characterizing perchlorate exposure - citeseerx

192
CHARACTERIZING PERCHLORATE EXPOSURE AND EFFECTS IN MAMMALS by QIUQIONG CHENG, B.S., M.S. A DISSERTATION IN ENVIRONMENTAL TOXICOLOGY Submitted to the Graduate Faculty of Texas Tech University in Partial Fulfillment of the Requirements for the Degree of DOCTOR OF PHILOSOPHY Approved Todd T. Anderson Chairperson of the Committee Michael J. Hooper W. Andrew Jackson Scott T. McMurry Ernest E. Smith Accepted John Borrelli Dean of the Graduate School May, 2006

Upload: khangminh22

Post on 19-Feb-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

CHARACTERIZING PERCHLORATE EXPOSURE

AND EFFECTS IN MAMMALS

by

QIUQIONG CHENG, B.S., M.S.

A DISSERTATION

IN

ENVIRONMENTAL TOXICOLOGY

Submitted to the Graduate Faculty of Texas Tech University in

Partial Fulfillment of the Requirements for

the Degree of

DOCTOR OF PHILOSOPHY

Approved

Todd T. Anderson Chairperson of the Committee

Michael J. Hooper

W. Andrew Jackson

Scott T. McMurry

Ernest E. Smith

Accepted

John Borrelli

Dean of the Graduate School

May, 2006

ACKNOWLEDGMENTS

I would like to express my sincerest gratitude here to many people who

provided help, guidance, and encouragement not only in my academic study, but

also in my everyday life over the last three and half years. Their help, patience,

guidance, and encouragement made it possible for me to complete this

dissertation as well as my education in Environmental Toxicology at The Institute

of Environmental and Human Health, Texas Tech University. I am deeply

grateful to each of them for the great help.

Firstly, I would like to thank my committee members: Drs. Todd Anderson,

Ernest Smith, Andrew Jackson, Michael Hooper, and Scott McMurry, for their

expert advice and mentorship. I really appreciate Dr. Todd Anderson, my

advisor, for providing space, time, and financial support for me to conduct

experiments relevant to my dissertation, letting me have the opportunity to work

independently, and giving me advice and encouragement when I had trouble with

my experiments. Todd is not only a mentor for me during studying and working

in the lab and classes, he is also a good friend in my everyday life. He made my

life in Lubbock colorful and exciting by inviting us to watch many sport games

such as college football and Cotton King hockey. I would like to sincerely thank

Todd for all his time, guidance, support, encouragement, and help over the

years.

Dr. Ernest Smith also provided space and support for me to work in his

lab, and I am particularly grateful for his generosity and kindness. I especially

appreciate his patience, guidance, and encouragement while training me in

molecular biology. I want to thank Dr. Andrew Jackson for his guidance, support,

and help over the years, particularly in the qualifying exams and providing an IC-

MS/MS for sample analysis; Dr. Hooper, for his enthusiasm and help to solve

and discuss problems in inside and outside of the classroom, and for providing

deer mice for my research; and Dr. McMurry for his kindness, impressive science

ii

attitude, and personality. Dr. McMurry also provided a lot of help and support for

perchlorate exposure assessment in beef cattle and the distribution experiment.

I appreciate all my committee members for dedicating their time, ideas, and

advice on my dissertation.

Secondly, I would like to thank all my Anderson lab mates over the last

three and half years for their help, support, encouragement, and advice: Lu Yu,

Jaclyn Cañas, Baohong Zhang, and Christina Freitag. Especially, I would like to

thank Lu Yu and Jaclyn Cañas for teaching me how to use instruments in the lab,

making me comfortable in the lab, and maintaining the lab neat and organized. I

would also like to thank them for their encouragement, support, suggestions, and

friendship in my research and life.

I would also like to thank all my colleagues out of the Anderson lab for

their friendship and help over the years. I would like to thank Bharath Ram,

Norka Malpartida, Jingqiu Zhu, Rene Viñas, and Colton Wilson in Dr. Smith’s lab

for their help, encouragement, and friendship. I had a very good time working

with them in the same lab. I would also like to thank Dr. Wang’s lab for their help

in both research and life. I would like especially to thank Fujun Liu, Haitao Luo,

and Hongmei Wu for their help during animal exposure studies, Toby McBride for

providing deer mice, and Jordan Smith for providing information about deer mice.

Thirdly, I would like to thank all those people who provided samples and

technical training for my dissertation research: Joanna Johnson and Dr. Leon

Glass from United Chemical Technologies, Inc (Bristol, PA) for providing SPE

cartridge samples; Cassie Abney and Dr. Michael Galyean from the Department

of Animal and Food Science, Texas Tech University, for cow urine samples, and

Dr. Sam Jackson for sheep urine samples; Dr. Michael McCarty and Linxia Dong

from the Department of Human Development and Family Studies, Texas Tech

University for providing human breast milk samples; Lisa Perlmutter, Scott

Severt, and John Isanhart for field sample collection; Andrew Kirk for providing

goat milk samples; Brad Thornhill from the Department of Civil Engineering for

iii

training on IC-MS/MS operation; and Dr. Kang Tian for

preconcentration/preelution IC and LC-MS/MS training. I also want to thank Drs.

Benjamin Blount and Liza Valentin from the Centers for Disease Control and

Prevention for help in urine sample analysis using IC-MS/MS.

I would like to thank all the faculty and stuff at The Institute of

Environmental and Human Health for providing a supportive and rich

environment during my research. Especially I would like to Dr. Angela Gentles

for her kindness and technical support during my time in Dr. Smith’s lab; Dr.

Jaclyn Cañas for reviewing my dissertation draft; Dr. George Cobb for his

knowledge and guidance in analytical instrumentation, allowing me to access to

the LC-ESI-MS, attending my defense, and reading my dissertation; Dr. Steven

Cox for his advice on statistical analysis; Dr. Phil Smith for his support and help

in the cattle study and vole distribution experiment; Kimberly Pruitt for patience

and help during ordering and purchasing supplies; and Tammy Henricks for the

help with international student paperwork.

Finally, I would like to thank my husband and family. Without their love,

support, and understanding, I would not be where I am today, and my

dissertation and education would not have been possible. I am grateful for the

love and support from my husband: Fujun Liu. We learned to take care of each

other and cherish each other over the years. Fujun also provided a lot of help,

encouragement, advice, and confidence in my research over the last years. I

deeply appreciate having him in my life. I am sincerely grateful for the love and

understanding of my great parents. They have always been supportive and

understanding of my decisions and desires. I always feel guilty not being able to

stay with them and take care of them, especially over the last three and half

years. I would like to thank my younger sisters and brother for being so

supportive of my board education, for taking care of our parents, and for being

my best sisters and brother. I am very proud of them and grateful to have them

iv

as family. I would like to dedicate this work to my great husband, parents,

sisters, and brother.

This research was supported in part by the U.S. Department of Defense

contract CU1141, through the Strategic Environmental Research and

Development Program (SERDP) under a Cooperative Agreement IERA-99-001

with the USAF, Institute for Environment, Safety, and Occupational Health,

Brooks AFB, TX

v

TABLE OF CONTENTS

ACKNOWLEDGMENTS ........................................................................................ii

ABSTRACT .......................................................................................................... x

LIST OF TABLES ............................................................................................... xiii

LIST OF FIGURES ..............................................................................................xv

CHAPTER

I. GENERAL INTRODUCTION................................................................ 1

Perchlorate Occurrence and Fate ................................................... 1 Potential Health Effects of Perchlorate Exposure............................ 3 Study Objectives, Background, and Hypotheses ............................ 7 Dissertation Format and Contents................................................. 10

II. PERCHLORATE DETERMINATION IN PLASMA, URINE,

AND MILK – METHOD DEVELOPMENT ........................................... 12

Introduction ................................................................................... 12 Cleanup Method for Perchlorate Determination in Plasma and urine .......................................................................... 17

Materials and Method .............................................................. 17 Chemicals........................................................................... 17 Sample sources and treatment........................................... 17 Sample preparation procedure for urine cleanup ............... 18 Sample analysis ................................................................. 19

Results and Discussion ........................................................... 20 Recovery and precision of alumina-neutral plus C18 as a cleanup method for plasma................................. 20 Efficiency of various cleanup procedures for urine ............. 20 Accuracy and precision of NAX plus Al-N for cleanup of different sources of urine .................................. 23 Application of cleanup method (NAX plus Al-N) to urine samples from field and laboratory studies ................. 24

Conclusions ............................................................................. 27 Perchlorate Determination in Urine, Milk, and Plasma Using Liquid Chromatography Electrospray Ionization Mass Spectrometry (LC-ESI-MS) and Comparison with Suppressed Conductivity Detection (IC-SCD)............................... 32

Materials and Method .............................................................. 32

vi

Reagents and standards .................................................... 32 Sample sources and preparation........................................ 32 IC-SCD analysis ................................................................. 34 LC-ESI-MS analysis ........................................................... 34

Results and Discussion ........................................................... 35 Linearity of response and detection limit for LC-ESI-MS ......................................................................... 35 Comparison of LC-ESI-MS with IC-SCD ............................ 37

Conclusions ............................................................................. 39 Perchlorate Determination in Urine Using Ion Chromatography Couples with Atmospheric Pressure Ionization Tandem Mass Spectrometry (IC-MS/MS) ..................... 48

Materials and Method .............................................................. 48 Reagents and standards .................................................... 48 Ion chromatography ........................................................... 48 Mass spectrometry ............................................................. 49 Samples preparation .......................................................... 50 Quality control .................................................................... 50

Results and Discussion ........................................................... 50 Selectivity and sensitivity of IC-MS/MS .............................. 50 Linearity of calibration curves ............................................. 52 Accuracy and precision of the IC-MS/MS method .............. 53

Conclusions ............................................................................. 53

III. ASSESSING PERCHLORATE EXPOSURE TO

MAMMALS IN THE FIELD ................................................................. 61

Introduction ................................................................................... 61 Materials and Methods.................................................................. 63

Study description and sample collection in TX......................... 63 Determination of perchlorate.................................................... 64

Results and Discussion................................................................. 66 Perchlorate in drinking water, blood plasma, and tissues of beef cattle in TX....................................................... 66 Perchlorate in contaminated water, blood plasma, and urine from cattle in Kansas ............................................... 67

Conclusions .................................................................................. 69

IV. PERCHLORATE DISTRIBUTION AND EXCRETION IN

LABORATORY ANIMALS .................................................................. 71

vii

Introduction ................................................................................... 71 Materials and Methods.................................................................. 73

Chemicals ................................................................................ 73 Animals .................................................................................... 74 Distribution experiment ............................................................ 74 Excretion experiment ............................................................... 75 Depuration experiment ............................................................ 76 Perchlorate determination method........................................... 76 Statistical analysis ................................................................... 79

Results and Discussion................................................................. 80 Perchlorate distribution in voles ............................................... 80 Perchlorate excretion pattern via urine in deer mice................ 82 Perchlorate depuration via urine in deer mice ......................... 86

Conclusions .................................................................................. 89

V. EFFECTS OF PERCHLORATE EXPOSURE ON

SODIUM-IODIDE SYMPORTER (NIS) AND PENDRIN

GENE EXPRESSION IN DEER MICE................................................ 97

Introduction ................................................................................... 97 Materials and Methods................................................................ 102

Animal exposure to perchlorate ............................................. 102 Partial deer mice NIS cDNA sequence development............. 103 Characterization of gene expression in deer mice with real-time PCR................................................................. 106 Statistical analysis ................................................................. 109

Results and Discussion............................................................... 109 Partial sequence of NIS gene ................................................ 109 NIS gene expression pattern in deer mice............................. 110 Effects of perchlorate exposure on NIS gene expression in deer mice......................................................... 113 Effects of perchlorate exposure on pendrin gene expression in deer mice......................................................... 115

Conclusions ................................................................................ 117

VI. PERCHLORATE EFFECTS ON FATTY ACID

PROFILES IN MILK.......................................................................... 126

Introduction ................................................................................. 126 Materials and Methods................................................................ 129

viii

Experimental design and samples collection ......................... 129 Fatty acid analysis in milk ...................................................... 130 Perchlorate determination in human milk............................... 131 Statistical analysis ................................................................. 132

Results and Discussion............................................................... 132 Fatty acids profile in goat milk dosed with perchlorate........... 132 Human milk survey ................................................................ 137

Conclusions ................................................................................ 139

VII. CONCLUSIONS AND FUTURE RESEARCH .................................. 147

Study Summary and Conclusions ............................................... 147 Future Studies............................................................................. 150

REFERENCES................................................................................................. 153

APPENDEX...................................................................................................... 171

ix

ABSTRACT

Perchlorate contamination in the U.S. appears to be more widespread

than originally thought. Perchlorate is naturally occurring, in addition to its

anthropogenic sources. Animals including humans have been exposed to

perchlorate through drinking water and/or trophic transfer; perchlorate has been

detected in dairy milk, some food items, human milk, and urine. Perchlorate

exposure and effects in mammals were characterized in the current study.

The ability to detect perchlorate in exposed animals is critical for effects

studies and risk assessments; perchlorate residues in biological fluids such as

plasma, urine, and milk can serve as biomarkers for perchlorate exposure. A

well-developed method for perchlorate determination in these matrices would

contribute greatly to perchlorate exposure assessment. In this study, analytical

methods for perchlorate determination in these matrices were explored.

Alumina-neutral cartridges combined with C18 cartridges and NAX combined

with alumina-neutral provided the best cleanup with significantly reduced

background signal of plasma and urine, respectively, and relatively high recovery

of perchlorate using conventional ion chromatography with suppressed

conductivity detection (IC-SCD). However, the cleanup method was not robust

enough to be used on some urine matrices such as deer mouse urine collected

in perchlorate excretion and elimination experiments. As an alternative to IC-

SCD, IC-MS/MS provides an excellent method with high selectivity and sensitivity

for part per trillion perchlorate determination in both aqueous and deer mice urine

matrices.

A study on perchlorate exposure and absorption in beef cattle showed that

constant exposure to 25 ng/mL perchlorate in water over 14 weeks did not result

in measurable residues in blood plasma or edible tissues of cattle at the first test

site (McLennan County, Texas). However, perchlorate was detected in 4 of 33

and 17 of 26 cattle at the two Kansas farms with the highest plasma perchlorate

x

concentrations of 43 and 32 ng/mL, respectively. Compared to perchlorate

residues in urine, perchlorate residues in plasma may not be a proper biomarker

for perchlorate exposure assessment.

A study on perchlorate distribution, excretion, and depuration in rodents

showed that urine was the major pathway for perchlorate fate in the body.

Higher levels of perchlorate exposure corresponded well to high levels of

perchlorate excreted in the urine. Perchlorate excretion via urine reached a

steady state after one day in the 28-day exposure experiment. An average of 46,

46, and 61 % of perchlorate was recovered in urine over the exposure period in

high, medium, and low dose groups, respectively. Metabolism of perchlorate

may occur in the body based in part on the 40% perchlorate unaccounted for in

this study. Endogenous perchlorate may also exist. Perchlorate exposure

through dosed drinking water for 28 days increased sodium-iodide symporter

(NIS) gene expression in the kidney and stomach, and pendrin gene expression

in the kidney, without significant difference. No significant difference was

observed neither between the low and high dose groups in the depuration

experiment regarding either gene expression in the kidney or stomach. No

significant linear relationship was found between perchlorate urinary excretion

and either gene expressions in the kidney.

A partial sequence of deer mice NIS gene cDNA with 425 bps was

discovered in the current study for the first time. Quantitative analysis of NIS

mRNA expression in various tissues was also studied for the first time in the

current study with expression levels from highest to lowest in deer mice tissues in

the following order: stomach, testes, brain, large intestine, and barely expression

in the lung, kidney, heart, and liver.

The effect of perchlorate exposure on the fatty acid profile in milk was

observed in lactating goats dosed with perchlorate from Monday to Friday each

week for 4.5 weeks. ω-6 fatty acids and total polyunsaturated fatty acids (PUFA)

at the 1 mg/kg treatment were significantly reduced at day 10 (p = 0.0113 and

xi

0.0053, respectively), day 17 (p < 0.001), and day 24 (p < 0.05), but not at day 2

and 31. Monounsaturated fatty acid (MUFA) was significantly reduced only at

day 17 (p = 0.0130). Significant reductions in short- and long-chain fatty acids

were observed at day 24 only (p = 0.0431 and 0.0097, respectively) in the high

and low dose groups, respectively. Additionally, a weak negative correlation

between milk perchlorate concentrations and total PUFA levels was found in

human breast milk samples collected from Lubbock, TX or nearby counties. To

our knowledge, this is the first report on the effect of perchlorate exposure on

fatty acid profiles in milk. Further study is urged to investigate mechanistic

aspects of the effect. This work will contribute to the human risk assessment of

perchlorate, particularly for the development of infants with maternal exposure to

perchlorate.

xii

LIST OF TABLES

2.1 Accuracy and precision of perchlorate determination in cow plasma using ion chromatography with suppressed conductivity detection .................................................................................................. 28

2.2 Recoveries of perchlorate in perchlorate-spiked cow urine

(100 ng/mL) cleaned using various solid phase extraction (SPE) Cartridges ................................................................................................ 29

2.3 Accuracy and precision for NAX combined with Al-N as cleanup

cartridges for perchlorate determination in different types of urine using ion chromatography............................................................... 31

2.4 Physical properties (pH and conductivity) of the different

sources of urine ....................................................................................... 31 2.5 Limit of detection and method detection limit for perchlorate

in standard solutions, sheep urine, dairy milk, and cow plasma analyzed by LC-ESI-MS........................................................................... 42

2.6 Accuracy and precision of perchlorate determination using

LC-ESI-MS in sheep urine, dairy milk, and cow plasma matrices............ 42 2.7 Precision of perchlorate determination in spiked Milli-Q water,

dairy milk, and cow plasma matrices using IC-SCD and LC-ESI-MS ...... 47 2.8 Method detection limit study for perchlorate using IC-MS/MS ................. 55 2.9 Accuracy and precision of the IC-MS/MS method ................................... 60 4.1 Changes in perchlorate recovery in different tissues of prairie

voles (Microtus ochrogaster) after 4- or 8-h exposure ............................. 91 5.1 Sequence of sense and antisense primers designed by

VectorNTI based on Mus musculus NIS mRNA for PCR amplification of deer mice NIS cDNA..................................................... 119

5.2 Sequence of sense primers, antisense primers, and

TaqMan probes designed for real-time PCR amplification of pendrin and NIS mRNA equivalents in deer mouse............................... 119

6.1 Results of two-way ANOVA test for fatty acid contents

xiii

in lactation goats dosed with perchlorate ............................................... 141

A.1 Perchlorate uptake and distribution in various studies........................... 172 A.2 Perchlorate excretion in various studies ................................................ 173

xiv

LIST OF FIGURES

2.1 Chromatograms of water contaminated with perchlorate and plasma spiked with 100 μg/L perchlorate using ion chromatography with suppressed conductivity detection (IC-SCD) .................................... 28

2.2 Ion chromatography analysis of cow urine samples (with

no perchlorate) after being processed via various cleanup procedures ................................................................................. 29

2.3 Efficiencies of four cleanup procedures (NAX combined with Al-N,

NAX with PSA, C18 with Al-N, and PSA with Al-N) for perchlorate determination in urine spiked with and without perchlorate...................... 30

2.4 Linearity of MS response to perchlorate in prepared standard

solutions and spiked biological matrices, including dairy milk, cow plasma, and sheep urine .................................................................. 41

2.5 Comparison of the analysis of dairy milk by IC-SCD and LC-ESI-MS ..... 43 2.6 Comparison of the determination of perchlorate in cow plasma

by IC-SCD and LC-ESI-MS...................................................................... 44 2.7 Comparison of the determination of perchlorate in sheep

urine by IC-SCD and LC-ESI-MS............................................................. 45 2.8 Calibration curves of perchlorate spiked into Milli-Q water and

milk matrices analyzed by IC-SCD and LC-ESI-MS ................................ 46 2.9 IC-MS/MS analysis of standard solution with 0.02 ng/mL perchlorate ..... 56 2.10 IC-MS/MS analysis of deer mice urine (diluted 500-fold) ......................... 57 2.11 Analysis of a standard solution and deer mice urine diluted

500-fold by ion chromatograph with suppressed conductivity detection (IC-SCD) .................................................................................. 58

2.12 Calibration curves for the two MRM transitions of perchlorate

in IC-MS/MS ............................................................................................ 59 3.1 The relationship between perchlorate exposure in drinking

water and perchlorate in heifer plasma.................................................... 70

xv

4.1 Perchlorate concentrations in tissues in prairie voles (Microtus ochrogaster) after 4- or 8-h exposure ...................................................... 91

4.2 Perchlorate excretion patterns in deer mice (Peromyscus

maniculatus) exposed to perchlorate through drinking water for 28 days ............................................................................................... 92

4.3 Perchlorate (% of total perchlorate intake through dosed drinking

water) excreted via urine over the 28-day exposure period ..................... 93 4.4 Perchlorate concentration in urine in the depuration experiment ............. 94 4.5 Exponential non-linear regression models for the depuration patterns .... 95 4.6 Comparison of perchlorate excretion and elimination patterns

over a 96-h exposure period in deer mice (Peromyscus maniculatus) .... 96 5.1 RNA integrity test on a 1% denaturing formaldehyde agarose gel......... 119 5.2 PCR product (more than 500 bp) from small intestine and

stomach mixture cDNA samples with primers shown in Table 5.1......... 120 5.3 Partial sequence of deer mouse NIS gene cDNA (425 bps) .................. 120 5.4 NIS gene expression pattern in various deer mice tissues .................... 121 5.5 Effect of perchlorate exposure through drinking water on

NIS gene expression in the stomach of deer mice................................. 122 5.6 Effect of perchlorate exposure through drinking water on

NIS gene expression in the kidney of deer mice.................................... 123

5.7 Effect of perchlorate exposure through drinking water on pendrin gene expression in the stomach of deer mice........................... 124

5.8 Effect of perchlorate exposure through drinking water on pendrin gene expression in the kidney of deer mice.............................. 125

6.1 Proposed relationship between perchlorate and fatty acid

composition in milk fat............................................................................ 140 6.2 The content of total polyunsaturated fatty acids (PUFA) in goat

milk during perchlorate exposure........................................................... 141

xvi

6.3 The content of ω-6 fatty acids in goat milk during perchlorate Exposure................................................................................................ 142

6.4 The content of mono-unsaturated fatty acids (MUFA) in goat

milk during perchlorate exposure........................................................... 142 6.5 The content of medium-chain fatty acids in goat milk during

perchlorate exposure ............................................................................. 143 6.6 The content of long-chain fatty acids in goat milk during

perchlorate exposure ............................................................................. 144 6.7 Changes in PUFA and ω-6 fatty acid content with time

in the three different dosing groups ....................................................... 145 6.8 Correlation between polyunsaturated fatty acid (PUFA) and

perchlorate concentrations in human breast milk from mothers in Lubbock; Texas or nearby counties ................................................... 146

6.8 Plot for docosahexaenoic acid (DHA; C22:6n3) as a function of

perchlorate concentration in human milk from Lubbock, Texas or nearby counties ................................................................................. 146

A.1 Linear relationships between gene expression (NIS and pendrin) at day 28 and perchlorate urinary excretion........................................... 174

xvii

CHAPTER I

GENERAL INTRODUCTION

1.1 Perchlorate Occurrence and Fate

Perchlorate (ClO4-) is a tetrahedral anion with four oxygen atoms oriented

around a central chlorine atom (VII) (Urbansky and Schock, 1999). It is

commonly associated with the solid salts of ammonium, potassium, and sodium.

Perchlorate salts are widely used as ingredients in pyrotechnics, matches,

blasting agents, automobile air bag inflators, nuclear reactors, lubricating oils,

textile dye fixing, electronic tubes, tanning and finishing leather, rubber

manufacturing, electroplating, aluminum refinishing, paint and enamel

production, and pharmaceuticals (Motzer, 2001). However, the majority of

manufactured perchlorate is used as a solid oxidizer for propellants in military

munitions items such as rockets and missiles (Urbansky, 1998; Motzer, 2001).

Large-scale production of perchlorate salts began in the mid-1940s and large

volumes have been disposed of in various states since the 1950s (Urbansky,

1998; Fisher et al., 2000; Motzer, 2001; Winkler et al., 2004).

In addition to anthropogenic sources of perchlorate, it may also naturally

occur. As early as 1958, perchlorate was detected in seawater (Bass Becking et

al., 1958). Four decades later, around 1% perchlorate was found in fertilizers

derived from the well-known naturally occurring Chilean caliche (Susarla et al.,

1999; Urbansky et al., 2001; Urbansky and Collette, 2001). Recently,

perchlorate was found in the Texas southern high plains aquifer system

(Dasgupta et al., 2005; Jackson et al., 2005a), where there is no historical record

or evidence of manufacturing or use of perchlorate salts or known sources of

fertilizers derived from Chilean caliche. Perchlorate was also reported to be

present in many rain and snow samples collected in Lubbock, TX, and Cocoa

Beach, FL with hurricane Frances (Dasgupta et al., 2005). In situ

electrochemical production of perchlorate within a public water distribution

1

system has also been documented (Jackson et al., 2004). It also has been

shown that perchlorate is readily formed by a variety of stimulated atmospheric

processes such as from chloride aerosol by electrical discharge and by exposing

aqueous chloride to high concentrations of ozone (Dasgupta et al., 2005).

Once released into the environment, perchlorate can persist for decades

in aquatic systems under normal conditions because it is extremely water soluble

and kinetically stable. Perchlorate contamination in water was discovered as

early as 1985 in wells located at California superfund sites; however, nationwide

contamination of perchlorate in water was not recognized until 1997 with the

development of new analytical methods. Today, perchlorate contamination in the

environment is widespread in the U.S. and has been found in water and/or soil

samples from over 35 states to date (EPA, 2005a)

In most aqueous systems perchlorate concentrations are low, but there

are also some highly contaminated sites across the U.S. In the Las Vegas,

Nevada area (near Henderson), perchlorate concentrations ranged from 630 to

3,700 µg/mL in contaminated groundwater (Motzer, 2001) with concentrations up

to 1,700 ng/mL in Lake Mead inlet surface water (EWG, 2001). At the Naval

Weapons Industrial Reserve Plant (NWIRP) in McGregor, Texas, perchlorate

was detected at concentrations ranging from 4 to 97,000 ng/mL in the

groundwater and 5,600 ng/mL in tributary surface water (Motzer, 2001). In

California, the maximum perchlorate level detected in contaminated drinking

water reached 820 ng/mL (EWG, 2003). It has been estimated that more than 11

million people could be exposed to perchlorate through public drinking water

supplies at concentrations of at least 4 ng/mL (NAS, 2005).

It has been well reported that perchlorate can be taken up and

accumulated by plants and edible vegetation (in both the laboratory and the

field), including salt cedar (Urbansky et al., 2000b); bulrush, crabgrass, cupgrass,

and goldenrod (Smith et al., 2001), tobacco plants (Ellington et al., 2001);

smartweed, watercress, ash, chinaberry, elm, willow, mulberry, and hackberry

2

(Tan et al., 2004); cucumber, lettuce, and soybean (Yu et al., 2004); soybean,

alfalfa, wheat, cucumber, cantaloupe, and tomato (Jackson et al., 2005b); lettuce

(Sanchez et al., 2005). Therefore, there are concerns regarding the potential

exposure of animals and humans to perchlorate through trophic transfer such as

ingestion of plants or food containing perchlorate in addition to drinking water.

Perchlorate has been detected in tissues of aquatic plants and animals in

the vicinity of contaminated sites. Aquatic organisms have been found to be at a

relatively high risk for perchlorate exposure. In streams and ponds near the

Longhorn Army Ammunition Plant (LHAAP) near Karnack, Texas, perchlorate

was detected in aquatic insects, fish, and frogs at concentrations ranging from

811 to 2038 µg/kg, below detection to 207 µg/kg, and below detection to 580

µg/kg, respectively (Smith et al., 2001). Smith et al. (2001) also reported

perchlorate concentrations ranging from below detection to 2328 µg/kg in small

mammals (e.g., rodents) collected from LHAAP. In addition, perchlorate

contamination has been found in supermarket milk samples, human breast milk,

and human urine (Kirk et al., 2003; Krynitsky et al., 2004; Kirk et al., 2005;

Valentin-Blasini et al., 2005), which indicates that perchlorate exposure and

environmental contamination in the U.S. are much more widespread than

originally thought.

1.2 Potential Health Effects of Perchlorate Exposure

Perchlorate has the same ionic charge and a similar ionic radius as iodide,

which allows perchlorate to competitively inhibit iodide uptake by the thyroid via

the sodium-iodide symporter (NIS) (Wolff, 1998; Motzer, 2001; Soldin et al.,

2001). The inhibitory effects of perchlorate on thyroidal uptake of iodide have

been reported in laboratory animals (Yu et al., 2002; Merrill et al., 2003; Merrill et

al., 2005) and humans (Lawrence et al., 2000; Greer et al., 2002; Clewell et al.,

2004). Perchlorate is also known to reduce the transfer of radioiodine from

plasma to milk (Cline et al., 1969; Djurdjevic and Lengemann, 1970), and

3

recently, perchlorate concentrations in milk above 10 ng/mL were reported to be

associated with low iodine content in human milk samples (Kirk et al., 2005). In

addition, perchlorate can reduce the deposition of iodine in the chicken egg

(Peña et al., 1976). Less iodine in milk and/or egg may eventually cause iodide

deficiency in the thyroid, especially for infants using milk as the only food source.

As an essential element for thyroid hormone production, iodide deficiency in

the thyroid caused by perchlorate exposure may eventually result in disruption of

normal thyroid function: reduced production of thyroid hormones, triiodothyronine

(T3) and thyroxine (T4), and increased production of thyroid stimulating hormone

(TSH) via a negative feedback mechanism triggered by the hypothalamus-

pituitary-thyroid axis. Based on this rationale, perchlorate was once clinically

used to treat Grave’s disease (hyperthyroidism) (Wolff, 1998; Soldin et al., 2001).

However, perchlorate is no longer used therapeutically in the U.S. because of its

potential adverse effects and toxicity (Wolff, 1998).

Many studies have shown the effects of perchlorate exposure on thyroid

functions. Following perchlorate exposure, increased levels of thyroid stimulating

hormone and/or decreased levels of thyroid hormones were reported in rats

(Siglin et al., 2000; Yu et al., 2002), deer mice (Thuett et al., 2002), quail chicks

(McNabb et al., 2004), fish (Bradford et al., 2005; Crane et al., 2005), and

humans (Lamm et al., 1999; Brechner et al., 2000; Lawrence et al., 2000; Greer

et al., 2002; Braverman et al., 2005). Alteration of thyroid gland morphology,

such as thyroid follicle hypertrophy, colloid depletion, and angiogenesis, was also

assessed in many studies and was reported as a biomarker for perchlorate

exposure and effect in animals (Thuett et al., 2002; Patino et al., 2003; York et

al., 2003; Bradford et al., 2005; Gentles et al., 2005; Mukhi et al., 2005). On the

other hand, the absence of an effect of perchlorate exposure on thyroid

hormones was also observed in some epidemiology (Crump et al., 2000; Li et al.,

2000a; Li et al., 2000b) and occupational (Gibbs et al., 1998; Lamm and

Doemland, 1999) studies.

4

Thyroid hormones are essential for many physiological processes,

including development, growth, and metabolism. Therefore, concerns about

potential effects of perchlorate exposure include carcinogenic, developmental,

neurodevelopmental, reproductive, and immunotoxic effects. In some studies,

perchlorate exposure did produce adverse effects during critical periods of

development, such as alteration of gonadal differentiation during amphibian

metamorphosis (Goleman et al., 2002a); precocious metamorphosis (Manzon

and Youson, 1997); delay in metamorphosis (Goleman et al., 2002b; Tietge et

al., 2005); and developmental retardation in the early life stages of fish (Crane et

al., 2005). Developmental toxicity of perchlorate was also investigated in rabbits

(York et al., 2001a) and rats (York et al., 2003) with no observable adverse

developmental effect at the test concentrations. The developmental no-

observable-adverse-effect-level (NOAEL) was set at 100 mg/kg-day for rabbits,

which was the highest dose tested in the study; the NOAEL was 1.0 mg/kg-day in

the rat study with developmental delays in ossification occurred in the 30.0

mg/kg-day group.

Neurodevelopmental effects are also a consideration for perchlorate

exposure because normal production of thyroid hormones is integrally involved in

the development of brain, central nervous system, and neurobehavioral

capacities (Bekkedal et al., 2004). Neurodevelopmental toxicity of perchlorate

exposure was evaluated in rats with no behavioral effects in the offspring

exposed as high as 10.0 mg/kg-day (the highest dose) as evaluated by passive

avoidance, swimming watermaze, motor activity, and auditory startle, but with

effects on thyroid hormone (T3, T4, and TSH) levels and thyroid morphometric

and histopathology (York et al., 2004). Pediatric neurobehavioral diseases (i.e.,

attention deficit-hyperactivity disorder (ADHD) and autism) and school

performance were also evaluated in an ecological study (Chang et al., 2003), but

no evidence was found that children from an area with perchlorate in drinking

5

water (up to 24 ng/mL) had either an increase in pediatric neurobehavioral

diseases or a decrease in school performance.

The reproductive toxicity of perchlorate has also been assessed.

Perchlorate exposure during a critical period of development was found to disrupt

ovarian follicle maturation and reduce the number of preantral and antral follicles

in certain size classes of rats (Baldridge et al., 2004). Perchlorate did show

adverse effects on cocoon production and hatch in earthworm (Landrum et al.,

2005). Perchlorate was also found to accumulate in quail eggs (Gentles et al.,

2005). In fish, 18 µg/mL perchlorate, which is a high environmentally relevant

concentration, affected the histological condition of adult zebrafish thyroid

follicles but not their reproductive performance, although 677 µg/mL perchlorate

reduced spawn volume within one week and the effect became negligible after

four weeks (Patino et al., 2003). On the other hand, perchlorate was suggested

as not a reproductive toxicant in rats with doses up to 30 mg/kg-day through

drinking water (York et al., 2001b).

There is concern about the potential human health effects of low-level

perchlorate exposure in drinking water (Siglin et al., 2000). The U.S. EPA

originally asked for a regulatory level of 1 ng/mL in drinking water for perchlorate

in 2002 and several states set an advisory level of perchlorate in drinking water

ranging from 1 to 51 ng/mL (EPA, 2002; EPA, 2005b). In contrast, a much less

conservative reference dose equivalent to 200 ng/mL was proposed by the

Perchlorate Study Group, which is composed of perchlorate producers and

industrial consumers (Dahl 2004). Since then, debate on what level of

perchlorate in drinking water is safe has been ongoing (Dahl, 2004; Stokstad,

2005). No National Drinking Water Standard is set for perchlorate currently, but

the National Research Council of the National Academies (NRC) issued a

reference dose for chronic oral exposure (oral RfD) of 0.7 μg/kg/day (NAS, 2005)

in January 2005 and EPA set this as its safe dose for perchlorate. The safe dose

corresponds to 24.5 ng/mL perchlorate in drinking water for a 70-kg adult

6

consuming 2 L of water every day. This value is 23 times higher than EPA’s draft

reference dose, but 7 times lower than that recommended by the Perchlorate

Study Group.

1.3 Study Objectives, Background, and Hypotheses

The first objective of this study is to develop analytical methods for

perchlorate determination in biological matrices. Even though new more

sensitive analytical methods have recently been developed for perchlorate

determination (Tian et al., 2003; Krynitsky et al., 2004; Winkler et al., 2004; Li

and George, 2005; Martinelango et al., 2005; Valentin-Blasini et al., 2005),

perchlorate determination in complex biological matrices using traditional ion

chromatography (IC) is still a challenge. The unique characteristics of the

perchlorate anion and complexity of the matrices results in high background and

interference problems during routine IC analysis. However, the ability to detect

perchlorate in exposed animals or humans is critical for effects studies and risk

assessments. Perchlorate residues in biological fluids such as plasma, urine,

and milk can serve as biomarkers for perchlorate exposure, and therefore, a well-

developed method for perchlorate determination in these matrices would

contribute greatly to perchlorate exposure assessment. In this study, analytical

methods for perchlorate determination in these matrices were explored.

The second objective was to assess perchlorate exposure in mammals in

both the laboratory and the field using developed analytical methods for

perchlorate determination. There is evidence for perchlorate exposure and effect

on plants, animals (both aquatic and terrestrial), and humans in contaminated

sites as mentioned above, however to our knowledge, there is little data on

perchlorate exposure to large mammals (such as dairy and beef cattle). Large

mammals could potentially ingest perchlorate through both contaminated drinking

water and forage crops and thus present a potential pathway of perchlorate

exposure to humans. There is increasing concern regarding potential

7

perchlorate exposure and effects on animals and humans through trophic

transfer especially since perchlorate has been detected in dairy milk, human

breast milk, and some food items (Erickson, 2003; Kirk et al., 2003; Krynitsky et

al., 2004; Capuco et al., 2005; Kirk et al., 2005). Therefore, perchlorate

exposure to large mammals from contaminated sites was assessed.

Perchlorate exposure in laboratory animals was assessed through

investigating perchlorate distribution and excretion in prairie vole (Microtus

ochrogaster) and deer mice (Peromyscus maniculatus), respectively. Several

studies on the uptake, distribution, and excretion of perchlorate in animals

including humans showed that most perchlorate was not metabolized in the body

and excreted primarily in urine (Anbar et al., 1959; Goldman and Stanbury, 1973;

Peña et al., 1976; Batjoens et al., 1993; Yu et al., 2002). Although the highest

perchlorate concentrations were reported in the thyroid in many studies, the

perchlorate distribution pattern in other tissues was not consistent. To a large

degree, adverse effects of perchlorate exposure on animals and humans depend

on its uptake, distribution, and excretion in the body. Whether perchlorate

accumulates or not, how perchlorate distributes among tissues, and what are the

excretion and depuration patterns via urine are fundamental questions. That will

help determine whether perchlorate exposure causes adverse human health

effects or ecological impacts. Therefore, a tissue distribution study of perchlorate

in prairie vole (Microtus ochrogaster) and perchlorate excretion and depuration

studies in deer mice (Peromyscus maniculatus) were conducted. The

hypotheses we tested included (1) urine is the major pathway through which

perchlorate is excreted; (2) perchlorate excretion via urine is dose-dependent:

high levels of perchlorate exposure correspond to high levels of perchlorate

urinary excreted; (3) perchlorate is depurated rapidly via urine after removal of

perchlorate exposure. Developed methods were used to determine perchlorate

residues in tissues and urine.

8

The third objective of this study was to characterize perchlorate effects in

mammals. The effect of perchlorate exposure was investigated in terms of

transporter gene expression. The sodium-iodide symporter (NIS) is well

documented as a basolaternal membrane transporter for iodide in many organs

including human kidney. Since perchlorate has a similar size and same charge

as iodide, NIS may play an important role in perchlorate uptake in the thyroid; it is

also a potential transporter for perchlorate influx and efflux in other organs such

as the kidney. Pendrin has been reported as a transporter of several ions in the

thyroid, kidney, and lung. Perchlorate exposure in deer mice was associated

with changes in pendrin gene expression in the kidney and lung (Ramachandran,

2005). The relationship between the perchlorate excretion pattern via urine and

the level of these transporters in the kidney can provide a potential perchlorate

excretion mechanism and a potential adaptation mechanism at the molecular

level. Therefore, the relationship between pendrin and NIS gene expression in

the kidney and perchlorate excretion pattern via urine was explored. It was

hypothesized that the level of the transporter gene(s) would increase upon

perchlorate exposure, and in turn, perchlorate excretion via urine would increase.

