

n (u

M)

Time (hour)

Figure 4.18. Comparison of doxorubicin levels post-dose in liver dialysate samples from in vitro (), anesthetized rat (Δ) and awake rat () experiments.

211

Page 212: ~Chapter 1~ Introduction 1.1. Biomarkers

212

significantly, at the 95% confidence level. Additionally, it can be seen from the

logarithmic scale that doxorubicin exhibits biphasic behavior both in vitro and in vivo

– a distribution and elimination phase is identifiable in the plot. This biphasic

behavior is likely from doxorubicin sticking to the probe membrane after dosing and

subsequently releasing from the membrane over time. This hypothesis was tested in

vitro by “dosing” both doxorubicin and antipyrine through the same microdialysis

probe for one hour. Antipyrine does not stick to PAN membrane, so if the biphasic

behavior of doxorubicin was due to membrane sticking, antipyrine levels should fall

rapidly in the dialysate after dosing. Figure 4.19 shows the results from this

experiment. This hypothesis was confirmed, as antipyrine did not linger in the

dialysate after dosing. The pharmacokinetic parameters of the doxorubicin dose were

calculated by fitting the doxorubicin concentration curves on a logarithmic scale to a

biexponential decay, according to the following equation:

(1)

Doxorubicin pharmacokinetic parameters were then calculated from the

coefficients27, and the results are shown in Table 4.3. No statistically significant

differences in distribution and elimination between awake and anesthetized animals

were found. This was unexpected, as anesthesia has been shown to alter the

pharmacokinetics of many other drugs. However, these drugs have been dosed

systemically, usually either intravenously or intraperitoneally. Very few

pharmacokinetics studies have been performed examining drug dosage through a

microdialysis probe. The use of microdialysis probes as a dosing vehicle may lead to

btat BAeC −− +=

Page 213: ~Chapter 1~ Introduction 1.1. Biomarkers

-0.5 0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0

0.01

0.1

1

Nor

mal

ized

Con

cent

ratio

n (u

M)

Time (Hour)

Figure 4.19. Comparison of doxorubicin () and antipyrine () levels in dialysate samples from an in vitro microdialysis experiment.

213

Page 214: ~Chapter 1~ Introduction 1.1. Biomarkers

Parameter In Vitro (n=2)

Anesthetized (n=3)

Awake (n=3)

t1/2 (distribution)

0.219 +/- 0.018 0.883 +/- 0.339 0.849 +/- 0.221

t1/2 (elimination)

3.07 +/- 0.05 17.6 +/- 13.2 11.4 +/- 9.0

Cmax (μM) 2540 +/- 370 1180 +/- 480 1300 +/- 1050 (693)

AUC 915 +/- 182 1280 +/- 450 1740 +/- 1700 (755)

Clearance (x 10-10)

1.78 +/- 0.33 0.864 +/- 0.288 0.952 +/- 0.677 (1.34)

Table 4.3. Doxorubicin pharmacokinetic calculations. In the awake rat data, one rat appeared to be an outlier. The numbers in parentheses correspond to the average parameter value when the possible outlier point is removed.

214

Page 215: ~Chapter 1~ Introduction 1.1. Biomarkers

215

experiments in which the results are less susceptible to anesthesia effects, perhaps

because such a small portion of the animal is affected.

4.4.3.2. Dual Probe Experiments in Anesthetized Animals

Doxorubicin was first administered to anesthetized animals. Although strong

metabolic trends were observed as a result of the anesthesia, as described above, it

was initially not known whether the magnitude of the metabolic changes induced by

doxorubicin would be greater than those induced by the anesthesia. Three hours of

basal dialysate were collected from each probe prior to dosing doxorubicin (2 mg/mL

perfusate concentration) for one hour through probe B. Samples were collected for 8

hours post-dose from both probes.

Figure 4.20 shows the scores plot of the first two principal components in the

PCA analysis of data for anesthetized animals dosed with doxorubicin. The data

segregates into dosed and undosed animals, with both data sets grouping in the top

left of the plot. Each rat trends away from the starting point, with undosed rats

trending downward along principal component 2 and dosed rats trending to the right

along principal component 1. Samples from rats taken at hours 6 and 12 prior to

doxorubicin dosing are marked with a star. In the dosed animals, there is no

segregation between probe A and B, as would be expected if doxorubicin only

perturbed the metabolism of the tissue around probe B. The timecourse data for

individual rats from the PCA is shown in Figure 4.21. (This is the same PCA as

Figure 4.20, but the plots are broken up to highlight individual rats’ metabolic trends.)

There is good consistency for both the dosed and control groups of data. It should be

Page 216: ~Chapter 1~ Introduction 1.1. Biomarkers

216

represents samples from Probe A of undosed rats, represents samples from Probe B of undosed rats, represents samples from Probe A of dosed rats and Δ represents samples from Probe B of dosed rats (dosed probes). Samples points with an * are samples acquired before doxorubicin was dosed.

0 12 14

Figure 4.20. Doxorubicin liver metabolic effects in anesthetized rats.

-4 -2 0 2 4 6 8 1-8

-6

-4

-2

0

2

4

Prin

cipa

l Com

pone

nt 2

Principal Component 1

***

***

**

* ***

-4 -2 0 2 4 6 8 1-8

-6

-4

-2

0

2

4

0 12 14

Prin

cipa

l Com

pone

nt 2

Principal Component 1

***

***

**

* ***

Page 217: ~Chapter 1~ Introduction 1.1. Biomarkers

217

Figure 4.21. Doxorubicin liver metabolic effects in anesthetized rats. This is the same PCA as Figure 4.18, but displayed by individual rat. represents samples from Probe A of undosed rats, represents samples from Probe B of undosed rats, represents samples from Probe A of dosed rats and Δ represents samples from Probe B of dosed rats (dosed probes). A, B, and C show individual anesthetized rats dosed with doxorubicin and D shows time trends from undosed anesthetized rats (n=4).

-4 -2 0 2 4 6 8 10 12

-8

-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

PC2

PC1-4 -2 0 2 4 6 8 10 12

-8

-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

PC2

PC1

-4 -2 0 2 4 6 8 10 12

-8

-6

-4

-2

0

2

4

PC

2

PC1-4 -2 0 2 4 6 8 10 12

-8

-6

-4

-2

0

2

4

PC

2

PC1

A

D

B

C

-4 -2 0 2 4 6 8 10 12

-8

-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

PC2

PC1-4 -2 0 2 4 6 8 10 12

-8

-7

-6

-5

-4

-3

-2

-1

0

1

2

3

4

PC2

PC1

-4 -2 0 2 4 6 8 10 12

-8

-6

-4

-2

0

2

4

PC

2

PC1-4 -2 0 2 4 6 8 10 12

-8

-6

-4

-2

0

2

4

PC

2

PC1

A

D

B

C

Page 218: ~Chapter 1~ Introduction 1.1. Biomarkers

noted that the trends observed in dosed animals are unidirectional, meaning that the

changes observed to the tissue from perfusing doxorubicin through the probe are

permanent and irreversible (at least as observed over a 12 hour period).

From the scores plot in Figure 4.20, it appears that doxorubicin is causing a

metabolic change in the dosed rats, but it is possibly a global effect rather than

localized at the dosage site. This is because the metabolic profiles from both Probe A

and B change in the same way in response to the doxorubicin dose. Histological

analysis did confirm that no necrosis was observed around Probe A and LC-UV

analysis did not detect doxorubicin in probe A. Principal component 1 is the

component responsible both for separating control and dosed samples, as well as

defining the time trend observed in dosed animals. The loadings for principal

component 1 show the following metabolites as important: lysine, alanine, valine,

leucine, isoleucine, creatine and β-hydroxybutyrate. Metabolic implications of this

will be discussed further in Section 4.4.3.5.

4.4.3.3. Glutathione: Biomarker of Oxidative Stress

Metabonomics in general is an exploratory analytical technique, with one of

the primary goals being to identify new biomarkers of pathophysiological conditions.