The second effect was the correlation between perchlorate exposure and

milk fatty acid profile. Based on preliminary data that showed a weak negative

correlation between perchlorate concentrations and total polyunsaturated fatty

acids (PUFA) in 39 human breast milk samples collected from Lubbock, TX and

nearby counties (unpublished data), a theoretically supported relationship was

proposed: disruption of thyroid hormone production by perchlorate exposure

ultimately could alter the fatty acid profile in milk. Therefore, we investigated the

effects of perchlorate exposure on the milk fatty acid profile in lactating goats that

were dosed with perchlorate for 4.5 weeks. We hypothesized that perchlorate

exposure would change the fatty acid profile in the goat milk. Fatty acids,

particularly PUFA, are important functional components in milk; therefore, effects

on fatty acids may alter their health benefits to offspring. Thus, this work will

9

contribute to the environmental and human health risk assessment of

perchlorate, particularly for normal development of offspring with maternal

exposure to perchlorate.

1.4 Dissertation Format and Contents

This dissertation is composed of seven chapters. Each chapter is about a

sub-study and can be an individual manuscript for publication, except Chapter I,

which is a general introduction for perchlorate and the background of this study,

and Chapter VII, which is a general conclusion for this study. Each chapter, from

Chapter II to VI, includes introduction, methods and materials, results and

discussion, and conclusions sections.

Chapter I includes a general review for perchlorate occurrence and fate in

the environment, and its potential toxicity (potential health effect) of exposure,

which gives readers a general idea on what perchlorate is and why it is important

to do research on this chemical. It also briefly introduces the background,

objectives, and hypotheses of the sub-studies.

Chapter II is about analysis methodology development for perchlorate

determination. It includes a review of perchlorate analysis methods

(introduction), and three sub-chapters, each contain methods and materials,

results and discussion, and conclusions sections. The first sub-chapter is about

the cleanup methods developed for perchlorate determination in plasma and

urine matrices, which has been published in Talanta (Cheng, Q. F. Liu, J. E.

Cañas, and T. A. Anderson. 2006, A cleanup method for perchlorate

determination in urine. 68:1457-1462); the second is about a LC-ESI-MS method

developed for perchlorate determination in biological fluids (plasma, urine, and

milk); and the third is about an IC-MS/MS method for perchlorate determination

in urine.

Chapter III is about a study on perchlorate exposure and absorption in

mammals, beef cattle specifically. It has been published in Journal of Agricultural

10

and Food Chemistry (Q. Cheng, L. Perlmutter, P. N. Smith, S. T. McMurry, W. A.

Jackson, and T. A. Anderson. 2004. A Study on Perchlorate Exposure and

Absorption in Beef Cattle. 52, 3456-3461)

Chapter IV is about a study designed to elucidate perchlorate distribution,

excretion via urine, and depuration pattern in small rodents. It includes three

experiments: perchlorate distribution in prairie vole (Microtus ochrogaster),

excretion, and depuration in deer mice (Peromyscus maniculatus).

Chapter V is about the effect of perchlorate exposure on gene expression

in deer mice. The genes were focused on two transporters: sodium-iodide

symporter (NIS) and pendrin. The relationships between perchlorate exposure

and/or excretion patterns and the level of these gene expressions in the kidney

and stomach are discussed.

Chapter VI is about the effect of perchlorate exposure on fatty acid profiles

in goat milk. That perchlorate may alter fatty acid profiles in milk was

hypothesized and the alternation was observed in the lactating goats which were

dosed with perchlorate for 4.5 weeks.

Chapter VII is a general conclusion for this dissertation (this study).

Results and concerns from characterizing perchlorate exposure and effects in

mammals in the current study are summarized briefly.

11

CHAPTER II

PERCHLORATE DETERMINATION IN PLASMA, URINE, AND MILK

- METHOD DEVELOPMENT

2.1 Introduction

Many methods have been historically used to determine perchlorate

including gravimetry, spectrophotometry, electrochemistry (ion selectivity

electrodes (ISEs) and membrane field transistors (MEMFETs)), capillary

electrophoresis, and ion pair high performance liquid chromatography (HPLC)

(Urbansky, 1998; Urbansky, 2000; Motzer, 2001; Urbansky, 2002). However,

there are problems with these methods such as interferences from phosphate,

nitrate, chlorate, and other anions, low sensitivity, and high detection limits. An

extraction-spectrophotometric method was recently developed for the

determination of trace amounts of perchlorate based on an ion pair with thionine;

the reported detection limit (23 ng/mL) was still higher than other reported

detection limits (Pourreza and Mousavi, 2005).

Ion chromatography (IC) is currently the recommended method by the

U.S. EPA for routine determination of perchlorate (Hautman et al., 1999). It is a

sensitive and reliable technique, especially for drinking water, surface water, and

groundwater analysis for perchlorate. Dionex Corporation was a pioneer in the

development of methods for trace perchlorate determination in ground and

drinking water by IC (Hautman et al., 1999; Jackson et al., 1999; Jackson et al.,

2000a; Jackson et al., 2000b; Jackson and Chassaniol, 2002), and has marketed

a commercial IC instrument. With the improved method developed by Jackson et

al. (2000b), the detection limit for perchlorate in drinking water can be as low as

0.15 ng/mL. Perchlorate determination was performed on a DX-500 IC System

(Dionex Corporation) consisting of a GP 50 gradient pump, EG 40 eluent

generator, AS 40 automated sampler, LC 30 chromatography compartment, and

CD 20 conductivity detector. Dionex IonPac AS16 analytical column and IonPac

12

AG 16 guard column were used to separate the analytes. Anions were detected

by suppressed conductivity. It is these components that are the basis for EPA

Method 314.0 (Hautman et al., 1999).

Perchlorate determination by IC is greatly hindered when samples contain

high total dissolved solids (TDS), such as high salinity water, fertilizer, plant, and

animal matrices. Conductivity detection responds to any ion with sufficient

conductivity and therefore lacks specificity for perchlorate. Quantitative

determination of perchlorate in biological fluids (e.g. milk, urine, and plasma) is

often problematic with ion chromatography using suppressed conductivity

detection (IC-SCD) because biological fluids usually contain additional ions,

proteins, lipids, sugars, and other biomolecules that may confound accurate

determination of perchlorate. Without effective sample preparation and cleanup

methods, which are typically costly, time consuming, and labor intensive,

problems such as high background conductivity and interferences in biological

fluids are always encountered using IC-SCD. The higher background and

interferences may lead to a higher likelihood of both false positives and false

negatives. Therefore, the sensitivity, precision, and accuracy of IC-SCD are not

satisfactory in these complicated matrices. The desire to eliminate TDS has led

to development of a preconcentration method for perchlorate prior to IC analysis

(Tian et al., 2003; Tian et al., 2005; Cañas et al., 2006). With an online

preconcentration/preelution technique based on IC, perchlorate is concentrated

on a preconcentration column, undesirable matrix is preeluted with a low

concentration eluent and then perchlorate is injected into the IC separation and

detection system. This method has proved to be effective in detecting trace

perchlorate in milk with a LOD of 0.5 ng/mL (Kirk et al., 2003). The method was

also used to determine perchlorate in complex matrices such synthetic high ionic

strength solutions, bird egg extracts, hydroponic nitrate fertilizer samples, animal

tissues, and lettuce (Tian et al., 2005; Cañas et al., 2006). This method was

highly effective in removing most of the matrix anions and was resistant to the

13

interferences commonly encountered in a high ionic strength background (Tian et

al., 2005). An evaporative preconcentration technique has also been reported to

determine trace perchlorate in drinking water using ion chromatography with a

detection limit of 0.2 ng/mL (Liu et al., 2002).

There are other options to eliminate TDS and some interferences in

samples, for example, improvement of the sample preparation method before

instrumental analysis. It was reported that exposure of plant tissue extracts to

alumina could significantly reduce the high background conductivity during IC

analysis (Ellington and Evans, 2000). The minimum reporting level of

perchlorate in lettuce and tomato was found to be about 250 ng/mL (based on

wet weight) with this preparation method. Anderson and Wu (2002) also

evaluated cleanup methods for perchlorate determination in animal tissues.

Cleanup of animal tissue extracts using polar sorbents (Al and Si), an ion

exchange sorbent (SCX), or an ion exchange membrane (Nafion®) reduced the

background conductivity in IC and led to high perchlorate recovery (≥ 85%). In

addition, granular barium chloride, Dionex OnGuard cartridges (silver, barium,

and hydrogen), and pH adjustment with HCl were used as cleanup methods for

environmental water samples to remove sulfate, chloride, and carbonate ions

which usually contribute to high total conductivity and interferences for

perchlorate determination using ion chromatography or mass spectrometry

(Winkler et al., 2004; Medina et al., 2005).

Confirmatory testing is another primary way to minimize the interferences

and provide unambiguous results for perchlorate determination, especially in

complicated sample matrices, such as biological fluids. Confirmation with

Raman spectroscopy has been used for qualitative identification of perchlorate in

fertilizers and plant tissues (Williams et al., 2001). Infrared spectroscopy using

an attenuated total reflectance crystal (ATR-FTIR) was also used for perchlorate

identification in fertilizers (Collette et al., 2003). However, mass spectrometry

(MS) is a major technique that has been recently reported for perchlorate

14

determination in a variety of matrices. MS has become an alternative to IC-SCD

and is a preferred technique in accurate qualitative and quantitative

determination of perchlorate.

Methods based on coupling ion chromatography with electrospray

ionization mass spectrometry (IC-ESI-MS) have been used to determine

perchlorate in milk (Kirk et al., 2003; Kirk et al., 2005), and extracts of whole fish

(Dodds et al., 2004). LC/IC-ESI-MS/MS also has been applied for perchlorate

determination in water, groundwater, soil, milk, some food items, and human

urine (Koester et al., 2000; Krynitsky et al., 2004; Winkler et al., 2004; Li and

George, 2005; Snyder et al., 2005; Valentin-Blasini et al., 2005). Without the

assistance of chromatography, ESI-MS itself can also be used for perchlorate

confirmation. Following ion-pair extraction, quantitative determination of

perchlorate in water samples and commercial fertilizers using flow injection ESI-

MS has been reported (Urbansky et al., 1999; Magnuson et al., 2000b;

Magnuson et al., 2000a; Urbansky et al., 2000a; Collette et al., 2003). ESI-

FAIMS (high-field asymmetric waveform ion mobility) -MS with flow injection was

also used for perchlorate determination in water and human urine (Ells et al.,

2000; Handy et al., 2000).

To enhance the sensitivity and selectivity for measuring perchlorate by

MS, some specialized techniques involving improved sample preparation and/or

instrumentation have been reported. For instance, an online pre-concentration

and pre-elution (PC-PE) technique, reported by (Tian et al., 2003), has been

applied to the analysis of milk by IC-MS (Kirk et al., 2003; Kirk et al., 2005). Gas

phase ion association with a long chain dipositive cationic agent was applied in

IC-ESI-MS to increase selectivity and sensitivity of the perchlorate measurement

(Martinelango et al., 2005). An isotopically labeled internal standard has also

been used to correct ionization suppression (Krynitsky et al., 2004; Li and

George, 2005; Valentin-Blasini et al., 2005).

15

Due to the widespread occurrence of perchlorate contamination in the

environment and the potential hazards of perchlorate exposure and effect in

animals and humans, perchlorate contamination monitoring is important and

critical for environmental and human risk assessment. Perchlorate residues in

biological fluids such as urine, milk, and plasma can serve as non-lethal

biomarkers for monitoring environmental contamination of perchlorate, especially

since the anion is excreted via urine and milk in animals and humans (Anbar et

al., 1959; Goldman and Stanbury, 1973; Peña et al., 1976; Batjoens et al., 1993;

Greer et al., 2002; Yu et al., 2002; Kirk et al., 2003; Capuco et al., 2005; Kirk,

2005; Kirk et al., 2005; Valentin-Blasini et al., 2005), and blood delivers

perchlorate to the target tissues. Therefore, it is desirable to monitor perchlorate

residues in these biological fluids and thus, assess its risk to the environment,

animals, and humans.

In some instances, it may be more preferable to develop a good

preparation method prior to conventional IC analysis because of the high cost to

setup a MS system and maintain its normal performance. However, information

on preparation methods, especially cleanup methods, is limited. Since the ability

to detect perchlorate in exposed animals or humans is critical to effects studies

and risk assessments, a well-developed preparation method would contribute

greatly to the more accurate detection of trace perchlorate using conventional IC.

Cleanup methods were developed for blood plasma and urine in order to

eliminate interferences for perchlorate determination using ion chromatography.

Perchlorate determination in complex matrices, including urine, milk, and plasma,

using an LC-ESI-MS system was also explored. The characteristics and

efficiency of IC-SCD and LC-ESI-MS for the determination of perchlorate in these

types of samples were also compared. An IC-MS/MS method was also explored

and applied to urine analysis for perchlorate.

16

2.2 Cleanup Methods for Perchlorate Determination in Plasma and Urine†

† Part of this work has been published in the journal of Talanta (Q. Cheng, F. Liu, J.E. Cañas,

T.A. Anderson, Talanta. 2006 68:1457-1462).

2.2.1 Materials and Methods

Chemicals

A custom perchlorate (ClO4-) standard solution was obtained from

AccuStandard, Inc. (New Haven, CT). Sodium hydroxide (50%, w/w) aqueous

solution and ethanol (pesticide grade) were purchased from Fisher Scientific

(Fair Lawn, NJ). Acetonitrile (HPLC grade) was from EMD (Gibbstown, NJ). All

solutions were prepared in 18.2 MΩ Milli-Q water.

Sample source and treatment

Plasma was collected from heifer calves inhabiting a reference site near

McGregor, McLennan County, Texas during spring 2003. The reference site was

a pasture that was a sufficient distance from a perchlorate manufacture and

usage facility such that perchlorate was not present in available on-site surface

or groundwater utilized by the cattle, nor was it present in vegetation. Plasma

samples were processed using methods similar to those previously described

(Fisher et al., 2000; Anderson and Wu, 2002; Narayanan et al., 2003). First, 1

mL of plasma was precipitated with 4 mL of ethanol (ice-cold) and then

centrifuged (4 °C) at 3750 rpm for 5 min. The supernatant was removed,

evaporated to dryness under nitrogen, and reconstituted in 5 mL Milli-Q water.

Samples were then cleaned using alumina and C18 solid phase extraction (SPE)

cartridges, and filtered (0.45 μm) prior to ion chromatography (IC) analysis.

Perchlorate standard was spiked into the plasma samples at final concentrations

of 2.5, 5, 10, 50, and 100 ng/mL to determine the recovery and precision of the

cleanup method.

Urine samples used in the current study were from prairie voles, cattle,

and humans (female). Cow urine was provided by the Department of Animal and

17

Food Science, Texas Tech University (Lubbock, TX). Vole urine was obtained

from a breeding colony housed at Texas Tech University. Human urine was

obtained from volunteers in Lubbock, TX. Perchlorate was not detected in the

cattle and human urine. However, trace perchlorate was detected in vole urine.

In order to determine the efficiency of different cleanup methods for urine,

a perchlorate standard solution was spiked into cattle urine at a final

concentration of 100 µg/L. The samples were analyzed for perchlorate using ion

chromatography after preparation by different cleanup methods, and the

efficiency and recovery of perchlorate in the samples was determined. Based on

these preliminary results, the most efficient method was chosen for further

evaluation of its applicability to different urine sources (vole and human) with

spiked perchlorate (2.5, 10, and 100 µg/L). In addition, urine samples collected

from cows inhabiting a perchlorate-contaminated site were used to evaluate the

applicability of the cleanup method for field samples.

Sample preparation procedure for urine cleanup

Solid phase extraction (SPE) cartridges were used in the cleanup process

of urine for ion chromatography analysis. Ten types of SPE cartridges were

evaluated, individually or in combination, to determine cleanup efficiency. The

SPE cartridges tested included quaternary amine (CUQAX 100 mg), quaternary

amine with hydroxide (CHQAX 100 mg), quaternary amine-acetate (CAQAX 100

mg), strong anion exchange (Strata SAX 100 mg), N-2 aminoethyl (PSA 500 mg)

combined with alumina-neutral (Al-N 1 g), octadecyl (C18 1 g) combined with

PSA (500 mg), hydrophobic and aminopropyl (NAX 1 g), NAX combined with Al-

N, NAX combined with PSA, and C18 combined with Al-N. CUQAX, CHQAX,

CAQAX, PSA, and NAX were obtained from United Chemical Technologies, Inc.

(Bristol, PA). Strata SAX was purchased from Phenomenex (Torrance, CA).

C18 cartridges were purchased from Honeywell B&J (Muskegon, MI), and Al-N

was purchased from J.T. Baker (Phillipsburg, NJ).

18

Depending on the sorbent, SPE cartridges were conditioned as

appropriate prior to use. For CUQAX, CHQAX, and CAQAX, 0.4 mL of urine was

loaded, 1 mL Milli-Q water (>18MΩ) was used to elute the sample through the

cartridge, and the eluate was diluted to a final volume of 2 mL with Milli-Q water.

For SAX, 0.5 mL of urine was loaded, then 2.5 mL NaOH (20 mM in 15%

acetonitrile solution) was added to elute the sample through the cartridge

following consecutive washing with 1 mL Milli-Q water and 1.5 mL NaOH (20 mM

in 15% acetonitrile solution). For NAX, following the loading of 0.8 mL urine and

washing with 0.6 mL DI water, 4 mL Milli-Q water was used to elute the cartridge.

For the combination cartridges with NAX and Al-N or NAX and PSA, the 4 mL

Milli-Q water eluted from NAX was further processed through Al-N or PSA. In the

case of C18 in combination with PSA or Al-N, 0.8 mL urine was loaded onto the

cartridge, followed by 4 mL Milli-Q water to elute the cartridge. The eluate then

was loaded and eluted through PSA or Al-N cartridges. In the case of PSA

combined with Al-N, 0.8 mL urine was first diluted to 4 mL with Milli-Q water and

then eluted through the PSA cartridge, followed by an Al-N cartridge. All final

eluates were filtered (0.45 µm) and analyzed by ion chromatography without

further dilution.

Sample analysis

A method similar to EPA Method 314.0 (Hautman et al., 1999) was

followed to determine perchlorate in all samples. The analysis was performed on

a Dionex DX-500 Ion Chromatography System equipped with a GP50 gradient

pump, a CD20 conductivity detector, and an AS40 automated sampler (Dionex

Corp.). PeakNet® chromatography software was used to control the system. Ion

separation was conducted with a Dionex IonPac AS16 (250 mm x 4.0 mm i.d.)

analytical column after a Dionex guard column (AG16). Conditions for the

system were as follows: flow rate = 1.0 mL/min; eluent = 50 mM sodium

hydroxide; injection volume = 1000 µL. Ion detection was by suppressed

19

conductivity in the external water mode. Computer-generated peak areas were

used to measure sample concentrations in an external standard mode.

2.2.2 Results and Discussion

Recovery and precision of alumina-neutral plus C18 as a cleanup method for

plasma

Several different sorbent materials and solid phase extraction (SPE)

cartridges were tested as cleanup methods before IC analysis. An alumina-

neutral (Al-N) cartridge combined with a C18 cartridge provided the best cleanup

for plasma in terms of reduced background conductivity and interference (Figure

2.1). Table 2.1 shows perchlorate recoveries of samples spiked with 2.5 to 100

μg/L. The total average recovery was 84% ± 2.3 (mean ± SD). In addition, the

detection limit for perchlorate in blood plasma (ratio of signal and noise (S/N) = 3)

was estimated as 2.74 μg/L after 5X dilution.

Efficiency of various cleanup procedures for urine

Among the tested SPE cartridges as individuals or in combination, NAX

and its combination with other cartridges reduced the background conductivity

and interference dramatically (Figure 2.2). For a 5X-diluted urine sample,

cleanup by NAX in combination with PSA or Al-N cartridges resulted in very low

background conductivity and much less interference compared with other

cleanup procedures. As illustrated in Figure 2.2, cleanup with NAX combined

with Al-N gave the best result in terms of reducing interference and background

signal. Furthermore, cleanup by NAX, NAX plus PSA, and NAX plus Al-N

cartridges showed much higher perchlorate recoveries among all tested cleanup

procedures (Table 2.2). The sorbent in NAX cartridges is composed of a silica

backbone with an anion exchanger (aminopropyl) and a hydrophobic carbon

chain (C8). When a urine sample, which contains abundant positively charged

ions, is applied to the NAX cartridge, the positively charged ions are not retained

20

by the anion exchanger (the amine groups) and are eluted, whereas the

negatively charged ions can interact with the amine groups. Hydrophobic

molecules in the urine, such as organic-based compounds (i.e., carbohydrates,

proteins, etc.), bind to the C8 phase. Therefore, the negatively charged and

hydrophobic compounds can be held in the cartridge after application of urine.

Because amine groups are weak anion exchangers, perchlorate anions do not

strongly interact with them. Thus, perchlorate anions can be eluted by water

following the application of urine while hydrophobic compounds and other

stronger anions are left in the cartridge through interaction with C8 and amine

groups, respectively. However, cleanup with the NAX cartridge alone gave a

relatively high background signal due to the high total conductivity. The

combination of NAX with PSA and particularly with Al-N showed significant

improvement (Figure 2.2). Work in our lab has shown that Al-N was effective in

reducing background and interference in a variety of other matrices (unpublished

data). Here, we also found this type of cartridge works efficiently for urine

cleanup as well when combined with NAX. Although the combination of C18 and

PSA has a similar functional composition as NAX, this combination did not show

similar results as NAX in terms of background signal and perchlorate recovery:

two functional groups in one cartridge work more effectively to reduce

background and maintain high recovery than the combination of the two

functional groups in two separate cartridges.

CAQAX, CHQAX, CUQAX (i.e., QAXs), and SAX cartridges contain

quaternary amine anion exchangers. In the current study, the way we used them

as cleanup cartridges for perchlorate analysis is based on a preelution principle:

loading the sample on the cartridge, washing the cartridge with water or other

solution, and then eluting perchlorate from the cartridge with a proper solution.

These cartridges were not as effective as either NAX alone or in combination

with Al-N or PSA in reducing background signal and interferences (Figure 2.2).

In addition, they gave poor perchlorate recovery (Table 2.2). The QAX-type

21

cartridges do not bind perchlorate strongly, and thus perchlorate may be eluted

by the washing solution. Therefore, we skipped the washing step, and directly

eluted perchlorate with water without washing after loading the sample on the

cartridges. However, co-eluting ions in the eluate and high background signal

interfered with perchlorate determination by ion chromatography. Compared to

QAX, SAX is a much stronger anion exchanger. However, the anion exchange

capacity appears to vary depending on the vendor. In the current study, we

found that perchlorate can be eluted from SAX cartridges from Phenomenex

(Torrance, CA), but not from another provider/brand by using 20 mM (or higher)

NaOH as an eluent. Because of the relatively small mass of the sorbent bed in

the cartridge (100 mg), it was not easy to separate perchlorate from other

interferences. As a result, high background conductivity and interferences were

observed. It was difficult to elute perchlorate if larger SAX cartridges with more

sorbent bed were used. Furthermore, perchlorate recovery was low when using

SAX cartridges. SAX is a strong anion exchanger, which holds perchlorate

tightly, causing difficulty in eluting perchlorate using water. Even with 20 mM or

higher NaOH (a stronger eluent than water), perchlorate could not be eluted with

a limited amount of eluent, and therefore, low recovery of perchlorate was

observed. In addition, poor reproducibility was observed using this cleanup

method. This may be caused by inconsistent elution rates. Therefore,

controlling the flow rate of eluent at a constant value would probably improve the

reproducibility of the SAX cartridge in cleanup of urine.

The combinations of C18 with Al-N and PSA with Al-N produced high

perchlorate recoveries similar to NAX in combination with PSA or Al-N (Table

2.2), but they were less effective in reducing background conductivity and

interferences. Cleanup efficiency for these four types of cleanup procedures for

both blank and spiked urine are presented in Figure 2.3. For the combination of

either C18 with Al-N or PSA with Al-N, the perchlorate peak (peak A in Figure

2.3a) was very close to an interference peak (peak B in Figure 2.3b), leading to

22

the appearance of a shoulder-peak near the retention time for perchlorate

(retention times less than 0.5 min difference). Consequently, it would be easy to

mistakenly identify the interference as perchlorate in samples which contain no

perchlorate (false positive). In contrast, no interference peak was observed in

samples processed by a combination of NAX with Al-N or with PSA. The

cleanup of urine using NAX combined with Al-N or PSA produced high recovery,

low background conductivity, and no adjacent interference peak.

Accuracy and precision of NAX plus Al-N for cleanup of different sources of urine

Since NAX combined with Al-N proved to be the most efficient for cleanup

of cow urine for the determination of perchlorate by ion chromatography, we

further evaluated the accuracy and precision of this cleanup procedure for

different types of urine samples spiked with perchlorate (Table 2.3). Perchlorate

recoveries (± SD) were 67% ± 2.51, 77% ± 3.56, and 81% ± 1.66 for cow urine

spiked with perchlorate at 2.5, 10, and 100 μg/L, respectively. The highest

perchlorate recovery was found for vole urine, with 97% ± 4.13, 83% ± 5.13, and

86% ± 1.62 in the samples spiked with 2.5, 10, and 100 μg/L perchlorate,

respectively. Recovery was lowest in human urine, with 76% ± 0.82, 46% ± 0.96,

and 56% ± 3.40, respectively. Since Al-N (without conditioning) does not adsorb

perchlorate, the reduced perchlorate recovery in human urine samples is

probably caused by the NAX cartridge. We tested the effect of different elution

volumes of Milli-Q water on perchlorate recovery, and found that perchlorate was

eluted primarily within the first 4 mL of water, with 92% and 63% recovery of

spiked perchlorate (100 μg/L) in cow and human urine, respectively.

The pH profile of different urine samples (Table 2.4) may cause differential

perchlorate recovery. The pH of human and cow urine was 7.10 and 9.05,

respectively. The pH may alter perchlorate elution from NAX by affecting the

amine groups. Perchlorate recovery was increased dramatically if 20 mM NaOH

was used as an eluent, but with interferences. In addition, conductivity of urine

23

samples (Table 2.4) may also have an influence on cleanup using NAX. Human

urine (with higher conductivity) may have additional ions which could compete

with perchlorate interaction with the amine groups and cause a wide disperse

pattern of perchlorate during wash and elution processes: some perchlorate may

be lost in the wash step, some may still interact with the amine groups even after

4 mL of water elution. Consequently, unsatisfactory recovery was observed.

Considering the recoveries and reduction of background signal, the limit of

detection (LOD) for perchlorate in cow, vole, and human urine samples was 12.6,

12.3, 18.7 μg/L, respectively (based on S/N = 3).

Application of cleanup method (NAX plus Al-N) to urine samples from field and

laboratory studies

It is well known that perchlorate is excreted primarily via urine (Anbar et

al., 1959; Goldman and Stanbury, 1973; Peña et al., 1976; Batjoens et al., 1993;

Yu et al., 2002; Valentin-Blasini et al., 2005); therefore, monitoring perchlorate

residues in urine should be a sensitive biomarker for perchlorate exposure. The

current study provided a promising cleanup method for perchlorate determination

using ion chromatography in a variety of urine types, including cattle, vole, and

human urine. In addition, the application of this cleanup method provides for the

detection of perchlorate in urine as low as 12.6 μg/L. To our knowledge, this is

the lowest reported detection limit of perchlorate in urine or a similar biological

matrix using conventional ion chromatography.

We attempted to use this cleanup method to evaluate perchlorate in urine

samples from the field. Urine (n = 4) was collected from cattle inhabiting two

different pastures where perchlorate was detected in ponds which serve as

drinking water for the cattle at concentrations as high as 100 μg/L. Cattle on

these pastures were not restricted to water supplies containing perchlorate; most

of the ponds did not contain perchlorate and there was some anecdotal evidence

that the cattle were avoiding the perchlorate-contaminated water. Using the

24

cleanup method described, we did not detect perchlorate in any of the urine

samples, which was consistent with analysis results of the corresponding plasma

samples in which no perchlorate was detected (data not shown here and

unpublished). Confirmation analysis using LC-MS revealed that one urine

sample contained perchlorate (concentration = 3.45 μg/L after 8X dilution).

Perchlorate was detected in all 4 samples using IC-MS/MS (the minimal report

detection limit is 10 ng/L) but concentrations in three samples are lower than the

detection limit of LC-MS (2.28 ng/mL, S/N = 3) after 8X dilution, and therefore not

detected using the LC-MS. The other urine sample with detectable perchlorate

using LC-MS was very dirty with many interferences and high background

conductivity even after cleanup; thus perchlorate was not detected by

conventional IC.

Compared to the cow urine provided by the Department of Animal and

Food Science, Texas Tech University, used in the laboratory portion of this study,

urine collected from the field was much darker in color. The average pH of these

cow urine samples was 7.86 ± 0.11 (± SD), and the conductivity ranged from

11.74 mS to 68.68 mS. These field urine samples had much higher background

and poor recovery if 0.8 mL sample was loaded onto the NAX and eluted with 4

mL Milli-Q water. These more complex urine samples appeared to exceed the

capacity of NAX, resulting in only partial interaction of sample with the NAX.

Therefore, we had to revise the procedure slightly (only 0.4 mL or less sample

was loaded onto NAX and eluted with 4 mL Milli-Q water or 5 mM NaOH after

washing with 0.6 mL Milli-Q water). Nonetheless, perchlorate was not detected

in these field animal urine samples by conventional ion chromatography.

We also applied this cleanup method to deer mouse urine from a

laboratory study, but the result was not satisfactory. The urine was concentrated,

and mixed with some feces because of limited separation ability of the metabolic

cages. An interference that may have come from food (rodent chow) confounded

perchlorate determination using conventional IC. Therefore, an online

25

preconcentration/preelution method described in Tian et al (2003; 2005) was

used to analyze this urine matrix. Various types of cartridges and sorbents were

used before IC analysis to get rid of the interferences, and NAX plus DD6

alumina (Alcoa, Pittsburgh, PA) plus Al-N produced the best result regarding

interference elimination. Urine (0.4 mL) was loaded onto a preconditioned NAX,

eluted with 5 mL of Milli-Q water after washing with 1 mL Milli-Q water, 2 g

granular DD6 was added into the eluate and allowed sit at room temperature for

36 h, and then the eluate was passed through Al-N. The final volume was 4 mL

in order to maintain a 10X dilution. Perchlorate recovery was 68% ± 10.6 and

83% ± 22.1 for 20 and 100 μg/L spiked perchlorate samples, respectively.

However, recovery was not good (below 50 %) for low concentrations of

perchlorate (i.e. below 10 μg/L).

To monitor perchlorate exposure in wild animals or humans, two

categories of endpoints are widely used: biomarkers of exposure and biomarkers

of effect. Perchlorate determination in blood or tissue matrices are examples of

the former (Smith et al., 2001; Cheng et al., 2004; Smith et al., 2004a); the latter

includes iodide uptake, thyroid hormone status/profile, and thyroid histopathology

(Lawrence et al., 2000; Greer et al., 2002; Yu et al., 2002; Merrill et al., 2003;

Clewell et al., 2004; Merrill et al., 2005). However, it appears that these

biomarkers are useful primarily in cases of high perchlorate

contamination/exposure. At environmentally relevant exposures, these

biomarkers are not as effective (Li et al., 2000a; Li et al., 2000b; Li et al., 2001;

Cheng et al., 2004; Smith et al., 2005). For biomarkers of exposure, urine

analysis may be more sensitive than blood residue analysis since urine is a

primary excretion pathway for perchlorate elimination by animals. Therefore, an

efficient cleanup method for urine analysis for perchlorate is useful and should

contribute to environmental monitoring of perchlorate contamination. However,

urine is a challenging matrix for perchlorate determination using conventional IC,

and confirmatory testing is mandatory even with an extremely efficient cleanup

26

method. Consequently, using MS to determine perchlorate in these matrices has

become more prevalent, as indicated by a recent report of successful perchlorate

determination in human urine (Valentin-Blasini et al., 2005).

2.2.3 Conclusions

The presence of ions and other biomolecules in matrices like plasma and

urine usually confounds accurate determination of perchlorate by ion

chromatography; therefore, efficient cleanup before IC analysis is necessary for

perchlorate determination in these matrices. Several different cleanup methods

of plasma and urine were tested and compared, in terms of reduced background

conductivity, interference, perchlorate recovery, and precision. For plasma

samples, alumina-neutral cartridges combined with C18 cartridges provided the

best cleanup with an average perchlorate recovery of 84% ± 2.3 (mean ± SD,

spiked concentration ranging from 2.5 to 100 ng/mL), and the detection limit

(based on S/N = 3) was estimated to be 2.74 μg/L after 5X dilution. For urine

samples, NAX combined with alumina-neutral showed the best effect with

significantly reduced background signal of urine and relatively high recovery of

perchlorate. In cow urine samples spiked with perchlorate at 2.5, 10, and 100

μg/L, perchlorate recoveries were 67% ± 2.51, 77% ± 3.56, and 81% ± 1.66

(mean ± SD), respectively. In addition, the detection limit was as low as 12.6,

12.3, and 18.7 μg/L in cow, vole, and human urine samples, respectively.

However, this cleanup method is not robust enough to be used on some dirty

urine matrices such as deer mice urine collected in perchlorate excretion and

depuration studies (Chapter 4).

27

0

0.5

1

6 8 10 12 14 16Time (min)

cond

uctiv

ity (u

S/cm

)

w ater

plasma

Figure 2.1. Chromatograms of water contaminated with perchlorate and plasma

spiked with 100 ng/mL perchlorate using ion chromatography with suppressed conductivity detection (IC-SCD). Plasma samples were cleaned with alumina-neutral and C18 SPE cartridges, producing low background conductivity and less interferences. Perchlorate retention time was around 14.5 min.

Table 2.1. Accuracy and precision of perchlorate determination in cow plasma

using ion chromatography with suppressed conductivity detection (IC-SCD). Plasma was cleaned with Alumina-N and C18 SPE cartridges.

Sample matrices

Replicates

Spiked perchlorate

concentration (μg/L)

Perchlorate concentration

detected (μg/L) ± SD

CV (%)

Recovery (%) ± SD

2 2.5 2.6 ± 0.01 0.5 105 ± 0.5

2 5 4.1 ± 0.21 5.1 82 ± 4.2

2 10 9.0 ± 0.04 0.5 90 ± 0.5

2 50 40.3 ± 1.48 3.7 81 ± 3.0

Cow

plasma

2 100 87.4 ± 0.52 0.6 87 ± 0.5

28

0

1

2

3

4

5

7 8 9 10 11 12 13 14 15 16 17Time (min)

uS

CAQAXCHQAXCUQAXC18 AlNSAXC18 PSAPSA AlNNAXNAX PSA NAX AlN

Figure 2.2. Ion chromatography analysis of cow urine samples (with no

perchlorate) after being processed via various cleanup procedures. Different cartridges or combinations of cartridges were tested for cleanup efficiency in terms of reduced background signal and interferences.

Table 2.2. Recoveries of perchlorate in perchlorate-spiked cow urine (100 µg/L) cleaned using various solid phase extraction (SPE) cartridges.

Phase % Recovery (mean ± SD) Phase % Recovery (mean ± SD)

NAX 74 ± 7.5 PSA Al-N 70 ± 1.9

NAX Al-N 70 ± 6.0 CUQAX 29 ± 2.8

NAX PSA 73 ± 3.9 CHQAX 25 ± 5.5

C18 Al-N 75 ± 1.1 CAQAX 24 ± 3.6

C18 PSA 43 ± 3.4 SAX 30 ± 11.7

29

0

1

2

3

4

5

7 8 9 10 11 12 13 14 15 16 17Time (min)

uS

C18 AlN S100

PSA AlN S100

NAX PSA S100

NAX AlN S100

0

1

2

3

4

5

7 8 9 10 11 12 13 14 15 16 17Time (min)

uS

C18 AlN

PSA AlN

NAX PSA

NAX AlN

a

a

b

(A)

b

b

(B)

Figure 2.3. Efficiencies of four cleanup procedures (NAX combined with Al-N, NAX with PSA, C18 with Al-N, and PSA with Al-N) for perchlorate determination in urine spiked with perchlorate (100 µg/L) (A) and without perchlorate (B). Peak a in (A) is perchlorate. When samples are cleaned with combination of C18 and Al-N or PSA and Al-N there is a shoulder peak (Peak b) adjacent to peak a. The adjacent peak b is an interference peak more clearly shown in (B).

30

Table 2.3. Accuracy and precision for NAX combined with Al-N as cleanup cartridges for perchlorate determination in different types of urine using ion chromatography.

% Recovery (mean ± SD) Perchlorate spike

(ng/mL) Cow Vole Human

2.5 67 ± 2.5 97 ± 4.1 76 ± 0.8

10 77 ± 3.6 83 ± 5.1 46 ± 1.0

100 81 ± 1.7 86 ± 1.6 56 ± 3.4

Table 2.4. Physical properties (pH and conductivity) of the different sources of urine.

Property Cow Vole Human

pH 9.05 8.37 7.10

Conductivity (mS) 15.39 19.29 23.04

31

2.3 Perchlorate Determination in Urine, Milk, and Plasma Using Liquid

Chromatography Electrospray Ionization Mass Spectrometry (LC-ESI-MS) and

Comparison with Ion Chromatography with Suppressed Conductivity Detection

(IC-SCD)

2.3.1 Materials and Methods

Reagents and standards

A custom perchlorate (ClO4-) standard solution was purchased

AccuStandard, Inc. (New Haven, CT). Sodium hydroxide (50%, w/w) aqueous

solution and acetonitrile (HPLC grade) were purchased from Fisher Scientific

(Fair Lawn, NJ). Ammonium acetate (99%) was obtained from J.T. Baker

(Phillipsburg, NJ). Ammonium hydroxide, 28.0-30.0% as NH3, was purchased

from Sigma-Aldrich (St. Louis, MO). All solutions were prepared in 18.2 MΩ Milli-

Q water.

Sample sources and preparation

Urine was collected from sheep raised at the Texas Tech University

Agriculture Research Farm. Urine samples were prepared using NAX2 SPE

cartridges (United Chemical Technologies, Inc. Bristol, PA). After 0.3 mL of urine

was loaded onto the preconditioned cartridge and washed with 1.5 mL of DI

water, 3 mL Milli-Q water was used to elute the cartridge, and then the 3 mL

eluate was processed through an alumina-neutral cartridge (J. T. Baker,

Phillipsburg, NJ). Several prepared samples were pooled, divided into 7 aliquots,

and spiked with perchlorate to obtain final perchlorate concentrations of 0, ~ 2, ~

5, ~ 10, ~ 50, ~ 100, and ~ 200 ng/mL.

Dairy milk (1% fat) and soymilk were purchased from a local supermarket,

and human breast milk was provided by healthy lactating mothers residing in

Lubbock, TX or nearby counties that was collected by the Department of Human

Development and Family Studies, Texas Tech University (Lubbock, TX). Milk

32

samples (48 mL) were precipitated using acetonitrile (96 mL), and centrifuged at

3750 rpm for 10 min (Beckman Allegra 6R Centrifuge, USA). The supernatant

was evaporated to 24 mL under nitrogen gas, and then passed through

preconditioned C18 SPE cartridges with 2 mL per cartridge (Fisher Science,

USA). The eluate was collected, and each cartridge was then washed with 5 mL

of Milli-Q water which was combined with the eluate. The pooled eluate was

evaporated to a final volume of 48 mL, and then cleaned using alumina-neutral

cartridges (4 mL per cartridge). The eluate was collected and divided into nine

aliquots, and then spiked with perchlorate to achieve final concentrations of 0, ~

2, ~ 5, ~ 10, ~ 50, ~ 100, ~ 200, ~ 500, and ~ 1000 ng/mL. Because of

inadequate sample volume, only 25 mL of human breast milk were used to

prepare the spiked samples with final concentrations of 0, ~ 5, ~ 10, and ~ 100

ng/mL.