However, biomarkers have been previously established for many conditions, although

their reliability and relevance to the condition being studied may be questionable

(thus necessitating these exploratory techniques for additional and/or better

biomarkers). It would be prudent to perform concurrent studies of established

biomarkers of the condition of interest with exploratory metabonomics studies. This

218

Page 219: ~Chapter 1~ Introduction 1.1. Biomarkers

could both aid in interpretation of the metabonomics data and help to validate

potential newly identified biomarkers.

Oxidative stress in particular has many previously established biomarkers,

both small molecules and enzymes. Oxidative stress can be gauged by products of

oxidative damage, including malondialdehyde, a product of lipid peroxidation, and

spin trapping of the actual free radicals. It can also be gauged by the presence of

repair products or alterations in antioxidant levels. Glutathione is one of the most

prevalent antioxidants in the body, serving a variety of purposes. It is well

established that both the levels of glutathione and the ratio of the thiol to its disulfide

are markers of oxidative stress. Doxorubicin when administered systemically causes

a decrease in liver glutathione concentration28. Glutathione also detoxifies

doxorubicin through conjugation, as catalyzed by glutathione-S-transferase28,29. The

conjugate is excreted into the bile.

Glutathione (GSH) and its disulfide (GSSG) were chosen as the biomarkers of

oxidative stress to monitor in conjunction with liver dialysate metabolic profiles.

GSH and GSSG were chosen for a couple of reasons. First, they are present in the

extracellular fluid in the high nanomolar to low micromolar range, making

quantitation amenable to a variety of analytical techniques. Second, the response of

glutathione to oxidative stress is rapid. Biomarkers such as 8-hydroxy-2’-

deoxyguanosine from the base excision repair process can have a significant lag time

in response to an oxidative stress event. For these experiments, it would be difficult

219

Page 220: ~Chapter 1~ Introduction 1.1. Biomarkers

to correlate more immediate metabolic changes to biomarker fluctuations occurring

several hours later.

These studies were initially performed in anesthetized animals to establish

patterns that could be correlated to the metabolic profile changes described in the

previous section. Initially, glutathione was quantitated using an established method

in our laboratory, CE/UV. However, this could only monitor total glutathione levels.

An LC/EC method was then developed to quantitate GSH and GSSG separately in

liver dialysate samples. This method was used to compare trends in rats anesthetized

with ketamine and isoflurane inhalation, as well as to examine trends in awake and

anesthetized rats dosed with doxorubicin.

4.4.3.3.1. CE/UV: “Total” Glutathione

Total glutathione in liver dialysate samples was quantified by a CE/UV

method previously established in our laboratory, as described in Section 4.3.4. This

method was unable to detect both GSH and GSSG, due to on-capillary oxidation

effects previously noted in our laboratory30. As a result, all of the GSH was oxidized

to GSSG, resulting in a measure of total glutathione through the concentration of

GSSG. (The GSH and GSSG did not comigrate in the separation – any GSH present

in the sample would be detected. However, none was detected in the samples.) The

detection limit for GSSG was 500 nM, with a precision of 3% and linear range of

0.75 – 50 μM.

The experiments dosing doxorubicin to anesthetized animals through the

microdialysis probe were repeated, but at a higher time resolution than the

220

Page 221: ~Chapter 1~ Introduction 1.1. Biomarkers

221

metabonomics experiments. In these studies, two probes were implanted in the liver,

and doxorubicin was dosed through Probe B after 3 hours of sampling basal dialysate.

Samples were collected every 15 minutes, rather than every hour as in the

metabonomics experiments. Three sets of experiments were performed – rats not

dosed with doxorubicin, rats dosed with doxorubicin after 3 hours of basal dialysate

collection and rats dosed with doxorubicin after 6 hours of basal dialysate collection.

For all rats dosed with doxorubicin, Probe B was perfused with 2 mg/mL doxorubicin

in Ringer’s solution for 60 minutes.

Figure 4.22 shows the results of the CE/UV analysis of the total glutathione

(as measured by GSSG) in liver dialysate samples. The basal dialysate showed a high

level of glutathione at the beginning of the experiment that fell and eventually leveled

around 5 hours. Ideally, doxorubicin would be dosed after basal glutathione levels

reached a steady state. However, the metabonomics experiments performed

previously dosed doxorubicin 3 hours after the start of sampling. Therefore,

doxorubicin was dosed at two time points in the experiment – at hour 3, to mimic the

metabonomics experiments and at hour 6, after the total glutathione levels had

steadied. In both dosing regimes, doxorubicin had no effect on total glutathione

levels. There was no significant difference between glutathione levels in Probe A and

B, as well as between experiment sets of dosed and control animals.

4.4.3.3.2. LC/EC: Reduced and Oxidized Glutathione

The CE/UV results suggested that total glutathione was not a good marker of

oxidative stress in liver dialysate. It was hypothesized that measuring reduced and

Page 222: ~Chapter 1~ Introduction 1.1. Biomarkers

222

Figure 4.22. Basal levels of total glutathione in liver dialysate over time. represents samples from Probe A (control probe) and represents samples from Probe B (dosed probe). A shows glutathione levels in basal liver dialysate (n=3). B shows glutathione levels in liver dialysate from rats dosed with doxorubicin at hour 3, as represented by a dashed line (n=3). C shows glutathione levels in liver dialysate from rats dosed with doxorubicin at hour 6, as represented by a dashed line (n=2).

4 6 8 10 12

0 2

0

20

40

60

80

100

120

140

Nor

mal

ized

[GS

SG

] (uM

)

Time (

0 2

0

20

40

60

80

100

120

140

Hour)

4 6 8 10 12

Nor

mal

ized

[GS

SG

] (uM

)

Time (

0 2

0

20

40

60

80

100

120

140

Hour)

4 6 8 10 12

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

A

B

C

4 6 8 10 120 2

0

20

40

60

80

100

120

140

Nor

mal

ized

[GS

SG

] (uM

)

Time (

0 2

0

20

40

60

80

100

120

140

Hour)

4 6 8 10 12

Nor

mal

ized

[GS

SG

] (uM

)

Time (

0 2

0

20

40

60

80

100

120

140

Hour)

4 6 8 10 12

A

B

C

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

Page 223: ~Chapter 1~ Introduction 1.1. Biomarkers

223

oxidized glutathione separately would be a better gauge of the oxidative stress of the

tissue, as the ratio of the reduced to oxidized form has been shown to indicate the

oxidative stress level of the tissue.

One common method of thiol analysis is LC/EC with dual gold-mercury

amalgam electrodes (Figure 4.23). In this method, the two electrodes are placed in

series configuration relative to the flow from the LC column. The upstream electrode

is used as a reducing, or “generator”, electrode. The potential applied to this

electrode is sufficient to reduce any disulfide bonds to thiols. As thiols (either in the

sample or from reduction at the first electrode) pass over the downstream or working

electrode, the slightly positive potential is sufficient to form a thiol-mercury complex,

creating a detectable signal. Thus, every compound is detected in the system as a

thiol, but they can be quantified separately because they are separated

chromatographically. One advantage of using gold-mercury electrodes over glassy

carbon or plain gold for thiol detection is that a much lower potential can be used,

resulting in lower background current, better selectivity and improved detection

limits16. This setup also allows for the easier identification of thiols in the sample –

simply turning off the upstream electrode will remove signal from the disulfides in

the chromatogram.

An LC/EC method was developed to quantify GSH and GSSG in liver

dialysate. The final method is described in Section 4.3.5. One important

consideration for this type of analysis is the presence of dissolved oxygen in the

mobile phase. The reducing electrode is set at such a strongly negative potential that

Page 224: ~Chapter 1~ Introduction 1.1. Biomarkers

224

Figure 4.23. Dual-electrode LC/EC analysis of thiols with gold-mercury electrodes.

LC Flow

W2W1

GeneratingElectrode-1.000 V

DetectionElectrode0.150 V

2 GSH Hg(S2H+

GSSG + 2H+ + 2 e-

G)2 + + 2e-

LC Flow

W2W1

GeneratingElectrode-1.000 V

DetectionElectrode0.150 V

2 GSH Hg(S2H+

GSSG + 2H+ + 2 e-

G)2 + + 2e-

Page 225: ~Chapter 1~ Introduction 1.1. Biomarkers

225

current is generated from dissolved oxygen in the mobile phase. This can create high

background currents, as well as erratic baselines in the chromatogram. Consequently,

the mobile phase was continuously sparged with helium and the mobile phase inlet

tubing was composed of PEEK, rather than PTFE. PEEK is only slightly permeable

to oxygen, and the use of PEEK tubing resulted in much lower and stable background

currents for both electrodes (upstream: -2 μA, downstream: -1.5 nA). An oxygen

scrubber was also placed in the helium gas flow. The precision of the method was 1-

2%. The limit of detection (LOD) of GSH was 100 nM, with a limit of quantitation

(LOQ) of 250 nM. The LOD of GSSG was 500 nM, with a LOQ of 900 nM. The

dynamic range for GSH was 250 nM – 50 μM, and the dynamic range for GSSG was

900 nM – 90 μM. Sample stability is an important concern in thiol analysis, as thiols

are easily oxidized, particularly in Ringer’s solution. Acidification of the sample

stabilizes the thiol/disulfide ratio31. As a result, all samples were diluted by 50% in

the mobile phase buffer (0.12 M monochloroacetic acid, pH = 3.0) without the ion-

pairing agent. The samples were shown to be stable for two days at -20°C and

through one freeze-thaw cycle.

4.4.3.3.3. Anesthetized Rats

The effects of anesthesia on basal levels of GSH and GSSG were

characterized, as shown in Figure 4.24. (In all the glutathione plots, the levels were

normalized to the % delivery of the microdialysis probe.) GSH levels were high at

the start of the experiment, but eventually dropped and leveled around 6 hours after

sampling began (about 7 hours post-implantation). The initial high levels of GSH

Page 226: ~Chapter 1~ Introduction 1.1. Biomarkers

226

Figure 4.24. Comparison of levels of GSH (A) and GSSG (B) in rats anesthetized with ketamine (n=3). represents samples taken from Probe A, and represents samples taken from Probe B.

8 10 12

0 2 4 6

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hou

0 2 4 6

0

2

4

6

8

10

12

14

16

18 r)

8 10 12

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hou

0 2 4 6

0

10

20

30

40

50

60

70

80

90

100

110

120

r)

A

B8 10 12

Nor

mal

ized

[GS

H] (

uM)

Time (Hou

0 2 4 6

0

2

4

6

8

10

12

14

16

18 r)

8 10 12

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

A

B

Page 227: ~Chapter 1~ Introduction 1.1. Biomarkers

227

may have been due to tissue trauma from probe implantation that eventually cleared

as the wound healed. Basal GSSG levels were fairly stable but level over the course

of sampling.

The effect of anesthesia on the basal levels of GSH and GSSG was examined

by comparing results from rats anesthetized with ketamine and rats anesthetized with

isoflurane inhalation. Figure 4.25 shows a comparison of GSH levels, and Figure

4.26 shows a comparison of GSSG levels. For both analytes, similar time trends were

noted in GSH and GSSG levels between rats anesthetized by ketamine or isoflurane.

A student’s t test was performed to determine if any differences were statistically

significant. None were found at 95% confidence.

4.4.3.3.4. Awake Rats

Comparison studies were then performed between glutathione levels in

anesthetized and awake rats. The results are shown in Figure 4.27. Both GSH and

GSSG are at low levels and stable at the start of sampling (24 hours post-probe

implantation). GSSG was below the LOQ of the method in awake animals, with

occasional quantifiable levels. GSH levels were low in awake animals, but still

within the dynamic range of the method. It should be noted that neither GSH nor

GSSG levels changed significantly over the course of three days of sampling,

showing that immunological changes from probe implantation do not alter glutathione

levels in the extracellular fluid of liver tissue.

Doxorubicin was dosed in awake animals at sampling hour 25 and the levels

of both GSH and GSSG were monitored. The results are shown in Figure 4.28. No

Page 228: ~Chapter 1~ Introduction 1.1. Biomarkers

228

Figure 4.25. Comparison of GSH levels from rats anesthetized with isoflurane and ketamine. represents samples from Probe A of rats anesthetized with ketamine; represents samples from Probe A of rats anesthetized with isoflurane; represents samples from Probe B of rats anesthetized with ketamine; represents samples from Probe B of rats anesthetized with isoflurane. A compares GSH levels in Probe A and B compares GSH levels in Probe B. C shows an average of GSH levels from each experiment group.

0 2 4 6 8 10 12-10

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)

0 2 4 6 8 10 12-10

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)0 2 4 6 8 10 12

-10

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)

A B

C

0 2 4 6 8 10 12-10

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)

0 2 4 6 8 10 12-10

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)0 2 4 6 8 10 12

-10

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)

A B

C

Page 229: ~Chapter 1~ Introduction 1.1. Biomarkers

229

Figure 4.26. Comparison of GSSG levels from rats anesthetized with isoflurane and ketamine. represents samples from Probe A of rats anesthetized with ketamine; represents samples from Probe A of rats anesthetized with isoflurane; represents samples from Probe B of rats anesthetized with ketamine; represents samples from Probe B of rats anesthetized with isoflurane. A compares GSSG levels in Probe A and B compares GSSG levels in Probe B. C shows an average of GSSG levels from each experiment group.

0 2 4 6 8 10 12

0

5

10

15

20

25

30

35

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

0 2 4 6 8 10 12

-202468

10121416182022242628303234

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)0 2 4 6 8 10 12

-202468

10121416182022242628303234

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

A

C

B

0 2 4 6 8 10 12

0

5

10

15

20

25

30

35

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

0 2 4 6 8 10 12

-202468

10121416182022242628303234

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)0 2 4 6 8 10 12

-202468

10121416182022242628303234

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

A

C

B

Page 230: ~Chapter 1~ Introduction 1.1. Biomarkers

230

Figure 4.27. Comparison of GSH (A) and GSSG (B) levels from rats anesthetized with isoflurane (n=3) and ketamine (n=3), as well as awake rats. The axis represents time past probe implantation represents samples from Probe A of rats anesthetized with ketamine; represents samples from Probe A of rats anesthetized with isoflurane; represents samples from Probe B of rats anesthetized with ketamine; represents samples from Probe B of rats anesthetized with isoflurane; represents samples from Probe A of awake rats; Δ represents samples from Probe B of awake rats.

70 80 90 100

0 10 20 30 40 50 60-10

0

10

20

30

40

50

60

70

80

90

100

110

120

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)

0 20 40 6-5

0

5

10

15

20

25

30

35

40

0 80 100

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

0 10 20 30 40 50 60-10

0

10

20

30

40

50

60

70

80

90

100

110

120

A

B70 80 90 100

Nor

mal

ized

[GS

H] (

uM)

Time (Hour)

0 20 40 6-5

0

5

10

15

20

25

30

35

40

0 80 100

A

B

Nor

mal

ized

[GS

SG

] (uM

)

Time (Hour)

Page 231: ~Chapter 1~ Introduction 1.1. Biomarkers

231

Figure 4.28. Comparison of GSH (A) and GSSG (B) levels from awake rats dosed and undosed with doxorubicin (at t=0). represents samples from Probe A of awake rats undosed with doxorubicin; Δ represents samples from Probe B of awake rats undosed with doxorubicin; ♦ represents samples from Probe A of awake rats dosed with doxorubicin; ◊ represents samples from Probe B of awake rats dosed with doxorubicin.