Plasma was collected from cows inhabiting a perchlorate-contaminated

site in Kansas. These cows were not restricted to the contaminated water

source, and perchlorate was not detected in the plasma samples used in this

study. The plasma sample (8 mL) was precipitated with acetonitrile (16 mL) and

centrifuged at 3750 rpm for 10 min. The supernatant was evaporated to 8 mL,

and then passed through three preconditioned C18 cartridges (i.e., about 3 mL

per cartridge). The eluate was collected and each cartridge was then washed

with 5 mL of Milli-Q water which was combined with the eluate. The pooled

eluate was diluted to a final volume of 40 mL with Milli-Q water. After cleanup

through aluminum-neutral cartridges (4 mL per cartridge), all eluates were pooled

and divided into seven aliquots, and spiked to obtain final perchlorate

concentrations of 0, ~ 2, ~ 5, ~ 10, ~ 50, ~ 100, and ~ 200 ng/mL.

Prepared samples were then separated into two aliquots, one for ion

chromatography analysis with suppressed conductivity detection (IC-SCD) and

the other for liquid chromatography electrospray ionization mass spectrometry

(LC-ESI-MS). Because perchlorate could not be separated from the

33

interferences using IC-SCD, sheep urine samples were only analyzed by LC-ESI-

MS.

IC-SCD analysis

A method similar to EPA Method 314.0 (Hautman et al., 1999) was

followed to determine perchlorate in all samples. The analysis was performed on

Dionex DX-500 Ion Chromatography System equipped with a GP50 gradient

pump, a CD20 conductivity detector, and an AS40 automated sampler (Dionex

Corp., Sunnyvale, CA). PeakNet® chromatography software was used to control

the system. Ion separation was conducted with a Dionex IonPac AS16 (250 mm

x 4.0 mm i.d.) analytical and AG16 (50 mm x 4.0 mm i.d.) guard columns.

Conditions for the system were as follows: flow rate = 1.0 mL/min; eluent = 50

mM sodium hydroxide; injection volume = 1000 µL. Ion detection was by

suppressed conductivity in the external water mode. Computer-generated peak

areas were used to measure sample concentrations in an external standard

mode.

LC-ESI-MS analysis

Perchlorate determination by mass spectrometry was conducted by LC-

MS (LCQ Advantage, Thermo-Finnigan Inc., San Jose, CA, USA) operated in the

electrospray ionization mode (ESI). An IC-Pak Anion HR analytical column (75

mm × 4.6 mm i.d) from Waters (Milford, MA) was eluted with 20 mM ammonium

acetate in 55:45 (v/v) acetonitrile/water (pH = 10 with ammonium hydroxide) at

500 μL/min. An injection loop volume of 25 μL was used for MS analysis.

Selected ion monitoring (SIM) mode was used to monitor ClO4- at m/z 99.0 ± 0.5

and 101.0 ± 0.5 corresponding to 35ClO4- and 37 ClO4

-, respectively. A diversion

valve was used during each run to direct flow to waste for the first 10.0 min, after

which the eluent was directed into the electrospray interface for 6.0 min.

Conditions for negative ion ESI - MS were as follows: 0.5 kV ion spray voltage, -4

34

V capillary voltage, and 150 ºC capillary temperature. Other parameters such as

sheath gas flow rate and aux/sweep gas flow rate were optimized with tune.

Peak area produced at m/z 99.0 ± 0.5 was used for quantitative analysis. The

isotopic ratio of 35ClO4- and 37ClO4

- ( m/z 99.0 ± 0.5 and 101.0 ± 0.5) at the

retention time within ± 50% of 3.08 (i.e., 1.5 to 4.6) was used for perchlorate

confirmation.

2.3.2 Results and Discussion

Linearity of response and detection limit for LC-ESI-MS

For 25 μL injections, the MS response to perchlorate was found to be

linear over the spiked concentration range (2, 5, 10, 50, 100, and 200 ng/mL) in

water, sheep urine, and dairy milk. Good linearity for perchlorate was found over

the concentration range from 2.0 to100.0 ng/mL in spiked cow plasma samples.

Milli-Q water: A (counts) = (0.93 ± 0.06) × 105 [ClO4-, ng/mL] + (4.27 ± 0.68) ×

105, r2 = 0.9948, n = 6…(1)

Sheep urine: A (counts) = (1.65 ± 0.09) × 105 [ClO4-, ng/mL] – (2.69 ± 4.67) ×

105, r2 = 0.9926, n = 5…(2)

Dairy milk: A (counts) = (1.09 ± 0.14) × 105 [ClO4-, ng/mL] + (6.72 ± 5.52) × 105,

r2 = 0.9971, n = 5…(3)

Cow plasma: A (counts) = (1.13 ± 0.04) × 105 [ClO4-, ng/mL] + (9.74 ± 1.71) ×

105, r2 = 0.9977, n = 5…(4)

In the current study, no statistical difference was observed in the MS

responses to perchlorate in dairy milk and Milli-Q water samples over the

concentration range from 2.0 to 200.0 ng/mL in terms of slopes of the linear

range (p = 0.058. t-test), but it was found between cow plasma and Milli-Q water,

sheep urine and Milli-Q water (p < 0.001) (Figure 2.4). Much higher MS

35

responses to perchlorate were found in sheep urine than in Milli-Q water,

particularly in the samples spiked with more perchlorate.

Both the limit of detection (LOD) based on S/N=3 and the method

detection limit (MDL) based on the definition in EPA Method 314.0 were

estimated (Table 2.5). In water, a MDL as low as 50 ng/L (Winkler et al., 2004),

and LOD (S/N = 3) as low as 25 ng/L with an ion association technique

(Martinelango et al., 2005) have been reported. In the current study, the LOD

(S/N = 3) was 2.49 ng/mL in a simple matrix such as Milli-Q water. The huge

difference regarding method sensitivity may be due to the limitation of our

instrument. Most mass spectrometers used in previous reports, in which a much

lower MDL or LOD for perchlorate was reported, used a quadrupole mass filter

as the mass analyzer, while a octapole ion trap was used as the mass analyzer

in our instrument. To our knowledge, this type of mass analyzer has not been

previously used to determine perchlorate. Ion traps are not as effective as

quadrupole mass filters in trapping low m/z ions. Additionally, because of mass

discrimination, low m/z ions are usually not transferred by ESI as efficiently as

higher m/z ions, and background noise also tends to be higher for low m/z ions

(Kempen and Brodbelt, 2000; Martinelango et al., 2005). In addition, high mass

molecules have been reported to suppress the signal of smaller molecules

(Sterner et al., 2000; Annesley, 2003). Ion-pair (or association) techniques were

therefore developed by some groups to improve sensitivity and selectivity of

perchlorate determination by MS (Urbansky et al., 1999; Magnuson et al., 2000a;

Martinelango et al., 2005). The optimum m/z range in the MS used in this study

was designed to be 200 - 2000. Therefore, since the perchlorate ion was

monitored at m/z of 99 and 101, which is outside the optimal range, the MDL and

LOD using this instrument in the current study were higher than those reported in

other studies.

Method accuracy and precision were evaluated in sheep urine, dairy milk,

and cow plasma by spiking perchlorate into these matrices at different levels and

36

evaluating the recovery (Table 2.6). Because of the difference in MS response

for perchlorate in different matrices, perchlorate recovery in sheep urine and cow

plasma matrices was calculated based on the calibration curve of its respective

matrix, and the recovery in dairy milk was based on the calibration curve of Milli-

Q water. Perchlorate recovery was 80 – 120% in the spiked sheep urine and

cow plasma matrices, while 75 – 140% recoveries were observed in the spiked

dairy milk samples. Good precision was revealed by a lower coefficient of

variation (CV % ≤ 4.88%) in a single-day experiment of perchlorate determination

in these types of matrices.

Comparison of LC-ESI-MS with IC-SCD

Interfering components, such as additional ions, proteins, lipids, sugars,

and other biomolecules in biological fluids, produced high background

conductivity and co-elution were routine phenomena encountered in perchlorate

determination by IC-SCD. As shown in Figures 2.5, 2.6, and 2.7, retention time

of interfering components in the samples was close (or the same) to that of

perchlorate during IC-SCD analysis, causing uncertainty for perchlorate

determination in the samples. In contrast, with the mass spectrometry method,

only perchlorate was eluted at the retention time using m/z 99.0 ± 0.5 SIM. As

seen from the chromatograms in Figures 2.5, 2.6, and 2.7, representing MS

chromatograms of the same sample with m/z 99.0 ± 0.5 SIM detection, no high

background and co-elution of interferences occurred. The matrices under study

did not show a difference in LC-ESI-MS in terms of baseline and interferences,

but a pronounced difference was observed using IC-SCD, unless effective

preparation and cleanup methods were applied to these matrices. On the other

hand, without information about the background difference in mass spectrometry,

dirty samples with high background would be analyzed by LC-ESI-MS, and as a

result, the separation column would be more easily impacted. In this cascade, a

37

guard column is necessary and important to protect the separation column from

potential damage caused by dirty samples.

Because of high background conductivity and the existence of

interferences in IC-SCD analysis, perchlorate LODs (S/N = 3) of the dairy milk

and cow plasma were not able to be calculated, and because perchlorate could

not be separated from the interferences, sheep urine matrix was not analyzed for

LOD determination using IC-SCD (Figure 2.7). Based on previous studies, LODs

(based on S/N = 3) of cow plasma and urine samples were 2.74 and 2.52 ng/mL,

respectively, in 5X diluted matrices using IC-SCD (Cheng et al., 2004; Cheng et

al., 2006). These reported LODs approximate the LOD by LC-ESI-MS in the

current study (Table 2.5). With extremely effective offline and online cleanup

methods, the LOD of milk using IC-SCD was reported as low as 0.4 ng/mL (Kirk

et al., 2005). However, the matrices used in the current study were not prepared

and cleaned as extensively as that in the previous studies, where the baseline

and conductivity background of the matrices were comparable to that of standard

solutions using IC-SCD. In addition, different types of matrices, even the same

type of matrix, but from different species, may have different properties for

perchlorate determination using IC-SCD because of different composition, which

can lead to different LODs using the same instrument. For example, cow urine

analyzed in the previous study was much cleaner than sheep urine in the current

study. The cleaner sample allowed a cow urine LOD (for IC-SCD), which was

close to that of LC-ESI-MS, to be lower than that of the sheep urine. Therefore, if

the LODs by IC-SCD could have been estimated, they would have been higher

than those by LC-ESI-MS.

For both IC-SCD and LC-ESI-MS analyses, mean peak areas for

standards and spiked milk matrices, including dairy milk, soymilk, and human

breast milk, were plotted as a function of perchlorate concentration (Figure 2.8).

There was no significant difference between standards, dairy milk, and soymilk

matrices regarding instrumental response to perchlorate using either IC-SCD or

38

LC-ESI-MS method (p > 0.05 in most comparisons over the spiked concentration

range using t-test). The MS response to perchlorate spiked in human milk was

lower than that in other matrices over the spiked concentration range, which may

be caused by ionization suppression due to more ion species in the human milk

samples. As described by Dodds et al. (2004), high background conductivity in

the case of IC-SCD corresponds to ionization suppression in the case of ESI-MS,

higher background conductivity was also observed in these human milk samples

using IC-SCD. Linearity of perchlorate response was observed from 2 to 1,000

ng/mL by IC-SCD (r2 > 0.99) whereas it was from 2 to 200 ng/mL by LC-ESI-MS

(r2 > 0.99). Deviation from linearity was observed above 200 ng/mL by LC-ESI-

MS; this may result from ionization suppression caused by high ionic strength of

perchlorate.

All perchlorate standards and spiked dairy milk samples were analyzed in

triplicate by both methods, and spiked cow plasma samples were analyzed by

IC-SCD in duplicate and LC-ESI-MS in triplicate, respectively. Coefficient of

variation (CV %) of these two methods are displayed in Table 2.7. Compared

with the IC-SCD method, the analysis precision was improved for the dairy milk,

especially at low levels of spiked perchlorate, but not for cow plasma and

standard solutions when using LC-ESI-MS. This implies that LC-ESI-MS has an

advantage over IC-SCD regarding precision for samples which have high

background conductivity in IC-SCD analysis, such as the dairy milk matrix

prepared in the current study.

2.3.3 Conclusions

High background conductivity and the existence of interfering components

are problems routinely encountered in perchlorate determination in biological

fluids by IC-SCD; the application of LC-ESI-MS avoids these problems. Although

the linear range in LC-ESI-MS is much narrower than that in IC-SCD and there is

a problem with ionization suppression in the case of ESI-MS corresponding to

39

high background conductivity in IC-SCD, the LC-ESI-MS methodology offers a

significant improvement over IC-SCD. LC-ESI-MS provides better precision,

especially at low perchlorate concentrations in complicated matrices, perchlorate

confirmation, and lower detection limits for perchlorate determination in matrices

with less sample cleanup. Therefore, the LC-ESI-MS method reported here can

provide a better quantitative and qualitative analysis of complicated matrices

tested in the current study and work as an efficient alternative analytical tool to

IC-SCD for perchlorate contamination monitoring in the environment.

40

0

5000000

10000000

15000000

20000000

25000000

30000000

35000000

40000000

0 50 100 150 200 250Perchlorate (ng/mL)

Res

pone

se (a

rea)

SUCPDMSTD

Figure 2.4. Linearity of MS response to perchlorate in prepared standard solutions (Milli-Q water, STD) and spiked biological matrices, including dairy milk (1% fat, DM), cow plasma (CP), and sheep urine (SU). The linear range of perchlorate was 2 – 200 ng/mL. There was no statistical difference between dairy milk samples and standard solutions in terms of the slopes of the linear range (p = 0.058, t-test), but there was a significant statistical difference between cow plasma and standard solutions, and between sheep urine and standard solutions (p < 0.001). Error bars represent the SD (n= 5 in spiked matrices and n = 6 in the standard solutions).

41

Table 2.5. Limit of detection (LOD, based on S/N=3) and method detection limit (MDL, determined according to EPA Method 314.0) for perchlorate in standard solutions prepared in Milli-Q water, sheep urine, dairy milk, and cow plasma analyzed by LC-ESI-MS.

LOD (ng/mL) MDL (ng/mL)

Standard solutions 2.49 1.17

Sheep urine 2.48 0.93

Dairy milk (1% fat) 3.00 1.56

Cow plasma 2.28 1.12

Table 2.6. Accuracy and precision of perchlorate determination using LC-ESI-MS in sheep urine, dairy milk, and cow plasma matrices.

Sample matrices

Replicates

Spiked perchlorate concentration (ng/mL)

Perchlorate concentration detected (ng/mL) ± SD

CV (%)

Recovery (%) ± SD

3 4.95 5.5 ± 0.10 1.9 111 ± 2.1

3 9.90 9.8 ± 0.48 4.9 99 ± 4.8 Sheep

urine 3 99.01 85.8 ± 2.00 2.3 87 ± 2.0

3 4.95 6.8 ± 0.31 4.5 137 ± 6.2

3 9.90 10.9 ± 0.33 3.0 110 ± 3.3 Dairy milk

(1% fat) 3 99.01 74.7 ± 0.54 0.7 75 ± 5.5

3 4.98 4.7 ± 0.11 2.3 95 ± 2.2 Cow

plasma 3 11.30 9.1 ± 0.26 2.8 81 ± 2.3

42

Figure 2.5. Comparison of the analysis of dairy milk (1% fat) by IC-SCD and LC-

ESI-MS. Dairy milk (1% fat) was spiked with 4.99 ng/mL perchlorate. The zoomed-in peak a and b represent perchlorate and an interference close to the perchlorate peak (less than 0.5 min difference in terms of retention time), respectively. The insert shows the LC-ESI-MS chromatogram of the same sample with selected ion monitoring at m/z 99 (35ClO4

-). The peak at 12.24 min corresponds to 4.99 ng/mL perchlorate. Unlike the IC-SCD analysis, no interference was found in the milk when analyzed by LC-ESI-MS.

43

Figure 2.6. Comparison of the determination of perchlorate in cow plasma by IC-

SCD and LC-ESI-MS. Cow plasma was spiked with 4.99 ng/mL perchlorate. The zoomed-in peak a and b are perchlorate and an interference which is very close to the perchlorate peak (less than 0.5 min difference in terms of retention time), respectively. The insert shows the LC-ESI-MS chromatogram of the same sample with selected ion monitoring at m/z 99 (35ClO4

-); the peak at 12.27 min corresponds to 4.99 ng/mL perchlorate. No interference was found in the plasma when analyzed by LC-ESI-MS.

44

Figure 2.7. Comparison of the determination of perchlorate in sheep urine by IC-SCD and LC-ESI-MS. Sheep urine was spiked with 50 ng/mL perchlorate (solid line) and water was spiked with 50 ng/mL to make a standard solution (dashed line). The insert is the chromatogram from the LC-ESI-MS analysis. Perchlorate peaks in sheep urine and the standard solution were indicated by a or (A), and b or (B), respectively. Peaks A and B in the LC-ESI-MS analysis overlap well whereas the area of peak a in the IC-SCD analysis is much bigger than that of peak b, which indicates that there are interferents with perchlorate in sheep urine samples analyzed using IC-SCD.

45

0

200000

400000

600000

800000

1000000

1200000

1400000

1600000

1800000

0 200 400 600 800 1000 1200

Perchlorate (ng/mL)

Mea

n pe

ak a

rea

(us)

aDM

HM

SM

DI

Figure 2.8. Calibration curves of perchlorate spiked into Milli-Q water (DI) and

milk matrices analyzed by IC-SCD (a) and LC-ESI-MS (b). DM: dairy milk; HM: human breast milk; SM: soymilk. Error bars represent SD (n=3). Linearity for perchlorate concentrations were up to 1000 ng/mL by IC-SCD and up to 200 ng/mL by LC-ESI-MS.

46

Table 2.7. Precision of perchlorate determination (n = 3) in spiked Milli-Q water, dairy milk, and cow plasma matrices using IC-SCD and LC-ESI-MS. Triplicates were analyzed for either sample matrix except for the spiked cow plasma, in which duplicates were analyzed using IC-SCD.

Coefficient of variation Spiked

samples Perchlorate

concentration (ng/mL) IC-SCD analysis LC-ESI-MS analysis

2.5 2.6% 4.4%

5 5.2% 3.0%

10 5.5% 4.5%

20 5.7% 5.8%

50 5.7% 8.4%

100 3.2% 3.6%

Milli-Q

water

200 4.5% 6.8%

1.99 16.2% 18.2%

4.98 18.4% 7.3%

9.95 46.1% 3.6%

49.75 2.5% 3.1%

99.5 4.1% 1.7%

Dairy

milk

199.01 7.9% 6.3%

1.99 3.3% 0.9%

4.99 2.0% 5.2%

9.98 0.0% 2.8%

49.9 6.0% 2.9%

99.8 0.9% 0.4%

Cow

plasma

199.6 1.1% 1.9%

47

2.4 Perchlorate Determination in Urine Using Ion Chromatography Coupled with

Atmospheric Pressure Ionization Tandem Mass Spectrometry (IC-MS/MS)

2.4.1 Materials and Methods

Reagents and standards

A custom perchlorate (ClO4-) standard solution was purchased from

AccuStandard, Inc. (New Haven, CT). Sodium hydroxide (50%, w/w) aqueous

solution and acetonitrile (HPLC grade) was purchased from Fisher Scientific (Fair

Lawn, NJ). Liquid nitrogen (high purity grade) was purchased from Aeriform

(Lubbock, TX). 18O-perchlorate internal standard (NaCl18O4, 1 mg/L) was

obtained from Dionex (Sunnyvale, CA). All solutions were prepared in 18.2 MΩ

Milli-Q water. Concentrations of the calibration standards ranged from 0.02

ng/mL to 2 ng/mL. 0.1 ng 18O-perchlorate internal standard (10 μL of 10 ng/mL

internal standard) was spiked into each calibration standard or urine sample (0.5

mL) before injection into the IC-MS/MS system.

Ion chromatography

Analyses were conducted with an ion chromatograph coupled with the API

2000TM MS/MS system. The ion chromatography system (Dionex, Sunnyvale,

CA) was equipped with a GP50 gradient pump, AS40 auto-sampler, CD25

conductivity detector, LC30 chromatography oven with rear-loading Rheodyne

injection valve, a 2-mm anion self-regenerating suppressor (ASRS Ultra II)

operating in the external water mode, Rheodyne 6-port valve for matrix diversion,

shielded conductivity cell, and a static mixing “Tee”. Chromeleon® software was

used for system control.

The separation was performed on an IonPac AS20 analytical column (250

mm x 2 mm i.d., Dionex) with an AG20 guard column (50 mm x 2 mm i.d.,

Dionex). Sample was injected using loop injection mode (100 μL loop). NaOH

eluent (45 mM) was used under isocratic conditions at a flow rate of 0.3 mL/min.

48

A suppressor (ASRS Ultra II, Dionex) in external water mode was used to

remove Na+ (54 mA). The retention time of perchlorate was 13.4 min with a total

run time of 16 min.

Mass spectrometry

Applied Biosystems/MDS Sciex API 2000™ triple quadrupole mass

spectrometer (Applied Biosystems/MDS Sciex, Concord, ON, Canada) with a

TurboIonSpray® source was used for the determination of perchlorate.

Operational parameters were as follows: 500 °C source temperature, negative

ion mode, 30.0 psi curtain gas, 50 psi gas supply 1, 75 psi gas supply 2, -4500 V

ion spray voltage, 6 collision gas thichness, -50 V declustering potential, -300 V

focusing potential, -10 V entrance potential, -35 V collision energy, -13.5 collision

exit potential, 150 msec dwell time for each transition, and a 16 min run time.

Multiple reaction monitoring (MRM) mode was used to acquire mass spectral

data. Three transitions were monitored, including 98.9/82.9 (35Cl16O4- →

35Cl16O3-), 100.9/84.9 (37Cl16O4

-→ 37Cl16O3-), and 107.0/89.0 (35Cl18O4

- → 35Cl18O3

-). Analyst® software was used for data acquisition. Highly pure nitrogen

was used as the collision gas.

An on-line 6-port diversion valve was kept in the divert position to divert

matrix ions to waste for the first 10 min, after which the column eluent was

switched into the TurboIonSpray® source for 5 min, during this time perchlorate

was eluted from the column, and then the valve was returned to the original

divert position for the last minute of the run. The matrix diversion technique

diverts non-analyte matrix salts to waste and helps to minimize contamination of

the downstream MS system components. An auxiliary pump supplied post-

column solvent (90% acetonitrile in water) at a flow rate of 0.3 mL/min through

the static mixing “Tee” to the MS throughout the run to improve the electrospray

process efficiency and sensitivity. The column effluent is switched at 10 min of

the run to combine with the post-column solvent about 3 min before perchlorate

49

elutes. The aqueous analytical stream combines and mixes with the solvent

stream inside the static mixing “Tee” before entering the MS.

Sample preparation

Deer mice urine was thawed at room temperature and centrifuged to

remove particulate material at 3750 rpm for 10 min (Beckman Allegra 6R

Centrifuge, USA). The supernatant was transferred to a clean tube and 10 μL

urine was diluted 500X with Milli-Q water. Diluted urine (0.5 mL) was transferred

to an auto-sampler vial, spiked with 0.1 ng of 18O-perchlorate internal standard

(10 μL of 10 ng/mL internal standard), and injected into the IC-MS/MS system.

Perchlorate standard was also spiked into the urine matrix pool to achieve

final concentrations of 0.02, 0.05, 0.1, 0.5, 1, 2, and 5 ng/mL. 18O-perchlorate

internal standard (10 μL of 10 ng/mL internal standard) was also added into each

matrix standard (0.5 mL) before injection.

Quality control

Quality control included blanks (true blank), spiked internal standard (IS),

Milli-Q water (DI), standards with different concentrations of perchlorate (check

standards), and perchlorate-spiked deer mice urine. A DI and a check standard

were analyzed after every 12 deer mice urine sample.

2.4.2 Results and Discussion

Selectivity and sensitivity of IC-MS/MS

Three multiple reaction monitoring (MRM) transitions were monitored for

perchlorate determination. 35Cl16O4- → 35Cl16O3

- (98.9/82.9) was used for

quantifying the main analyte. 37Cl16O4-→ 37Cl16O3

- (100.9/84.9) was used for

examining a proper isotopic abundance ratio of 37Cl/35Cl (1:3.08) and therefore

confirmation of perchlorate presence. 35Cl18O4- → 35Cl18O3

- (107.0/89.0) was

used for checking for ion suppression. As a result, highly improved selectivity

50

was achieved for perchlorate quantitation, especially in a complex matrix (urine),

by ion chromatography coupled with tandem mass spectrometry (IC-MS/MS)

(Figure 2.9 and 2.10).

The IC-MS/MS method was highly sensitive with an excellent limit of

detection (LOD). The calculated LOD (S/N = 3 and 5) in Milli-Q water was 0.067

ng/L and 0.005 ng/mL, respectively, both of which were lower than the lowest

standard (0.02 ng/mL), with a S/N exceeding 10. The instrument detection limit

(IDL) was determined by analyzing a 10 ng/L standard solution 8 times in five

separate days. The IDL was calculated by multiplying the standard deviation of

the replicate measurements by the appropriate Student’s t value (t = 2.99 for 7

degrees of freedom). The calculated IDL was 5.89 ng/L (Table 2.8). Since the

lowest calibration standard was 10 ng/L in the calibration range of 10 to 500 ng/L

(Equation 1), this conservative lowest calibration was used as the minimal

reporting limit (MRL). The low detection limit produced by this instrument

requires minimal sample preparation which usually is expensive, time-

consuming, and laborious. High sample dilution can significantly reduce high

background conductivity of complex matrices. Additionally, avoiding sample

cleanup using cartridges, which have been reported to contain trace levels of

perchlorate (Valentin-Blasini et al., 2005), also avoids exogenously introducing

perchlorate.

(1) Y (analyte area/IS area) = (7.46 ± 0.23) [ClO4-, ng/mL] + (0.04 ± 0.01), r2

= 0.9986 ± 0.0008 , n = 4

For complex biological matrices such as urine, good resolution of

perchlorate from other coeluants is not easily achieved, even with the AS20

column; therefore, interferences in ion chromatography are usually false positives

(Figure 2.11 (B), in which perchlorate concentration was shown more than that

detected by MS/MS). In addition, it is possible for coeluants to suppress

51

ionization of perchlorate and the 18O-perchlorate internal standard. More dilution

or sample cleanup is required if a severe ion suppression occurs, but minimal

suppression would not alter the ratio of perchlorate signal to internal standard

signal and therefore would not change quantitation (Valentin-Blasini et al., 2005).

Urine that was diluted 500-fold in the current study did not cause severe ion

suppression using the tandem mass spectrometry. However, ion

chromatography with conductivity detection showed exaggerated results of

perchlorate quantitation because of the coeluants.

Linearity of calibration curves

Excellent calibration curves (r2 > 0.999) were produced using the IC-

MS/MS method over a broad range from 0.02 to 5 ng/mL in Milli-Q water. Figure

2.12 shows two typical calibration curves in Milli-Q water ranging from 0.02 to 5

ng/mL spiked with 0.196 ng/mL internal standard. Both 98.9/82.9 and 100.9/84.9

transitions showed good linearity and sensitivity for perchlorate quantification,

although the former transition was used for perchlorate quantification in the

current study. Robust results of repeated calibrations were showed by this

method with minimal intraday and interday variability. On one day, the coefficient

of variation (CV) for 0.02 and 5 ng/mL were 13.5% and 0.3%, respectively.

Interday variability of calibration curve slopes was also minimal with CV = 1.3%

(n = 5 over a two week period, Equation 2). Good linearity was also obtained in a

narrow calibration range from 0.01 to 0.5 ng/mL with r2 > 0.99 over a three-week

period (Equation 1).

(2) Y (analyte area/IS area) = (6.82 ± 0.09) [ClO4-, ng/mL] + (0.03 ± 0.04), r2

= 0.9998 ± 0.0001 , n = 5

(3) Y (analyte area/IS area) = (6.80 ± 0.08) [ClO4-, ng/mL] + (0.22 ± 0.03), r2

= 0.9999 ± 0.00005 , n = 3

52

Considering potential matrix effects on the calibration curve, calibration

curve characteristics were compared between standards prepared in Milli-Q

water and spiked deer mice urine (Equations 2 and 3, respectively). Because of

existing perchlorate in the deer mice urine, a substantial y-intercept was

observed for the calibration curve of the spiked urine matrix. However, there was

no significant difference between slopes (6.82 and 6.80 in Milli-Q water and urine

matrix, respectively; p = 0.78, t-test). Therefore, calibration curves generated

from standards prepared in Mill-Q water were used for perchlorate quantitative

determination in all deer mice urine samples.

Accuracy and precision of the IC-MS/MS method

Accuracy and precision were evaluated for the IC-MS/MS method using

check standards and spiked urine matrix pools. Three quality control pools with

perchlorate levels of 0.01, 0.05, and 0.5 ng/mL in Mill-Q water, unspiked, and

spiked urine matrix (0.05, 0.5, and 1 ng/mL) were used in the evaluation (Table

2.9). Less variability was produced over a three-week period in the check

standards with high levels of perchlorate (CV < 11% for perchlorate concentration

> 50 ng/L). Based on the spiked amounts and existing perchlorate, perchlorate

recovery in the spiked urine matrix was within the range of 96.2 to 103.3% after

subtracting the endogenous amount of perchlorate. Repeated analysis of the

urine matrix pool over a 10-day period resulted in minimal deviation regarding the

measured concentrations (CV = 0.6 to 5.6%).

2.4.3 Conclusions

Ion chromatography coupled with the API 2000™ system (IC-MS/MS)

provide an excellent method for part per trillion of perchlorate determination in

both aqueous and deer mice urine matrices. The IC-MS/MS method produces

good selectivity for perchlorate determination through monitoring three MRM

transitions (98.9/82.9: 35Cl16O4- → 35Cl16O3

-, 100.9/84.9: 37Cl16O4-→ 37Cl16O3

-, and

53

107.0/89.0: 35Cl18O4- → 35Cl18O3

-), even in complex matrices such as urine. The

IC-MS/MS method is very sensitive for perchlorate determination with a limit of

detection (LOD) as low as 5.35 ng/L (S/N = 5) in Milli-Q water, which to our

knowledge is the lowest level reported to date. Excellent linearity (r2 = 0.999) of

the calibration curve was demonstrated by this method with a very broad

calibration range of 0.02 to 5 ng/mL. There was no difference in calibration curve

characteristics between the highly diluted urine matrix (500X) and Milli-Q water

regarding the calibration slope. The urine matrix with 500X dilution did not show

ion suppression during MS analysis. Robust accuracy and precision were also

achieved using the IC-MS/MS method.

54

Table 2.8. Instrument detection limit study for perchlorate using IC-MS/MS. Perchlorate concentration was 10 ng/L.

Analysis time Measurement (ng/L)

Replicate No. 1 12/26/2005 04:20:26 AM 9.62

Replicate No. 2 12/29/2005 04:19:41 AM 7.54

Replicate No. 3 12/30/2005 12:18:04 AM 7.53

Replicate No. 4 01/06/2006 12:12:45 AM 10.9

Replicate No. 5 01/06/2006 05:12:21 AM 11.4

Replicate No. 6 01/06/2006 08:11:09 PM 13

Replicate No. 7 01/07/2006 06:10:21 AM 9.77

Replicate No. 8 01/07/2006 11:09:56 AM 8.15

Standard deviation 1.96

Student’s t value 2.998

IDL 5.89

55

0

500

1000

0 2 4 6 8 10 12 14

Time (min)

Inte

nsity

(cps

)

MRM 98.9/82.935Cl16O3

-

16

0

200

400

0 2 4 6 8 10 12 14

Time (min)

Inte

nsity

(cps

)

MRM 100.9/84.937Cl16O3

-

16

0

1000

2000

3000

4000

5000

0 2 4 6 8 10 12 14 1

Time (min)

Inte

nsity

(cps

)

MRM 107/8935Cl18O3

-

6

Figure 2.9. IC-MS/MS analysis of standard solution with 0.02 ng/mL perchlorate

(35Cl16O4- and 37Cl16O4

-) and 0.196 ng/mL internal standard (35Cl18O4-).

56

0

1000

2000

3000

4000

0 2 4 6 8 10 12 14 1

Time (min)

Inte

nsity

(cps

)

MRM 98.9/82.935Cl16O3

-

6

0

400

800

1200

0 2 4 6 8 10 12 14 16

Time (min)

Inte

nsity

(cps

)

MRM 100.9/84.937Cl16O3

-

0

1000

2000

3000

4000

5000

0 2 4 6 8 10 12 14 1

Time (min)

Inte

nsity

(cps

)

MRM 107/8935Cl18O3

-

6

Figure 2.10. IC-MS/MS analysis of deer mice urine (diluted 500-fold). The

perchlorate (35Cl16O4- and 37Cl16O4

-) concentration was determined to be 0.097 ng/mL. The internal standard (35Cl18O4

-) concentration was 0.196 ng/mL.

57

-0.2

0

0.2

0.4

0.6

0.8

1

0 2 4 6 8 10 12 14

Time (min)

Con

duct

ivity

(us)

(A)

16

-0.5

0

0.5

1

1.5

2

0 2 4 6 8 10 12 14

Time (min)

Con

duct

ivity

(us)

ClO4- and

interferences

16

(B)

Figure 2.11. Analysis of a standard solution (A, 0.02 ng/mL) and deer mice urine

diluted 500-fold (B, perchlorate was detected as 0.097 ng/mL using MS/MS) by ion chromatograph with suppressed conductivity detection (IC-SCD). The peak around 13.45 min shown in chromatogram B represents perchlorate and interferences, because the area is much bigger than that of 0.097 ng/mL perchlorate. Therefore, a false positive was observed using IC-SCD. The column eluent was switched into the TurboIonSpray® source at 10 min and back to waste at 15 min.

58

y = 6.7673x + 0.0039r2 = 1

0

5

10

15

20

25

30

35

40

0 1 2 3 4 5Analyte concentration (ng/mL)

Ana

lyte

pea

k ar

ea/IS

pea

k ar

ea

(A)MRM 98.9/82.9 35Cl16O3

-

y = 2.2155x + 0.0003r2 = 1

0

2

4

6

8

10

12

0 1 2 3 4Analyte concentration (ng/mL)

Ana

lyte

pea

k ar

ea/IS

pea

k ar

ea

(B) MRM 98.9/82.9 37Cl16O3

-

5

Figure 2.12. Calibration curves for the two MRM transitions: (A) 98.9/82.9:

35Cl16O4- → 35Cl16O3

-, (B) 100.9/84.9: 37Cl16O4-→ 37Cl16O3

-. Calibration curve concentrations ranged from 0.02 to 5 ng/mL.

59

Table 2.9. Accuracy and precision of the IC-MS/MS method. Three quality control (QC) pools with perchlorate concentrations of 10, 50, 500 ng/mL, unspiked, and spiked urine matrix (spiked concentration was 50, 500, or 1000 ng/mL) were used for the accuracy and precision study.

Check standard N Theoretical concentration (ng/L)

Measurement (mean, ng/L)

SD CV% Diff. %

QC1 10 10 10.1 1.98 19.6 0.8

QC2 10 50 49.9 5.07 10.2 -0.1

QC3 7 500 525.6 33.43 6.3 5.1

Unspiked urine 4 35.9 2.02 5.6

Spiked 50 ng/L 4 86 88.01 0.99 1.1 2.5

Spiked 500 ng/L 4 536 515.8 3.30 0.6 -3.8

Spiked 1000 ng/L 3 1036 1070.0 10 0.9 2.9

60

CHAPTER III

ASSESSING PERCHLORATE EXPOSURE TO MAMMALS IN THE FIELD‡

‡: results of this work were published in the Journal of Agricultural and Food Chemistry (Q.

Cheng, L. Perlmutter, P. N. Smith, S. T. McMurry, W. A. Jackson, and T. A. Anderson, 2004, A

Study on Perchlorate Exposure and Absorption in Beef Cattle. 52: 3456-3461)

3.1 Introduction

Once released into the environment, perchlorate can persist for many

decades because of its unique characteristics: extremely water-soluble, mobile in

aqueous systems, and kinetically stable. Perchlorate can be taken up and

accumulated by plants and edible vegetation through irrigation with contaminated

water, fertilization with fertilizer derived from Chilean caliche, or from perchlorate-

contaminated soil (Susarla et al., 2000; Urbansky et al., 2000b; Ellington et al.,

2001; Tan et al., 2004; Yu et al., 2004; Jackson et al., 2005b; Sanchez et al.,

2005). Animals and human beings can be exposed to perchlorate through

ingestion of plants or food containing perchlorate (trophic transfer) and/or through

drinking contaminated water directly.

Perchlorate has been reported in tissues of animals in the vicinity of

contaminated sites. Aquatic organisms have been found to be at a relatively high

risk for perchlorate exposure. In streams and ponds near the Longhorn Army

Ammunition Plant (LHAAP) near Karnack, Texas, perchlorate was detected in

aquatic insects, fish, and frogs at concentrations ranging from 811 to 2038 µg/kg,

below detection to 207 µg/kg, and below detection to 580 µg/kg, respectively

(Smith et al., 2001). Smith et al. (2001) also reported perchlorate concentrations

ranging from below detection to 2328 µg/kg in small mammals (e.g., rodents)

collected from LHAAP. Perchlorate exposure and effects were also investigated

in raccoons at a contaminated site by monitoring perchlorate residues in plasma

and changes of thyroid hormones in the circulation system (blood) (Smith et al.,

2005).

61

Epidemiological studies of perchlorate exposure and effects on humans

have also been conducted (Lamm and Doemland, 1999; Brechner et al., 2000;

Crump et al., 2000; Li et al., 2000a; Li et al., 2000b; Li et al., 2001). In all these

cases, contaminated drinking water was investigated as the source of

perchlorate exposure for humans (neonates and/or school-age children as the

major subjects). However, recently, perchlorate contamination has been found in

supermarket milk samples, human breast milk, and some food items (Kirk et al.,

2003; Krynitsky et al., 2004; Kirk et al., 2005; Valentin-Blasini et al., 2005), which

indicates that humans may be exposed to perchlorate through trophic transfer in

addition to exposure to drinking water. The detection of perchlorate in

supermarket milk samples, human milk, and human urine (Kirk et al., 2003;

Krynitsky et al., 2004; Valentin-Blasini et al., 2005) indicates that environmental

contamination of perchlorate is much more widespread than what was originally

thought in the U.S. and animals and humans are being exposed.

Although many studies have been conducted to assess perchlorate

exposure and effect on plants, animals (both aquatic and terrestrial), and humans

at contaminated sites as mentioned above, there is scarce data on large

mammal (such as dairy and beef cattle) exposure to perchlorate. However, large

mammals present a potential pathway of perchlorate exposure to humans

through trophic transfer and it has been of concern since perchlorate was

reported in some commercial milk samples and other food items (Erickson, 2003;

Kirk et al., 2003; Krynitsky et al., 2004; Kirk et al., 2005). Cattle raised in

impacted areas may be of special importance due to their potential ingestion of

perchlorate both through impacted water and their large intake of forage crops,

which could also contain significant quantities of perchlorate. In some instances,

humans are exposed to perchlorate through similar ways of trophic transfer in

addition to contaminated drinking water. Therefore, an assessment of

perchlorate exposure to large mammals at contaminated sites is necessary and

would contribute to perchlorate risk assessment in the environment and humans.