30 40 50

-30 -20 -10 0 10 20

-2

0

2

4

6

8

10

12

14

Nor

mal

ized

[GSH

] (uM

)

Time (Hour)

-30 -20 -10 0 10 20-3

-2

-1

0

1

2

3

4

5

6

7

8

9

10

11

30 40 50

Nor

mal

ized

[GSS

G] (

uM)

Time (Hour)

-30 -20 -10 0 10 20

-2

0

2

4

6

8

10

12

14

A

B30 40 50

Nor

mal

ized

[GSH

] (uM

)

Time (Hour)

-30 -20 -10 0 10 20-3

-2

-1

0

1

2

3

4

5

6

7

8

9

10

11

30 40 50

A

B

Nor

mal

ized

[GSS

G] (

uM)

Time (Hour)

Page 232: ~Chapter 1~ Introduction 1.1. Biomarkers

232

significant and consistent changes were noted as a result of the doxorubicin dose,

either a decrease in GSH levels or increase in GSSG levels that would signify

oxidative stress conditions. In these studies, it appears that GSH and GSSG are not

good markers of the hepatic oxidative stress induced by doxorubicin, as they do not

change in response to the dose. The dose to the tissue is large, sufficient to induce a

large amount of oxidative stress in the liver tissue surrounding the probe. Therefore,

glutathione levels could not be used to correlate to changes observed in metabolic

profiles. Metabonomics studies were still performed in awake animals, as described

below, but no statistical comparisons were made between glutathione levels and

metabolite levels.

4.4.3.4. Dual Probe Experiments in Awake Animals

Doxorubicin was dosed in awake rats, removing the metabolic effects of

anesthesia from the experiment so that the metabolic impact of the doxorubicin dose

could be more clearly discerned. As in the anesthetized animal experiments,

doxorubicin was dosed through probe B, with probe A used as a marker for natural

changes in liver metabolism. In the awake animal experiments, sampling began 24

hours after probe implantation. Basal dialysate was collected from both probes for 24

hours. Doxorubicin was then dosed through probe B for one hour (2 mg/mL

perfusate concentration), and dialysate was collected for an additional 48 hours post-

dose.

The patterns observed in awake rats as a result of the doxorubicin dose are

shown in Figure 4.29. As in Figure 4.20, both the undosed and dosed rats show a

Page 233: ~Chapter 1~ Introduction 1.1. Biomarkers

233

Figure 4.29. Doxorubicin liver metabolic effects in awake rats. represents samples from Probe A of undosed rats, represents samples from Probe B of undosed rats, ♦ represents samples from Probe A of dosed rats and ◊ represents samples from Probe B of dosed rats (dosed probes).

15

**

**

**

* **

-5 0 5 10

-8

-6

-4

-2

0

2

4

Prin

cipa

l Com

pone

nt 3

Principal Component 1

**

**

**

* **

-5 0 5 10

-8

-6

-4

-2

0

2

4

15

Prin

cipa

l Com

pone

nt 3

Principal Component 1

Page 234: ~Chapter 1~ Introduction 1.1. Biomarkers

234

starting point in the top left corner of the scores plot. The undosed rat samples

continue down the principal component 2 axis, as in Figure 4.20. However, in Figure

4.29, a separation was observed between probe A and B in the dosed rats. The probe

A samples tracked along the principal component 2 axis, along with the samples from

the undosed animals. In contrast, the probe B samples tracked along the principal

component 1 axis, separating from the control animals and samples taken at probe A

in the dosed animals at the time point that doxorubicin was dosed (and subsequent

time points post-dose). Figure 4.30 shows the individual time courses of rats dosed

with doxorubicin. Variance in response to the dose can be visualized from the plots.

These results are consistent with a localized response to the doxorubicin dose,

in that the liver tissue metabolism around probe B was altered but the healthy tissue

around probe A maintained metabolic trends consistent with normal healthy tissue.

This is in stark contrast to the anesthetized animal results in Figure 4.20, where more

global changes from doxorubicin were observed. This suggests that anesthesia alters

the metabolic changes resulting from doxorubicin administration to the liver, causing

it to have a more global effect. Loadings from principal component 1 show lysine,

alanine, leucine, valine, glutamine, creatine and citrulline as significantly changing in

response to the doxorubicin dose. Citrulline and glutamine changes were not

observed in anesthetized animal experiments. Figure 4.31 shows a representative plot

for how each metabolite changed individually over time, with lysine used as an

example. All metabolites showed a similar time trend, increasing after the

doxorubicin dose and then falling back to near-basal levels. Metabolites were fairly

Page 235: ~Chapter 1~ Introduction 1.1. Biomarkers

235

Figure 4.30. Doxorubicin liver metabolic effects in awake rats. Plots A, B and C show the metabolic time course from individual rats dosed with doxorubicin. ♦ represents samples from Probe A of dosed rats and ◊ represents samples from Probe B of dosed rats (dosed probes).

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

Prin

cipa

l Com

pone

nt 3

Principal Component 1

Prin

cipa

l Com

pone

nt 3

Principal Component 1

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

Prin

cipa

l Com

pone

nt 3

Principal Component 1

A

B

C

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

Prin

cipa

l Com

pone

nt 3

Principal Component 1

Prin

cipa

l Com

pone

nt 3

Principal Component 1-5 0 5 10 15

-8

-6

-4

-2

0

2

4

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

Prin

cipa

l Com

pone

nt 3

Principal Component 1

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

Prin

cipa

l Com

pone

nt 3

Principal Component 1

Prin

cipa

l Com

pone

nt 3

Principal Component 1

-5 0 5 10 15

-8

-6

-4

-2

0

2

4

A

B

C

Prin

cipa

l Com

pone

nt 3

Principal Component 1

Page 236: ~Chapter 1~ Introduction 1.1. Biomarkers

236

Figure 4.31. Time course of lysine in liver dialysate. A shows the metabolite time course in undosed rats. represents samples from Probe A of undosed rats and represents samples from Probe B of undosed rats. The dashed line indicates the dosage of doxorubicin through Probe B. ♦ represents samples from Probe A of dosed rats and ◊ represents samples from Probe B of dosed rats (dosed probes). B shows the metabolite time course in dosed rats.

0 40 50

0 10 20 30

1

2

3

4

5

6

Nor

mal

ized

Inte

gral

Time (hou

0 10 20 30

1

2

3

4

5

6r)

0 40 50

Nor

mal

ized

Inte

gral

Time (hour)

A

B

0 40 500 10 20 30

1

2

3

4

5

6

Nor

mal

ized

Inte

gral

Time (hou

0 10 20 30

1

2

3

4

5

6r)

0 40 50

A

B

Nor

mal

ized

Inte

gral

Time (hour)

Page 237: ~Chapter 1~ Introduction 1.1. Biomarkers

stable in the control/undosed rats, as well as in the undosed microdialysis probes of

the dosed rats. However, the administration of a doxorubicin dose induced an

increase in the levels of the endogenous energy metabolite lysine (in this example). It

should be noted that no significant differences were noted in the rate of clearance of

doxorubicin between awake and anesthetized animals.

4.4.3.5. Metabolic Effects of Doxorubicin

The damage induced by doxorubicin is often measured by established

biomarkers of oxidative stress, such as malondialdehyde to measure the extent of

lipid peroxidation32. From the studies of these single biomarkers, it is apparent that

large changes occur in cellular small molecule levels in response to doxorubicin.

Therefore, it was anticipated that changes in the small molecules detected in the liver

extracellular fluid (primarily energy compounds) would be responsive to the

doxorubicin dose. Doxorubicin has not been previously studied by metabonomics,

although the metabolic consequences of the drug have been broached primarily by

investigations on the activity of enzymes pertinent to cell respiration.

Doxorubicin has been demonstrated to cause a depletion of energy stores in

the cell in a dose-dependent manner28,33, in the forms of both ATP and

phosphocreatine. In fact, it has been demonstrated that both ATP and

phosphocreatine levels fall dramatically within one hour of doxorubicin dosage, and

this depletion of energy stores can be sustained over a period of several days33. This

depletion occurs by several mechanisms, including the inhibition of creatine kinase

and transporters of ATP from the mitochondria to the cytosol, preventing the transfer

237

Page 238: ~Chapter 1~ Introduction 1.1. Biomarkers

of the high energy phosphate from ATP to creatine for longer term storage in the

cellular energy reserves. The accumulation of creatine in response to the doxorubicin

dose shows evidence that doxorubicin interferes in the energy stores of liver tissue.

The creatine is not phosphorylated for energy storage, causing it to accumulate in the

cell. This is reflected by the increase in creatine in the extracellular fluid levels,

which was observed in both the anesthetized and awake animals.