62

A study on perchlorate exposure and absorption in beef cattle was

conducted. Perchlorate residues in blood plasma and tissues were evaluated in

heifer calves raised on a site impacted by perchlorate. These animals inhabited

areas in central Texas near the Naval Weapons Industrial Reserve Plant

(NWIRP), a facility that produced many kinds of perchlorate-containing products

prior to 1995. Monitoring data indicated that perchlorate was present in some

streams and tributaries near the site (Motzer, 2001). This raised concerns that

wildlife and domestic animals consumed by humans may contain perchlorate

residues and contribute to perchlorate exposure to humans. Perchlorate

residues in blood plasma were also investigated in cattle from two Kansas farms

adjacent to facilities that used or handled perchlorate.

3.2 Materials and Methods

Study description and sample collection in TX

The study was conducted over 14 weeks during the spring of 2003 near

McGregor, McLennan County, Texas. Four Shorthorn heifer calves (~535 lbs.

each) were purchased commercially from an individual seller. All calves were

held for 1 week on the “Reference Site”. Two calves (E1 and E2) were then

separated and sent to a perchlorate-impacted “Treatment Site” while the other

two calves (R1 and R2) remained on the Reference Site. The Reference Site

was a pasture a sufficient distance from NWIRP such that perchlorate was not

present in available on-site surface or groundwater utilized by the cattle, nor was

it present in vegetation. The Treatment Site was a pasture near the NWIRP

boundary with a spring-fed stream bisecting the pasture. Previous monitoring

data for the spring (collected over a 2 year period) indicated that perchlorate was

consistently present in the water (range = 20-60 ng/mL) (unpublished data from

our laboratory). The spring-fed stream served as the only water source for

calves on the Treatment Site. Calves on the Treatment Site also had access to

vegetation (mostly grass) near the stream. We had previously monitored

63

vegetation near the stream (trees and aquatic plants) and showed that

perchlorate levels in leaves varied with species and distance from the stream

(Tan et al., 2004). However, supplemental forage (hay) used during the first 2

weeks of the study on both sites did not contain detectable perchlorate

(unpublished data from our laboratory).

Calves on both sites were visually monitored on a daily basis, and blood

was collected from each animal every 2 weeks. Blood samples were always

collected in the morning (before 9:00 a.m.). Approximately 50 mL of blood was

drawn at each sampling and placed into four sterile EDTA-coated blood

collection vials. The samples were then spun at approximately 3100 rpm for 15

min. Plasma was transferred off and placed in 15-mL falcon tubes. Samples

were stored on dry ice during transport back to the laboratory and frozen (-80 °C)

until analysis. At the time of each blood collection, drinking water that the

animals had access to was also collected and placed on ice during transport

back to the laboratory, where it was stored at 4 °C until analysis.

At the conclusion of the study, animals were processed in a manner

identical to a commercial beef processing operation. The following tissue

samples were obtained from each animal for residue analysis: liver, thyroid, and

various meat cuts (sirloin steak, round steak, T-bone steak, and roast).

Determination of perchlorate

A perchlorate (ClO4-) standard solution was obtained as a custom

standard from AccuStandard, Inc. (New Haven, CT). Sodium hydroxide (NaOH),

50% (w/w) aqueous solution was purchased from Fisher Scientific. All solutions

were prepared in 18.2 MΩ Milli-Q water. Ethanol was purchased from Fisher

Scientific.

Plasma samples were processed using methods similar to those

described previously (Fisher et al., 2000; Anderson and Wu, 2002; Narayanan et

al., 2003) prior to analysis. First, 1 mL of plasma was precipitated with 4 mL of

64

ethanol (ice-cold) and then centrifuged (4 °C) at 3750 rpm for 5 min. The

supernatant was removed, evaporated to dryness under nitrogen, and

reconstituted in 5 mL of Milli-Q water. Samples were then cleaned using alumina

and C18 solid-phase extraction (SPE) cartridges and filtered (0.45 μm) prior to

ion chromatography (IC) analysis.

Tissue samples were also processed prior to IC analysis. Samples (10-20

g) were air-dried and then extracted with Milli-Q water using accelerated solvent

extraction (ASE; Dionex Corp.). Extraction conditions were as follows: pressure

= 1500 psi, temperature = 100 °C, extraction time = 15 min. Sample extract

volumes were measured, diluted (5X), and cleaned with alumina and C18 SPE

cartridges. Eluates were filtered (0.45 μm) prior to IC analysis.

Surface water samples collected from the 2 sites during the course of the

study were filtered (0.45 μm) prior to IC analysis.

All samples (plasma extracts, tissue extracts, and surface water) were

analyzed by ion chromatography similar to EPA Method 314 (Hautman et al.,

1999). Analysis was carried out using a Dionex DX-500 Ion Chromatography

System equipped with a GP50 gradient pump, a CD20 conductivity detector, and

an AS40 automated sampler (Dionex Corp.). PeakNet chromatography software

was used to control the system. Ion separation was made with a Dionex IonPac

AS16 (250-mm X 4.0-mm) analytical column. Conditions for the system were as

follows: flow rate = 1.0 mL/min; eluent = 50 mM sodium hydroxide; injection

volume = 1000 μL. Ion detection was by suppressed conductivity in the external

water mode. A seven-point standard curve was constructed from constant

volume injections of calibration standards of 2.5, 5, 10, 20, 50, 100, 200, and 500

ppb (ng/mL). Computer-generated peak areas were used to measure sample

concentrations in an external standard mode. Quality control (QC) samples

included blanks, matrix spikes, and check standards. Using the analytical

method described above, the detection limits for perchlorate in water, plasma,

and muscle were 1.0 ng/mL, 13.7 ng/mL, and 23.2 ng/g, respectively. Good

65

perchlorate recovery (average recovery of 83.6% ± 2.3) of spiked blood plasma

has been shown in Chapter II (Table 2.1).

3.3 Results and Discussion

Perchlorate in drinking water, blood plasma, and tissues of beef cattle in TX

Perchlorate was not detected in surface water samples collected from the

Reference Site (detection limit = 1.0 ng/mL). In contrast, perchlorate was

detected throughout the study (14 weeks) in water samples from the Treatment

Site (mean ± standard error = 25.4 ± 1.3 ng/mL; n = 8) (Figure 3.1). The

concentrations of perchlorate in water from the Treatment Site during the 14-

week study were consistent with more than 2 years of monitoring data from that

location (unpublished data).

Perchlorate was not detected in blood plasma samples from either calf

occupying the Reference Site (Figure 3.1). Similar results were obtained for the

2 calves on the Treatment Site with one exception; perchlorate was detected (15

ng/mL and 22 ng/mL) in one of the calves on consecutive sampling periods 4 and

6 weeks after the beginning of the study. Perchlorate was not detected

(detection limit = 23.2 ng/g) in any of the tissue samples collected from calves

occupying the Reference Site. In addition, perchlorate was not detected in any of

the tissue samples collected from calves occupying the Treatment Site.

Our results indicate that despite the presence of perchlorate in drinking

water at the Treatment Site, there was little quantifiable perchlorate exposure

measured in blood plasma from heifer calves. Constant exposure to 25 ppb

perchlorate in water over 14 weeks did not result in measurable residues in blood

plasma or edible tissues.

Water intake of cattle is well understood (NRC, 2001). A beef heifer

consumes 20-55 liters of water per day and excretes 75% of that intake (2:1

feces/urine). On the basis of that assumption and the measured drinking water

concentrations, the heifers on the Treatment Site ingested 125-350 μg

66

perchlorate/day. If one makes the assumption that cattle have 60 mL of blood

per kilogram of body weight (Animal-Welfare-Advisory-Committee, 1996), the

expected perchlorate concentration in plasma should be 9-25 ng/mL. This range

is consistent with the 2 detections that were observed (15 and 22 ng/mL).

Perchlorate can be rapidly excreted in urine, with reported urinary

excretion half-lives ranging from 8 to 20 h in rats (Goldman and Stanbury, 1973;

Wolff, 1998; Fisher et al., 2000). Although Batjoens and co-workers (Batjoens et

al., 1993) reported that prolonged perchlorate administration (4 g/day for 10

days) in cows resulted in a longer excretion period in the urine than that with a

single dose, perchlorate accumulation did not occur because of the relatively

rapid excretion. The rapid half-life of perchlorate in mammals contributes to the

difficulty in assessing perchlorate exposure by monitoring blood. Although we

also collected urine from the animals opportunistically, this sample matrix proved

to be difficult for IC analysis by Method 314 at that time, due to a high signal

background.

Perchlorate in contaminated water, blood plasma, and urine from cattle in

Kansas

Perchlorate was detected with greater frequency and at slightly higher

concentrations in blood of cattle from two Kansas farms adjacent to facilities that

used or handled perchlorate (unpublished data from our laboratory). Cattle on

these farms were not restricted to water supplies containing perchlorate (as were

those on the pasture near the NWIRP site), although the water that was

contaminated had higher perchlorate concentrations (>100 ng/mL). Nonetheless,

perchlorate was detected in 4 of 33 and 17 of 26 cattle at the two Kansas farms.

The highest plasma perchlorate concentrations observed in the Kansas cattle

were 43 and 32 ng/mL, respectively. Although perchlorate was detected at

slightly higher concentrations in these animals, thyroid hormone levels were not

different from control (unexposed) cattle (Cheng et al., 2004).

67

There is an increasing concern regarding potential perchlorate exposure

and effect on animals and humans through trophic transfer. Cattle could

potentially ingest perchlorate through both impacted drinking water and forage

crops, which can accumulate perchlorate with bioconcentration factors ranging

from 1.4 to 620 in field and greenhouse studies (Jackson et al., 2005b). Drinking

water was the major source for perchlorate exposure to these cattle, especially

from TX with no perchlorate detectable in supplemental forage (hay) used during

the first 2 weeks of the study. Therefore, it is possible that exposure to

perchlorate through drinking water which contained ~25 ng/mL perchlorate rather

than food did not show high residues in blood plasma. However, it is likely that

overall perchlorate exposure in heifers from the Treatment Site (TX) was higher

than that measured in water as vegetation samples from the Treatment Site also

contained perchlorate (Tan et al., 2004).

The difference in perchlorate concentrations in whole blood vs. plasma

was tested in order to know the location of perchlorate in the blood and whether

the negative result of perchlorate residues in cow plasma was caused by less

perchlorate distributed into plasma. Equal amounts of perchlorate were spiked

into two aliquots of deer mouse whole blood (0.2 mL each). One aliquot was

precipitated with ethanol directly, centrifuged, and the supernatant was

evaporated to dryness, reconstituted with Milli-Q water, and finally was cleaned

with C18 and alumina-neutral cartridges. The other aliquot was centrifuged and

plasma was obtained. The plasma was then processed according to the

procedure used for the first aliquot (precipitated, evaporated, reconstituted, and

cleaned). The final volume of the two cleaned samples was the same.

Perchlorate concentrations determined using IC were 12% higher in the whole

blood sample than in the plasma. Considering the average recovery of 83.6% ±

2.3 with the cleanup method, 12% difference in perchlorate concentration

between whole blood and plasma would not contribute significantly to the

negative results of perchlorate residues in cow plasma.

68

Using the cleanup method and LC-ESI-MS described in Chapter II,

confirmation analysis revealed that one cattle urine sample (total n = 4) from

Kansas contained perchlorate (concentration = 3.45 μg/L after 8X dilution).

Perchlorate was detected in all 4 urine samples using IC-MS/MS described in

Chapter II. We did not detect perchlorate in any of the corresponding cattle

plasma samples (n = 4). Urine would be a more sensitive biomarker for

perchlorate exposure. In addition, perchlorate may deposit in its target tissue

(thyroid) and cause less perchlorate to be retained in the circulation system.

Therefore, perchlorate distribution, excretion, and depuration studies were

conducted and results are presented in next chapter.

3.4 Conclusions

As environmental contamination of perchlorate is more widespread, and

animals and humans have been exposed, an assessment of perchlorate

exposure to large mammals at contaminated sites becomes important and

necessary. Information gathered contributes to perchlorate risk assessment in

the environment and to humans. Constant exposure to 25 ng/mL perchlorate in

water over 14 weeks did not result in measurable residues in blood plasma or

edible tissues of cattle at the first test site (McLennan County, Texas). However,

perchlorate was detected in 4 of 33 and 17 of 26 cattle at the two Kansas farms

with the highest plasma perchlorate concentrations of 43 and 32 ng/mL,

respectively. Because of excretion via urine or other pathways, urine could be a

sensitive biomarker for perchlorate exposure to animals and humans. In

addition, perchlorate may deposit in its target tissue (thyroid) and thus less

perchlorate would remain in the circulation system. Therefore, perchlorate

residues in plasma may not be a proper biomarker for perchlorate exposure

assessment.

69

Date

2/1/2003 3/1/2003 4/1/2003 5/1/2003 6/1/2003 7/1/2003

PER

CH

LOR

ATE

(ng/

mL)

-10

0

10

20

30

40

Treatment Site WaterReference Site PlasmaTreatment Site PlasmaReference Site WaterWater Detection LimitPlasma Detection Limit

Figure 3.1. The relationship between perchlorate exposure in drinking water and

perchlorate in heifer plasma. Each datum point is the mean ± SD of duplicate measurements. For graphing and calculation purposes, non-detects were given a value of ½ the respective detection limit for perchlorate in water and plasma. The detection limit for perchlorate in water and plasma was 1.0 and 13.7 ng/mL, respectively.

70

CHAPTER IV

PERCHLORATE DISTRIBUTION AND EXCRETION IN LABORATORY

ANIMALS

4.1 Introduction

Environmental contamination of perchlorate (ClO4-) appears to be

widespread in the United States, especially in aqueous systems. Perchlorate

can be taken up by and accumulated in vegetation. Animals, including humans,

can be potentially exposed to perchlorate through drinking water and/or trophic

transfer. Evidence of the presence of perchlorate in commercial milk, human

milk, human urine, and some food items (Kirk et al., 2003; Krynitsky et al., 2004;

Valentin-Blasini et al., 2005) exists in the scientific literature.

Many studies have shown that perchlorate is taken up by the thyroid.

Using labeled perchlorate, Anbar et al. (1959) found the highest concentration in

the thyroid, followed by ova and blood in rats and rabbits. Peña et al. (1976)

reported the greatest concentration in the thyroid, followed by gizzard lining at 3

hours after a single injection of K36ClO4 in hens. Chow et al. (1969; 1970)

demonstrated relative thyroid concentrations of perchlorate were much greater

than other organs in rats and guinea pigs. The perchlorate ion was concentrated

in rat thyroid but not in skeletal muscle, and the radioactive ClO4- concentration in

the thyroid was negatively related to the doses of stable perchlorate received.

Yu et al. (2002) reported that perchlorate appeared to be actively sequestered

into the thyroid gland, gastrointestinal tract, and possibly the skin of rats.

Narayanan et al. (2003) also found that the highest concentration of perchlorate

was in the thyroid, followed by milk, amniotic fluid, and serum in pregnant and

lactating rats dosed with 10 mg/kg perchlorate through drinking water from

gestation day 2 until either gestation day 20 or postnatal day 10.

Perchlorate does not appear to be metabolized in the body and compared

to the portion taken up by the thyroid, a major portion of perchlorate can be

71

excreted from the body primarily into urine with half-lives varying from < 8 h to

~20 h in rats (Wolff, 1998). Goldman and Stanbury (1973) reported that

perchlorate was taken up by the thyroid in rats, reached a peak around 4 h, and

then fell to approximately 5% of the peak value within 95 h with a half-life of 20 h.

Peña et al (1976) found that 87.6% and 99.4% of 36ClO4- was recovered in

excreta 3 h and 24 h, respectively, after administration in hens. The largest total

deposition in the 3 h group was found in the blood (2.9%), followed by muscles,

internal organs, and ova. In a study by Yu et al. (2002), very high recovery of

perchlorate (99.5%) was obtained in urine from male rats; 95% of administered

perchlorate was cleared in 55 to 75 h. In cattle, perchlorate was also excreted in

the urine at a concentration-related elimination rate after oral administration of a

single dose (Batjoens et al., 1993). The study also showed that prolonged

perchlorate administration (4 g/day for 10 days) resulted in a longer excretion

period in the urine. In humans, Lamm et al. (1999) reported that urinary

perchlorate concentration increased during a “work” exposure time with a

decrease between the 12-hour work shifts. The average perchlorate elimination

half-lives of post-exposure for low- and medium-exposure groups were 7.9 and

8.2 hours, respectively. In a low-dose short-term study (10 mg/day perchlorate

for 14 days), urinary perchlorate markedly increased and reached a level of ~7.6

mg per 24 hours at 7 and 14 days (Lawrence et al., 2000). Greer et al. (2002)

determined that post-exposure half-lives ranged from 6.0 to 9.3 h for eight

volunteers who drank water at a dosage of 0.5 mg/kg-day for two weeks.

Perchlorate has also been observed to be excreted via milk. Perchlorate

has been found in both commercial milk samples and human breast milk (Kirk et

al., 2003; Kirk et al., 2005). In a perchlorate dosing study with lactating goats

(Kirk, 2005), perchlorate was detected in milk at higher concentrations 2-h post-

dose than 24-h post-dose. Generally, perchlorate concentrations in goat milk

collected at 24 h after dosing decreased after week one through week four.

Following a 20-h infusion interval of perchlorate into dairy cow rumen everyday

72

for 5 weeks, perchlorate was detected at a constant level in milk and urine during

the 5 weeks, with much higher levels of perchlorate in the urine (Capuco et al.,

2005).

In many of the aforementioned studies, the highest perchlorate

concentration was reported in the thyroid, but the perchlorate distribution pattern

in other tissues was not consistent. To a large degree, adverse effects of

perchlorate on vertebrates depend on uptake, distribution, and excretion in the

body. Fundamental questions such as whether perchlorate accumulates in the

body, how is perchlorate distributed among tissues, and what are the excretion

and elimination patterns via urine will help to determine whether perchlorate

exposure causes adverse human health effects or ecological impacts.

Therefore, a tissue distribution study of perchlorate in voles and perchlorate

excretion and elimination studies in deer mice were conducted. Prairie voles

(Microtus ochrogaster) provide a useful mammalian herbivore model of

contaminant exposure and toxicity. Deer mice (Peromyscus maniculatus) are the

most abundant and widely distributed rodents in North America (Joyner et al.,

1998) and are considered to be highly relevant to ecological risk assessment of

environmental contaminants. Both species are used as terrestrial wildlife

receptors for environmental contaminant risk assessment.

The hypotheses tested included (1) urine is the major pathway through

which perchlorate is excreted; (2) perchlorate excretion is dose-dependent: high

perchlorate exposure levels correspond to high excretion levels in urine; (3)

perchlorate is depurated rapidly via urine.

4.2 Materials and Methods

Chemicals

A custom perchlorate (ClO4-) standard solution was obtained from

AccuStandard, Inc. (New Haven, CT). Magnesium perchlorate, sodium

perchlorate (reagent grade anhydrous), and sodium hydroxide (NaOH, 50%

73

(w/w)) aqueous solution were purchased from Fisher Scientific. All solutions

were prepared in 18.2 MΩ Milli-Q water. Ethanol and acetonitrile (HPLC grade)

were also purchased from Fisher Scientific. 18O-perchlorate internal standard

(NaCl18O4, 1 mg/L) was obtained from Dionex (Sunnyvale, CA).

Animals

Male adult prairie voles (Microtus ochrogaster) (42.9 ± 7 g) used

throughout the distribution experiment were obtained from the breeding colony,

Texas Tech University (Lubbock, TX). Male adult deer mice (Peromyscus

maniculatus) (18.5 ± 1.8 g) used in the excretion and elimination studies were

provided by Dr. Michael Hooper’s breeding colony, Texas Tech University

(Lubbock, TX).

Distribution experiment

Ten voles were arbitrarily placed into 10 different metabolic cages at 8:00

pm on the first day with no food or water. The size of the cage was 233 mm D.I.

X 103 mm H with a wire-mesh bottom. Reference urine and feces were collected

from the metabolic cages between 7:30 am and 8:00 am on the second day. At

8:00 am, upon completion of collection of reference materials, all voles were

randomly assigned to either a 4-h or 8-h dosing period (five voles for each

group), with oral access to 250 μg/mL magnesium perchlorate in water via pre-

weighed water bottles. The 4-h group was dosed from 8:00 am to 12:00 pm. At

the end of the 4 hours, water bottles were removed from metabolic cages and

weighed. Voles were euthanized using a carbon dioxide chamber and

necropsied to obtain blood, liver, kidney, and thyroid tissues. The blood was

centrifuged to collect plasma. All samples were stored on dry ice until

transported to The Institute of Environmental and Human Health where they were

stored at -80 °C until analysis. Urine and feces were collected from the cages

upon completion of the necropsy. The other five voles (8-h group) were dosed

74

from 8:00 am to 4:00 pm and then processed as described above for the 4-h

group.

Excretion experiment

Twenty-four deer mice were arbitrarily assigned into four groups (one

control and three treatments: low, medium, and high). Siblings were placed in

different groups. Animals were housed individually in metabolic cages held by

two racks (4 groups X 3 cages per rack) and animals in the same treatment

group were placed arbitrarily on the racks. Animals were allowed to acclimate for

two days in the cages to test facility conditions of room temperature (ranging

from 65 – 78 °F), 25 – 75% relative humidity, and 12:12 h of dark:light prior to

initiation of dosing. Animals were exposed to perchlorate daily through drinking

water for four consecutive weeks (sodium perchlorate dissolved in Milli-Q water).

Mice in the control group were given Milli-Q water without perchlorate throughout

the study. Urine samples were initially collected at 8, 16, 24, 36, 48, 72, and 96

h, and then were collected every 24 hours up to day 8. After day 8, urine was

collected every two or three days until the end of the study. Reference urine was

collected before treatment initiation. Food and water were provided ad libitum

and checked daily. Water consumption was recorded at each urine collection.

Perchlorate concentrations in the dosed drinking water (low, medium, and high)

were 17, 165, and 1600 ng/mL, respectively. Deer mouse body weights were

recorded before treatment and each week during the exposure to monitor any

weight changes. Metabolic cages were replaced (cleaned) weekly.

At end of the exposure experiment (day 29), animals were weighed,

anesthetized with carbon dioxide, bled via heart puncture, and euthanized by

exsanguination. At necropsy, the following tissues were collected: kidney liver,

intestinal tract, and stomach. Blood samples were placed in EDTA-rinsed tubes,

centrifuged, and plasma was collected. All tissues, plasma, and carcasses were

placed on dry ice after collection and during transport to the laboratory, where

75

they were stored at – 80 °C until gene expression analysis (presented in Chapter

V).

Depurtion experiment

Twelve deer mice were arbitrarily assigned into two groups (low dose and

high dose). Siblings were placed in different groups. Animals were housed

individually in 12 metabolic cages placed arbitrarily on one rack. Animals were

allowed to acclimate for two days in the cages to test facility conditions of room

temperature (ranging from 65 – 78 °F), 25 – 75% relative humidity, and 12:12 h

of dark:light prior to initiation of dosing. Animals were dosed with perchlorate

through drinking water over 8 hours (sodium perchlorate dissolved in Milli-Q

water). After the exposure period and throughout the “elimination phase” of the

study, Milli-Q water rather than perchlorate-contaminated water was given to the

animals. Urine samples were collected at 8, 16, 24, 36, 48, 72, and 96 hours

after treatment. Reference urine was collected before treatment initiation. Food

and water were provided ad libitum and checked every day. Water consumption

was recorded at each urine collection. Perchlorate concentrations in the drinking

water were 17 and 1600 ng/mL in the low and high dose groups, respectively.

Body weights were recorded before treatment and at the end of the study (day

4).

Perchlorate determination method

An ion chromatography (IC) method similar to EPA Method 314 was used

to determine perchlorate residues in the distribution experiment. The IC method

was shown to be capable of detecting perchlorate in water, plasma, and muscle

at 1.0 ng/mL, 13.7 ng/mL, and 23.2 ng/g, respectively (Chapter II). The

chromatography was performed by a Dionex DX-500 Ion Chromatography

System equipped with a GP50 gradient pump, a CD20 conductivity detector, and

an AS40 automated sampler (Dionex Corp.). PeakNet® chromatography

76

software was used to control the system. Ion separation was made with a

Dionex IonPac AS16 (250 mm x 4.0 mm) analytical column. Conditions for the

system were as follows: flow rate = 1.0 mL/min; eluent = 50 mM sodium

hydroxide; injection volume = 1000 µL. Ion detection was by suppressed

conductivity in the external water mode.

Before IC analysis, tissue samples (liver, kidney, and thyroid) were air

dried and then extracted with Milli-Q water using Accelerated Solvent Extraction

(ASE; Dionex Corp.). Sample extract volumes were measured, diluted, and

cleaned with C18 and alumina SPE cartridges. Eluates were filtered (0.45 μm)

prior to IC analysis.

Plasma samples were also processed prior to analysis. Plasma was

precipitated with ethanol (ice-cold) and then centrifuged (4 °C) at 3750 rpm for 5

min. The supernatant was removed, evaporated to dryness under nitrogen, and

reconstituted in 5 mL Milli-Q water. Samples were cleaned with C18 and

alumina, and then filtered prior to ion chromatography (IC) analysis. Urine

samples were diluted 2500X with Milli-Q water, filtered, and then analyzed using

the IC.

Perchlorate determination in deer mice urine in the excretion and

depuration experiments was conducted with an ion chromatograph coupled with

the API 2000TM MS/MS system. The ion chromatography system (Dionex,

Sunnyvale, CA) was equipped with a GP50 gradient pump, AS40 auto-sampler,

CD25 conductivity detector, LC30 chromatography oven with rear-loading

Rheodyne injection valve, a 2-mm anion self-regenerating suppressor (ASRS

Ultra II) operated in the external water mode, Rheodyne 6-port valve for matrix

diversion, shielded conductivity cell, and a static mixing “T”. Chromeleon®

software was used for system control. The separation was performed with an

IonPac AS20 analytical column (250 mm x 2 mm i.d., Dionex) with an AG20

guard column (50 mm x 2 mm i.d., Dionex). Sample was injected using loop

injection mode (100 μL loop). NaOH eluent (45 nM) was used under isocratic

77

conditions at a flow rate of 0.3 mL/min. A suppressor (ASRS Ultra II, Dionex) in

external water mode was used to remove Na+ (54 mA). The retention time of

perchlorate was 13.4 min with a total run time of 16 min.

Applied Biosystems/MDS Sciex API 2000™ triple quadrupole mass

spectrometer (Applied Biosystems/MDS Sciex, Concord, ON, Canada) with a

TurboIonSpray® source was used for perchlorate determination. The operational

parameters were as follows: 500 °C source temperature, negative ion mode, 30.0

psi curtain gas, 50 psi gas supply 1, 75 psi gas supply 2, -4500 V ion spray

voltage, 6 collision gas thichness, -50 V declustering potential, -300 V focusing

potential, -10 V entrance potential, -35 V collision energy, -13.5 collision exit

potential, 150 msec dwell time for each transition, and 16 min as running time.

Highly pure nitrogen was used as the collision gas. Multiple reaction monitoring

(MRM) mode was used to acquire mass spectral data. Three transitions were

monitored, including 98.9/82.9 (35Cl16O4- → 35Cl16O3

-), 100.9/84.9 (37Cl16O4-→

37Cl16O3-), and 107.0/89.0 (35Cl18O4

- → 35Cl18O3-). Analyst® software was used

for data acquisition.

An on-line 6-port diversion valve was kept in the divert position to divert

matrix ions to waste for the first 10 min, after which the column eluent was

switched into the ionization source for the next 5 min, during this time perchlorate

eluted from the column, and then the valve switched back to the original divert

position for the last minute of the run. The matrix diversion technique diverts

non-analyte matrix salts to waste and helps to minimize contamination of the

downstream MS system components. An auxiliary pump supplies post-column

solvent (90% acetonitrile in water) at a flow rate of 0.3 mL/min through the static

mixing “Tee” to the MS throughout the run to improve the electrospray process

efficiency and sensitivity. The column effluent is switched at 10 min in the run to

combine with the post-column solvent about 3 min before perchlorate elutes.

The aqueous analytical stream combines and mixes with the solvent stream

inside the static mixing “Tee” before entering the MS.

78

Deer mouse urine was thawed at room temperature and centrifuged (3750

rpm for 10 min) to remove particulate material (Beckman Allegra 6R Centrifuge,

USA). The supernatant was transferred to a clean tube and 10 μL was diluted

(500 x) with Milli-Q water. Diluted urine (0.5 mL) was transferred to an auto-

sampler vial, spiked with 0.1 ng of 18O-perchlorate internal standard (10 μL of 10

ng/mL internal standard), and injected into the IC-MS/MS system. Quality control

samples included blanks, spiked matrix, and check standards. Blank and check

standards were analyzed after every 15 samples during the IC-MS/MS analyses

to assure that perchlorate recovery was between 80 – 120%; 96 – 103%

perchlorate was recovered in the spiked matrix (Table 2.9 in Chapter II).

Statistical analysis

Statistical analyses were performed using R software (version 2.1.1, Free

Software Foundation, Boston, MA). Normality and homogeneity of variance of

the data were tested using Sharpiro-Wilk normality test and Bartlett’s test,

respectively. Two-tailed t-tests were conducted to test differences in water

consumption, urine production, and perchlorate recovery in vole tissues, plasma,

and urine in the distribution experiment. Two-tailed t-tests were also conducted

to test differences in perchlorate intake and urine residues in the depuration

experiment. Correlation analysis was conducted to test the relationship between

perchlorate residues in tissues, plasma, or urine and perchlorate intake. One-

way ANOVA was used to test differences in water consumption, body weight,

perchlorate concentration in urine, and mass percentage of the total intake

through drinking water excreted into urine (within different treatments and at each

sampling time) in the excretion experiment. An α = 0.05 was used for all tests.

Exponential non-linear regressions were modeled using Excel®.

79

4.3 Results and Discussion

Perchlorate distribution in voles

There was no difference between groups regarding water consumption

and body weight, but there was an individual in the 4-h group that drank an

excessive amount of water which resulted in much higher perchlorate intake; that

individual was not included in data analysis. Excluding that individual datum,

perchlorate intake in the 4-h group was significantly lower than that in the 8-h

group (28.8 ± 6.5 μg/g bw vs. 31.1 ± 5.4 μg/g bw, p < 0.001). But the rate of

perchlorate intake was higher in the 4-h than in the 8-h group (7.2 μg/g bw/hr vs.

3.9 μg/g bw/hr), with the majority of water consumed in the first 4 hours. Higher

perchlorate was detected in urine with average concentrations of 351 and 490

μg/mL in the 4- and 8-h groups, respectively, followed by plasma (48.4 and 40.2

μg/mL, respectively), thyroid (12.4 and 8.1 μg/g, respectively), feces (2.1 and 2.2

μg/g, respectively), liver (1.5 and 1.8 μg/g, respectively), and kidney (0.9 and 0.7

μg/g, respectively) (Figure 4.1). Compared to the 4-h group, average perchlorate

concentration increased 39.6% in urine in the 8-h group. Average perchlorate

concentration also increased in liver and feces: 20.5% and 8.8% in the 8-h group,

respectively. However, average perchlorate concentrations decreased in

plasma, thyroid, and kidney in the 8-h group.

Perchlorate mass in kidney, thyroid, blood, and urine was significantly

correlated with perchlorate intake with r2 = 0.82, 0.78, 0.73, and 0.62,

respectively (p < 0.01). Perchlorate residues relative to perchlorate intake were

calculated in the fluids and tissues, and it was observed that perchlorate can be

excreted primarily through urine with an average recovery of 34% and 88% in 4-

and 8-h exposure groups, respectively (Table 4.1). There was a significant

difference of perchlorate recovery in urine between the two groups (p = 0.0129).

Higher perchlorate urinary excretion rate occurred between 4 and 8 h because

54% perchlorate was recovered at that period. Compared to perchlorate

recovery in urine, little perchlorate was observed in blood, liver, kidney, thyroid,

80

and feces; there were no significant differences between the two groups

regarding recovery of perchlorate in blood, liver, thyroid, kidney, and feces.

Perchlorate recovery in liver was increased slightly in the 8-h group when

compared to the 4-h group. However, perchlorate recovery decreased in blood,

thyroid, kidney, and feces in the 8-h group. It seems to indicate that a major

portion of perchlorate was depleted in these tissues and blood before 8 hours,

and maybe even before 4 hours. Among the tissues tested, more perchlorate

(expressed as mass percentage of perchlorate intake) was found in the liver

rather than the thyroid.

Our preliminary results are consistent with many other studies (Goldman

and Stanbury, 1973; Peña et al., 1976; Batjoens et al., 1993; Greer et al., 2002;

Yu et al., 2002). It is well established that a major portion of perchlorate can be

excreted from the body into the urine, and in a relatively short time. For example,

Batjoens et al. (1993) reported that perchlorate was excreted in the urine at a

concentration-related elimination rate after oral administration of a single dose in

cattle. Yu et al. (2002) reported 99.5% perchlorate was recovered in urine, and

95% of injected perchlorate was cleared in 55 to 75 h in rats. Greer and

coauthors (2002) documented that the half-life of perchlorate elimination in

humans ranged from 6 to 9.3 h following perchlorate withdraw. Peña et al.

(1976) reported that 87.6% and 99.4% of 36ClO4 were recovered in the excreta 3

h and 24 h, respectively, after administration in laying hens. Goldman and

Stanbury (1973) reported that 36ClO4 disappeared from the body with a half-life of

approximately 20 h in rat. The authors also reported that the disappearance

rates of 36ClO4 from the plasma and thyroid, and the appearance rate in the urine,

corresponded closely.

In this study, the thyroid had the highest perchlorate concentration among

the tested tissues, which was similar to findings of many other studies (Anbar et

al., 1959; Goldman and Stanbury, 1973; Peña et al., 1976; Yu et al., 2002).

However, the liver had a higher perchlorate residue, expressed as a mass

81

percentage of perchlorate intake, than thyroid and kidney. This phenomenon

was also observed in other studies. Peña et al. (1976) reported the highest

concentration of perchlorate in thyroids of laying hens at 3 h after a single

injection of K36ClO4. In their study, the largest total deposition (amount) in the 3-

h group was found in the blood, followed by muscle, internal organs, and ova.

Because of the limited number of tissues tested in this study, the perchlorate

distribution pattern among tissues was not always consistent with other studies;

however, there have also been inconsistencies among studies regarding

perchlorate distribution in body tissues. Yu et al. (2002) reported the highest

perchlorate concentration was in the thyroid, followed by plasma, red blood cells,

kidney, skin, liver, spleen, and muscle at 0.5 h post-dosing in rats; this is not

consistent with the Pena et al. study (1976), or the Goldman and Stanbury study

(1973). Variation may be caused by the differences in dosage, dose route,

tested animal, exposure duration, sampling time, and analysis method, which all

could change perchlorate toxicokinetics.

In this study, the perchlorate mass balance was not completely closed,

particularly in the 4-h group. There are at least two reasons for this observation.

First, the amounts of urine and blood collected were less than the total amount of

each fluid in the animals. Collected blood may only represent one quarter of the

total blood in the animals. For the 4-h group, urine volume was much less than

the 8-h group because there was less urine production. Since perchlorate

concentration was highest in urine, small volume differences can result in huge

differences in terms of perchlorate mass. Therefore, the total perchlorate

recovery in the 4-h group was only 36%, while 90% was recovered in the 8-h

group. Secondly, we only determined perchlorate in urine, blood, thyroid, liver,

kidney, and feces. Perchlorate may also exist in the gastrointestinal tract and

contents, especially in the 4-h group. Perchlorate may also be taken up by other

organs such as skin. Therefore, it is possible that more than 90% of the dose

82

could be recovered into urine if longer time (> 8 h) was used to excrete

perchlorate from the body.

Perchlorate excretion pattern via urine in deer mice

No significant difference was found among treatments regarding water

consumption (on average, 7.97 ± 2.7 g/day) and body weight (on average, 18.46

± 1.8 g) of the deer mice at each sampling time over the 28 d exposure.

Perchlorate daily dosage was estimated as 6.25 ± 0.6, 72.64 ± 5.8, and 778.06 ±

82.2 ng/g bw in the low, medium, and high dose groups, respectively.

Perchlorate concentrations in the urine increased rapidly after initiation of

exposure in the medium and high dose groups. Perchlorate concentration

appeared to reach a steady state within one day in all groups (Figure 4.2).

Higher concentrations of perchlorate were found at 16 h and day 2 in all

treatments, which is consistent with the fact that urine collected at 16 h and day 2

was mainly produced during the night and deer mice are nocturnal animals with a

higher excretion at night. Perchlorate concentrations in urine were significantly

correlated with perchlorate intake over the exposure period with an average of r2

> 0.6899 and p < 0.001. There was a significant treatment difference (p < 0.001)

among the three dose groups in terms of perchlorate concentration in urine over

the exposure period (28 days), with significant differences beginning at the first

urine collection time (8 h) (Figure 4.2). Even though perchlorate was detected in

the control group (31.4 ± 28.7 ng/mL), there was still a significant difference

between the control and low dose groups at days 7 and 20 (p < 0.005) but not on

days 14 and 28 (p > 0.1). Trace perchlorate was detected in the tap water (0.65

ng/mL) and rodent chow (1.44 ng/g). Perchlorate was also detected in the dish-

washing solution. A “background” intake of perchlorate has been previously

reported because of low-level perchlorate in some feed and water (Capuco et al.,

2005). Assuming a deer mouse ate 3 - 5 g food/day (personal communication),

the daily intake would be 4.32 – 7.20 ng perchlorate, much lower than the

83

amount excreted daily via urine (20.65 ± 12.9 ng) in the control animals. Other

sources of perchlorate in the deer mice are not clear, and an endogenous

perchlorate source may exist.

The amount of perchlorate excreted via urine was calculated based on

perchlorate concentrations in urine and the amount of urine collected (which is

assumed to be the amount of urine produced). Based on the amount of the dose

solution (water containing a certain concentration of perchlorate) the deer mice

consumed, the amount of perchlorate intake was similarly estimated. Therefore,

a mass balance of perchlorate intake and output was established. Perchlorate

output through urinary excretion as a mass percentage of perchlorate intake

through dosed drinking water in the deer mice over the 28-d exposure period is

illustrated in Figure 4.3. At 8 h, 22, 29, and 51% of the perchlorate intake

through drinking water was excreted into urine in the high, medium, and low dose

groups, respectively, and these percentages increased to a steady state after

one day. Average perchlorate output through urinary excretion was 46, 46, and

61% of the perchlorate intake through drinking water in the high, medium, and

low dose groups, respectively, over the 28-d exposure. There was no significant

treatment difference regarding perchlorate output through excretion via urine as a

mass percentage of perchlorate intake through dosed drinking water in the deer

mice at any sample collection time.

In the perchlorate distribution experiment in voles, 34 and 88% of

perchlorate was recovered in the urine in the 4- and 8-h exposure groups,

respectively. However, in deer mice only 22, 29, and 51% of the perchlorate

intake through drinking water was excreted into urine after 8 h exposure in the

three different dose groups, respectively. The difference regarding perchlorate

recovery in the urine may be caused by a species difference. Different species

show different absorption, distribution, metabolism, and excretion, which may

produce different excretion rates. Different life traits and other characteristics

may also contribute to different excretion rates of perchlorate. For example,

84

prairie voles usually produce more urine than deer mice, which may result in

more perchlorate recovery in the urine in terms of mass percentage.

Previous studies have shown that a major portion of perchlorate can be

excreted from the body in the urine in a relatively short time (Anbar et al., 1959;

Goldman and Stanbury, 1973; Peña et al., 1976; Batjoens et al., 1993; Greer et

al., 2002; Yu et al., 2002). Therefore a steady state, with more than 80% of

perchlorate excretion via urine in terms of mass percentage, was expected to be

reached in the current study. However, much lower perchlorate output via urine

as a mass percentage of perchlorate intake was found in the current excretion

experiment. Over the 28-d exposure, an average of 46% from the high and

medium and 61% from the low dose group of perchlorate intake were excreted

via urine. Several possible factors may help to explain these observations. First,

in this study, deer mice were exposed to perchlorate-treated drinking water ad

libitum over 28 days. In previous studies, perchlorate was injected as a one-time

dose. Exposure routes can influence uptake, and thus influence excretion of

perchlorate via urine. Continuous oral consumption of perchlorate may cause

accumulation in the body because of retarded elimination and excretion. It is

possible that perchlorate was present in the tissues such as liver and kidney, but

below the detection limit after high dilution during extraction and/or small mass of

these organs. In addition, skin represents a greater percentage of body weight of

rats than humans (19% vs. 3.7%) (NAS, 2005), therefore, more perchlorate could

be retained in deer mouse skin and cause less perchlorate to be excreted via

urine.

Secondly, metabolism of perchlorate within the gastrointestinal tract may

occur. Although there is no evidence for perchlorate metabolism in the body

(Anbar et al., 1959), perchlorate can be reduced to chlorate, chlorite, and

chloride through a series of enzymatic conversions under anaerobic conditions

(Coates et al., 1999). The existence of anaerobic bacteria which express

perchlorate reductase in the environment (Bruce et al., 1999; Coates et al.,

85

1999), isolated and purified perchlorate reductase (Kengen et al., 1999), and

identified gene encoding perchlorate reductase (Bender et al., 2005) support the

possibility that some metabolism takes place in the body. In addition, perchlorate

metabolic conversion has been proposed to occur in cow rumen (Capuco et al.,

2005), based in part on results where the total output of perchlorate was 58, 55,

33, and 17% of total intake for cows in the 0, 0.4, 4, and 40 mg/day infusion

groups, respectively. These output mass percentages were comparable with our

observations. Deer mice do not have a rumen, but this does not exclude the

possibility of perchlorate metabolism in the intestine and cecum by microbial

activities.

Finally, perchlorate may be lost because of incomplete urine collection or

via excretion in feces. Metabolic cages were used to hold these animals during

exposure. Deer mice can move rodent chow easily from the food container to

the mesh because of their small body size relative to the cages. The cage does

not isolate the chow and feces from the urine very well, and as a result, urine

may be absorbed by the chow and feces sticking to the wall of the cage, causing

less urine to be collected. Perchlorate also can be excreted via feces although it

contributes a small portion compared to the urine. Perchlorate was not

detectable in the feces collected at 8 h, which may be because of the high

dilution factor during sample extraction and resulting perchlorate below the

detection limit. Subsequent feces analysis was not continued. Assuming 15%

perchlorate loss because of incomplete urine collection, feces excretion, and

retained in the body, there is still about 40% of the perchlorate that is still

unaccounted for in this study.

Perchlorate intake from food and other sources resulted in a higher

perchlorate intake than intended in this study. The extra amount of perchlorate

accounted for a more significant portion of perchlorate in urine in the low dose

group, while the contribution from the extra perchlorate was negligible compared

to intake in the medium and high dose groups. As a result, slightly higher

86

recovery of perchlorate (expressed as a mass percentage of the total perchlorate

intake through dosed drinking water) was observed in the low dose group at each

collection time and overall. Similarly, much more perchlorate was “recovered” in

the low dose group than the high dose group in the depuration experiment.

Perchlorate depuration via urine in deer mice

Perchlorate depuration patterns were observed in deer mice via urinary

excretion during an 88 h depuration period following an 8 h exposure to

perchlorate in drinking water. No significant difference occurred between the low

and high dose groups in terms of perchlorate “background” concentration in urine

before exposure (21.6 ± 4.4 and 20.9 ± 6.5 ng/mL (mean ± SD) in the low and

high dose groups, respectively) (Figure 4.4). Perchlorate concentrations in urine

increased rapidly after the 8 h exposure to 2X and 122X that of background

perchlorate concentrations in the low and high dose groups, respectively. During

the 88 h depuration period, perchlorate concentrations in urine decreased

dramatically, especially in the high dose group. For the low dose group,

perchlorate concentrations in urine decreased to a steady “background” level

after 16 h of depuration. However, perchlorate concentrations in urine continued

to decrease after 88 h of depuration in the high dose group.

In the evaluation of perchlorate mass balance, it was found that 56.6 ±

14.3% of the perchlorate intake was excreted in the 88 h depuration period in the

high dose group, which was comparable to the percentage of total perchlorate

intake excreted into urine in the high and medium dose groups in our previous

excretion experiment (46%, on average). However, 234.6 ± 57.5% was found in

the low dose group. Daily perchlorate intake through food and other sources

introduced extra perchlorate in addition to the amount from dosed drinking water

and thus resulted in more than 200% recovery of perchlorate via urine. Daily

intake of perchlorate through food also may help explain the leveling of

87

perchlorate concentrations in urine to a steady “background” level after 16 hours

of depuration.

Exponential non-linear regression models were used to predict the half-life

and mean lifetime of perchlorate in deer mice. The models were developed

based on perchlorate concentrations in urine during the depuration period after

subtracting the “background” perchlorate levels (Figure 4.5). Mean background

perchlorate level in the low dose group minus SD (21.6 – 4.4 = 17.2 ng/mL) was

used as the subtracter in the low dose group to avoid negative concentrations in

the late depuration period. Mean background perchlorate level in the high dose

group (20.9 ng/mL) was used as the subtracter in the high dose group. The

models fit the depuration patterns well (r2 = 0. 0.38 and 0.84 in the low and high

dose groups, respectively). The half-lives of perchlorate in the low and high dose

groups were calculated as 37.9 and 18.2 hours, respectively. The mean lifetime

in the low and high dose groups was 54.6 and 26.3 hours, respectively. Our

results were comparable with other previous studies. The half-life of perchlorate

elimination has been reported differently for different species, ranging from 6 to

9.3 hours in humans (Lamm et al., 1999; Greer et al., 2002; Braverman et al.,

2005), and < 8 to 20 h in rats (Goldman and Stanbury, 1973; Wolff, 1998; Fisher

et al., 2000). Slightly longer half-lives obtained in the current experiment may be

caused by the different exposure routes (inhalation, oral, or injection) and one

dose or continuous exposure. In the current study, deer mice were exposed to

perchlorate for 8 h through drinking water. A shorter half-life and mean lifetime of

perchlorate were found in the high dose group when compared to the low dose

group. Greater perchlorate burdens in the body may cause a higher excretion

rate, and therefore a shorter half-life and mean lifetime.

Perchlorate excretion patterns during continuous 96 h exposure to

perchlorate and a depuration patterns with 88 h of depuration after 8 h exposure

were compared (Figure 4.6). No significant difference was found between

depuration and excretion at 8 h regarding perchlorate concentrations in urine in

88

both groups. However, a significant difference was observed between

depuration and excretion from 16 to 96 h in both groups (all p < 0.05).

Perchlorate concentrations in urine decreased after 8 h in the depuration

experiment in both groups, with a steady state reached after one day in the low

dose group and a continuous decrease in the high dose group. In contrast,

perchlorate concentrations in urine increased at 16 h and fluctuated within a

certain range in the excretion experiment in both groups after 16 h.

Mass balance evaluation showed differential perchlorate intake and output

in the excretion and depuration experiments. Perchlorate from the food was

lower than the daily excretion via urine in the control group in the excretion

experiment; more than 200% recovery of perchlorate was found in urine in the

low dose group in the depuration experiment. Thus, there must be other sources

of perchlorate and endogenous perchlorate may exist. On the other hand,

around 46 - 61% of the perchlorate was excreted via urine in the excretion

experiment, and 57% of perchlorate was recovered in urine in the high dose

group in the depuration experiment, resulting in the proposed concept of

metabolism of perchlorate in the body (gastrointestinal tract) (Capuco et al.,

2005). Perchlorate existence in the rodent chow exposed the animals to

perchlorate before initiation of exposure through dosed drinking water.

Therefore, adaptation to perchlorate may occur in these animals, which could

cause the ambiguity in the current study. Perchlorate existence in the food, other

potential sources of perchlorate, and perchlorate metabolism made perchlorate

fate in animals was not as straightforward as reported by other previous studies.

4.4 Conclusions

A study on perchlorate distribution was conducted in male adult prairie

voles (Microtus ochrogaster). Excretion via urine was the major pathway for

perchlorate fate in the body, with much higher concentrations of perchlorate

detected in urine in 4- and 8-hour exposure groups. Perchlorate mass in kidney,

89

thyroid, blood, and urine significantly correlated with perchlorate intake. Using

male adult deer mice (Peromyscus maniculatus), perchlorate excretion and

depuration patterns via urine were tested further. A significant treatment

difference was showed during the 28-day exposure period in terms of perchlorate

concentrations in urine. However, no difference was found among the three

treatments in terms of percentage of total perchlorate intake in urine at each

sampling time over the 28-day exposure period. Both concentrations of

perchlorate and mass percentage in urine leveled to a steady state after one day

in all treatments. On average 46, 46, and 61% of perchlorate were recovered in

urine over the exposure period in high, medium, and low dose groups,

respectively. Exponential non-linear regression models fit the depuration curve

well, particularly for the high dose group (r2 = 0.38 and 0.84 in the low and high

dose groups, respectively); half-lives of perchlorate in deer mice were predicted

as 37.9 and 18.2 hours in the low and high dose groups, respectively.

Metabolism of perchlorate may occur in the body, based in part on the 40%

perchlorate unaccounted for in this study. Other sources of perchlorate were not

clear and endogenous perchlorate may exist. Perchlorate existence in the food,

other potential sources of perchlorate, and perchlorate metabolism made

perchlorate fate in animals was not as straightforward as reported by other

previous studies.

90

100

1000

10000

100000

1000000

Liver Kidney Thyroid Urine Plasma Feces

Tissues

Perc

hlor

ate

conc

entr

atio

n (n

g/g

or n

g/m

L)

4 hr8 hr

Figure 4.1. Perchlorate concentrations (mean ± SE) in fluids and tissues in

prairie voles (Microtus ochrogaster) after 4- or 8-h exposure to 250 μg/mL perchlorate.

Table 4.1. Changes in perchlorate recovery in different tissues of prairie voles (Microtus ochrogaster) after 4- or 8-h exposure. “+” indicates an increase (%) of perchlorate recovery in the 8-h exposure, and “-“ indicates a decrease (%).

Recovery of perchlorate (%) (mean % ± SE) Tissue

4 hours 8 hours Increase or decrease (%)

Urine 33.563 ± 11.079 a 87.671 ±7.774 b +161.22

Plasma 2.087 ± 0.339 1.582 ± 0.267 -24.20

Liver 0.189 ± 0.090 0.223 ± 0.052 +18.15

Thyroid 0.084 ± 0.017 0.061 ± 0.005 -26.96

Kidney 0.035 ± 0.016 0.024 ± 0.013 -30.48

Feces 0.007 ± 0.001 0.006 ± 0.001 -22.44

Different letter: p=0.0129 (t-test)

91

1

10

100

1000

10000

0 5 10 15 20 25 30

Perchlorate exposure time (day)

Perc

hlor

ate

conc

entr

atio

n in

urin

e (n

g/m

L)

control Low Medium High

Figure 4.2. Perchlorate excretion patterns (concentration in urine (mean ± SD))

in deer mice (Peromyscus maniculatus) exposed to perchlorate through drinking water for 28 days. A significant difference was shown among the three treatments (p < 0.01). No significant difference was observed between the control and low dose groups at days 14 and 28. Higher doses corresponded well to higher concentrations of perchlorate detected in the urine. Dosages from low to high were 17, 165, and 1600 ng/mL, respectively.

92

0

0.2

0.4

0.6

0.8

1

1.2

1.4

0 5 10 15 20 25 30

Perchlorate exposure time (day)

Perc

hlor

ate

excr

etio

n (p

erce

ntag

e of

pe

rchl

orat

e in

take

)

LOW

MEDIUM

HIGH

Figure 4.3. Perchlorate urinary excretion as percentage of total perchlorate

intake through dosed drinking water over the 28-day exposure period (mean ± SD). Dosages from low to high were 17, 165, and 1600 ng/mL, respectively. No significant difference was observed among the three treatments at each sampling time over the exposure period.

93

1

10

100

1000

10000

low highGroup

Perc

hlor

ate

conc

entr

atio

n in

urin

e (n

g/m

L)

0 8 h 16 h 24 h 36 h 48 h 72 h 96 h

Figure 4.4. Perchlorate concentration in urine (mean ± SD) in the depuration

experiment in which deer mice (Peromyscus maniculatus) was exposed to perchlorate for 8 hours and followed by an 88 h depuration period. Low dose - 17 ng/mL; high dose - 1600 ng/mL.

94

y = 11.974e-0.0183x

r2 = 0.3759

-15

0

15

30

45

0 20 40 60 80

Depuration time (hour)

Perc

hlor

ate

conc

entr

atio

n in

urin

e (n

g/m

L)

(A)

100

y = 1217e-0.038x

r2 = 0.8384

-500

0

500

1000

1500

2000

2500

3000

3500

4000

4500

0 20 40 60 80Depuration time (hour)

Perc

hlro

ate

conc

entr

atio

n in

urin

e (n

g/m

L)

(B)

100

Figure 4.5. Exponentional non-linear regression models for the depuration

patterns in the low (A) and high (B) dose groups after subtracting the background levels of perchlorate. Predicted half-lives for perchlorate in the deer mice were 37.9 and 18.2 h in the low and high dose groups, respectively. Error bars represent the SD.

95

0

20

40

60

80

100

120

140

160

8 16 24 36 48 72 96

Time (hour)

Perc

hlor

ate

conc

entr

atio

n in

urin

e (n

g/m

L)

depurationexcretion

(A)

0

1000

2000

3000

4000

5000

6000

7000

8 16 24 36 48 72 96

Time (hour)

Perc

hlor

ate

conc

entr

atio

n in

urin

e (n

g/m

L)

depurationexcretion

(B)

Figure 4.6. Comparison of perchlorate excretion and depuration patterns within

96-h in deer mice (Peromyscus maniculatus ). Error bars represent the SD. Low dose (A) was 17 ng/mL. High dose (B) was 1600 ng/mL. For depuration experiments, deer mice were exposed to perchlorate for 8 h and then withdraw from perchlorate for 88 h; for excretion experiments, deer mice were exposed to perchlorate for 96 h.

96

CHAPTER V

EFFECTS OF PERCHLORATE EXPOSURE ON SODIUM-IODIDE

SYMPORTER AND PENDRIN GENE EXPRESSION IN DEER MICE

5.1 Introduction

The sodium-iodide symporter (NIS) is a basolateral plasma membrane

transporter which mediates active uptake of iodide into thyroid follicular cells.

Iodide uptake by the thyroid is the crucial first step in thyroid hormone

biosynthesis. NIS couples the inward transport of two sodium (Na+) ions with the

inward transport of one iodide (I-) ion into the cell (Eskandari et al., 1997; De La

Vieja et al., 2000). The translocation of Na+ into the cell occurs in favor of its

electrochemical gradient, which is maintained by the Na+/K+ ATPase. The

inwardly directed Na+ gradient drives iodide uptake against its electrochemical

gradient.

As I- can be transported and accumulated into many other tissues in

addition to the thyroid, such as skin, salivary glands, gastric mucosa, and

lactating mammary gland, the presence of NIS in extra-thyroidal tissues has also

been reported. NIS has been detected in rat gastric mucosa using

immunocytochemistry, and NIS mRNA has been detected in rat brain, stomach,

skin, mammary gland, placenta, uterus, and ovary using reverse-transcriptase

polymerase chain reaction (PCR) technique (Kotani et al., 1998). In humans,

immunohistochemical assays revealed positive NIS protein expression in salivary

glands and stomach but not in pancreas, skin, ovaries, spleen, esophagus, small

intestine, appendix, and kidney (Vayre et al., 1999). Northern blot analysis and

reverse transcription-PCR followed by Southern hybridization revealed NIS RNA

expression in several extrathyroidal tissues in humans, including the salivary

gland, parotid gland, submandibular gland, pituitary gland, pancreas, testis,

mammary gland, gastric mucosa, prostate, ovary, adrenal gland, heart, thymus,

and lung (Spitzweg et al., 1998; Spitzweg et al., 1999b). NIS gene expression

97

was also reported in human kidney and placenta with reverse transcription-PCR

(Mitchell et al., 2001; Spitzweg et al., 2001). Controversies were shown on the

localization of extrathyroidal NIS among different mammals (i.e. man, pig,

guinea-pig, rat and mouse) (Josefsson et al., 2002). In addition, the conflicting

reports on the location of NIS protein or gene expression may be caused by

different methodologies and their sensitivity (Josefsson et al., 2002).

Expression of NIS protein in the thyroid is upregulated by thyroid

stimulating hormone (TSH) in vivo and in the FRTL-5 cell (De La Vieja et al.,

2000; Dohan et al., 2000; Dohan et al., 2003). Besides TSH, I- has also been

considered as a main factor regulating NIS activity in the thyroid. The well-

known acute Wolff-Chaikoff effect shows that organic binding of I- in rat thyroid

was blocked when [I-] in plasma levels reached a crucially high threshold (Wolff

and Chaikoff, 1948). The ensuing escape from the acute effect, in which the

level of organification of I- is restored, was explained as a decrease in I- uptake

possibly caused by down-regulation of NIS expression (Braverman and Ingbar,

1963; Eng et al., 1999; Dohan et al., 2000). Administration of iodide has been

reported to decrease NIS mRNA and/or protein levels in in vivo and in vitro

studies although with different proposed mechanisms (Spitzweg et al., 1999a;

Eng et al., 2001; Dohan et al., 2003). In addition to TSH and I-, cytokines were

also reported to play a role in the modulation of NIS gene expression (Ajjan et al.,

1998a; Ajjan et al., 1998b; Spitzweg et al., 1999a; Eng et al., 2001). These

cytokines, including TNF-α, TNF-β, interferon-γ, IL-1α, IL-1β, and IL-6, exerted

an inhibitory effect on NIS expression and iodide uptake in thyroid cells.

Prolactin and insulin also stimulated NIS accumulation in mammary gland and

was associated with enhanced iodide accumulation in milk (Rillema et al., 2000;

Rillema et al., 2002).

NIS transports a wide variety of anions in addition to I-. These include

ClO3-, SCN-, SeCN-, NO3

-, Br-, BF4-, IO4

-, and BrO3-; NIS shows different affinity

for these anions with I- ≥ SCN- > ClO3- > Br- (Dohan et al., 2000; Van Sande et

98

al., 2003). Perchlorate has been characterized as the most widely known

inhibitor of thyroidal iodide uptake; it competitively inhibits iodide uptake by the

thyroid via NIS, and therefore disrupts normal thyroid function. Perchlorate was

previously proposed to be transported via NIS, but it action as a blocker rather

than a substrate for NIS was proposed as well (Eskandari et al., 1997; De La

Vieja et al., 2000).

Alternatively, a mechanism of perchlorate-induced toxicity in the thyroid

has been proposed in which perchlorate stimulates the discharge of iodide from

the follicles (Wolff, 1998). Pendrin, an iodide/chloride transporter found in the

apical membrane, has been suggested as a possible participant in the efflux of

iodide into the follicular lumen (Wolff, 2005). In the thyroid, perchlorate seems to

facilitate the efflux of iodide from the follicular lumen and cytoplasm (Wolff, 1998).

No report has described the mechanism of iodide transport at the apical

membrane. Therefore, it is possible that perchlorate alters the transport function

of pendrin at the apical membrane by an unknown mechanism.

Pendrin is a putative 780-amino acid transmembrane glycoprotein

containing 11 and 12 transmembrane domains and is encoded by the Pendrin

syndrome gene (PDS) (Everett et al., 1997; Royaux et al., 2000). Mutations of

the PDS gene are responsible for Pendred syndrome, which is characterized by

congenital hearing loss and goiter (Everett et al., 1997). Pendrin has been

demonstrated to transport iodide and chloride, but not sulfate, in a sodium-

independent fashion in Xenopus laevis ooctyes and Sf9 cells (Scott et al., 1999).

Pendrin also acts as chloride/formate and chloride/hydroxyl/bicarbonate

exchangers in the mammalian kidney, Xenopus laevis oocytes, and HEK-293 cell

expression systems (Scott and Karniski, 2000; Royaux et al., 2001; Soleimani et

al., 2001). In the thyroid, pendrin is an iodide-specific apical transporter and is

responsible for iodide efflux from thyroid cells (Royaux et al., 2000; Yoshida et

al., 2002).

99

Generally, pendrin displays a different pattern of expression than NIS in

extra-thyroidal tissues. Pendrin expression has been reported primarily in the

thyroid (Bidart et al., 2000b; Royaux et al., 2000; Mian et al., 2001), kidney

(Lacroix et al., 2001; Royaux et al., 2001; Soleimani et al., 2001), and inner ear

(Everett et al., 1999; Yoshino et al., 2004). It has also been found in other

tissues such as human sertoli cells (Lacroix et al., 2001), the endometrium of

humans, rats, and mice (Suzuki et al., 2002), lactating mammary glands (Rillema

and Hill, 2003), and the placenta during gestation (Bidart et al., 2000a). In

contrast to NIS, no transcription of the PDS gene was detectable in any digestive

tract tissue (Lacroix et al., 2001). In deer mice, a much higher expression of

pendrin was found in kidney, lungs, and reproductive tissues (including testes,

ovaries, and uterus) than in the thyroid (Ramachandran, 2005).

Pendrin expression was found to be significantly induced by low

concentrations of thyroglobulin (TG), but not by TSH, sodium, iodide, or insulin in

a rat thyroid cell line (FRTL-5) (Royaux et al., 2000). At physiological

concentrations, the combination of prolactin, insulin, and cortisol stimulated

pendrin expression in cultured mammary tissues taken from midpregnant mice

(Rillema and Hill, 2003). Prolactin stimulation of iodide transport was abolished

by pendrin exchange inhibitors in cultured mammary tissues from 12- to 14-day

pregnant mice (Rillema and Hill, 2003). In the kidney, pendrin expression was

regulated by the acid-base status with acid-loading or K+-depletion causing a

reduction and bicarbonate-loading producing an increase in pendrin expression

level (Wagner et al., 2002; Frische et al., 2003; Quentin et al., 2004). Chronic

changes in the chloride balance have played a role in the regulation of pendrin

expression in the kidney (Quentin et al., 2004). Pendrin was also reportedly

upregulated with aldosterone analogues (deoxycorticosterone pivalate) and

therefore may be critical in the pathogenesis of mineralocorticoid-induced

hypertension and metabolic alkalosis (Verlander et al., 2003) in addition to being

100

essential for acid-base balance. Pendrin also plays a vital role in the renal

conservation of water and Cl- during NaCl restriction (Wall et al., 2004).

Although perchlorate has been most commonly characterized as being

transported into the thyroid, it appears to be eliminated rapidly and primarily via

urine in humans, rats, and cows (Anbar et al., 1959; Goldman and Stanbury,

1973; Batjoens et al., 1993; Yu et al., 2002). It is uncertain whether NIS is

expressed in the kidney due to conflicting reports (Vayre et al., 1999; Spitzweg et

al., 2001), but pendrin has been found to be highly expressed in deer mice

kidney (Ramachandran, 2005). Therefore, it is possible that perchlorate

exposure may have an effect on transporter (i.e. NIS and/or pendrin) levels or

activities in the kidney; higher levels or activities of these transporters in the

kidney may correspond to high levels of perchlorate exposure and in turn, a

higher perchlorate excretion rate via urine. The relationship between perchlorate

exposure and transporters in the kidney (or other organs) would provide

additional insight about the perchlorate excretion pattern via urine and some

indication of perchlorate toxicokinetics at the molecular level. To our knowledge,

no information describes the relationship of perchlorate and (regulation of) NIS

expression. Therefore, the relationship between transporter gene expression

and perchlorate exposure was explored in the current study. Four questions

were addressed: (1) What is the expression pattern of NIS among deer mice

tissues? (2) How does perchlorate exposure affect NIS gene expression in the

kidney if the NIS gene is expressed in the kidney? (3) How does perchlorate

exposure affect pendrin gene expression in the kidney? (4) Is there a

relationship between urinary perchlorate excretion and NIS (and/or pendrin) gene

expression in the kidney? It was hypothesized that the transporter gene

expression would increase upon perchlorate exposure.

101

5.2 Materials and Methods

Animal exposure to perchlorate

Animals: Male adult deer mice (Peromyscus maniculatus) (18.5 ± 1.8 g)

were provided by Dr. Michael Hooper’s breeding colony at Texas Tech University

(Lubbock, TX). Perchlorate dosing solutions were prepared by dissolving sodium

perchlorate (reagent grade anhydrous, Fisher Scientific) in 18.2 MΩ Milli-Q

water. The concentration of perchlorate was determined using ion

chromatography.

Perchlorate excretion experiment: Twenty-four deer mice were arbitrarily

assigned into four groups (one control and three treatments: low, medium, and

high). Siblings were separated into different groups. Animals were housed

individually in metabolic cages which were placed on two racks (4 groups X 3

cages per rack). Cages were placed arbitrarily on the racks. Animals (around

70-d old) were allowed to acclimate for two days to test facility conditions of room

temperature (65 – 78 °F), relative humidity (25 – 75%), and 12:12 h dark:light

prior to the initiation of dosing. Food was provided ad libitum and checked daily.

Perchlorate was administered daily through drinking water ad libitum for four

consecutive weeks (sodium perchlorate dissolved in Milli-Q water). The mice in

the control group were given Milli-Q water without perchlorate throughout the

study. Water consumption was recorded at each urine collection. Perchlorate

concentrations in the dosed drinking water (low, medium, and high) were 17,

165, and 1600 ng/mL, respectively. Deer mice body weights were recorded

before treatment and each week during the exposure to monitor any weight

changes. Metabolic cages were replaced (cleaned) once per week.

Perchlorate elimination experiment: Twelve deer mice were arbitrarily

assigned into two groups (low dose and high dose). As in the excretion

experiment, siblings were placed in different groups. Animals were housed

individually in metabolic cages placed arbitrarily on one rack. Animals were

housed in the cages and allowed to acclimate for two days to test facility

102

conditions of room temperature (65 – 78 °F), relative humidity (25 – 75%), and

12:12 h dark:light prior to the initiation of dosing. Perchlorate was dosed through

drinking water over 8 hours (sodium perchlorate dissolved in Milli-Q water). After

dosing, Milli-Q water (no perchlorate) was provided to these animals throughout

the remainder of the study (88 hour, elimination phase). Food and water were

provided ad libitum and checked daily. Water consumption was recorded at each

urine collection. Perchlorate concentrations in the drinking water were 17 and

1600 ng/mL in the low and high dose groups, respectively. Deer mice body

weights were recorded before treatment and at the end of the study (day 4).

At end of the excretion and elimination experiments (days 29 and 4,

respectively), animals were weighed, anesthetized with carbon dioxide, bled via

heart puncture, and euthanized by exsanguination. At necropsy, the following

tissues were collected: kidney, liver, intestinal tract, stomach, and testes. Brain,

heart, and lung were also collected from the control animals to characterize the

NIS gene expression pattern among these tissues. Blood samples were placed

in EDTA-rinsed tubes, centrifuged, and plasma was collected. All tissues,

plasma, and carcasses were placed on dry ice after collection and during

transport back to the laboratory, where they were stored at – 80 °C until gene

expression analysis.

Partial deer mice NIS cDNA sequence development

Ribonucleic acid (RNA) isolation: Since NIS expression has been

reported in rat stomach and human gastric mucosa (Kotani et al., 1998; Spitzweg

et al., 1998; Spitzweg et al., 1999b), gastrointestinal track and stomach were

used to isolate total mRNA. TRIzol Reagent (Invitrogen, Carlsbad, CA) was used

to flush contents out from fresh gastrointestinal tract and stomach from a control

deer mouse. Tissue (50-100 mg) was homogenized in 1 mL TRIzol Reagent

using a PowerGen 125 homogenizer (Fisher Scientific, Pittsburgh, PA) and

incubated for 5 minutes at room temperature. Chloroform was added (0.2 mL

103

per mL of TRIzol Reagent), shaken vigorously, and incubated at room

temperature for 5 min. The samples were centrifuged at 12,000 rpm for 15 min

at 4 °C (Microfuge 22R Centrifuge, Beckman Coulter, Fullerton, CA). The top

aqueous phase was transferred to a fresh microtube. Isopropyl alcohol was

added (0.5 mL per mL of TRIzol Reagent), shaken, and incubated for 10 min at

room temperature. The samples were then centrifuged at 12,000 rpm for 10 min

at 4 °C. The supernatant was decanted and the pellet was washed with 75%

ethanol (at least 1 mL per mL TRIzol Reagent). The samples were centrifuged at

9,600 rpm for 5 min at 4 °C. The supernatant was decanted and the pellets were

air dried for 5 min and dissolved in 50 μL nuclease-free water (Ambion, Austin,

TX). Aliquots (1 μL) were used to spectrophotometrically measure RNA

concentration based on the absorbance (260 nm) using a SmartSpec 3000

spectrophotometer (Bio-Rad, Hercules, CA). The remaining RNA was aliquoted

and stored at -80 °C.

RNA purity and integrity: RNA of high quality (i.e. purified and intact RNA)

is the first and most critical requirement for gene expression analysis. Good

purity of isolated RNA was determined by the absorbance ratio at 260 nm and

280 nm (range = 1.8 - 2.1). Integrity of total RNA was assessed by running the

isolated RNA sample on a 1% formaldehyde agarose gel with ethidium bromide.

A sample solution containing 1 μg RNA, 8-10 μL of formaldehyde loading buffer,

and 0.3 μL ethidium bromide was prepared for each sample and incubated for 17

min at 75 °C. An 18S + 28S Ribosomal RNA marker from calf liver (Sigma-

Aldrich, St. Louis, MO) was also prepared in the same way. Sample or marker

solution (~ 10 µL) was loaded on a 1% denaturing formaldehyde agarose gel and

electrophoresis was performed at 80 volts for one hour in 1X MOPS-EDTA-

Sodium Acetate (MESA) buffer. A photograph of the gel was taken under ultra-

violet (UV) light with a DigiDoc-It imaging system in combination with TFM-20

transilluminator and Dot it 1-d analysis software (UVP, Inc., Upland, CA). Intact

104

total RNA on such a denaturing gel has two sharp and clear bands: 28S and 18S

ribosomal RNA (rRNA) with an intensity ratio of 1:2 as shown in Figure 5.1.

Deoxyribonuclease (DNase) treatment: A DNA-free kit (Ambion, Austin,

TX) was used to remove genomic deoxyribonucleic acid (DNA) from isolated

RNA. An aliquot of 10 µg of RNA sample was taken and nuclease-free water

was added to a total volume of 25 µL. DNase buffer (10X, 2.5 µL) and 1 µL

rDNase was added and the sample was incubated for 25 min at 37 °C. DNase

Inactivation Reagent (2.5 µL) was added and incubated for 2 min at room

temperature, mixing occasionally, to remove the DNase and divalent cations,

which will affect reverse transcription PCR. The sample was centrifuged at

10,000 rpm for 2 min at room temperature. The supernatant, DNA free RNA,

was transferred to a fresh tube without touching the pellet. Aliquoted sample was

stored at - 80 °C or used for reverse transcription.

Reverse transcription: DNA free RNA sample (2 μg) aliquot was used for

reverse transcription using a RETROscript Kit (Ambion, Austin, TX). Oligo (dT)

(2 μL) was added to the sample and nuclease-free water was added to a final

volume of 12 μL. Heat denaturation of RNA was conducted at 75 °C for 3 min.

RT buffer (10X, 2 μL), 4 μL of dNTP mix, 1 μL of RNase Inhibitor, and 1 μL of

Moloney Murine Leukemia Virus-Reverse Transcriptase (MMLV-RT) were added

and the sample was incubated at 43 °C for one hour for reverse transcription.

Finally, the sample was incubated at 92 °C for 10 min to inactivate the Reverse

Transcriptase. The generated complementary DNA (cDNA) was stored in -20

°C.

Polymerase chain reaction (PCR): PCR was conducted using the

RETROscript Kit (Ambion, Austin, TX) based on its protocol. Five μL of the

reverse transcription product (cDNA) was used as template, 5 μL 10X PCR

buffer, 2.5 μL dNTP mix, and nuclease-free water were added to a final volume

of 50 μL. Sense primer (1.25 μL), antisense primer (1.25 μL), and 2 units

SuperTaq polymerase were then added into the reaction solution. PCR was

105

conducted in a Biometra TGradient thermocycler (Whatman Biometra,

Goettingen, Germany) for 30 cycles of denaturation (94 °C, 30 sec.) annealing

(66.5 °C, 30 sec.), and extension (72 °C. 45 sec.), with an initial denaturation at

95 °C for 2 min and a final extension at 72 °C for 5 min. The PCR products were

analyzed on a native 2% agarose gel in the presence of ethidium bromide.

Electrophoresis was performed at 120 volts for 25 min in 1X tris-acetate-EDTA

(TAE) buffer. A photograph of the gel was taken under ultra-violet (UV) light with

DigiDoc-It imaging system combining TFM-20 transilluminator and Dot it 1-d

analysis software (UVP, Inc., Upland, CA).

Sense and antisense oligonucleotide primers were designed using

VectorNTI primer design software (Infomax, Bethesda, MD) with partial NIS

cDNA sequence in Mus musculus (AF235001), and synthesized by Invitrogen

(USA). The partial Mus musculus NIS sequence showed a high similarity to

other species including Homo sapiens and Rattus norvegicus. The generated

sense and antisense oligonucleotide primers are listed in Table 5.1.

Sequencing: Deer mice NIS PCR products were sequenced for

identification and verification using ABI PRISM 377 automated DNA sequencer

(Perkin-Elmer Applied Biosystems, Foster City, CA) at the DNA sequencing core

facility, Biotechnology Center, Texas Tech University. Deer mice specific partial

NIS sequence was then analyzed for homology to previously characterized

genes deposited in the National Center for Biotechnology Informatics (NCBI)

database using the Basic Local Alignment Tool (BLAST) program.

Characterization of gene expression in deer mice with real-time PCR

cDNA preparation: Total RNA was extracted from deer mice tissues as

described above using TRIzol Reagent. The isolated mRNA samples were

assessed for their purity and integrity. High quality RNA samples were DNase

treated and reverse transcribed to cDNA.

106

Primers and probe design for real-time PCR: With the deer mice specific

partial NIS cDNA sequence generated, the TaqMan specific probe and

oligonucleotide primers for NIS mRNA equivalents real-time PCR amplification

were designed by Primer Express software (Perkin-Elmer Applied Biosystems,

Foster City, CA) following Applied Biosystem’s probe and primers design

guidelines. Additional rules were also considered to determine probe and primer

sets such that no more than 2 Gs or Cs in the last 5 nucleotides at the 3’ end for

both primers, the melting temperature (Tm) of the primer was 10 ° lower than the

probe, and no more Cs than Gs in the probe. Designed TaqMan probes and

primers specific for deer mice NIS gene were synthesized by Applied Biosystem.

FAM (6-carboxyfluorescein) was used as the 5’ reporter dye and TAMRA (6-

carboxytetramethylrhodamine) as the 3’ quencher dye for the TaqMan probe.

Similarly, TaqMan probes and primers for pendrin mRNA equivalents real-

time PCR were also designed and synthesized based on the deer mice specific

partial pendrin cDNA sequence developed in Dr. Ernest Smith’s lab

(Ramachandran, 2005). TaqMan probes and primers for pendrin and NIS mRNA

equivalents real-time PCR amplification are shown in Table 5.2.

Real-time PCR: Quantitative real-time PCR analysis was performed using

an ABI PRISM 7000 Sequence Detection System (Perkin-Elmer Applied

Biosystems, Foster City, CA). Real-time fluorescence-based quantitative PCR

has been established as a benchmark technology for the quantification of nucleic

acids and/or comparison of RNA levels (Bustin, 2005; Bustin et al., 2005). Real-

time PCR monitors the fluorescence emitted during the reaction as an indicator

of amplification during each PCR cycle (Ginzinger, 2002; Dorak, 2005). It

quantitates the initial amount of the template specifically, sensitively, and

reproducibly, rather than just detecting the amount of final amplified product at

the endpoint as in conventional PCR. Therefore, it provides a highly sensitive

and accurate quantitative analysis of gene expression with cDNA as the reaction

template.

107

Since real-time PCR is a very sensitive method for gene quantification,

careful normalization is essential for accurate quantitative analysis. Therefore, a

housekeeping gene expression is usually used to normalize the quantification of

target gene expression. A good housekeeping gene should be expressed at a

consistent level under experimental conditions. There are several commonly

used internal housekeeping controls; glyceraldehyde-3-phosphate

dehydrogenase (GAPDH) is one of the most widely used. Most important is that

GAPDH is expressed at consistent levels across age and in various tissues of

deer mouse (Ramachandran, 2005). The probe of GAPDH has VIC as the 5’

reporter dye and TAMRA as the 3’ quencher dye.

A PCR reaction solution with a final volume of 50 μL was prepared that

contained TaqMan GAPDH (250 nM), sense and antisense GAPDH primers (20

nM x 20 nM), sense and antisense pendrin or NIS primers (300 nM x 300 nM), 50

ng of pendrin cDNA or 150 ng of NIS cDNA, and 25 μL of TaqMan Universal

PCR master mix (Perkin-Elmer Applied Biosystems, Foster City, CA). Four 10-

fold serial dilutions of pooled control cDNA samples were used as standards.

Reaction solution without cDNA template was also prepared as the No Template

Controls (NTC). All samples were prepared in duplicate and PCR reactions were

processed in sealed 96-well optical plates.

The program for the real-time thermal cycler was set as the following:

initial 2 min hold at 50 °C for optimal AmpErase UNG activity and a 10 min hold

at 95 °C for AmpliTaq Gold DNA polymerase activation, then 40 cycles of

denaturation (95 °C, 15 sec.) and annealing/extension ( 60 °C, 1 min.).

Fluorescence is released during the amplification reaction and a threshold level

is reached when the fluorescent signal is first statistically significantly above

background. The cycle threshold (CT) value, the cycle number at which the

threshold level is reached, represents the amount of product amplified that is

related to cDNA template concentration. This value was recorded for both the

FAM and VIC dyes in the standard and unknown samples. A calibration curve

108

between the CT values for the FAM or VIC dye of the standards and the amount

of their respective mRNA equivalents was established. Then, the amount of

mRNA equivalents of the target gene and GAPDH in the unknowns was

determined from their CT values based on the calibration curve. The ratio of the

mRNA equivalents of target gene and GAPDH was taken as target gene

expression relative to GAPDH.

NIS gene expression pattern among tissues (i.e. brain, liver, kidney, testis,

large intestine, stomach, heart, and lung) in deer mice was characterized first.

Because NIS gene was observed highly expressed in the stomach, and

perchlorate was mainly excreted via urine, NIS and pendrin gene expressions in

the stomach and kidney were then characterized in deer mice exposed to

perchlorate in the excretion and depuration experiments.