Another noteworthy metabolic effect of doxorubicin is the change in levels of

amino acids in the liver extracellular fluid, including lysine, alanine, glutamine,

valine, leucine and isoleucine. This is likely due to doxorubicin-induced alterations

in amino acid metabolism, specifically changes in aminotransferase levels and

activity in the liver, including alanine aminotransferase and aspartate

aminotransferase34. The liver is the major site of amino acid catabolism in the body,

because it is the first organ to receive amino acids after feeding via the portal vein. It

is also the only organ that can eliminate nitrogen from the amino acids via the urea

cycle. In general, amino acids are metabolized in the liver by transaminases, which

remove the amino group to make a keto acid that can go on to participate in cellular

respiration in either the liver itself or other tissues, such as muscle. (For example, the

transamination of alanine produces pyruvate.) This is an important link between

carbohydrate and amino acid metabolic pathways. The process is generally regulated

by substrate supply and glucagon and cortisol levels. However, because doxorubicin

alters the transamination pathways in the liver, significant changes in the levels of the

amino acids listed above are measured. All of the amino acids listed above showed

238

Page 239: ~Chapter 1~ Introduction 1.1. Biomarkers

an increase in level within one hour of the doxorubicin dose, and levels gradually

returned to normal as the doxorubicin was eliminated from the liver.

Doxorubicin may further alter amino acid metabolism by interfering in urea

cycle metabolic pathways, as judged by alterations in the levels of citrulline,

glutamine and creatine. The urea cycle is used to eliminate nitrogen by the

production of urea through a reaction cycle involving ornithine, citrulline and

arginine. Glutamine feeds into the urea cycle by its ultimate conversion to the urea

cycle intermediate argininosuccinate via conversion to glutamate. Creatine

production is also metabolically linked to the urea cycle through the conversion of

arginine to creatine.

Increases in β-hydroxybutyrate levels in the liver extracellular fluid were also

observed in response to the doxorubicin dose. Previous studies have shown

alterations in lipid metabolism in the liver in response to doxorubicin32. Specifically

in this study, increased lipid peroxidation products such as acetone were observed in

the urine, indicating increased β-oxidation of fatty acids or an accumulation of acetyl-

CoA by decreased metabolism of carbohydrates. Both acetone and β-

hydroxybutyrate are ketone bodies. Because doxorubicin has been shown to increase

acetone levels in previous studies, it seems logical that doxorubicin might also result

in an increase in β-hydroxybutyrate levels by a similar mechanism to acetone. (The

detection of acetone in these samples is precluded due to the speedvacuuming of the

samples prior to NMR analysis, thus removing volatile components like acetone.)

239

Page 240: ~Chapter 1~ Introduction 1.1. Biomarkers

Figures 4.20 and 4.29 demonstrate that doxorubicin induces different

metabolic effects in the liver tissue of awake and anesthetized rats. Anesthetized rats

experienced more of a global metabolic effect in the liver lobe, while awake animals

experienced more of a localized metabolic effect. The reason for this is unclear at

this time, although several possibilities can be considered. First, it is established in

literature that doxorubicin-induced oxidative stress is dependent on tissue oxygen

levels35. Depleted oxygen levels will decrease the observed toxicity of the drug.

Related to this, regions of the liver may experience differences in oxygen levels as a

result of their proximity to the portal vein36,37. Although no differences were

observed between probes under basal metabolic conditions, it is possible that changes

in blood flow and heart rate due to anesthesia could alter the liver’s metabolic

response to the doxorubicin dose. Second, changes in liver enzyme levels and

activity induced by the anesthesia could alter the liver’s response to the doxorubicin

dose. It should be noted that the metabolites that changed in response to doxorubicin

were similar in anesthetized and awake animals, indicating that anesthesia likely did

not change the mechanism of doxorubicin hepatotoxicity dramatically. However, two

new metabolites (citrulline and glutamine) were identified as significantly changing

in response to the doxorubicin dose when awake animals were used. This would

suggest that anesthesia can influence the identification of key metabolites in tissue-

targeted metabonomics studies, perhaps through alterations in metabolic pathways

caused by the anesthesia itself.

240

Page 241: ~Chapter 1~ Introduction 1.1. Biomarkers

4.5. Conclusions

The purpose of the studies presented in this chapter was to illustrate the

advantages of using tissue-targeted metabonomics to study a pathophysiological

condition targeting the metabolism of a specific tissue, such as oxidative stress.

Studies (of both ischemia/reperfusion and doxorubicin) were initially performed in

the heart, but were ultimately moved to the liver, as the heart is less amenable to

metabonomics studies. The biomarker of oxidative stress, glutathione, was

characterized in both awake and anesthetized animals. Surprisingly, this biomarker

did not respond to doxorubicin-induced oxidative stress (although evidence for

oxidative stress induction by doxorubicin was shown in histological analysis of the

liver). In future studies, however, the concurrent analysis of biomarkers with

metabolic profiles could help to both interpret the metabolic changes induced by the

metabolic stressor and to validate new biomarkers identified by the metabonomics

analysis.

The primary metabolic stressor of this experiment, doxorubicin, was used to

monitor perturbations in patterns of small molecules in the extracellular fluid of the

liver in response to oxidative stress conditions. This chapter presents the first

metabonomics study of doxorubicin-induced oxidative stress. Data indicated that

multiple metabolic pathways were affected by the doxorubicin dose, including

depletion of energy stores and alterations in amino acid and fatty acid metabolism.

Thus, the utility of combining microdialysis sampling with NMR analysis is

illustrated by these results: many metabolic pathways can be monitored

241

Page 242: ~Chapter 1~ Introduction 1.1. Biomarkers

simultaneously to characterize conditions best observed through the metabolism of

specific tissues rather than biofluids.

4.6. References

1. Prabakaran, S., Swatton, J. E., Ryan, M. M., Huffaker, S. J., Huang, J. T.-J., Griffin, J. L., Wayland, M., Freeman, T., Dudbridge, F., Lilley, K. S., Karp, N. A., Hester, S., Tkachev, D., Mimmack, M. L., Yolken, R. H., Webster, M. J., Torrey, E. F. and Bahn, S. (2004). Mitochondrial dysfunction in schizophrenia: evidence for compromised brain metabolism and oxidative stress. Molec. Psychiat. 9, 684-697.

2. Viant, M. R., Lyeth, B. G., Miller, M. G. and Berman, R. F. (2005). An

NMR metabolomic investigation of early metabolic disturbances following traumatic brain injury in a mammalian model. NMR Biomed. 18, 507-516.

3. Soga, T., Baran, R., Suematsu, M., Ueno, Y., Ikeda, S., Sakurakawa, T.,

Kakazu, Y., Ishikawa, T., Robert, M., Nishioka, T. and Tomita, M. (2006). Differential metabolomics reveals ophthalmic acid as an oxidative stress biomarker indicating hepatic glutathione consumption. J. Biol. Chem. 281, 16768-16776.

4. Klenø, T. G., Kiehr, B., Baunsgaard, D. and Sidelmann, U. G. (2004).

Combination of 'omics' data to investigate the mechanism(s) of hydrazine-induced hepatotoxicity in rats and to identify potential biomarkers. Biomarkers 9, 116-138.

5. Griffin, J. L., Scott, J. and Nicholson, J. K. (2007). The influence of

pharmacogenetics on fatty liver disease in the Wistar and Kyoto rats: a combined transcriptomic and metabonomic study. J. Proteom. Res. 6, 54-61.

6. Fardet, A., Llorach, R., Martin, J.-F., Besson, C., Lyan, B., Pujos-Guillot, E.

and Scalbert, A. (2008). A liquid chromatography-quadrupole time-of-flight (LC-QTOF)-based metabolomic approach reveals new metabolic effects of catechin in rats fed high-fat diets. J. Proteom. Res. 7, 2388-2398.

7. Serkova, N., Fuller, T. F., Klawitter, J., Freise, C. E. and Niemann, C. U.

(2005). 1H-NMR-based metabolic signatures of mild and severe ischemia/reperfusion injury in rat kidney transplants. Kidney Intern. 67, 1142-1151.