Statistical analyses

Statistical analyses were performed using R software (version 2.1.1, Free

Software Foundation, Boston, MA). Normality and homogeneity of variance of

the data were tested using Sharpiro-Wilk normality test and Bartlett’s test,

respectively. Differences of target gene (i.e. pendrin and NIS) expression in

specific tissues among different perchlorate treatment groups were tested using

one way analysis of variance (ANOVA) followed by Tukey Honest Significant

Difference (Tukey HSD) multiple comparison if a significant difference was

observed. Liner regression analysis was conducted between gene expression in

the kidney and perchlorate excretion via urine. Alpha was set at 0.05 for the

statistical analyses.

5.3 Results and Discussion

Partial sequence of NIS gene

With the sense and antisense primers designed by Vector NIT from partial

NIS cDNA sequence of Mus musculus (Table 5.1), a PCR product with 425 base

109

pairs was amplified from the cDNA of deer mouse small intestine and stomach

(Figure 5.2). The deer mice specific NIS partial sequence (Figure 5.3) shows

87% similarity to the NIS gene in Rattus norvegicus, more than 86% similarity to

the NIS gene in Mus musculus, and 84% similarity to Homo sapiens NIS gene.

To our knowledge, this is the first discovery of a partial NIS cDNA

sequence specific for deer mice. Cloned NIS cDNA was reported in several

species including rat, mouse, and human (Dai et al., 1996; Smanik et al., 1996;

Perron et al., 2001; Pinke et al., 2001), but no complete or partial deer mice NIS

gene sequence has been reported before. To design the primers for PCR

amplification of NIS cDNA in deer mice, complete NIS cDNA sequences of

Rattus norvegicus, Mus musculus, and Homo sapiens were compared using

Vector NIT, and then a specific Mus musculus partial sequence with the highest

similarity to other species was used as a template for the design of sense and

antisense primers.

NIS gene expression pattern in deer mice

The NIS gene was expressed at relatively different levels in various deer

mice tissues (Figure 5.4). The highest expression of NIS was found in the

stomach, which was 11.59 times higher than in the testes, followed by testes,

brain, and large intestine. NIS gene expression levels in the brain and large

intestine were 3.7 and 4.29 times, respectively, lower than that in the testes.

Barely was any NIS gene expression observed in the lung, kidney, heart, and

liver, which was 215, 272, 1070, and 2315 times lower than the expression

observed in testes, respectively. This is the first time to quantitatively analyze

NIS mRNA expression in various deer mice tissues to date.

To some degree, our results were consistent with previous reports on NIS

expression in various rat or human tissues. In extrathyroidal tissues, previous

studies showed NIS mRNA expression in rat brain, stomach, skin, mammary

gland, placenta, uterus, and ovary using the reverse-transcription polymerase

110

chain reaction technique (Kotani et al., 1998). In humans, NIS mRNA was

expressed in salivary gland, parotid gland, submandibular gland, pituitary gland,

pancreas, testis, mammary gland, gastric mucosa, prostate, ovary, adrenal

gland, heart, thymus, and lung using Northern blot analysis and reverse

transcription PCR followed by Southern hybridization (Spitzweg et al., 1998;

Spitzweg et al., 1999b). NIS gene expression was also observed in human

kidney using reverse transcription PCR followed by Southern hybridization

(Spitzweg et al., 2001). .

Previous reports showed NIS mRNA expression in rat stomach and

human gastric mucosa (Kotani et al., 1998; Vayre et al., 1999). Study also

showed that NIS mediated (at least partly) iodide active transport into the rat

gastric mucosa, and perchlorate attenuated gastric iodide transport from the

serosal to the mucosal side (Josefsson et al., 2006). The result that NIS was

highly expressed in the stomach in the current study suggests that iodide could

be secreted into the stomach. Therefore, perchlorate, which has a similar size

and the same charge as iodide, might also be excreted into the stomach. NIS

protein was not detected in human small intestine with an immunohistochemical

method (Vayre et al., 1999). Because of a processing problem, the quality of

isolated small intestine RNA in the current study was not adequate for

quantification, no data can be presented here to characterize or support the

published data on NIS expression level in the small intestine.

In the current study, barely expression of NIS mRNA was found in the

kidney of deer mice. Previous findings on expression of NIS mRNA and proteins

in this tissue have been inconsistent. Absence of NIS proteins was reported in

the human kidney by immunohistochemical methods (Vayre et al., 1999), but

NIS mRNA expression was demonstrated in human kidney with reverse

transcription PCR followed by Southern hybridization (Spitzweg et al., 2001).

Different methodology and corresponding sensitivity may be responsible for

these inconsistent observations. Expression of NIS mRNA in the kidney may

111

also be different among species. Nevertheless, in the current study, NIS mRNA

expression was found at a very low level in the deer mice kidney using real-time

PCR methodology. In contrast, high pendrin mRNA expression in the kidney and

its function as an anion exchanger to maintain electrolyte balance and acid-base

homeostasis has been reported (Royaux et al., 2001; Soleimani et al., 2001;

Ramachandran, 2005). Therefore, pendrin may play a more essential role in the

kidney to reabsorb and excrete base and acid equivalents and maintain

electrolyte balance and acid-base homeostasis in deer mice.

Previous reports revealed that NIS mRNA was expressed in female rat

reproductive tissues including the uterus and ovary (Kotani et al., 1998), but NIS

protein was not found in the human ovary (Vayre et al., 1999). NIS mRNA was

expressed in the human testes (Spitzweg et al., 1998; Spitzweg et al., 1999b). In

the current study, real-time PCR analysis showed that NIS is second highly

expressed in the testes of deer mice, with much higher expression than in other

tissues except the stomach. Regulation of intracellular ions might be important

during spermatogenesis. Thus, studies evaluating the regulation of NIS in the

testes might help to determine NIS function in the testes and therefore, the

relationship of NIS to spermatogenesis.

NIS mRNA was expressed in the deer mice brain although at a low level.

This is consistent with a previous report in which NIS mRNA expression was

demonstrated in rat brain (Kotani et al., 1998). It has been reported that cells

expressing the thyroid stimulating hormone receptor also expressed the NIS

gene (Slominski et al., 2002). Thyroid-stimulating hormone receptor is not

thyroid gland specific; its mRNA or protein expression has been detected in

extrathyroidal tissues such as orbital and adipose tissues (Kishihara et al., 1979;

Endo et al., 1993; Bell et al., 2000). Therefore, it is reasonable that thyroid-

stimulating hormone receptor and in turn, the NIS gene might be expressed in

the brain. As perchlorate can competitively inhibit iodide uptake by the thyroid as

a substrate or blocker, it may also influx into the neuron or block the influx (or

112

efflux) of other ions into (or from) the brain cells. This could affect the brain

especially during brain development. Therefore, studies on the relationship

between perchlorate exposure and NIS gene expression regulation in the brain

would provide some insight on NIS function in the brain and relationship to brain

development.

Effects of perchlorate exposure on NIS gene expression in deer mice

NIS gene expression in the stomach was higher in the three dosed groups

in a dose-dependent manner, but the difference was not statistically significant, in

the excretion experiment in which deer mice were dosed with perchlorate through

drinking for 28 consecutive days (Figure 5.5A). There was no statistically

significant difference between the low and high dose groups in the depuration

experiment in which the mice were exposed to perchlorate for 8 h and then Milli-

Q water for 88 h (Figure 5.5B).

In the kidney, change of NIS gene expression following perchlorate

exposure was similar to the stomach. NIS expression increased in the three

dose groups without a statistically significant difference or dose-dependent

manner in the deer mice exposed to perchlorate for 28 days in the excretion

experiment (Figure 5.6A). No statistically significant difference was observed

between the low and high dose groups in the depuration experiment (Figure

5.6B).

The lack of a significant difference of NIS gene expression in the kidney

was not completely unexpected. Almost all previous studies reported no NIS

gene or protein expression in rat or human kidneys, except one study which

demonstrated NIS expression in human kidney using reverse transcription PCR

(Spitzweg et al., 2001). In the current study, NIS gene expression was barely

detectable in the kidney. With little or no expression in the kidney, perchlorate

exposure and excretion via urine may not affect NIS gene expression in the

kidney. However, increased NIS gene expression in the dosing groups seem to

113

indicate some response to perchlorate exposure and excretion via urine,

although no dose-response relationship or significant difference among doses

was observed.

It is possible that there is no relationship between perchlorate exposure

and/or urinary excretion and NIS gene expression in the stomach and kidney,

(i.e., perchlorate exposure does not affect NIS gene expression, particularly in

the kidney). To our knowledge, there is no published data on perchlorate

exposure and NIS gene expression in extrathyroidial tissues. In addition, NIS is

not the only transporter for perchlorate translocation, especially in extrathyroidial

tissues. In contrast to NIS with a very low expression level in the kidney,

pendrin, a very important anion transporter, is highly expressed in the kidney

(Royaux et al., 2001; Soleimani et al., 2001). Therefore, it is possible that

perchlorate exposure has a more significant effect on pendrin, rather than NIS

gene expression in extrathyroidial tissues, particularly the kidney.

Effects on pendrin gene expression in deer mice

In the excretion experiment, low and high dose perchlorate exposures for

28 days increased pendrin gene expression in the stomach, however, pendrin

gene expression in the medium group was lower than the control (p = 0.064)

(Figure 5.7 A). A large variance within groups was observed in the low and high

dose groups. No statistically significant difference was revealed between the low

and high dose groups in the depuration experiment (Figure 5.7 B).

In the kidney, pendrin gene expression increased in the perchlorate-

treated groups in the excretion experiment, with relatively higher levels in the low

and high dose groups (p = 0.056 and 0.073, respectively) and a relatively lower

level in the medium dose group (Figure 5.8 A). No statistically significant

difference was observed either in the low or high dose groups in the depuration

experiment (Figure 5.8 B).

114

Pendrin expression among different tissues in deer mice has been

previously characterized (Ramachandran, 2005) but without information on

pendrin expression in the stomach. Transcription of the pendrin gene was not

detectable in any human digestive tract tissue (Lacroix et al., 2001). Therefore,

low or trace pendrin gene expression in the stomach could occur, and cause

large variance within groups.

In the depuration experiment, after 88-h depuration with clear Milli-Q

water, perchlorate concentration in urine decreased dramatically, especially in

the high dose group, with average of 22 and 87 µg/mL in the low and high dose

groups, respectively. No significant difference was observed between the low

and high dose groups regarding both NIS and pendrin gene expression in the

kidney, which were also comparable to (no significant difference from) that in the

control group in the excretion experiment. It indicates that these two transporters

gene expression in the kidney came down to a normal level after 88-h depuration

if they were induced at 8 h in the depuration experiment. It is also possible that

these two transporters still are able to handle perchlorate ion urinary excretion

even with high perchlorate burden in the body such as in the high dose group, if

perchlorate was excreted into urine via these two transporters.

Ammonium perchlorate exposure has been shown to alter pendrin gene

expression in deer mice kidney (Ramachandran, 2005), with different alteration

patterns at different mice ages. It seems likely that an animal’s age, dosing

period, and dosage would contribute to different effects of perchlorate on pendrin

gene expression in the kidney. However, compared to the present study, much

higher concentrations of ammonium perchlorate (58.5 and 117 µg/mL) were

used. In addition, high concentrations of ammonium perchlorate also contain

high levels of ammonium ion, which has been shown to have an effect on acid-

base balance and induction of metabolic acidosis (Tizianello et al., 1982; Tanner,

1984; Lina and Kuijpers, 2004). Therefore, use of ammonium perchlorate in the

exposure could complicate attempts to elucidate the effect of perchlorate ion on

115

pendrin gene expression in the kidney. In the current study, deer mice were

exposed to sodium perchlorate, which may have contributed to the different

effect pattern of perchlorate on pendrin gene expression in the kidney and lack of

statistical significance when compared to the study conducted by Ramachandran

(2005).

Several factors maybe have contributed to responsible for the lack of a

significant difference in both NIS and pendrin gene expression in the stomach or

kidney. The first is the fact that these animals were exposed to perchlorate

through food and water, which contained trace levels of perchlorate, before

initiation of the exposure study. Trace amounts of perchlorate exposure before

the dosing experiment may have allowed the deer mice to develop a tolerance to

perchlorate. Therefore, the effect of perchlorate exposure to a higher level

and/or urinary excretion on NIS gene expression may have been compromised.

The second is the fact that only 45 – 61% of the total perchlorate intake from

dosed drinking water was excreted via urine. Perchlorate metabolism or

organification may occur in the body and then result in less perchlorate excreted

via urine. Besides, no statistically significant difference was found among the

three treatments with regards to mass percentage of total perchlorate intake

through drinking water that was excreted via urine over the 28-day exposure.

Finally, high individual variance may result in non-significant differences among

the four dosing groups. Individual differences and a relatively small sample size

resulted in high standard deviations within groups. Thus, no statistically

significant difference was revealed, even though NIS gene expression in the

treatments was higher than the control and varied in a dose-dependent manner

in the stomach.

Is a 28-day exposure period long enough to induce NIS gene expression

and reveal a significant difference among the three treatments in the excretion

experiment? Longer exposure period was used in the study conducted by

Ramachandran (2005), and significant changes of pendrin gene expression was

116

observed in the deer mice kidney. While perchlorate urinary excretion rate was

found higher in the first two day during exposure period, and therefore

transporters gene expression could change significantly in a short time as well.

In the excretion experiment, gene expression was only investigated at day

28, at that time perchlorate urinary excretion rate has been decreased and

reached a steady state. Thus, NIS and pendrin gene expression in the kidney

could also turn back to the normal level and cause no significant increase

observed in the dose groups. Therefore, temporal pattern of NIS and (or)

pendrin gene expression in the kidney along perchlorate exposure period may

reveal a significant increase in certain time, such as at the first two days, in which

perchlorate urinary excretion rate was high. Transporters and their

corresponding mRNA may have different half-lives in the body (or specific

tissues) and then have different response to perchlorate exposure and urinary

excretion. Therefore, if the transporters (proteins) level or activity could be

measured in the current study, it would provide more information and insight on

the mechanism of perchlorate urinary excretion and potential health effect in the

animals.

Linear regression analysis between gene expression in the kidney at day

28 and perchlorate excretion did not show a significant relationship between

gene expression (either NIS or pendrin) in the kidney and perchlorate urinary

excretion mass at day 28, average daily excretion mass, or total excretion mass

over the 28 day exposure. No significant linear relationship was found neither

between NIS in the stomach and kidney, pendrin in the stomach and kidney, or

NIS in the stomach and pendrin in the kidney. It is possible that transporters

gene expression in the kidney and perchlorate urinary excretion at corresponding

time interval could show a significant relationship. However, there is a possibility

as well that perchlorate excretion via urine was facilitated by other transporters

rather than NIS and pendrin.

117

5.4 Conclusions

A partial sequence of deer mice NIS gene cDNA with 425 bps was

elucidated in the current study, which to our knowledge is the first discovery of

the deer mice NIS gene. Quantitative analysis of NIS mRNA expression in

various tissues was also studied for the first time with expression levels from

highest to lowest in deer mice tissues in the following order: stomach, testes,

brain, large intestine, lung, kidney, heart, and liver. Perchlorate exposure

through dosed drinking water for 28 days did not cause a significant effect on NIS

gene expression in the kidney and stomach, or pendrin gene expression in the

kidney, although gene expression increased in the dosing groups and a dose-

dependent manner was observed in NIS gene expression in the stomach. In the

depuration experiment, no significant difference was found between the low and

high dose groups (8-h exposure to perchlorate followed by an 88-h depuration

period) in terms of NIS or pendrin gene expression in the kidney or stomach.

Furthermore, no significant linear relationship was observed between both genes

expression in the kidney and perchlorate urinary excretion in the excretion

experiment. Several factors could complicate the effect of perchlorate exposure

on NIS and pendrin gene expression in the stomach and kidney. In the current

study, these include (1) pre-exposure to trace perchlorate through food and

water, which may have resulted in adaptation (or tolerance) in these animals; (2)

metabolism of perchlorate in deer mice, which may be the reason for only 46 –

61% perchlorate excreted into urine; (3) large differences among individuals and

relatively small sample size. It is also possible that there is no effect of

perchlorate exposure and/or excretion via urine on these two transporters gene

expression, particularly in the kidney.

118

B C DA

28s 18s

Figure 5.1. RNA integrity test on a 1% denaturing formaldehyde agarose gel.

(A) represents 18S + 28S Ribosomal RNA markers from calf liver. (B), (C), and (D) represent RNAs isolated from deer mice tissues (stomach and small intestine mixture, stomach, and large intestine, respectively).

Table 5.1. Sequence of sense and antisense primers designed by VectorNTI

based on Mus musculus NIS mRNA for PCR amplification of deer mice NIS cDNA.

Sequence type Designed sequence

Sense primer CTCATCCTGAACCAAGTGACCGG

Antisense primer TGTAGGCACAGGCCAGGAAGAG

Table 5.2. Sequence of sense primers, antisense primers, and TaqMan probes designed for real-time PCR amplification of pendrin and NIS mRNA equivalents in deer mouse. Gene Sequence type Designed sequence

Sense primer CAGAACCATTCCCGGATCAA

Antisense primer CACGAACGTCCAGAAGGTGTAG NIS

TaqMan probe TGATGGACTTTGACCCCGACCCC

Sense primer GTCCCCAAAGTGCCAATCC

Antisense primer ACTCCTACCACATCCAGGAAGGA Pendrin

TaqMan probe AGCCTGGTGCTGGACTGTGGAGCT

119

A B C2000 bp 1000 bp 500 bp 200 bp 100 bp

Figure 5.2. PCR product (more than 500 bp) from small intestine and stomach

mixture cDNA samples with primers shown in Table 5.1. Lanes (A) and (B) are deer mice specific cDNA samples, and (C) is a marker.

TGGGCGGTATGAAGGCCGTGGTCTGGACAGATGTGTTCCAGGTGGTGGTGATGC

TCAGTGGCTTCTGGGTGATCCTGGCCCGAGGCGCCGTGCTCATGGGGGGCCCCT

GGAACGTGCTCCGGCTCGCCCAGAACCATTCCCGGATCAACCTGATGGACTTTGA

CCCCGACCCCCGGAGCCGCTACACCTTCTGGACGTTCGTGGTGGGCGGCACGCT

GGTGTGGCTCTCCATGTACGGCGTGAACCAGGCCCAGGTGCAGCGCTACGTGGC

CTGCGGCTCGGAGGCCAAGGCCAAGCTGGCCCTGCTCGTCAACCAGCTGGGCCT

CTTCCTGATCGTGGTCAGCGCGGCCGGCTGTGGCGTCGTCATGTTCGTCTTCTACA

AGGACTGCGACCCCCTCCTCAGGGGTCGCATCTCGGCGCCAGAC

Figure 5.3. Partial sequence of deer mouse NIS gene cDNA (425 bps).

120

Figure 5.4. NIS gene expression pattern in various deer mice tissues (L.I.: large

intestine). Insert figure showed NIS gene expression pattern among various tissues except the stomach in deer mice. Boxplots represent the median (M), upper and lower quartiles (Q1 and Q3); whiskers include data points within 3 times the interquartile range (IQR).

Brain Heart Kidney L.I. Liver Lung Stomach Testes

010

2030

40

Tissues

NIS

gen

e/G

AP

DH

gen

e

Brain Heart Kidney L.I. Liver Lung Testes

0.0

0.5

1.0

1.5

2.0

2.5

3.0

Tissues

NIS

gen

e/G

AP

DH

gen

e

121

0

0.5

1

1.5

2

2.5

control Low Medium High

NIS

exp

ress

ion/

GA

PDH

ex

pres

sion

in s

tom

ach

(A)

0

0.5

1

1.5

2

2.5

Low -8h High-8hNIS

exp

ress

ion/

GA

PDH

exp

ress

ion

in s

tom

ach

(B)

Figure 5.5. Effect of perchlorate exposure through drinking water on NIS gene

expression in the stomach of deer mice. A: In the excretion experiment, deer mice were exposed to perchlorate-treated drinking water for 28 days; B: In the depuration experiment, deer mice were exposed to perchlorate for 8 h and then given Milli-Q water for 88 h.

122

0

0.5

1

1.5

2

control Low Medium HighNIS

exp

ress

ion/

GA

PDH

exp

ress

ion

in k

idne

y

(A)

0

0.5

1

1.5

2

Low -8h High-8hNIS

exp

ress

ion/

GA

PDH

exp

ress

ion

in k

idne

y

(B)

Figure 5.6. Effect of perchlorate exposure through drinking water on NIS gene

expression in the kidney of deer mice. A: In the excretion experiment, deer mice were exposed to perchlorate through dosed drinking water for 28 days; B: In the depuration experiment, deer mice were exposed to perchlorate for 8 h and then given Milli-Q water for 88 h.

123

0

1

2

3

4

control Low Medium High

Pend

rin e

xpre

ssio

n/G

APD

H

expr

essi

on in

sto

mac

h

(A)

*

0

0.5

Low -8h High-8h

Pend

rin e

xpre

ssio

n/G

APD

H

expr

essi

on in

sto

mac

h (B)

Figure 5.7. Effect of perchlorate exposure through drinking water on pendrin

gene expression in the stomach of deer mice. A: In the excretion experiment, deer mice were exposed to perchlorate for 28 days (*: p = 0.064 with difference between the control and medium dose groups); B: In the depuration experiment, deer mice were exposed to perchlorate for 8 h and then given Milli-Q water for 88 h.

124

0

0.5

1

1.5

2

control Low Medium High

Pend

rin e

xpre

ssio

n/G

APD

H

expr

essi

on in

kid

ney

(A) †*

0

0.5

1

1.5

Low -8h High-8h

Pend

rin e

xpre

ssio

n/G

APD

H

expr

essi

on in

kid

ney (B)

Figure 5.8. Effect of perchlorate exposure through drinking water on pendrin

gene expression in the kidney of deer mice. A: In the excretion experiment, deer mice were exposed to perchlorate for 28 days (*: p = 0.056 with difference between the control and low dose groups; †: p = 0.073 with difference between the control and high dose groups); B: In the depuration experiment, deer mice were exposed to perchlorate for 8 h and then given Milli-Q water for 88 h.

125

CHAPTER VI PERCHLORATE EFFECTS ON FATTY ACID PROFILES IN MILK

6.1 Introduction

Milk fat is important for growing newborn(s) by acting as an energy source

as well as involvement in some other biological processes (Koletzko and

Rodriguez-Palmero, 1999; Jensen and Heird, 2002; Alessandri et al., 2004;

German and Dillard, 2006). For example, lipids are structural components of all

tissues, and fatty acids are indispensable for cell membrane synthesis. Lipids

comprise 50-60% of the dry weight of the adult brain, of which approximately

35% are in the form of long chain polyunsaturated fatty acids (PUFA), with

arachidonic acid (AA, C20:4n6) and docosahexaenoic acid (DHA, C22:6n3) as

the majority (Wainwright, 2002). Therefore, the early development of highly

specialized organs, such as brain and retinal tissue, requires adequate intake of

fatty acids, especially PUFA (Koletzko and Rodriguez-Palmero, 1999; Alessandri

et al., 2004). PUFA are incorporated into nerve cell membranes, modulate

membrane biophysical properties, and contribute to the functional maturation of

the central nervous system. In addition, PUFA are also involved in the regulation

of neurotransmitter release, synthesis of biologically active oxygenated

derivatives, and gene expression at the transcriptional level during adipocyte

differentiation (Uauy et al., 2000; Uauy et al., 2001; Wainwright, 2002; Alessandri

et al., 2004; Lapillonne et al., 2004).

Lower PUFA intake through milk causes deficiencies in PUFA and could

arrest the development of the immune system, brain, and visual system in infants

(Neville and Picciano, 1997; Heird, 2001; Alessandri et al., 2004). Essential fatty

acid (EFA) deficiency impairs physical and behavioral development, extreme

dietary fatty acid imbalance such as high DHA levels with low ω-6 fatty acids

levels causes growth retardation in mice, and dietary deficiency of ω-3 fatty acids

during development leads to characteristic changes in fatty acid composition in

the brain (Wainwright, 2002). Deficiency of PUFA has been implicated in

126

schizophrenia and attention-deficit hyperactivity disorder in humans (Stevens et

al., 1996; Wainwright, 2002). Alteration of PUFA composition in membranes may

also affect a variety of membrane functions such as effects on ion channels,

transporters, protein binding, cellular and intracellular signal transduction, and

neurotransmission (Wainwright, 2002). As the only food source to infants, milk

quality in terms of fatty acid composition is very important for normal infant

growth and development.

Milk fat synthesis is a multiple step process with the involvement of many

enzymes, such as fatty acid synthase (FAS), which is the key enzyme for de

novo synthesis of fatty acids within mammary epithelial cells for short- and

medium-chain fatty acids (C ≤ 14) (Hadley, 2000). Thyroid hormone (i.e.

triiodothyronine) was reported to stimulate transcription of the fatty acid synthase

gene (Goodridge, 1986; Wilson et al., 1986; Stapleton et al., 1990; Kameda,

1995). Therefore, as a thyroid hormone disruptor, perchlorate may affect de

novo fatty acid synthesis in mammary gland. In addition to the fatty acids de

novo synthesized in mammary gland, other origins of fatty acids for milk fat

synthesis include: from endogenous stored lipids (and/or fatty acids), from

synthesis of lipids (and/or fatty acids) by liver or other tissues, and from the diet

(Neville and Picciano, 1997). Fatty acids for milk fat synthesis, especially the

long chain fatty acids with C ≥ 16, can be derived from these lipids from the

circulation system through hydrolysis regulated by lipoprotein lipase (LPL),

another important enzyme for milk fat synthesis (Hamosh et al., 1972; Iverson et

al., 1995). It has been reported that de novo fatty acid synthesis in the liver and

triglycerides synthesis in both the liver and kidney were reduced in the presence

of KClO4 in sea lamprey (Kao et al., 1999). Therefore, circulating lipids or non-

esterified fatty acids could decrease and result in less fatty acids available to the

mammary epithelial cells for milk fat synthesis, particularly the long chain fatty

acids (C ≥ 16).

127

Hormones such as prolactin also play important roles in mammary gland

development and milk synthesis and secretion. Prolactin has been postulated to

regulate activation of LPL in the mammary gland (Neville and Picciano, 1997;

Hadley, 2000). Prolactin may also affect the activity of FAS. Induction of

prolactin requires thyroid hormones (Hadley, 2000). Therefore, as a thyroid

hormone disruptor, perchlorate may eventually have an influence on the fatty

acid composition of milk fat through interference with prolactin and in turn, the

function of LPL and (or) fatty acid synthase. Interruption in thyroid hormone

production by perchlorate may also change fatty acid profile in milk fat through

affecting fatty acid de novo synthesis in other tissues such as the liver in addition

to the mammary gland. The proposed relationship between fatty acid profile in

milk fat and perchlorate is illustrated in Figure 6.1.

Perchlorate has been well documented as a thyroid hormone disruptor

since it competitively inhibits iodide uptake by the thyroid gland, and as a result,

interferes with normal thyroid hormone production. In mammals, this interference

affects development, growth, and metabolism (Wolff, 1998; Soldin et al., 2001).

Environmental contamination of perchlorate in the United States appears to be

widespread, especially in aqueous systems, because of its use in military

munitions and other industrial products, such as rocket propellant, fireworks, and

air bags, and also its natural occurrence (Susarla et al., 1999; Urbansky et al.,

2001; Dasgupta et al., 2005; Jackson et al., 2005a). Perchlorate can be taken up

into plants and animals (Siglin et al., 2000; Smith et al., 2001; Smith et al.,

2004b; Tan et al., 2004; Yu et al., 2004; Jackson et al., 2005b; Sanchez et al.,

2005). Humans can be exposed to perchlorate through drinking water or trophic

transfer. Recently, reports indicated that perchlorate occurs in supermarket milk

samples and human milk, and that perchlorate can be excreted into cow milk

(Kirk et al., 2003; Krynitsky et al., 2004; Capuco et al., 2005). This suggests that

perchlorate contamination in the U. S. is more widespread than was originally

thought and human exposure to perchlorate has likely occurred for several years.

128

There is a concern about the potential effect of perchlorate exposure on

milk quality, particularly because perchlorate has been found to exist in milk. Our

preliminary data revealed a weak negative relationship between perchlorate

residue levels and PUFA levels in human breast milk samples collected locally.

Therefore, maternal exposure to perchlorate may affect normal growth and brain

development in infants by altering fatty acid composition, particularly PUFA in

milk fat, since fatty acids play important roles in brain development, physical

growth, behavioral development, and many other biological processes (Stevens

et al., 1996; Koletzko and Rodriguez-Palmero, 1999; Uauy et al., 2001;

Wainwright, 2002; Alessandri et al., 2004).

In the current study, we evaluated the effect of perchlorate on fatty acid

composition in milk fat in lactating goats exposed to different levels of perchlorate

at a variety of exposure intervals. The hypothesis tested in our study was that

exposure of the lactating goats to perchlorate would change the fatty acid profile

in the goat milk. Additionally, the correlation between perchlorate concentrations

and fatty acid profile in human breast milk was investigated to further evaluate

the potential risk of human exposure to perchlorate.

6.2 Materials and Methods

Experimental design and sample collection

Eighteen lactating goats from the Texas Tech University Agriculture

Research Farm were assigned to three different perchlorate dosing groups,

including 0, 0.1, and 1 mg/kg body weight (i.e., six goats in each dosing group).

Each group consisted of goats of similar weight (and presumably age). For

example, four “heavier” (presumably older) goats were paired with two “light”

(presumably younger) goats to form each group. The goats were orally dosed

from Monday to Friday of each week (May 19th - June 21st 2003). Milk was

collected daily before perchlorate dosing. Milk samples were placed on ice after

collection and during transport back to the laboratory, where they were stored at -

129

20ºC until analysis. Fatty acid analyses were conducted for milk samples

collected on May 20th (day 2), May 28th (day 10), June 4th (day 17), June 11th

(day 24), and June 18th (day 31). Because some samples were unavailable for

fatty acid analysis, n ranged from 3 – 6 for different groups at different times.

Thirty-nine human milk samples were provided by the Department of

Human Development and Family Studies, Texas Tech University. These

samples were collected from healthy lactating mothers residing in Lubbock, TX or

nearby counties (from July 2004 to June 2005). Lactation stages were around

either 6 weeks or 3 months. Samples were stored at -20ºC after collection until

analysis.

Fatty acid analysis in milk

Frozen samples were thawed at room temperature and then heated at 85

ºC for 15 minutes to inactivate milk lipases. Each sample was prepared in

triplicate. Lipids were extracted from milk samples with chloroform/methanol

(2:1) containing 0.01% butylated hydroxytoluene (BHT) (Folch et al., 1957), and

then transesterified with 12% boron trifluoride (BF3) in methanol into fatty acid

methyl esters (Morrison and Smith, 1964). Fatty acid methyl esters were

extracted into hexane and analyzed using a Hewlett-Packard gas chromatograph

(HP 6890 series, USA) equipped with a flame ionization detector and a fused

silica capillary DB-WAX column (30 m × 0.32 mm ID, 0.25 μm film thichness)

from J&W Scientific (Folsom, CA). The oven temperature program was: initial

temperature 35 ºC, held for 2 min; raised to 210 ºC at a rate of 20 ºC/min, and

held at 210 ºC for 5 min; raised to 215 ºC at a rate of 1 ºC/min; raised again to

230 ºC at 8 ºC/min and held at 230 ºC for 5 min. The injector and detector

temperatures were 250 ºC. Helium was used as the carrier gas at a flow velocity

of 44 cm/sec. Identification and quantification of fatty acids in samples were

based on a 37-component FAME mixture standard (AccuStandard Inc. New

130

Haven, CT). Recovery of an internal standard (C19:0) was 84 ± 13% (mean ±

SD).

Monounsaturated fatty acids (MUFA), PUFA, medium-chain fatty acids,

and long-chain fatty acids were investigated as endpoints. MUFA consisted of

C16:1n9, 18:1n9, 20:1n9, and 22:1n9. PUFA included ω-6 series (C18:2n6,

18:3n6, 20:3n6, and 20:4n6), and ω-3 series (C18:3n3, 20:3n3, 20:5n3, and

22:6n3). Medium-chain fatty acids were composed of fatty acids with carbons ≤

14 (C8:0, 10:0, 12:0, and 14:0), and long-chain fatty acids included fatty acids

with carbons ≥ 16 (C16:0, 16:1n9, 18:0, 18:1n9, 18:2n6, 18:3n6, 18:3n3, 20:0,

20:1n9, 20:3n3, 20:3n6, 20:4n6, 20:5n3, 22:0, 22:1n9, and 22:6n3). Fatty acid

was expressed as a percentage of extracted lipid from the goat milk, and as a

percentage of total fatty acids for the human milk samples.

Perchlorate determination in human milk

An aliquot of human breast milk sample (1.5 mL) and 6 mL of acetonitrile

were combined into a 15-mL centrifuge tube followed by centrifugation at 3750

rpm for 10 min (Beckman Allegra 6R Centrifuge, USA). The supernatant was

evaporated to ~ 0.5 mL using nitrogen gas, and then passed through a

preconditioned C18 solid phase extraction (SPE) cartridge (Fisher). The eluate

was collected and the cartridge was then washed with 4 mL of 18.3 MΩ Milli-Q

water, which was combined with the eluate. Two replicates from one sample

were pooled and evaporated to a final volume of 3 mL. After cleanup through an

Alumina-neutral SPE cartridge (J.T. Baker, Phillipsburg, NJ), the eluate was

syringe-filtered (0.45 μm) before ion chromatography analysis.

A method similar to EPA Method 314.0 (Hautman et al., 1999) was

followed to determine perchlorate in human breast milk samples. The milk

analysis was performed on a Dionex DX-500 Ion Chromatography System

equipped with a GP50 gradient pump, a CD20 conductivity detector, and an

AS40 autosampler (Dionex Corp.). PeakNet® chromatography software was

131

used to control the system. Ion separation was made with a Dionex IonPac

AS16 (250 mm × 4.0 mm) analytical column following a Dionex guard column

(AG16). Conditions for the system were as follows: flow rate = 1.0 mL/min;

eluent = 50 mM sodium hydroxide; injection volume = 1000 µL.

Quantification of perchlorate was accomplished using an external

standard method. An instrument calibration curve was constructed by analyzing

standards at 2.5, 5, 10, 50, 100 μg/L perchlorate. Quality control samples

included blanks, matrix spikes, and check standards. The limit of detection for

perchlorate was 1 μg/L by IC. Recovery of spiked perchlorate (including cleanup

and IC analysis) was 81 – 115%.

Statistical analysis

Statistical analyses were conducted using R software (R 2.0.1 vision, Free

Software Foundation, Boston, MA). Two-way ANOVA was conducted to test the

effects of two factors: dosage and dosing time, and/or their interaction.

Differences in fatty acid composition of milk among the three treatments at either

exposure interval in goats were tested using one-way ANOVA followed by

Tukey’s multiple comparison. A linear regression analysis between perchlorate

content and total PUFA levels in the human milk samples was also conducted.

Student’s t-test was used to test the difference in fatty acid profiles between two

different lactation stages in humans (6-week and 3-month).

6.3 Results and Discussion

Fatty acid profile in goat milk dosed with perchlorate

Results of the two-way ANOVA are shown in Table 6.1. No significant

effect was found in terms of the interaction of dosage and dosing time regarding

all end-points. Time had a significant effect on fatty acid profiles in terms of all

end-points, and dose showed a significant effect on fatty acid profile (PUFA, ω-6,

ω-3, and MUFA content). Total PUFA content and ω-6 fatty acids in goat milk

132

were significantly reduced at day 10 (p = 0.0053 and 0.0113, respectively), day

17 (p < 0.001), and day 24 (p < 0.05), but not at days 2 and 31 in the high dose

treatment (Figures 6.2 and 6.3). ω-3 fatty acids showed no apparent dose-

dependent response during perchlorate exposure (data not shown). MUFA

content was significantly reduced only at day 17 (p = 0.0130) in the high dose

treatment (Figure 6.4). Although dose did not show a significant effect on

medium-chain and long-chain fatty acids (MCFA and LCFA) in milk (Table 6.1),

differences in both MCFA and LCFA among the three treatments were still tested

using one-way ANOVA. Significant reductions in medium- and long-chain fatty

acids were observed at day 24 only (p = 0.0431 and 0.0097, respectively)

(Figures 6.5 and 6.6). No significant difference was observed over the dosing

time in terms of PUFA and ω-6 fatty acids in both control and low dose groups

(Figure 6.7), but there was a significant difference between day 17 and 31

regarding both PUFA and ω-6 in the high dose group.

PUFA play key roles in the early development of young and are involved

in organogenesis (e.g. brain and retinal formation and development) (Koletzko

and Rodriguez-Palmero, 1999; Uauy et al., 2000; Uauy et al., 2001; Alessandri et

al., 2004). Therefore, adequate intake of PUFA is necessary for normal

development. In the current experiment, total PUFA was decreased at days 10,

17, and 24 for both perchlorate exposure scenarios, with a particularly significant

reduction occurring in the 1 mg/kg treatment (Figure 6.2). This indicates that

perchlorate exposure depressed total PUFA in milk of lactating goats, causing a

deficiency in PUFA available to the young. Maternal PUFA is the major PUFA for

younglings during early development. Therefore, supplementation of PUFA to

the diet of lactating goats could be required if perchlorate exposure occurs,

although some PUFA can be synthesized in and secreted from the mammary

gland (Marangoni et al., 2004) in addition to fatty acids derived from the diet.

Similar to the response of total PUFA upon perchlorate exposure, ω-6

fatty acids were reduced at days 10, 17, and 24 for both perchlorate treatments

133

(Figure 6.3). ω-6 fatty acids account for a major portion of total PUFA in goat

milk. In the current study, a significant linear correlation was found between ω-6

fatty acids and total PUFA content in all goat milk samples (r2 = 0.9929) at day

17. Therefore, ω-6 fatty acids and total PUFA in milk responded similarly to

perchlorate exposure. Among the ω-6 fatty acids examined in the current study,

linoleic acid (LA, C18:2n6) was the dominant fatty acid. Also, a significant linear

correlation existed between the LA content and total PUFA content within all

perchlorate treatments (r2 = 0.9873) at day 17. LA was proposed as a precursor

for long-chain PUFA, such as arachidonic acid (AA, C20:4n6) and

docosahexaenoic acid (DHA, C22:6n3) which are indispensable nutrients for

early development and growth of the brain and retina (Innis, 2000; Uauy et al.,

2000; Uauy et al., 2001; Alessandri et al., 2004; Marangoni et al., 2004; Innis,

2005). In the current study, AA and DHA in the high dose group decreased to

79.4 ± 12.82 % of the control at day 24 and 63.2 ± 16.80 % of the control at day

31, respectively. This is probably due to the decrease in the precursor of long-

chain PUFA, i.e. LA, during exposure, which decreased to 61.4 ± 15.63 % of the

control at day 17 and 64.8 ± 6.47 % of the control at day 24.

As seen in Figures 6.2 and 6.3, the maximum reduction of both PUFA and

ω-6 fatty acids in the 0.1 mg/kg perchlorate treatment occurred later than at the 1

mg/kg treatment. The delayed response of goats to the lower perchlorate

treatment indicates that the higher perchlorate concentrations appear to act on

and alter PUFA and ω-6 fatty acid content with greater efficiency.

The lack of a significant change in the endpoints evaluated for all

exposure scenarios at day 2 (Figures 6.2 - 6.6) indicates that a 2-day exposure

to perchlorate, up to 1 mg/kg, was not sufficient to alter the fatty acid profile in

goat milk. This was not completely unexpected because milk fat biosynthesis is

a complex process in which many molecular-level activities are involved (Figure

6.1) and some time is needed for an effect to be manifested. The reduction in

most fatty acids that occurred at days 10, 17, and/or 24 was reversed; no

134

significant difference was found when compared with the control at day 31

(Figures 6.2 - 6.6). This is possibly due to adaptation mechanisms which might

be induced to compensate for the effect of perchlorate exposure, such that the

goats increased the removal of perchlorate via milk and/or urine. Kirk et al

(2005) found that the perchlorate content in milk 24 hours after perchlorate

dosing was significantly reduced when compared with levels at 2 hours post-

dose (perhaps because less perchlorate was available for excretion with time).

Excretion of perchlorate via urine is a major pathway for mammals following

perchlorate exposure (Anbar et al., 1959; Goldman and Stanbury, 1973; Peña et

al., 1976; Batjoens et al., 1993; Yu et al., 2002; Capuco et al., 2005). A study on

the temporal pattern of urine excretion of perchlorate could provide an answer to

whether animals can reduce perchlorate effects by increasing excretion via urine

upon exposure to perchlorate. Perchlorate excretion via urine in deer mice,

whether based on perchlorate concentration or mass percentage of the total

intake through dosed drinking water, increased and achieved a steady state soon

(24 h) after perchlorate exposure (Chapter IV). Inconsistent temporal patterns

between urine excretion of perchlorate and changes in fatty acid profiles may be

a result of the complex and multiple-step process of fatty acid synthesis, which

needs some time to manifest the effect of perchlorate.

The reverse effect described earlier may also be caused by the increased

production of thyrotropin-releasing hormone (TRH), resulting from decreased

thyroid hormone negative feedback. As a potent prolactin-releasing factor, TRH

can stimulate prolactin secretion from the pituitary (Cooke et al., 2004), and as a

result, alter the fatty acid profile in milk by reversing the effect of perchlorate.

It was observed that there was no significant difference between 0.1

mg/kg and 1 mg/kg perchlorate treatments after week 1 through week 4 in terms

of mean perchlorate residues in goat milk collected 24 hours after dosing (Kirk,

2005), suggesting that perchlorate did not accumulate in milk. In the current

study, perchlorate exposure caused pronounced effects on all endpoints at day

135

10, day 17, and/or day 24 (Figures 6.2 - 6.6). This discrepancy between reduced

perchlorate residues in milk (i.e. week 1 through week 4) and the effect of

perchlorate on the fatty acid profile in milk (i.e. days 10, 17, and/or 24) suggests

that perchlorate does not act directly on fatty acids in milk. Fatty acid synthesis

and secretion in milk is partly regulated by prolactin and involves activation (or

function) of lipoprotein lipase (LPL) and fatty acid synthase, which are the

principle enzymes of fatty acid mobilization and synthesis in mammary epithelial

cells (Hamosh et al., 1972; Hadley, 2000). Alternatively, thyroid hormones are

necessary for the induction of prolactin and can stimulate transcription of the fatty

acid synthase gene (Goodridge, 1986; Wilson et al., 1986; Stapleton et al., 1990;

Kameda, 1995; Hadley, 2000). Therefore, theoretically, disruption of thyroid

hormone production by perchlorate exposure can ultimately alter the fatty acid

profile in milk (Figure 6.1), as was observed in the current study. A mechanistic

study needs to be conducted to determine what specific events occur during this

cascade.

Diet has a major influence on fatty acid composition in milk (Finley et al.,

1985; Neville and Picciano, 1997; Francois et al., 1998; Chilliard et al., 2003;

Chilliard and Ferlay, 2004), but milk and (or) milk fat composition also changes

with lactation stage (Arumughan and Narayanan, 1981; Zanartu et al., 1983;

Neville and Picciano, 1997). In the current study, the goats had access to the

same food and water, but their stage of lactation was unknown (i.e. they may not

have been at the same stage of lactation despite the food, water, and weight

grouping). Therefore, differences in lactation stage could have been a

confounding factor in the altered fatty acid profile in milk observed in this study.

Nevertheless, changes in fatty acid profiles in goat milk observed in the current

study were consistent with previous reports to a certain degree. Reduction in

milk fat was observed in a cow study with perchlorate exposure, although the

value was within the normal range for good health (Capuco et al., 2005). In

addition, it was also reported that de novo fatty acid synthesis in the liver and

136

triglyceride synthesis in both the liver and kidney were reduced in the presence

of KClO4 in sea lamprey (Kao et al., 1999).

Human milk survey

No significant statistical difference was observed between the 6-week and

3-month lactation stages in terms of either each fatty acid, total saturated fatty

acids, MUFA, PUFA, ω-3, ω-6, medium-chain fatty acids (C ≤14), or long-chain

fatty acids (C ≥ 16) (t-test, p > 0.05). A weak negative relationship (r2 = 0.033, p

= 0.267) between perchlorate concentrations and total PUFA levels in the 39

human milk samples was observed (Figure 6.8). As was the case for the goat

milk samples, there were good linear relationships between linoleic acid

(C18:2n6) and total PUFA, and ω-6 and total PUFA (r2= 0.9598 and 0.9769,

respectively). The difference in total PUFA among samples resulted mainly from

differences in linoleic acid. However, no significant relationship (either positive or

negative with p < 0.1) was found between AA, DHA, MUFA, medium- or long-

chain fatty acids and perchlorate concentrations. Given an average body weight

of 5.8 kg and an average daily breast milk intake of 712 mL for a 1 to 4-month old

baby (EPA, 1996), the reference dose of 0.7 μg/kg/day recommended by the

National Academy of Science (NAS, 2005) can be converted into a perchlorate

concentration in breast milk of 5.70 μg/L. When the ranges of observed

perchlorate and DHA were divided into two groups, four quadrants were formed

(Figure 6.9); not a single datum with high DHA (> 0.1%) – high perchlorate (>

5.70 μg/L) was found for these samples.

Perchlorate was detected in all human milk samples, ranging from 0.84 to

8.18 μg/L with an average of 3.43 μg/L. During the first 6 months of lactation

with exclusive breast-feeding, 24 h milk production from each breast is 453.6 ±

201 g (Kent et al., 1999). Assuming 5% of the perchlorate taken up by a 50-kg

lactating mother was excreted via milk, then the perchlorate exposure level to the

mother would be about 1.48 μg/kg body weight with a maximum of 2.14 μg/kg

137

(corresponding to average and high 24 h milk production with the maximum

perchlorate residues in milk, i.e. 8.18 μg/L). The fact that the predicted

perchlorate exposure level in humans was much lower than that administered in

the goat dosing study could explain why no significant relationship was found

between either endpoint and perchlorate concentrations in the human milk. In

addition, nutritional and physical factors which can influence fatty acid

composition in milk were not incorporated into the data analyses. Furthermore,

due to the unavailability of information about perchlorate exposure conditions

such as time, frequency, and source, the effect of perchlorate on fatty acid

composition in human milk samples were not conclusive. An epidemiological

study with large sample sizes would contribute more to evaluating the potential

risk of perchlorate on pregnant and/lactating women and their infants.

Both thyroid hormones and PUFA are very important in the early

development of infants, especially for brain development. Thyroid hormones

regulate the processes of neuronal proliferation, migration, synpatogenesis, and

myelination, as well as promote the uptake of amino acids, glucose, and other

necessary nutrients by brain tissues (Chan and Kilby, 2000; Brown, 2003;

Zoeller, 2003). Brain structure requires complex lipids, especially PUFA

(Koletzko and Rodriguez-Palmero, 1999; Alessandri et al., 2004). As a sole

source of nutrients for the infant, high breast milk quality is critical for normal

development and growth. In the current study, we found that perchlorate

depressed the total PUFA content in goat milk. This could also occur in human

milk if perchlorate exposure occurs at higher levels. In addition, it was reported

that perchlorate at concentrations above 10 µg/L in milk was associated with low

iodine content in human breast milk (Kirk et al., 2005). Adequate iodide intake is

necessary for pregnant and lactating women. If maternal exposure to

perchlorate occurs, it is possible that insufficient maternal thyroid hormones,

PUFA, and/or iodide is available for the normal development and growth of the

infants. Malnutrition, difficulty with motor coordination and balance, and attention

138

deficit/hyperactivity disorder (ADHA) in children (Brown, 2003) could be the likely

outcomes.

6.4 Conclusions

An effect of perchlorate on fatty acid composition in milk fat was found in

dosed goats, but it was not persistent. PUFA, MUFA, medium- and long-chain

fatty acids decreased significantly in the high treatment at days 10, 17, and/or 24,

but were reversed by day 31. Adaptation may be induced in these goats to

compensate for the perchlorate effect. Although a weak negative correlation was

observed between perchlorate concentrations and total PUFA in 39 human milk

samples, no conclusive statement can be made about perchlorate effect on

PUFA profile in human milk since the predicted perchlorate exposure level was

much lower than that used in the goat dosing study, and nutritional and physical

factors, which can affect the fatty acid profile in milk, were not incorporated into

the data analysis. Due to the widely reported adverse effects of perchlorate on

thyroid hormones, iodide content in milk, and PUFA profiles in milk (from the

current study), infant development during maternal exposure to perchlorate

should be of concern. To our knowledge, this is the first report on the effect of

perchlorate on fatty acid profiles in milk. Further study is urged to investigate the

mechanistic aspects of the effect.

139

Milk Fat Globule

Figure 6.1. Proposed relationship between perchlorate and fatty acid

composition in milk fat. Fatty acids are derived from circulating triglycerides (TGs) through hydrolysis of chylomicron triglyceride or very low density lipid by lipoprotein lipase (LPL). Fatty acids, especially short- and medium- chain fatty acids, can also be synthesized by fatty acid synthase (FAS). The fatty acids are used to synthesize triglycerides, which fuse to form lipid droplets and are secreted into milk as a milk fat globule. Prolactin was postulated to regulate activation of LPL, and may also affect the activity of FAS (Hadley, 2000). Induction of prolactin requires the existence of thyroid hormones (Hadley, 2000). Thyroid hormone (i.e. triiodothyronine) was also reported to stimulate transcription of the fatty acid synthase gene (Stapleton et al., 1990; Goodridge, 1986). As a thyroid hormone disrupter, perchlorate may affect LPL and FAS functions and eventually influence fatty acid composition in milk fat since thyroid hormone is associated directly or indirectly with these two enzymes which are important for milk synthesis and secretion. Interruption in thyroid hormone production by perchlorate may also change the fatty acid profile in milk fat by affecting de novo synthesis in other tissues such as the liver in addition to the mammary gland.

Fatty Acid Synthase

FA: C6-C14

Acetyl CoA

LPL

TGs

+ glycerol-3-P

TGs from Chylomicrons & Very Low Density Lipid

Milk

FA: ≥C16

Fatty Acids

ClO4-

Mam

mar

y ep

ithel

ial c

ell

Thyroid Hormones

Prolactin

Maternal Blood

Fatty acid de novo synthesis in other tissues such as liver

140

Table 6.1. p-values in two-way ANOVA test for fatty acid content in lactating goats dosed with perchlorate from Monday to Friday for 4.5 weeks. Two factors are time and factor. TimeXdose includs potential interaction in the statistical test; time+dose does not include potential interaction in the statistical test. ***: p < 0.001; **: p < 0.01; *: p < 0.05.

Two-way ANOVA p-value Test Factor

MUFA PUFA ω-6 ω-3 MCFA LCFA timeXdose time 3.19E-8*** 0.022* 0.041* 1.81E-5*** 0.0002*** 1.93E-5***

dose 0.008** 2.23E-6*** 1.82E-5*** 4.0E-4*** 0.263 0.157 t*d 0.588 0.171 0.22 0.229 0.403 0.584

time+dose time 1.311E-8*** 0.028* 0.047* 8.02E-6*** 1.4E-4*** 1.20E-5*** dose 0.007** 3.13E-6*** 2.23E-5*** 4.0E-4*** 0.264 0.151

0.0

0.5

1.0

1.5

2.0

day2 day10 day17 day24 day31Time

PUFA

(%lip

id)

0 mk/kg 0.1 mg/kg 1 mg/kg

a

a

b

a

b b b

Figure 6.2. Content of total polyunsaturated fatty acids (PUFA) in goat milk

during perchlorate exposure (mean ± SD) (n = 3 and 4 at 0.1 mg/kg at days 17 and 24, respectively, n = 3 at 1 mg/kg at day 24, and n = 5 or 6 at all other time points). Different letters indicate a significant difference at the 0.05 level.

141

0.0

0.4

0.8

1.2

1.6

day2 day10 day17 day24 day31Time

w-6

(%lip

id)

0 mk/kg 0.1 mg/kg 1 mg/kg

a

b

a

bb

a

b

Figure 6.3. Content of ω-6 fatty acids in goat milk during perchlorate exposure

(mean ± SD) (n = 3 and 4 at 0.1 mg/kg at days 17 and 24, respectively, n = 3 at 1 mg/kg at day 24, and n = 5 or 6 at all other time points). Different letters indicate a significant difference at the 0.05 level.

0

4

8

12

16

day2 day10 day17 day24 day31Time

MU

FA(%

lipid

)

0 mk/kg 0.1 mg/kg 1 mg/kg

a

b

Figure 6.4. Content of mono-unsaturated fatty acids (MUFA) in goat milk during

perchlorate exposure (mean ± SD) (n = 3 and 4 at 0.1 mg/kg at days 17 and 24, respectively, n = 3 at 1 mg/kg at day 24, and n = 5 or 6 at all other time points). Different letters indicate a significant difference at the 0.05 level.

142

0

5

10

15

20

25

day2 day10 day17 day24 day31

Time

Mid

ium

cha

in F

A (%

lipid

)

0 mk/kg 0.1 mg/kg 1 mg/kg

a

b

Figure 6.5. Content of medium-chain fatty acids (MUFA) in goat milk during

perchlorate exposure (mean ± SD) (n = 3 and 4 at 0.1 mg/kg at days 17 and 24, respectively, n = 3 at 1 mg/kg at day 24, and n = 5 or 6 at all other time points). Different letters indicate a significant difference between the control and high dose groups at day 24 (0.05 level).

143

0

10

20

30

40

day2 day10 day17 day24 day31Time

Long

cha

in F

A (%

lipid

)

0 mk/kg 0.1 mg/kg 1 mg/kg a

b

Figure 6.6. Content of long-chain fatty acids (LCFA) in goat milk during

perchlorate exposure (mean ± SD) (n = 3 and 4 at 0.1 mg/kg at days 17 and 24, respectively, n = 3 at 1 mg/kg at day 24, and n = 5 or 6 at all other time points). Different letters indicate a significant difference between the low dose group and control on day 24 (0.05 level). Small sample size (n = 3) in the high dose group, with generally higher LCFA content in these three individuals, may cause the LCFA content to be higher than the low dose group on day 24, and as a result, no significant difference was observed between the high dose and control groups.

144

0.0

0.3

0.6

0.9

1.2

1.5

1.8

0 mk/kg 0.1 mg/kg 1 mg/kgDosage

PUFA

(% li

pid)

day2 day10 day17 day24 day31

a

b

0.0

0.4

0.8

1.2

1.6

0 mk/kg 0.1 mg/kg 1 mg/kg

Dosage

w-6

(% li

pid)

day2 day10 day17 day24 day31

a

b

Figure 6.7. Changes in PUFA (top) and ω-6 fatty acid (bottom) content (% of

lipid) with time in three different dosing groups. Different letters indicate significant difference between day 17 and 31 in the high dose group (0.05 level).

145

y = -0.3234x + 21.757r2 = 0.0331

0

10

20

30

40

0 2 4 6 8

Perchlorate (ug/L)

PUFA

(% o

f tot

al fa

tty a

cids

10

Figure 6.8. Correlation (r2 = 0.0331; p = 0.267) between polyunsaturated fatty

acid (PUFA) and perchlorate concentrations in human breast milk from mothers in Lubbock, Texas or nearby counties (July 2004 – June 2005). PUFA levels were expressed as percentage of total fatty acids.

0

0.05

0.1

0.15

0.2

0 2 4 6 8Perchlorate (ug/L)

DH

A (C

22:6

n3, %

of t

otal

fatty

aci

d

10

s

Figure 6.9. Docosahexaenoic acid (DHA; C22:6n3) as a function of perchlorate

concentration in human milk from Lubbock, Texas or nearby counties (collected from July 2004 to June 2005). DHA levels were expressed as a percentage of total fatty acids. Four quadrants were obtained when dividing the ranges of the observed DHA and perchlorate into two.

146

CHAPTER VII

CONCLUSIONS AND FUTURE RESEARCH

7.1 Study Summary and Conclusions

Methods development for perchlorate determination in biological fluids

was explored firstly in this study. The presence of ions and other biomolecules in

matrices like plasma and urine usually confounds accurate determination of

perchlorate by ion chromatography with suppressed conductivity detection (IC-

SCD); therefore, efficient cleanup before IC analysis is necessary for perchlorate

determination in these matrices. Several different cleanup methods for plasma

and urine were tested and compared in the current study, in terms of reduced

background conductivity, interference, perchlorate recovery, and precision.

Effective sample preparation and cleanup process are usually expensive, time

consuming, and labor intensive. High background conductivity and the existence

of interfering components are still routine problems encountered for perchlorate

determination by conventional IC (IC-SCD). In addition, IC-SCD cannot provide

unambiguous confirmation for perchlorate determination, particularly in biological

samples. Therefore, the application of mass spectrometry (MS) in perchlorate

determination in biological fluids was also explored in the current study. MS was

able to provide a better quantitative and qualitative analysis of complicated

matrices and served as an efficient alternative analytical tool to IC-SCD for

perchlorate monitoring. Particularly, ion chromatography coupled with MS/MS

provided an excellent method for part per trillion perchlorate determination in

both aqueous and deer mice urine matrices, with high selectivity, sensitivity, and

robust accuracy and precision.

The study on perchlorate exposure and absorption in beef cattle showed

that constant exposure to 25 ng/mL perchlorate in water over 14 weeks did not

result in measurable residues in blood plasma or edible tissues of cattle at the

first test site (McLennan County, Texas). Perchlorate was detected in 4 of 33

147

and 17 of 26 cattle at two Kansas farms with the highest plasma perchlorate

concentrations of 43 and 32 ng/mL, respectively. However, perchlorate was

detected in all four urine samples from Kansas using IC-MS/MS. Perchlorate

residues in urine rather than plasma may be a more sensitive biomarker for

perchlorate exposure assessment.

The study on perchlorate distribution, excretion, and depuration in rodents

showed that (1) excretion via urine was the major pathway for perchlorate fate in

the body; (2) perchlorate urinary excretion was dose-dependent, higher dose

level corresponded well to high perchlorate concentration in urine; (3) perchlorate

urinary excretion reached to a steady state after one day in all treatments over

the 28-d exposure; and (4) perchlorate was depurated rapidly via urine.

Perchlorate mass in kidney, thyroid, blood, and urine was significantly correlated

with perchlorate intake. On average of 46, 46, and 61% of perchlorate were

recovered in urine over the 28-d exposure period in high, medium, and low dose

groups, respectively. Metabolism of perchlorate may occur in the body, based in

part on the 40% perchlorate unaccounted for in this study. Other sources of

perchlorate were not clear, and endogenous perchlorate may exist. Perchlorate

fate in animals was not as straightforward as reported by previous studies.

Perchlorate exposure through dosed drinking water for 28 days increased

NIS gene expression in the kidney and stomach, and pendrin gene expression in

the kidney, without a significant difference. A dose-dependent manner was

observed in NIS gene expression in the stomach. In the depuration experiment,

no significant difference was found between the low and high dose groups (8-h

exposure to perchlorate followed by an 88-h depuration period) in terms of NIS or

pendrin gene expression in the kidney or stomach. Linear regression analysis

did not show a significant relationship between perchlorate urinary excretion and

either gene expression in the kidney at day 28.

Several factors could complicate the effect of perchlorate exposure on NIS

and pendrin gene expression in the stomach and kidney in the current study,

148

including (1) pre-exposure to trace perchlorate through food and water, which

may have resulted in adaptation (or tolerance) in these animals; (2) metabolism

of perchlorate in deer mice, which may cause 45 – 61% perchlorate recovery via

urine; (3) short-term exposure, which may be insufficient to induce gene

expression in the tissues; and (4) large differences among individuals. It is also

possible that there is no effect of perchlorate exposure and/or excretion via urine

on NIS gene expression, particularly in the kidney. Nevertheless, alteration of

pendrin gene expression in the kidney produced by perchlorate exposure may

cause abnormalities in acid-base homeostasis and sodium-chloride balance,

metabolic alkalosis and hypertension, and alteration of blood pressure.

A partial sequence of deer mice NIS gene cDNA with 425 bps was

elucidated in the current study for the first time. Quantitative analysis of NIS

mRNA expression in various tissues was also studied for the first time with

expression levels from highest to lowest in deer mice tissues in the following

order: stomach, testes, brain, large intestine, and barely expression in lung,

kidney, heart, and liver.

An effect of perchlorate on fatty acid composition in milk fat was observed

in dosed goats during lactation, but it was not persistent. PUFA, MUFA, medium-

and long-chain fatty acids decreased significantly in the high treatment at days

10, 17, and/or 24, but were reversed by day 31. Adaptation may be induced in

these goats to compensate for the perchlorate effect. Although a weak negative

correlation was observed between perchlorate concentrations and total PUFA in

39 human milk samples, no conclusive statement can be made about perchlorate

effect on the PUFA profile in human milk since the predicted perchlorate

exposure level was much lower than that used in the goat dosing study, and

nutritional and physical factors, which can affect the fatty acid profile in milk, were

not incorporated into the data analysis. Due to the widely reported adverse

effects of perchlorate on thyroid hormones, iodide content in milk, and PUFA

profiles in milk (from the current study), infant development during maternal

149

exposure to perchlorate should be of concern. To our knowledge, this is the first

report on the effect of perchlorate on fatty acid profiles in milk. Further study is

urged to investigate the mechanistic aspects of the effect.

7.2 Future Studies

One major suggestion for future study on assessment of perchlorate

exposure in the environment and human is using mass spectrometry to

determine perchlorate in various matrices. Perchlorate was seldom detected in

cattle plasma samples in two contaminated sites (Chapter III), which may be

caused by the limitation of IC-SCD for perchlorate determination in complicate

biological matrices in terms of detection limit and selectivity. With IC-MS/MS

methodology, perchlorate can be detected in various matrices sensitively and

accurately. Additionally, perchlorate residues in urine may be a more sensitive

indicator for perchlorate exposure. Therefore, IC-MS/MS application for

perchlorate determination in environmental and biological samples would

contribute significantly to assessing perchlorate exposure in the environment and

human in the future, and determining perchlorate residue in urine rather than

plasma would provide a more sensitive biomarker for perchlorate exposure

assessment in animals including humans.

Inconsistence of perchlorate intake through drinking water and excretion

via urine in terms of mass balance was revealed in the excretion and depuration

experiments (Chapter IV), which presented a question: where did ~ 40%

perchlorate go in those animals? Perchlorate metabolism and in the body were

proposed in the current study, but more research is needed to provide evidence

and support it. A study could be conducted in the future to test the existence of

anaerobic bacteria which express perchlorate reductase, perchlorate reductase,

and/or the gene encoding perchlorate reductase in deer mice body, especially

the gastrointestinal tract, which would indicate the possibility of perchlorate

transition to chlorate, chlorite, and/or chloride. Isotopic labeled perchlorate can

150

be used as dosing solution to test fate, transport, and transition of perchlorate in

the body and provide evidence for perchlorate toxicokinetics in animals.

Regarding effects of perchlorate exposure on transporter gene

expression, protein expression should be analyzed as well. Usually, protein is

expressed because of its encoding gene expression. However, if perchlorate

was transported through these transporters (i.e. NIS and pendrin), protein levels

or activities rather than gene expression levels would be directly related to

perchlorate excretion or uptake by the cells. In addition, evaluation on protein

expression level is very important and necessary, especially for mechanism

study involving post-transcription. West blotting or immunohistochemical

methodologies could be used to evaluate effects of perchlorate exposure on

these transporter expression levels and activities in the interested tissues, such

as the stomach and kidney in the current study, and therefore, provide more

insight about the perchlorate excretion pattern via urine and some indication of

perchlorate toxicokinetics at the molecular level.

A study could be conducted to address the question about effects of

perchlorate exposure on profile of fatty acid in milk at molecular levels regarding

the mechanistic aspects. Effects of perchlorate exposure could be explored with

some enzymes and hormones levels as endpoints. These enzymes and

hormones could be thyroid hormone, prolactin, lipoprotein lipase, fatty acid

synthase, and etc., which are involved in milk fatty acid synthesis process and

related to thyroid hormone directly or indirectly. In terms of relationship between

perchlorate residue levels and PUFA levels in human breast milk collected

locally, perchlorate levels in milk confirmed using mass spectrometry would

provide an unambiguous evidence for the conclusion, and an epidemiological

study with large sample size and more information on subjects nutrition and

physic status would contribute more to evaluating the potential risk of perchlorate

on pregnant and/or lactating women and their infants.

151

Future studies also could be conducted to address questions including,

but not limited to, (1) species difference regarding perchlorate uptake,

distribution, excretion, and effects; (2) influence of pre-exposure to perchlorate

on perchlorate uptake, distribution, excretion, and effects; (3) induction of gene

expression caused by perchlorate exposure; and (4) diet confounding influence

on the relationship between perchlorate exposure and the profile of fatty acids in

milk.

152

REFERENCES

Ajjan, R.A., Kamaruddin, N.A., Crisp, M., Watson, P.F., Ludgate, M. and Weetman, A.P.: Regulation and tissue distribution of the human sodium iodide symporter gene. Clin Endocrinol (Oxf) 49 (1998a) 517-23.

Ajjan, R.A., Watson, P.F., Findlay, C., Metcalfe, R.A., Crisp, M., Ludgate, M. and Weetman, A.P.: The sodium iodide symporter gene and its regulation by cytokines found in autoimmunity. J Endocrinol 158 (1998b) 351-8.

Alessandri, J.M., Guesnet, P., Vancassel, S., Astorg, P., Denis, I., Langelier, B., Aid, S., Poumes-Ballihaut, C., Champeil-Potokar, G. and Lavialle, M.: Polyunsaturated fatty acids in the central nervous system: evolution of concepts and nutritional implications throughout life. Reprod Nutr Dev 44 (2004) 509-38.

Anbar, M., Guttmann, S. and Lewitus, Z.: The mode of action of perchlorate ions on the iodine uptake of the thyroid gland. Int J Appl Radiat Isot 7 (1959) 87-96.

Anderson, T.A. and Wu, T.H.: Extraction, cleanup, and analysis of the perchlorate anion in tissue samples. Bull Environ Contam Toxicol 68 (2002) 684-91.

Animal-Welfare-Advisory-Committee: Guidelines for the Welfare of Livestock from which Blood is Harvested for Commercial and Research Purposes, http://www.biosecurity.govt.nz/animal-welfare/codes/blood/index.htm. Animal Welfare Advisory Committee, 1996.

Annesley, T.M.: Ion suppression in mass spectrometry. Clin Chem 49 (2003) 1041-4.

Arumughan, C. and Narayanan, K.M.: Influence of stage of lactation on the triacylglycerol composition of buffalo milk fat. Lipids 16 (1981) 155-64.

Baldridge, M.G., Stahl, R.L., Gerstenberger, S.L., Tripoli, V. and Hutz, R.J.: In utero and lactational exposure of Long-Evans rats to ammonium perchlorate (AP) disrupts ovarian follicle maturation. Reprod Toxicol 19 (2004) 155-61.

Bass Becking, L.G.M., Haldane, A.D. and Izard, D.: Perchlorate, an important constituent of seawater. Nature 182 (1958) 645-647.

Batjoens, P., Debrabander, H.F. and Tkindt, L.: Ion Chromatographic Determination of Perchlorate in Cattle Urine. Analytica Chimica Acta 275 (1993) 335-340.

Bekkedal, M.Y., Arfsten, D. and Mattie, D.: An evaluation of neurobehavioral tests used to assess the neurodevelopmental effects of early ammonium perchlorate exposure. J Toxicol Environ Health A 67 (2004) 835-44.

153

Bell, A., Gagnon, A., Grunder, L., Parikh, S.J., Smith, T.J. and Sorisky, A.: Functional TSH receptor in human abdominal preadipocytes and orbital fibroblasts. Am J Physiol Cell Physiol 279 (2000) C335-40.

Bender, K.S., Shang, C., Chakraborty, R., Belchik, S.M., Coates, J.D. and Achenbach, L.A.: Identification, characterization, and classification of genes encoding perchlorate reductase. J Bacteriol 187 (2005) 5090-6.

Bidart, J.M., Lacroix, L., Evain-Brion, D., Caillou, B., Lazar, V., Frydman, R., Bellet, D., Filetti, S. and Schlumberger, M.: Expression of Na+/I- symporter and Pendred syndrome genes in trophoblast cells. J Clin Endocrinol Metab 85 (2000a) 4367-72.

Bidart, J.M., Mian, C., Lazar, V., Russo, D., Filetti, S., Caillou, B. and Schlumberger, M.: Expression of pendrin and the Pendred syndrome (PDS) gene in human thyroid tissues. J Clin Endocrinol Metab 85 (2000b) 2028-33.

Bradford, C.M., Rinchard, J., Carr, J.A. and Theodorakis, C.: Perchlorate affects thyroid function in eastern mosquitofish (Gambusia holbrooki) at environmentally relevant concentrations. Environ Sci Technol 39 (2005) 5190-5.

Braverman, L.E., He, X., Pino, S., Cross, M., Magnani, B., Lamm, S.H., Kruse, M.B., Engel, A., Crump, K.S. and Gibbs, J.P.: The effect of perchlorate, thiocyanate, and nitrate on thyroid function in workers exposed to perchlorate long-term. J Clin Endocrinol Metab 90 (2005) 700-6.

Braverman, L.E. and Ingbar, S.H.: Changes in Thyroidal Function During Adaptation to Large Doses of Iodide. J Clin Invest 42 (1963) 1216-31.

Brechner, R.J., Parkhurst, G.D., Humble, W.O., Brown, M.B. and Herman, W.H.: Ammonium perchlorate contamination of Colorado River drinking water is associated with abnormal thyroid function in newborns in Arizona. J Occup Environ Med 42 (2000) 777-82.

Brown, V.J.: Disrupting a delicate balance: environmental effects on the thyroid. Environ Health Perspect 111 (2003) A642-9.

Bruce, R.A., Achenbach, L.A. and Coates, J.D.: Reduction of (per)chlorate by a novel organism isolated from paper mill waste. Environ Microbiol 1 (1999) 319-29.

Bustin, S.A.: Real-time, fluorescence-based quantitative PCR: a snapshot of current procedures and preferences. Expert Rev Mol Diagn 5 (2005) 493-8.

Bustin, S.A., Benes, V., Nolan, T. and Pfaffl, M.W.: Quantitative real-time RT-PCR--a perspective. J Mol Endocrinol 34 (2005) 597-601.

Cañas, J.E., Cheng, Q., Tian, K. and Anderson, T.A.: Optimization of operating conditions for the determination of perchlorate in biological samples using

154

preconcentration/preelution ion chromatography. Journal of Chromatography A In press (2006).

Capuco, A.V., Rice, C.P., Baldwin, R.L.t., Bannerman, D.D., Paape, M.J., Hare, W.R., Kauf, A.C., McCarty, G.W., Hapeman, C.J., Sadeghi, A.M., Starr, J.L., McConnell, L.L. and Van Tassell, C.P.: Fate of dietary perchlorate in lactating dairy cows: Relevance to animal health and levels in the milk supply. Proc Natl Acad Sci U S A 102 (2005) 16152-7.

Chan, S. and Kilby, M.D.: Thyroid hormone and central nervous system development. J Endocrinol 165 (2000) 1-8.

Chang, S., Crothers, C., Lai, S. and Lamm, S.: Pediatric neurobehavioral diseases in Nevada counties with respect to perchlorate in drinking water: an ecological inquiry. Birth Defects Res A Clin Mol Teratol 67 (2003) 886-92.

Cheng, Q., Liu, F., Cañas, J.E. and Anderson, T.A.: A cleanup method for perchlorate determination in urine. Talanta 68 (2006) 1457-1462.

Cheng, Q., Perlmutter, L., Smith, P.N., McMurry, S.T., Jackson, W.A. and Anderson, T.A.: A study on perchlorate exposure and absorption in beef cattle. J Agric Food Chem 52 (2004) 3456-61.

Chilliard, Y. and Ferlay, A.: Dietary lipids and forages interactions on cow and goat milk fatty acid composition and sensory properties. Reprod Nutr Dev 44 (2004) 467-92.

Chilliard, Y., Ferlay, A., Rouel, J. and Lamberet, G.: A review of nutritional and physiological factors affecting goat milk lipid synthesis and lipolysis. J Dairy Sci 86 (2003) 1751-70.

Clewell, R.A., Merrill, E.A., Narayanan, L., Gearhart, J.M. and Robinson, P.J.: Evidence for competitive inhibition of iodide uptake by perchlorate and translocation of perchlorate into the thyroid. Int J Toxicol 23 (2004) 17-23.

Cline, T.R., Plumlee, M.P., Christian, J.E. and Kessler, W.V.: Effect of potassium perchlorate and sodium chloride given orally upon radioiodine deposition into milk of goats. J Dairy Sci 52 (1969) 1124-6.

Coates, J.D., Michaelidou, U., Bruce, R.A., O'Connor, S.M., Crespi, J.N. and Achenbach, L.A.: Ubiquity and diversity of dissimilatory (per)chlorate-reducing bacteria. Appl Environ Microbiol 65 (1999) 5234-41.

Collette, T.W., Williams, T.L., Urbansky, E.T., Magnuson, M.L., Hebert, G.N. and Strauss, S.H.: Analysis of hydroponic fertilizer matrixes for perchlorate: comparison of analytical techniques. Analyst 128 (2003) 88-97.

155

Cooke, P.S., Holsberger, D.R., Witorsch, R.J., Sylvester, P.W., Meredith, J.M., Treinen, K.A. and Chapin, R.E.: Thyroid hormone, glucocorticoids, and prolactin at the nexus of physiology, reproduction, and toxicology. Toxicol Appl Pharmacol 194 (2004) 309-35.

Crane, H.M., Pickford, D.B., Hutchinson, T.H. and Brown, J.A.: Effects of ammonium perchlorate on thyroid function in developing fathead minnows, Pimephales promelas. Environ Health Perspect 113 (2005) 396-401.

Crump, C., Michaud, P., Tellez, R., Reyes, C., Gonzalez, G., Montgomery, E.L., Crump, K.S., Lobo, G., Becerra, C. and Gibbs, J.P.: Does perchlorate in drinking water affect thyroid function in newborns or school-age children? J Occup Environ Med 42 (2000) 603-12.

Dahl, R.: Perchlorate debate grows. Environmental Health Perspectives 112 (2004) A546.

Dai, G., Levy, O. and Carrasco, N.: Cloning and characterization of the thyroid iodide transporter. Nature 379 (1996) 458-60.

Dasgupta, P.K., Martinelango, P.K., Jackson, W.A., Anderson, T.A., Tian, K., Tock, R.W. and Rajagopalan, S.: The origin of naturally occurring perchlorate: the role of atmospheric processes. Environ Sci Technol 39 (2005) 1569-75.

De La Vieja, A., Dohan, O., Levy, O. and Carrasco, N.: Molecular analysis of the sodium/iodide symporter: impact on thyroid and extrathyroid pathophysiology. Physiol Rev 80 (2000) 1083-105.

Djurdjevic, D. and Lengemann, F.W.: Effect of perchlorate on the milk-plasma radioiodine ratio of intact and thyroidectomized goats. J Dairy Sci 53 (1970) 808-12.

Dodds, E.D., Kennish, J.M., Von Hippel, F.A., Bernhardt, R. and Hines, M.E.: Quantitative analysis of perchlorate in extracts of whole fish homogenates by ion chromatography: comparison of suppressed conductivity detection and electrospray ionization mass spectrometry. Anal Bioanal Chem 379 (2004) 881-7.

Dohan, O., De la Vieja, A. and Carrasco, N.: Molecular study of the sodium-iodide symporter (NIS): a new field in thyroidology. Trends Endocrinol Metab 11 (2000) 99-105.

Dohan, O., De la Vieja, A., Paroder, V., Riedel, C., Artani, M., Reed, M., Ginter, C.S. and Carrasco, N.: The sodium/iodide Symporter (NIS): characterization, regulation, and medical significance. Endocr Rev 24 (2003) 48-77.

Dorak, M.T.: Real time PCR, http://www.dorak.info/genetics/realtime.html, 2005.

156

Ellington, J.J. and Evans, J.J.: Determination of perchlorate at parts-per-billion levels in plants by ion chromatography. Journal of Chromatography A 898 (2000) 193-199.

Ellington, J.J., Wolfe, N.L., Garrison, A.W., Evans, J.J., Avants, J.K. and Teng, Q.C.: Determination of perchlorate in tobacco plants and tobacco products. Environmental Science & Technology 35 (2001) 3213-3218.

Ells, B., Barnett, D.A., Purves, R.W. and Guevremont, R.: Trace level determination of perchlorate in water matrices and human urine using ESI-FAIMS-MS. Journal of Environmental Monitoring 2 (2000) 393-397.

Endo, T., Ohno, M., Kotani, S., Gunji, K. and Onaya, T.: Thyrotropin receptor in non-thyroid tissues. Biochem Biophys Res Commun 190 (1993) 774-9.

Eng, P.H., Cardona, G.R., Fang, S.L., Previti, M., Alex, S., Carrasco, N., Chin, W.W. and Braverman, L.E.: Escape from the acute Wolff-Chaikoff effect is associated with a decrease in thyroid sodium/iodide symporter messenger ribonucleic acid and protein. Endocrinology 140 (1999) 3404-10.

Eng, P.H., Cardona, G.R., Previti, M.C., Chin, W.W. and Braverman, L.E.: Regulation of the sodium iodide symporter by iodide in FRTL-5 cells. Eur J Endocrinol 144 (2001) 139-44.

EPA: Volume II Food ingestion factors; chapter 13 breast milk intake., http://www.epa.gov/ordntrnt/ORD/WebPubs/exposure/chap13.pdf, 1996.

EPA: Perchlorate Environmental Contamination: Toxicological Review and Risk Characterization, http://www.epa.gov/swerffrr/documents/perchlorate.htm, 2002.

EPA: Perchlorate Occurrences, http://www.epa.gov/fedfac/documents/perchlorate_links.htm#occurrences, 2005a.

EPA: State Perchlorate Advisory Levels, http://www.epa.gov/swerffrr/pdf/stateadvisorylevels.pdf, 2005b.

Erickson, B.E.: Perchlorate found in milk in Texas. Environmental Science & Technology 37 (2003) 377A-378A.

Eskandari, S., Loo, D.D., Dai, G., Levy, O., Wright, E.M. and Carrasco, N.: Thyroid Na+/I- symporter. Mechanism, stoichiometry, and specificity. J Biol Chem 272 (1997) 27230-8.

Everett, L.A., Glaser, B., Beck, J.C., Idol, J.R., Buchs, A., Heyman, M., Adawi, F., Hazani, E., Nassir, E., Baxevanis, A.D., Sheffield, V.C. and Green, E.D.: Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS). Nat Genet 17 (1997) 411-22.

157

Everett, L.A., Morsli, H., Wu, D.K. and Green, E.D.: Expression pattern of the mouse ortholog of the Pendred's syndrome gene (Pds) suggests a key role for pendrin in the inner ear. Proc Natl Acad Sci U S A 96 (1999) 9727-32.

EWG: Rocket Science, http://www.ewg.org/reports/rocketscience/chap1.html, 2001.

EWG: Perchlorate-contaminated drinking water sources in California., http://www.ewg.org/reports/rocketwater/table1.php, 2003.