242

Page 243: ~Chapter 1~ Introduction 1.1. Biomarkers

8. Mally, A., Amberg, A., Hard, G. C. and Dekant, W. (2007). Are 4-hydroxy-2(E)-nonenal derived mercapturic acids and 1H NMR metabonomics potential biomarkers of chemically induced oxidative stress in the kidney? Toxicol. 230, 244-255.

9. Mayr, M., Siow, R., Y.-L., C., Mayr, U., Griffiths, J. R. and Xu, Q. (2004).

Proteomic and metabolomic analysis of vascular smooth muscle cells: role of PKCδ. Circ. Res. 94, e87-e96.

10. Mayr, M., Chung, Y.-L., Mayr, U., Yin, X., Ly, L., Troy, H., Fredericks, S.,

Hu, Y., Griffiths, J. R. and Xu, Q. (2005). Proteomic and metabolomic analyses of atherosclerotic vessels from apolipoprotein E-deficient mice reveal alterations in inflammation, oxidative stress, and energy metabolism. Arterioscler. Thromb. Vasc. Biol. 25, 2135-2142.

11. Chu, C. Y., Xiao, X., Zhou, X. G., Lau, T. K., Rogers, M. S., Fok, T. F., Law,

L. K., Pang, C. P. and Wang, C. C. (2006). Metabolomic and bioinformatic analyses in asphyxiated neonates. Clin. Biol. 39, 203-209.

12. Davies, M. I. and Lunte, C. E. (1996). Simultaneous microdialysis sampling

from multiple sites in the liver for the study of phenol metabolism. Life. Sci. 59, 1001-1013.

13. Hoque, M. E., Arnett, S. D. and Lunte, C. E. (2005). On-column

preconcentration of glutathione and glutathione disulfide using pH-mediated base stacking for the analysis of microdialysis samples by capillary electrophoresis. J. Chromatogr. B 827, 51-57.

14. Allison, L. A., Keddington, J. and Shoup, R. E. (1983). Liquid

chromatographic behavior of biological thiols and the corresponding disulfides. J. Liq. Chromatogr. 6, 1785-1798.

15. Allison, L. A. and Shoup, R. E. (1983). Dual electrode liquid

chromatography detector for thiols and disulfides. Anal. Chem. 55, 8-12. 16. Lunte, S. M. and Kissinger, P. T. (1985). Detection of thiols and disulfides in

liver samples using liquid chromatography/electrochemistry. J. Liq. Chromatogr. 8, 691-706.

17. Stein, A. F., Dills, R. L. and Klaassen, C. D. (1986). High-performance

liquid chromatographic analysis of glutathione and its thiol and disulfide degradation products. J. Chromatogr. 381, 259-270.

243

Page 244: ~Chapter 1~ Introduction 1.1. Biomarkers

18. Bosanquet, A. G. (1986). Stability of solutions of antineoplastic agents during preparation and storage for in vitro assays. II. Assay methods, adriamycin and the other antitumour antibiotics. Cancer Chemother. Pharmacol. 17, 1-10.

19. Wood, M. J., Irwin, W. J. and Scott, D. K. (1990). Photodegradation of

doxorubicin, daunorubicin and epirubicin measured by high-performance liquid chromatography. J. Clin. Pharm. Ther. 15, 291-300.

20. Zhao, Y., Liang, X. and Lunte, C. E. (1995). Comparison of recovery and

delivery in vitro for calibration of microdialysis probes. Anal. Chim. Acta 316, 403-410.

21. Altschuld, R. A., Gibb, L. and Weissler, A. M. (1970). Metabolism of the

ischemic myocardium. Heart Bull. 19, 109-112. 22. Chaiswing, L., Cole, M. P., St. Clair, D. K., Ittarat, W., Szweda, L. I. and

Oberley, T. D. (2004). Oxidative damage precedes nitrative damage in adriamycin-induced cardiac mitochondrial injury. Toxicol. Pathol. 32, 536-547.

23. Gewirtz, D. A. (1999). A critical evaluation of the mechanisms of action

proposed for the antitumour effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem. Pharmacol. 57, 727-741.

24. Hong, Y. M., Kim, H. S. and Yoon, H.-R. (2002). Serum lipid and fatty acid

profiles in adriamycin-treated rats after administration of L-carnitine. Ped. Res. 51, 249-255.

25. Minotti, G., Menna, P., Salvatorelli, E., Cairo, G. and Gianni, L. (2004).

Anthracyclines: molecular advances and pharmacologic developments in antitumour activity and cardiotoxicity. Pharmacol. Rev. 56, 185-229.

26. Lévi, F. (1994). Chronotherapy of cancer: biological basis and clinical

application. Path. Biol. 42, 338-341. 27. Shargel, L. and Yu, A. (1999). Applied Biopharmaceutics and

Pharmacokinetics. McGraw-Hill, St. Louis. 28. Akaza, K., Nonami, T., Kurokawa, T., Kobayashi, H., Harada, A., Nakao, A.,

Sugiyama, S., Ozawa, T. and Takagi, H. (1996). Doxorubicin-induced disturbance of energy metabolism after hepatectomy. J. Surg. Res. 61, 454-458.

244

Page 245: ~Chapter 1~ Introduction 1.1. Biomarkers

29. Asakura, T., Sawai, T., Hashidume, Y., Ohkawa, Y., Yokoyama, S. and Ohkawa, K. (1999). Caspase-3 activation during apoptosis caused by glutathione-doxorubicin conjugate. Brit. J. Canc. 80, 711-715.

30. Hoque, M. E. (2007). Development of Analytical Methods Coupled to

Microdialysis Sampling for Studying Biomarkers of Oxidative Stress. Doctoral dissertation, Department of Chemistry, University of Kansas.

31. Reyes, M. G. (1997). Capillary Electrophoresis with Electrochemical

Detection for the Determination of Glutathione in Biological Samples. Master's Thesis, Department of Chemistry, University of Kansas.

32. Bagchi, D., Bagchi, M., Hassoun, E. A., Kelly, J. and Stohs, S. J. (1995).

Adriamycin-induced hepatic and myocardial lipid peroxidation and DNA damage, and enhanced excretion of urinary lipid metabolites in rats. Toxicology 95, 1-9.

33. Tokarska-Schlattner, M., Wallimann, T. and Schlattner, U. (2006).

Alterations in myocardial energy metabolism induced by the anti-cancer drug doxorubicin. C. R. Biol. 329, 657-668.

34. Deepa, P. R. and Varalakshmi, P. (2003). Protective effect of low molecular

weight heparin on oxidative injury and cellular abnormalities in adriamycin-induced cardiac and hepatic toxicity. Chem. Biol. Interact. 146, 201-210.

35. Ganey, P. E., Kauffman, F. C. and Thurman, R. G. (1988). Oxygen-

dependent hepatotoxicity due to doxorubicin: role of reducing equivalent supply in perfused rat liver. Mol. Pharmacol. 134, 695-701.

36. Liu, Y. and Thurman, R. G. (1992). Potentiation of adriamycin toxicity by

ethanol in perfused rat liver. J. Pharmacol. Exp. Ther. 263, 651-656. 37. Matsumara, T. and Thurman, R. G. (1983). Measuring rates of O2 uptake in

periportal and pericentral regions of liver lobule: stop-flow experiments with perfused liver. Am. J. Physiol. 244, G656-G659.

245

Page 246: ~Chapter 1~ Introduction 1.1. Biomarkers

~Chapter 5~

Conclusions and Future Work

5.1. Dissertation Overview

Tissue-targeted metabonomics was successfully developed as an analytical

tool to probe localized metabolic changes over time in an organism. This is a novel

approach to metabolic profiling experiments, offering a superior alternative to tissue

biopsy sampling studies. Multiple samples can be taken from the same tissue site

over time, making this approach uniquely suited to studying time trends. Most

metabonomics studies simply focus on group classification, but following time trends

opens the possibility of evaluating metabolic perturbations in a unique light.

Additionally, this analytical approach allows for simultaneous time trend studies of

multiple tissues, as microdialysis probes can be implanted in several locations in the

body during a single experiment. Dialysate samples also are much cleaner than

typical plasma or tissue samples. This improves sample stability by removing

enzymatic degradation, improves throughput by reducing the sample preparation

requirements and simplifies NMR and LC analysis.

246

Page 247: ~Chapter 1~ Introduction 1.1. Biomarkers

The primary limitation of tissue-targeted metabonomics analytical

methodology is throughput. NMR analysis time limits the number of samples that

can be analyzed per rat, with nearly 2 hours per sample required on the magnet used

in these experiments, a 600 MHz spectrometer. Dialysate samples are dilute,

requiring more signal averaging than biofluid samples like urine. Also, dialysate

samples are small in volume. This also makes NMR analysis challenging. Because

samples are already dilute, it is not feasible to dilute the samples to the appropriate

volume. In these studies, tube inserts were used rather than a specialized capillary

probe. Although this approach gives more flexibility in the magnet that can be used

(no special probe is required), the fill factor is low and more signal averaging is

required.

In addition (and related) to throughput, tissue-targeted metabonomics also

faces limitations because of the n value required to make confident conclusions about

the biological processes underlying the changes observed in the metabolic profiles.

Animal-to-animal variability is a challenge in any in vivo experiment, and although

some of this variability can be removed through data normalization, it is impossible to

completely remove it. One of the best ways to separate animal-to-animal variability

from an actual response to a metabolic perturbation is to increase the number of

animals studied. An adequate number of samples must be analyzed to sufficiently

characterize the response to the metabolic perturbation. However, the number of

replicates must be balanced with the analysis time required.

247

Page 248: ~Chapter 1~ Introduction 1.1. Biomarkers

While tissue-targeted metabonomics offers some interesting possibilities for

the study of metabolic changes in vivo, this technology is still developing and has

obstacles to be overcome. Throughput of the method must be increased, allowing for

more samples to be analyzed and more animals to be studied. This in turn would

improve the quality of the data set analyzed and allow more significant conclusions to

be made about biological changes observed.

Metabonomics studies have presented an interesting challenge for analytical

chemists. There is a switch in focus from the traditional pursuit of selectivity to the

new focus on detector nonselectivity. With this comes a new set of methodological

problems to tackle, mostly centering on the data set complexity. Tissue-targeted

metabonomics fills a void in the metabonomics community by offering the possibility

of continuous sampling of tissue-specific metabolism while still maintaining the

profiling/nonselective detection scheme. This dissertation presents in-depth studies

of the analytical methods required to perform these investigations and shows the

potential for monitoring a tissue-specific metabolic perturbation that develops and

changes over a period of time.

5.2. Summary by Section

5.2.1. Tissue-Targeted Metabonomics: Analytical Considerations

The analytical methodology for tissue-targeted metabonomics studies was

optimized. Microdialysis sampling and subsequent sample preparation was

examined. It was determined that speedvacuuming the samples to remove water and

248

Page 249: ~Chapter 1~ Introduction 1.1. Biomarkers

reconstituting in a deuterated solution improved water suppression in the NMR

experiment, allowed for sample concentration and dramatically improved sample

stability. As with most choices made in analytical methods, speedvacuuming is not

without a sacrifice. Volatile solutes are lost, and sample composition changes

slightly. However, these changes appear to be consistent among samples. Much

larger changes in sample composition result from directly freezing the dialysate

samples. Thus, the improvements achieved with speedvacuuming were deemed to

outweigh the cost of slight changes in sample composition.

Much of the important details in the analytical methodology for tissue-

targeted metabonomics lies in the data treatment and analysis. The number of

variables is quite large and complex statistical techniques are utilized. As with all

statistics, the quality of the data set analyzed will determine the quality of the

conclusions that can be reached after applying statistics. The goal of the data

treatment is to minimize the noise in the data set from variation unrelated to the

metabolic perturbation. It was determined for liver microdialysis experiments that the

NMR analysis and probe extraction efficiency are primary contributors to the noise in

the data. However, significant noise sources may vary depending on the experiment,

and it is important to establish these prior to experimentation.

5.2.2. Tissue-Targeted Metabonomics: Biological Considerations

With such complex data sets, it is important to determine the changes

expected in the metabolic profiles of tissue extracellular fluid under basal conditions.

Many factors contribute to metabolic changes under normal metabolism, some from

249

Page 250: ~Chapter 1~ Introduction 1.1. Biomarkers

the animal itself and some as artifacts of the experimental conditions. These studies

showed that anesthesia in particular causes significant alterations in basal

metabolism. While this is not unexpected, it is important to characterize because its

use is unavoidable in microdialysis experiments. While some microdialysis

experiments maintain the rat under anesthesia for the duration of sampling, the results

suggest that tissue-targeted metabonomics experiments should be confined to awake

animals, at least in the study of liver metabolism.

Another important characteristic of in vivo experiments is natural cycles in

metabolism due to cycles such as circadian rhythms, feeding, activity and estrus

cycles (this can be avoided by using males). In these studies, cycles were observed

in animal activity. However, these were not reflected in the metabolic profiles of

liver dialysate. It was concluded that at the time scale being observed and the tissue

extracellular fluid being sampled for these experiments, endogenous rhythms in

metabolism did not consistently or significantly contribute to variance in the

metabolic profiles. This dissertation presents studies of endogenous rhythms in

metabolic profiles of tissue extracellular fluid.

5.2.3. Tissue-Targeted Metabonomics Application: Oxidative Stress

There are many potential applications for tissue-targeted metabonomics

methodology. This dissertation presents the study of hepatic oxidative stress, as

induced locally by the perfusion of doxorubicin through the microdialysis probe. The

microdialysis setup in these experiments provided a unique opportunity to

differentiate basal changes in metabolic conditions from those induced by

250

Page 251: ~Chapter 1~ Introduction 1.1. Biomarkers

doxorubicin by the implantation of two probes in the liver lobe. This approach could

be utilized in other microdialysis experiments, particularly those performed in

homogenous tissues.

This study was the first time doxorubicin was investigated by metabonomics.

Studies have been performed previously characterizing the effects of doxorubicin on

liver tissue homogenate. However, these studies have centered on enzyme levels and

activity with very little or no attention paid to the levels of endogenous metabolites.

This dissertation shows that doxorubicin has been directly associated with changes in

the levels of creatine, lysine, alanine, glutamine, valine, leucine, isoleucine, citrulline

and β-hydroxybutyrate in the rat liver. The simultaneous changes in these

metabolites are also presented, providing another dimension to the study of

doxorubicin effects in the liver over time. The results obtained in this study do not

contradict the existing literature on the changes in enzyme levels and activity from a

doxorubicin systemic dose (no previous studies have been performed on the direct

perfusion of doxorubicin through the liver). It should be emphasized that these

results are preliminary and are used to illustrate the possibilities that tissue-targeted

metabonomics methodology offers. The data presented represents observations from

the principal components analysis performed on the dialysate metabolic profiles.

Experiments with additional animals would be needed to make any biological claims

on the effects of doxorubicin in the liver with confidence.

It was hypothesized that monitoring an established marker of oxidative stress

would aid in the identification and interpretation of relevant changes in the metabolic

251

Page 252: ~Chapter 1~ Introduction 1.1. Biomarkers

profile of liver extracellular fluid. Thus, glutathione (GSH) and glutathione disulfide

(GSSG), as established biomarkers of oxidative stress, were chosen to perform this

study. Unfortunately, GSH and GSSG was not an ideal oxidative stress marker in this

particular experimental setup, as no dose-dependent response was noted. Although

this approach was not successful here, it could still be applied in other studies.

5.3. Future Directions

5.3.1. Tissue-Targeted Metabonomics: Analytical Considerations

As stated previously, the primary limitation to tissue-targeted metabonomics

methodology is throughput. The poor sensitivity of NMR combined with dilute,

small volume dialysate samples requires long analysis times in the NMR experiment.

Future work on analytical methodology for tissue-targeted metabonomics should

focus on increasing throughput. One way to do this is by improving detection limits

and/or sensitivity. The NMR analysis parameters could be explored that aim to

decrease the number of spectra required for coaddition, but this may prove difficult.