Finley, D.A., Lonnerdal, B., Dewey, K.G. and Grivetti, L.E.: Breast milk composition: fat content and fatty acid composition in vegetarians and non-vegetarians. Am J Clin Nutr 41 (1985) 787-800.

Fisher, J., Todd, P., Mattie, D., Godfrey, D., Narayanan, L. and Yu, K.: Preliminary development of a physiological model for perchlorate in the adult male rat: a framework for further studies. Drug Chem Toxicol 23 (2000) 243-58.

Folch, J., Lees, M. and Sloane Stanley, G.H.: A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem 226 (1957) 497-509.

Francois, C.A., Connor, S.L., Wander, R.C. and Connor, W.E.: Acute effects of dietary fatty acids on the fatty acids of human milk. Am J Clin Nutr 67 (1998) 301-8.

Frische, S., Kwon, T.H., Frokiaer, J., Madsen, K.M. and Nielsen, S.: Regulated expression of pendrin in rat kidney in response to chronic NH4Cl or NaHCO3 loading. Am J Physiol Renal Physiol 284 (2003) F584-93.

Gentles, A., Surles, J. and Smith, E.E.: Evaluation of adult quail and egg production following exposure to perchlorate-treated water. Environ Toxicol Chem 24 (2005) 1930-4.

German, J.B. and Dillard, C.J.: Composition, structure and absorption of milk lipids: a source of energy, fat-soluble nutrients and bioactive molecules. Crit Rev Food Sci Nutr 46 (2006) 57-92.

Gibbs, J.P., Ahmad, R., Crump, K.S., Houck, D.P., Leveille, T.S., Findley, J.E. and Francis, M.: Evaluation of a population with occupational exposure to airborne ammonium perchlorate for possible acute or chronic effects on thyroid function. J Occup Environ Med 40 (1998) 1072-82.

Ginzinger, D.G.: Gene quantification using real-time quantitative PCR: an emerging technology hits the mainstream. Exp Hematol 30 (2002) 503-12.

Goldman, S.J. and Stanbury, J.B.: The metabolism of perchlorate in the rat. Endocrinology 92 (1973) 1536-8.

Goleman, W.L., Carr, J.A. and Anderson, T.A.: Environmentally relevant concentrations of ammonium perchlorate inhibit thyroid function and alter sex ratios in developing Xenopus laevis. Environ Toxicol Chem 21 (2002a) 590-7.

158

Goleman, W.L., Urquidi, L.J., Anderson, T.A., Smith, E.E., Kendall, R.J. and Carr, J.A.: Environmentally relevant concentrations of ammonium perchlorate inhibit development and metamorphosis in Xenopus laevis. Environ Toxicol Chem 21 (2002b) 424-30.

Goodridge, A.G.: Regulation of the gene for fatty acid synthase. Fed Proc 45 (1986) 2399-405.

Greer, M.A., Goodman, G., Pleus, R.C. and Greer, S.E.: Health effects assessment for environmental perchlorate contamination: the dose response for inhibition of thyroidal radioiodine uptake in humans. Environ Health Perspect 110 (2002) 927-37.

Hadley, M.E.: Endocrinology, fifth ed. Prentice Hall, Upper Saddle River, NJ, 2000.

Hamosh, M., Chernick, S.S. and Scow, R.O.: Lipoprotein lipase activity during pregnancy and lactation. Isr J Med Sci 8 (1972) 834-5.

Handy, R., Barnett, D.A., Purves, R.W., Horlick, G. and Guevremont, R.: Determination of nanomolar levels of perchlorate in water by ESI-FAIMS-MS. Journal of Analytical Atomic Spectrometry 15 (2000) 907-911.

Hautman, D.P., Munch, D.J., Eaton, A.D. and Haghani, A.W.: Method 314.0 Determination of Perchlorate in Drinking Water Using Ion Chromatography, Revision 1.0, EPA 815-B-99-003. U.S. Environmental Protection Agency, Cincinnati, OH, 1999.

Heird, W.C.: The role of polyunsaturated fatty acids in term and preterm infants and breastfeeding mothers. Pediatr Clin North Am 48 (2001) 173-88.

Innis, S.M.: The role of dietary n-6 and n-3 fatty acids in the developing brain. Developmental Neuroscience 22 (2000) 474-80.

Innis, S.M.: Essential fatty acid transfer and fetal development. Placenta 26 Suppl A (2005) S70-5.

Iverson, S.J., Hamosh, M. and Bowen, W.D.: Lipoprotein lipase activity and its relationship to high milk fat transfer during lactation in grey seals. J Comp Physiol [B] 165 (1995) 384-95.

Jackson, P.E. and Chassaniol, K.: Advances in the determination of inorganic ions in potable waters by ion chromatography. Journal of Environmental Monitoring 4 (2002) 10-15.

Jackson, P.E., Gokhale, S. and Rohrer, J.S.: Chapter 5: recent developments in the analysis of perchlorate using ion chromatography. In: Urbansky, E.T. (Ed.), Perchlorate in the Environment, New York, 2000a, pp. 37-44.

159

Jackson, P.E., Gokhale, S., Streib, T., Rohrer, J.S. and Pohl, C.A.: Improved method for the determination of trace perchlorate in ground and drinking waters by ion chromatography. Journal of Chromatography A 888 (2000b) 151-158.

Jackson, P.E., Laikhtman, M. and Rohrer, J.S.: Determination of trace level perchlorate in drinking water and ground water by ion chromatography. Journal of Chromatography A 850 (1999) 131-135.

Jackson, W.A., Anandam, S.K., Anderson, T., Lehman, T., Rainwater, K., Rajagopalan, S., Ridley, M. and Tock, R.: Perchlorate occurrence in the Texas southern high plains aquifer system. Ground Water Monitoring and Remediation 25 (2005a) 137-149.

Jackson, W.A., Arunagiri, S., Tock, R., Anderson, T. and Rainwater, K.: Technical note: electrochemical generation of perchlorate in municipal drinking water systems. Journal AWWA 96 (2004) 103-108.

Jackson, W.A., Joseph, P., Laxman, P., Tan, K., Smith, P.N., Yu, L. and Anderson, T.A.: Perchlorate accumulation in forage and edible vegetation. Journal of Agricultural and Food Chemistry 53 (2005b) 369-373.

Jensen, C.L. and Heird, W.C.: Lipids with an emphasis on long-chain polyunsaturated fatty acids. Clin Perinatol 29 (2002) 261-81, vi.

Josefsson, M., Evilevitch, L., Westrom, R., Grunditz, T. and Ekblad, E.: Sodium-iodide symporter mediates iodide secretion in rat gastric mucosa in vitro. Experimental Biology and Medicine 231 (2006) 277-281.

Josefsson, M., Grunditz, T., Ohlsson, T. and Ekblad, E.: Sodium/iodide-symporter: distribution in different mammals and role in entero-thyroid circulation of iodide. Acta Physiol Scand 175 (2002) 129-37.

Joyner, C.P., Myrick, L.C., Crossland, J.P. and Dawson, W.D.: Deer Mice As Laboratory Animals. Ilar J 39 (1998) 322-330.

Kameda, K.: Thyroid hormone inhibits fatty acid synthase gene transcription in chicken liver. Mol Cell Biochem 144 (1995) 105-8.

Kao, Y., Manzon, R.G., Sheridan, M.A. and Youson, J.H.: Study of the relationship between thyroid hormones and lipid metabolism during KClO4-induced metamorphosis of landlocked lamprey, Petromyzon marinus. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 122 (1999) 363-73.

Kempen, E.C. and Brodbelt, J.S.: A method for the determination of binding constants by electrospray ionization mass spectrometry. Anal Chem 72 (2000) 5411-6.

160

Kengen, S.W., Rikken, G.B., Hagen, W.R., van Ginkel, C.G. and Stams, A.J.: Purification and characterization of (per)chlorate reductase from the chlorate-respiring strain GR-1. J Bacteriol 181 (1999) 6706-11.

Kent, J.C., Mitoulas, L., Cox, D.B., Owens, R.A. and Hartmann, P.E.: Breast volume and milk production during extended lactation in women. Exp Physiol 84 (1999) 435-47.

Kirk, A.B.: Perchlorate in human and ruminant milk, Department of Environmental Toxicology. Texas Tech University, Lubbock, 2005.

Kirk, A.B., Martinelango, P.K., Tian, K., Dutta, A., Smith, E.E. and Dasgupta, P.K.: Perchlorate and iodide in dairy and breast milk. Environ Sci Technol 39 (2005) 2011-7.

Kirk, A.B., Smith, E.E., Tian, K., Anderson, T.A. and Dasgupta, P.K.: Perchlorate in milk. Environmental Science & Technology 37 (2003) 4979-4981.

Kishihara, M., Nakao, Y., Baba, Y., Matsukura, S., Kuma, K. and Fujita, T.: Interaction between thyroid-stimulating immunoglobulins and thyrotropin receptors in fat cell membranes. J Clin Endocrinol Metab 49 (1979) 706-11.

Koester, C.J., Beller, H.R. and Halden, R.U.: Analysis of perchlorate in groundwater by electrospray ionization mass spectrometry/mass spectrometry. Environmental Science & Technology 34 (2000) 1862-1864.

Koletzko, B. and Rodriguez-Palmero, M.: Polyunsaturated fatty acids in human milk and their role in early infant development. J Mammary Gland Biol Neoplasia 4 (1999) 269-84.

Kotani, T., Ogata, Y., Yamamoto, I., Aratake, Y., Kawano, J.I., Suganuma, T. and Ohtaki, S.: Characterization of gastric Na+/I- symporter of the rat. Clin Immunol Immunopathol 89 (1998) 271-8.

Krynitsky, A.J., Niemann, R.A. and Nortrup, D.A.: Determination of perchlorate anion in foods by ion chromatography-tandem mass spectrometry. Analytical Chemistry 76 (2004) 5518-5522.

Lacroix, L., Mian, C., Caillou, B., Talbot, M., Filetti, S., Schlumberger, M. and Bidart, J.M.: Na(+)/I(-) symporter and Pendred syndrome gene and protein expressions in human extra-thyroidal tissues. Eur J Endocrinol 144 (2001) 297-302.

Lamm, S.H., Braverman, L.E., Li, F.X., Richman, K., Pino, S. and Howearth, G.: Thyroid health status of ammonium perchlorate workers: a cross-sectional occupational health study. J Occup Environ Med 41 (1999) 248-60.

Lamm, S.H. and Doemland, M.: Has perchlorate in drinking water increased the rate of congenital hypothyroidism? J Occup Environ Med 41 (1999) 409-11.

161

Landrum, M., Canas, J.E., Coimbatore, G., Cobb, G.P., Jackson, W.A., Zhang, B. and Anderson, T.A.: Effects of perchlorate on earthworm (Eisenia fetida) survival and reproductive success. Sci Total Environ (2005).

Lapillonne, A., Clarke, S.D. and Heird, W.C.: Polyunsaturated fatty acids and gene expression. Curr Opin Clin Nutr Metab Care 7 (2004) 151-6.

Lawrence, J.E., Lamm, S.H., Pino, S., Richman, K. and Braverman, L.E.: The effect of short-term low-dose perchlorate on various aspects of thyroid function. Thyroid 10 (2000) 659-63.

Li, F.X., Byrd, D.M., Deyhle, G.M., Sesser, D.E., Skeels, M.R., Katkowsky, S.R. and Lamm, S.H.: Neonatal thyroid-stimulating hormone level and perchlorate in drinking water. Teratology 62 (2000a) 429-31.

Li, F.X., Squartsoff, L. and Lamm, S.H.: Prevalence of thyroid diseases in Nevada counties with respect to perchlorate in drinking water. J Occup Environ Med 43 (2001) 630-4.

Li, Y.T. and George, E.J.: Analysis of perchlorate in water by reversed-phase LC/ESI.MS/MS using an internal standard technique. Analytical Chemistry 77 (2005) 4453-4458.

Li, Z., Li, F.X., Byrd, D., Deyhle, G.M., Sesser, D.E., Skeels, M.R. and Lamm, S.H.: Neonatal thyroxine level and perchlorate in drinking water. J Occup Environ Med 42 (2000b) 200-5.

Lina, B.A. and Kuijpers, M.H.: Toxicity and carcinogenicity of acidogenic or alkalogenic diets in rats; effects of feeding NH(4)Cl, KHCO(3) or KCl. Food Chem Toxicol 42 (2004) 135-53.

Liu, Y.J., Mou, S. and Heberling, S.: Determination of trace level bromate and perchlorate in drinking water by ion chromatography with an evaporative preconcentration technique. Journal of Chromatography A 956 (2002) 85-91.

Magnuson, M.L., Urbansky, E.T. and Kelty, C.A.: Determination of perchlorate at trace levels in drinking water by ion-pair extraction with electrospray ionization mass spectrometry. Analytical Chemistry 72 (2000a) 25-29.

Magnuson, M.L., Urbansky, E.T. and Kelty, C.A.: Microscale extraction of perchlorate in drinking water with low level detection by electrospray-mass spectrometry. Talanta 52 (2000b) 285-291.

Manzon, R.G. and Youson, J.H.: The effects of exogenous thyroxine (T4) or triiodothyronine (T3), in the presence and absence of potassium perchlorate, on the incidence of metamorphosis and on serum T4 and T3 concentrations in larval sea lampreys (Petromyzon marinus L.). Gen Comp Endocrinol 106 (1997) 211-20.

162

Marangoni, F., Colombo, C., De Angelis, L., Gambaro, V., Agostoni, C., Giovannini, M. and Galli, C.: Cigarette smoke negatively and dose-dependently affects the biosynthetic pathway of the n-3 polyunsaturated fatty acid series in human mammary epithelial cells. Lipids 39 (2004) 633-7.

Martinelango, P.K., Anderson, J.L., Dasgupta, P.K., Armstrong, D.W., Al-Horr, R.S. and Slingsby, R.W.: Gas-phase ion association provides increased selectivity and sensitivity for measuring perchlorate by mass spectrometry. Analytical Chemistry 77 (2005) 4829-4835.

McNabb, F.M., Larsen, C.T. and Pooler, P.S.: Ammonium perchlorate effects on thyroid function and growth in bobwhite quail chicks. Environ Toxicol Chem 23 (2004) 997-1003.

Medina, V.F., Larson, S.L., Extine, B. and Bednar, A.: Perchlorate analysis using solid-phase extraction cartridges. Journal of Chromatographic Science 43 (2005) 195-200.

Merrill, E.A., Clewell, R.A., Gearhart, J.M., Robinson, P.J., Sterner, T.R., Yu, K.O., Mattie, D.R. and Fisher, J.W.: PBPK predictions of perchlorate distribution and its effect on thyroid uptake of radioiodide in the male rat. Toxicol Sci 73 (2003) 256-69.

Merrill, E.A., Clewell, R.A., Robinson, P.J., Jarabek, A.M., Gearhart, J.M., Sterner, T.R. and Fisher, J.W.: PBPK model for radioactive iodide and perchlorate kinetics and perchlorate-induced inhibition of iodide uptake in humans. Toxicol Sci 83 (2005) 25-43.

Mian, C., Lacroix, L., Alzieu, L., Nocera, M., Talbot, M., Bidart, J.M., Schlumberger, M. and Caillou, B.: Sodium iodide symporter and pendrin expression in human thyroid tissues. Thyroid 11 (2001) 825-30.

Mitchell, A.M., Manley, S.W., Morris, J.C., Powell, K.A., Bergert, E.R. and Mortimer, R.H.: Sodium iodide symporter (NIS) gene expression in human placenta. Placenta 22 (2001) 256-8.

Morrison, W.R. and Smith, L.M.: Preparation of Fatty Acid Methyl Esters and Dimethylacetals from Lipids with Boron Fluoride--Methanol. J Lipid Res 5 (1964) 600-8.

Motzer, W.E.: Perchlorate: Problems, detection, and solutions. Environmental Forensics 2 (2001) 301-311.

Mukhi, S., Carr, J.A., Anderson, T.A. and Patino, R.: Novel biomarkers of perchlorate exposure in zebrafish. Environ Toxicol Chem 24 (2005) 1107-15.

Narayanan, L., Buttler, G.W., Yu, K.O., Mattie, D.R. and Fisher, J.W.: Sensitive high-performance liquid chromatography method for the determination of low levels of

163

perchlorate in biological samples. Journal of Chromatography B-Analytical Technologies in the Biomedical and Life Sciences 788 (2003) 393-399.

NAS: Health Implications of Perchlorate Ingestion. THE NATIONAL ACADEMIES PRESS, Washington, D. C., 2005.

Neville, M.C. and Picciano, M.F.: Regulation of milk lipid secretion and composition. Annu Rev Nutr 17 (1997) 159-83.

NRC: Nutrient Requirements of Dairy Cattle. National Research Council, National Academy Press, Washingdon DC, 2001.

Patino, R., Wainscott, M.R., Cruz-Li, E.I., Balakrishnan, S., McMurry, C., Blazer, V.S. and Anderson, T.A.: Effects of ammonium perchlorate on the reproductive performance and thyroid follicle histology of zebrafish. Environ Toxicol Chem 22 (2003) 1115-21.

Peña, H.G., Kessler, W.V., Christian, J.E., Cline, T.R. and Plumlee, M.P.: A comparative study of iodine and potassium perchlorate metabolism in the laying hen. 2. Uptake, distribution, and excretion of potassium perchlorate. Poult Sci 55 (1976) 188-201.

Perron, B., Rodriguez, A.M., Leblanc, G. and Pourcher, T.: Cloning of the mouse sodium iodide symporter and its expression in the mammary gland and other tissues. J Endocrinol 170 (2001) 185-96.

Pinke, L.A., Dean, D.S., Bergert, E.R., Spitzweg, C., Dutton, C.M. and Morris, J.C.: Cloning of the mouse sodium iodide symporter. Thyroid 11 (2001) 935-9.

Pourreza, N. and Mousavi, H.Z.: Extraction spectrophotometric determination of trace amounts of perchlorate based on ion-pair formation with thionine. Journal of Analytical Chemistry 60 (2005) 816-818.

Quentin, F., Chambrey, R., Trinh-Trang-Tan, M.M., Fysekidis, M., Cambillau, M., Paillard, M., Aronson, P.S. and Eladari, D.: The Cl-/HCO3- exchanger pendrin in the rat kidney is regulated in response to chronic alterations in chloride balance. Am J Physiol Renal Physiol 287 (2004) F1179-88.

Ramachandran, B.R.: Characterization of pendrin gene expression in developing and adult deer mice following exposure to ammonium perchlorate during gestation and post-gestation, Environemtal Toxicology. Texas Tech University, Lubbock, TX, 2005.

Rillema, J.A. and Hill, M.A.: Prolactin regulation of the pendrin-iodide transporter in the mammary gland. Am J Physiol Endocrinol Metab 284 (2003) E25-8.

164

Rillema, J.A., Williams, C.H., Moulden, J. and Golden, K.L.: Effect of insulin on iodide uptake in mouse mammary gland explants. Exp Biol Med (Maywood) 227 (2002) 32-5.

Rillema, J.A., Yu, T.X. and Jhiang, S.M.: Effect of prolactin on sodium iodide symporter expression in mouse mammary gland explants. Am J Physiol Endocrinol Metab 279 (2000) E769-72.

Royaux, I.E., Suzuki, K., Mori, A., Katoh, R., Everett, L.A., Kohn, L.D. and Green, E.D.: Pendrin, the protein encoded by the Pendred syndrome gene (PDS), is an apical porter of iodide in the thyroid and is regulated by thyroglobulin in FRTL-5 cells. Endocrinology 141 (2000) 839-45.

Royaux, I.E., Wall, S.M., Karniski, L.P., Everett, L.A., Suzuki, K., Knepper, M.A. and Green, E.D.: Pendrin, encoded by the Pendred syndrome gene, resides in the apical region of renal intercalated cells and mediates bicarbonate secretion. Proc Natl Acad Sci U S A 98 (2001) 4221-6.

Sanchez, C.A., Krieger, R.I., Khandaker, N., Moore, R.C., Holts, K.C. and Neidel, L.L.: Accumulation and perchlorate exposure potential of lettuce produced in the Lower Colorado River region. J Agric Food Chem 53 (2005) 5479-86.

Scott, D.A. and Karniski, L.P.: Human pendrin expressed in Xenopus laevis oocytes mediates chloride/formate exchange. Am J Physiol Cell Physiol 278 (2000) C207-11.

Scott, D.A., Wang, R., Kreman, T.M., Sheffield, V.C. and Karniski, L.P.: The Pendred syndrome gene encodes a chloride-iodide transport protein. Nat Genet 21 (1999) 440-3.

Siglin, J.C., Mattie, D.R., Dodd, D.E., Hildebrandt, P.K. and Baker, W.H.: A 90-day drinking water toxicity study in rats of the environmental contaminant ammonium perchlorate. Toxicol Sci 57 (2000) 61-74.

Slominski, A., Wortsman, J., Kohn, L., Ain, K.B., Venkataraman, G.M., Pisarchik, A., Chung, J.H., Giuliani, C., Thornton, M., Slugocki, G. and Tobin, D.J.: Expression of hypothalamic-pituitary-thyroid axis related genes in the human skin. J Invest Dermatol 119 (2002) 1449-55.

Smanik, P.A., Liu, Q., Furminger, T.L., Ryu, K., Xing, S., Mazzaferri, E.L. and Jhiang, S.M.: Cloning of the human sodium lodide symporter. Biochem Biophys Res Commun 226 (1996) 339-45.

Smith, P.N., Theodorakis, C.W., Anderson, T.A. and Kendall, R.J.: Preliminary assessment of perchlorate in ecological receptors at the Longhorn Army Ammunition Plant (LHAAP), Karnack, Texas. Ecotoxicology 10 (2001) 305-13.

165

Smith, P.N., Utley, S.J., Cox, S.B., Anderson, T.A. and McMurry, S.T.: Monitoring perchlorate exposure and thyroid hormone status among raccoons inhabiting a perchlorate-contaminated site. Environ Monit Assess 102 (2005) 337-47.

Smith, P.N., Yu, L., McMurry, S.T. and Anderson, T.A.: Perchlorate in water, soil, vegetation, and rodents collected from the Las Vegas Wash, Nevada, USA. Environmental Pollution 132 (2004a) 121-127.

Smith, P.N., Yu, L., McMurry, S.T. and Anderson, T.A.: Perchlorate in water, soil, vegetation, and rodents collected from the Las Vegas Wash, Nevada, USA. Environ Pollut 132 (2004b) 121-7.

Snyder, S.A., Vanderford, B.J. and Rexing, D.J.: Trace analysis of bromate, chlorate, iodate, and perchlorate in natural and bottled waters. Environmental Science & Technology 39 (2005) 4586-4593.

Soldin, O.P., Braverman, L.E. and Lamm, S.H.: Perchlorate clinical pharmacology and human health: a review. Ther Drug Monit 23 (2001) 316-31.

Soleimani, M., Greeley, T., Petrovic, S., Wang, Z., Amlal, H., Kopp, P. and Burnham, C.E.: Pendrin: an apical Cl-/OH-/HCO3- exchanger in the kidney cortex. Am J Physiol Renal Physiol 280 (2001) F356-64.

Spitzweg, C., Dutton, C.M., Castro, M.R., Bergert, E.R., Goellner, J.R., Heufelder, A.E. and Morris, J.C.: Expression of the sodium iodide symporter in human kidney. Kidney Int 59 (2001) 1013-23.

Spitzweg, C., Joba, W., Eisenmenger, W. and Heufelder, A.E.: Analysis of human sodium iodide symporter gene expression in extrathyroidal tissues and cloning of its complementary deoxyribonucleic acids from salivary gland, mammary gland, and gastric mucosa. J Clin Endocrinol Metab 83 (1998) 1746-51.

Spitzweg, C., Joba, W., Morris, J.C. and Heufelder, A.E.: Regulation of sodium iodide symporter gene expression in FRTL-5 rat thyroid cells. Thyroid 9 (1999a) 821-30.

Spitzweg, C., Joba, W., Schriever, K., Goellner, J.R., Morris, J.C. and Heufelder, A.E.: Analysis of human sodium iodide symporter immunoreactivity in human exocrine glands. J Clin Endocrinol Metab 84 (1999b) 4178-84.

Stapleton, S.R., Mitchell, D.A., Salati, L.M. and Goodridge, A.G.: Triiodothyronine stimulates transcription of the fatty acid synthase gene in chick embryo hepatocytes in culture. Insulin and insulin-like growth factor amplify that effect. J Biol Chem 265 (1990) 18442-6.

Sterner, J.L., Johnston, M.V., Nicol, G.R. and Ridge, D.P.: Signal suppression in electrospray ionization Fourier transform mass spectrometry of multi-component samples. J Mass Spectrom 35 (2000) 385-91.

166

Stevens, L.J., Zentall, S.S., Abate, M.L., Kuczek, T. and Burgess, J.R.: Omega-3 fatty acids in boys with behavior, learning, and health problems. Physiol Behav 59 (1996) 915-20.

Stokstad, E.: Environmental health. Debate continues over safety of water spiked with rocket fuel. Science 307 (2005) 507.

Susarla, S., Bacchus, S.T., Harvey, G. and McCutcheon, S.C.: Phytotransformations of perchlorate contaminated waters. Environmental Technology 21 (2000) 1055-1065.

Susarla, S., Collette, T.W., Garrison, A.W., Wolfe, N.L. and McCutcheon, S.C.: Perchlorate identification in fertilizers. Environmental Science & Technology 33 (1999) 3469-3472.

Suzuki, K., Royaux, I.E., Everett, L.A., Mori-Aoki, A., Suzuki, S., Nakamura, K., Sakai, T., Katoh, R., Toda, S., Green, E.D. and Kohn, L.D.: Expression of PDS/Pds, the Pendred syndrome gene, in endometrium. J Clin Endocrinol Metab 87 (2002) 938.

Tan, K., Anderson, T.A., Jones, M.W., Smith, P.N. and Jackson, W.A.: Accumulation of perchlorate in aquatic and terrestrial plants at a field scale. J Environ Qual 33 (2004) 1638-46.

Tanner, G.A.: Renal regulation of acid-base balance: ammonia excretion. Physiologist 27 (1984) 95-7.

Thuett, K.A., Roots, E.H., Mitchell, L.P., Gentles, B.A., Anderson, T., Kendall, R.J. and Smith, E.E.: Effects of in utero and lactational ammonium perchlorate exposure on thyroid gland histology and thyroid and sex hormones in developing deer mice (peromyscus maniculatus) through postnatal day 21. J Toxicol Environ Health A 65 (2002) 2119-30.

Tian, K., Canas, J.E., Dasgupta, P.K. and Anderson, T.A.: Preconcentration/preelution ion chromatography for the determination of perchlorate in complex samples. Talanta 65 (2005) 750-755.

Tian, K., Dasgupta, P.K. and Anderson, T.A.: Determination of trace perchlorate in high-salinity water samples by ion chromatography with on-line preconcentration an preelution. Analytical Chemistry 75 (2003) 701-706.

Tietge, J.E., Holcombe, G.W., Flynn, K.M., Kosian, P.A., Korte, J.J., Anderson, L.E., Wolf, D.C. and Degitz, S.J.: Metamorphic inhibition of Xenopus laevis by sodium perchlorate: effects on development and thyroid histology. Environ Toxicol Chem 24 (2005) 926-33.

167

Tizianello, A., Deferrari, G., Garibotto, G., Robaudo, C., Acquarone, N. and Ghiggeri, G.M.: Renal ammoniagenesis in an early stage of metabolic acidosis in man. J Clin Invest 69 (1982) 240-50.

Uauy, R., Hoffman, D.R., Peirano, P., Birch, D.G. and Birch, E.E.: Essential fatty acids in visual and brain development. Lipids 36 (2001) 885-95.

Uauy, R., Mena, P. and Rojas, C.: Essential fatty acids in early life: structural and functional role. Proc Nutr Soc 59 (2000) 3-15.

Urbansky, E.T.: Perchlorate Chemistry: Implications for Analysis and Remediation. Bioremediation Journal 2 (1998).

Urbansky, E.T.: Quantitation of perchlorate ion: Practices and advances applied to the analysis of common matrices. Critical Reviews in Analytical Chemistry 30 (2000) 311-343.

Urbansky, E.T.: Perchlorate as an environmental contaminant. Environmental Science and Pollution Research 9 (2002) 187-192.

Urbansky, E.T., Brown, S.K., Magnuson, M.L. and Kelty, C.A.: Perchlorate levels in samples of sodium nitrate fertilizer derived from Chilean caliche. Environmental Pollution 112 (2001) 299-302.

Urbansky, E.T. and Collette, T.W.: Comparison and evaluation of laboratory performance on a method for the determination of perchlorate in fertilizers. Journal of Environmental Monitoring 3 (2001) 454-462.

Urbansky, E.T., Gu, B.H., Magnuson, M.L., Brown, G.M. and Kelty, C.A.: Survey of bottled waters for perchlorate by electrospray ionization mass spectrometry (ESI-MS) and ion chromatography (IC). Journal of the Science of Food and Agriculture 80 (2000a) 1798-1804.

Urbansky, E.T., Magnuson, M.L., Freeman, D. and Jelks, C.: Quantitation of perchlorate ion by electrospray ionization mass spectrometry (ESI-MS) using stable association complexes with organic cations and bases to enhance selectivity. Journal of Analytical Atomic Spectrometry 14 (1999) 1861-1866.

Urbansky, E.T., Magnuson, M.L., Kelty, C.A. and Brown, S.K.: Perchlorate uptake by salt cedar (Tamarix ramosissima) in the Las Vegas Wash riparian ecosystem. Science of the Total Environment 256 (2000b) 227-232.

Urbansky, E.T. and Schock, M.R.: Issues in managing the risks associated with perchlorate in drinking water. Journal of Environmental Management 56 (1999) 79-95.

168

Valentin-Blasini, L., Mauldin, J.P., Maple, D. and Blount, B.C.: Analysis of perchlorate in human urine using ion chromatography and electrospray tandem mass spectrometry. Analytical Chemistry 77 (2005) 2475-2481.

Van Sande, J., Massart, C., Beauwens, R., Schoutens, A., Costagliola, S., Dumont, J.E. and Wolff, J.: Anion selectivity by the sodium iodide symporter. Endocrinology 144 (2003) 247-52.

Vayre, L., Sabourin, J.C., Caillou, B., Ducreux, M., Schlumberger, M. and Bidart, J.M.: Immunohistochemical analysis of Na+/I- symporter distribution in human extra-thyroidal tissues. Eur J Endocrinol 141 (1999) 382-6.

Verlander, J.W., Hassell, K.A., Royaux, I.E., Glapion, D.M., Wang, M.E., Everett, L.A., Green, E.D. and Wall, S.M.: Deoxycorticosterone upregulates PDS (Slc26a4) in mouse kidney: role of pendrin in mineralocorticoid-induced hypertension. Hypertension 42 (2003) 356-62.

Wagner, C.A., Finberg, K.E., Stehberger, P.A., Lifton, R.P., Giebisch, G.H., Aronson, P.S. and Geibel, J.P.: Regulation of the expression of the Cl-/anion exchanger pendrin in mouse kidney by acid-base status. Kidney Int 62 (2002) 2109-17.

Wainwright, P.E.: Dietary essential fatty acids and brain function: a developmental perspective on mechanisms. Proc Nutr Soc 61 (2002) 61-9.

Wall, S.M., Kim, Y.H., Stanley, L., Glapion, D.M., Everett, L.A., Green, E.D. and Verlander, J.W.: NaCl restriction upregulates renal Slc26a4 through subcellular redistribution: role in Cl- conservation. Hypertension 44 (2004) 982-7.

Williams, T.L., Martin, R.B. and Collette, T.W.: Raman spectroscopic analysis of fertilizers and plant tissue for perchlorate. Applied Spectroscopy 55 (2001) 967-983.

Wilson, S.B., Back, D.W., Morris, S.M., Jr., Swierczynski, J. and Goodridge, A.G.: Hormonal regulation of lipogenic enzymes in chick embryo hepatocytes in culture. Expression of the fatty acid synthase gene is regulated at both translational and pretranslational steps. J Biol Chem 261 (1986) 15179-82.

Winkler, P., Minteer, M. and Willey, J.: Analysis of perchlorate in water and soil by electrospray LC/MS/MS. Analytical Chemistry 76 (2004) 469-473.

Wolff, J.: Perchlorate and the thyroid gland. Pharmacol Rev 50 (1998) 89-105.

Wolff, J.: What is the role of pendrin? Thyroid 15 (2005) 346-8.

Wolff, J. and Chaikoff, I.L.: Plasma inorganic iodide as a homeostatic regulator of thyroid function. Journal of Biological Chemistry 174 (1948) 555–564.

York, R.G., Barnett, J., Jr., Brown, W.R., Garman, R.H., Mattie, D.R. and Dodd, D.: A rat neurodevelopmental evaluation of offspring, including evaluation of adult and

169

neonatal thyroid, from mothers treated with ammonium perchlorate in drinking water. Int J Toxicol 23 (2004) 191-214.

York, R.G., Brown, W.R., Girard, M.F. and Dollarhide, J.S.: Oral (drinking water) developmental toxicity study of ammonium perchlorate in New Zealand White rabbits. Int J Toxicol 20 (2001a) 199-205.

York, R.G., Brown, W.R., Girard, M.F. and Dollarhide, J.S.: Two-generation reproduction study of ammonium perchlorate in drinking water in rats evaluates thyroid toxicity. Int J Toxicol 20 (2001b) 183-97.

York, R.G., Funk, K.A., Girard, M.F., Mattie, D. and Strawson, J.E.: Oral (drinking water) developmental toxicity study of ammonium perchlorate in Sprague-Dawley rats. Int J Toxicol 22 (2003) 453-64.

Yoshida, A., Taniguchi, S., Hisatome, I., Royaux, I.E., Green, E.D., Kohn, L.D. and Suzuki, K.: Pendrin is an iodide-specific apical porter responsible for iodide efflux from thyroid cells. J Clin Endocrinol Metab 87 (2002) 3356-61.

Yoshino, T., Sato, E., Nakashima, T., Nagashima, W., Teranishi, M.A., Nakayama, A., Mori, N., Murakami, H., Funahashi, H. and Imai, T.: The immunohistochemical analysis of pendrin in the mouse inner ear. Hear Res 195 (2004) 9-16.

Yu, K.O., Narayanan, L., Mattie, D.R., Godfrey, R.J., Todd, P.N., Sterner, T.R., Mahle, D.A., Lumpkin, M.H. and Fisher, J.W.: The pharmacokinetics of perchlorate and its effect on the hypothalamus-pituitary-thyroid axis in the male rat. Toxicol Appl Pharmacol 182 (2002) 148-59.

Yu, L., Canas, J.E., Cobb, G.P., Jackson, W.A. and Anderson, T.A.: Uptake of perchlorate in terrestrial plants. Ecotoxicol Environ Saf 58 (2004) 44-9.

Zanartu, D., Polan, C.E., Ferreri, L.E. and McGilliard, M.L.: Effect of stage of lactation and varying available energy intake on milk production, milk composition, and subsequent tissue enzymic activity. J Dairy Sci 66 (1983) 1644-52.

Zoeller, R.T.: Challenges confronting risk analysis of potential thyroid toxicants. Risk Anal 23 (2003) 143-62.

170

171

APPENDIX

Table A.1. Perchlorate uptake and distribution in various studies. Test Species Dose Observation Reference Rats and rabbits 36Cl18O4

- The highest concentration was in the thyroid, followed by ova and blood

Anber et al., 1959

Hen 100 μCi0 K36ClO4 The greatest concentration was in thyroid, followed by gizzard lining in hens at 3 hours after a single injection

Pena et al., 1976

Rats and guinea pigs

0.5 μCi0 K36ClO4/rat or pig

Much higher perchlorate concentration in thyroid, and the 36ClO4

- concentration in the thyroid was closely negative related to the doses of the stable perchlorate received

Chow et al., 1969; 1970

Rat (male) 3.3 mg/kg 36ClO4- Perchlorate was actively sequestered into the

thyroid gland, gastrointestinal tract, and possibly the skin

Yu et al., 2002

Rats (pregnant and lactating)

10 mg/day ClO4- from

gestation day 2 until either gestation day 20 or postnatal day 10

The highest concentration was in thyroid, followed by milk, amniotic fluid, and serum

Narayanan et al., 2003

172

Table A.2. Perchlorate excretion in various studies. Test Species Dose Observation Reference Human 784 mg NaClO4 ClO4

- diffused rapidly in the organism and appeared in the urine 10 minutes after ingestion, with 30, 50, 85, and 95% of the perchlorate at 3, 4, 24, and 48 hours, respectively

Durand, 1938

Rat 0.1 μCi0 K36ClO4 Perchlorate concentration in thyroid reached a peak around 4 h, and then fell to approximately 5% of the peak value within 95 h with a half-life of 20 h

Goldman et al., 1973

Hen 100 μCi0 K36ClO4 87.6% and 99.4% of 36ClO4- was recovered in

excreta 3 h and 24 h after administration, and the largest total deposition in the 3-hour group was found in the blood (2.9%), followed by muscles, internal organs, and ova

Pena et al., 1976

Cattle 2, 4, or 6 g/day for one day or 4g/day for 10 day

Perchlorate was excreted in the urine at a concentration-related elimination rate after oral administration of a single dose, and prolonged perchlorate administration resulted in a longer excretion period

Batjoens et al., 1993

Rat (male) 3.3 mg/kg 36ClO4- 99.5% perchlorate recovery in urine, and 95% of

injected perchlorate was cleared in 55 to 75 h

Yu et al., 2002

Human (9 male)

10 mg/day ClO4- for

14 days Urinary perchlorate markedly increased and reached a level of ~7.6 mg per 24 hours at 7 and 14 days

Lawrence et al., 2000

Human (8) 0.5 mg/kg/day for 2 weeks

Perchlorate elimination rate with a half-life ranged from 6.0 to 9.3 h following perchlorate withdrawal

Geer et al., 2002

173

y = -2E-06x + 1.274R2 = 0.0039

y = 2E-06x + 1.1056R2 = 0.0014

0

0.5

1

1.5

2

2.5

3

10 100 1000 10000 100000Perchlorate excretion at dy 28 (ng)

Gen

e ex

pres

sion

at d

ay 2

8

NIS

pendrin

y = -2E-05x + 1.3096R2 = 0.0266

y = -4E-06x + 1.1402R2 = 0.0005

0

0.5

1

1.5

2

2.5

3

10 100 1000 10000 100000average perchlorate excretion (ng/day)

Gen

e ex

pres

sion

at d

ay 2

8

NIS

pendrin

y = -1E-06x + 1.3047R2 = 0.0223

y = -3E-07x + 1.1389R2 = 0.0004

0

0.5

1

1.5

2

2.5

3

100 1000 10000 100000 1000000

Total perchlorate excretion (ng)

Gen

e ex

pres

sion

at d

ay 2

8 NIS

pendrin

Figure A.1. Linear relationships between gene expression (NIS and pendrin) at

day 28 and perchlorate urinary excretion at day 28, average excretion, and total excretion over the 28-d exposure. No significant relationship was observed.

173

PERMISSION TO COPY

In presenting this dissertation in partial fulfillment of the requirements for a

PhD degree at Texas Tech University or Texas Tech University Health Sciences

Center, I agree that the Library and my major department shall make it freely

available for research purposes. Permission to copy this thesis for scholarly

purposes may be granted by the Director of the Library or my major professor. It

is understood that any copying or publication of this thesis for financial gain shall

not be allowed without my further written permission and that any user may be

liable for copyright infringement.

Agree (Permission is granted.)

Qiuqiong Cheng 4/20/2006

Student Signature Date

Disagree (Permission is not granted.)

_______________________________________________ _________________

Student Signature Date