Studies could also be performed using capillary probes, which could allow for greater

concentration of the sample and thus lower analysis time. Another type of probe that

could improve detection limits is a cryoprobe. A cryoprobe, with electrical

components such as the rf coil and preamplifier cooled with liquid helium, yields

improved sensitivity and detection limits through the reduction of thermal noise.

Larger modifications to the method to improve detection limits could include moving

to a higher field magnet. Because signal to noise (S/N) scales with Bo3/2, increasing

252

Page 253: ~Chapter 1~ Introduction 1.1. Biomarkers

the field strength of the magnet will increase S/N. For example, if a 900 MHz magnet

was utilized with the same number of coadded spectra, the S/N would increase by

almost a factor of two. This could be used to either increase the S/N obtained in the

same amount of time as the acquisition parameters utilized in this research or be used

to increase throughput by achieving the same S/N in a shorter period of time.

Detection limits could also be improved by using a different analytical

method. Metabonomics studies using LC/MS1,2 and CE/MS3 have grown in

popularity in recent years, as discussed in Chapter 1. The exploration of these

techniques as detection schemes for dialysate samples could prove advantageous for

tissue-targeted metabonomics studies. Both mass spectrometry techniques have

improved detection limits when compared to NMR detection, albeit with a new set of

analytical challenges. The metabolites detected by these techniques may differ from

NMR due to some molecules not ionizing efficiently. Also, mass spectrometry is

generally more amenable to automation than NMR detection. While run times for

these techniques may still be somewhat high (an hour or more), they will likely be

lower than the NMR analysis time required for dialysate samples. Additionally,

throughput would be improved by opening the possibility for automation, an option

not usually found on NMR spectrometers for the analysis of small volume dialysate

samples. However, it should be noted that the high salt content in dialysate samples

may present an obstacle for MS studies by creating ionization problems.

Sensitivity is not the only limitation on the NMR analysis of dialysate

samples. Dynamic range is also an important constraint on the method. The overlap

253

Page 254: ~Chapter 1~ Introduction 1.1. Biomarkers

of metabolite resonances places limitations on the number and concentration of

metabolites that can be detected. Due to overlap, even if sensitivity is improved by

any of the mechanisms discussed above, metabolites at lower concentrations may still

not be detected. Metabolites at higher concentrations, typically energy metabolites,

will prevent the detection of the lower intensity resonances they overlap at any

sensitivity level. This can be improved slightly by moving to higher field magnets to

increase signal dispersion. Two dimensional NMR experiments can also improve

signal dispersion but are not practical for the routine analysis of dialysate samples.

The limitations on sensitivity and dynamic range faced by tissue-targeted

metabonomics using 1H-NMR analysis may be challenging to improve upon. With

this in mind, the use of class-specific analytical methods should be explored and

compared to tissue-targeted metabonomics methods. Class-specific methods, such as

the profiling of endogenous thiols by gold-mercury electrodes to characterize

oxidative stress, retain some of the nonselectivity of metabonomics detection

schemes, thus still allowing for biomarker discovery. However, these analyses are

more focused than conventional metabonomics approaches, so sensitivity and

dynamic range will likely be improved. These methodologies may represent the best

compromise between specific, highly sensitive assays of single biomarkers and the

very nonselective but poorly sensitive metabonomics analytical approaches.

5.3.2. Tissue-Targeted Metabonomics: Biological Considerations

The studies presented in this research focused on metabolic processes

observed in extracellular fluid in liver tissue. Each tissue has its own distinct

254

Page 255: ~Chapter 1~ Introduction 1.1. Biomarkers

metabolic processes, which could be affected differently by endogenous rhythms.

Therefore, it is important that each new tissue studied be fully characterized for basal

metabolic rhythms before studying the effects of a metabolic stressor.

The effects of different anesthetic agents should also be examined for their

effects on basal metabolic profiles. The agents studied in this dissertation were those

readily available, typically used and approved for use in the laboratory. However,

other anesthetic agents are available that could affect the metabolic profiles in

different ways. The optimum anesthetic agent used for tissue-targeted metabonomics

studies may also be tissue-dependent.

5.3.3. Tissue-Targeted Metabonomics Application: Oxidative Stress

The results of these studies suggest that tissue-targeted metabonomics with

NMR detection is most suited to the study of metabolic conditions that impact the

levels of energy metabolites in the extracellular fluid of the tissue of interest. These

compounds tend to be higher in concentration and are thus detected by the NMR

analysis. There are many potential applications of tissue-targeted metabonomics

methodology.

One potential application is the study of diabetes, metabolic syndrome and

obesity. All of these conditions, which can be interconnected, alter energy

metabolism in the body4,5. These diseases are increasing in prevalence in the United

States and many questions remain regarding the development and treatment of these

conditions. Multiple tissues are involved, including the muscle, brain, liver, pancreas

and kidney. Each tissue is uniquely impacted by changes in levels of or response to

255

Page 256: ~Chapter 1~ Introduction 1.1. Biomarkers

metabolic regulatory hormones such as insulin. Additionally, energy metabolites are

involved in intertissue communication regarding the metabolic status of the

organism4. In this way, tissues impact other tissues’ response to metabolic disorders

via metabolite levels in the extracellular matrix. Time trends in energy metabolism

can also be used as predictors for the development of these metabolic disorders.

Alterations in circadian rhythms of adipocyte metabolism have been linked to the

development of metabolic syndrome5. These factors combined make these metabolic

conditions an ideal application for tissue-targeted metabonomics.

Another potential application of tissue-targeted metabonomics is the study of

sepsis6. In many cases, it is difficult to detect the presence of this infection until it is

fairly well established. Previous studies in our research have shown that infection

causes alterations in the metabolic profiles of liver tissue extracellular fluid (Section

2.4.5). Additionally, other studies have also demonstrated the alteration in energy

metabolism observed during sepsis, but these have only focused on one or two

analytes7. Tissue-targeted metabonomics has the unique potential to identify new

markers of sepsis so that it can be detected and therefore treated more quickly.

5.4. References

1. Plumb, R. S., Stumpf, C. L., Gorenstein, M. V., Castro-Perez, J. M., Dear, G. J., Anthony, M., Sweatman, B. C., Connor, S. C. and Haselden, J. N. (2002). Metabonomics: the use of electrospray mass spectrometry coupled to reversed-phase liquid chromatography shows potential for the screening of rat urine in drug development. Rapid Commun. Mass Spectrom. 16, 1991-1996.

2. Plumb, R. S., Stumpf, C. L., Granger, J. H., Castro-Perez, J., Haselden, J. N.

and Dear, G. J. (2003). Use of liquid chromatography/time-of-flight mass

256

Page 257: ~Chapter 1~ Introduction 1.1. Biomarkers

257

spectrometry and multivariate statistical analysis shows promise for the detection of drug metabolites in biological fluids. Rapid Commun. Mass Spectrom. 17, 2632-2638.

3. Soga, T., Ohashi, Y., Ueno, Y., Naraoka, H., Tomita, M. and Nishioka, T.

(2003). Quantitative metabolome analysis using capillary electrophoresis mass spectrometry. J. Proteom. Res. 2, 488-494.

4. Yamuda, T., Oka, Y. and Katagiri, H. (2008). Inter-organ metabolic

communication involved in energy homeostasis: potential therapeutic targets for obesity and metabolic syndrome. Pharmacol. Ther. 117, 188-198.

5. Zvonic, S., Floyd, Z. E., Mynatt, R. L. and Gimble, J. M. (2007). Circadian

rhythms and the regulation of metabolic tissue function and energy homeostasis. Obesity 15, 539-543.

6. Deitch, E. A. (2005). Rodent models of intra-abdominal infection. Shock 24,

19-23. 7. Levy, B., Sadoune, L.-O., Gelot, A.-M., Bollaert, P.-E., Nabet, P. and Larcan,

A. (2000). Evolution of lactate/pyruvate and arterial ketone body ratios in the early course of catecholamine-treated septic shock. Crit. Care Med. 28, 114-119.