W35 Illillllllllllll\llllllllllllllllllllll ;) £100 3 1293 02046 7258 LIBRARY Michigan State University This is to certify that the dissertation entitled Instrumental and Bioanalytical Measures of ' Endocrine Disruptors in Water presented by Shane Allen Snyder has been accepted towards fulfillment of the requirements for Doctor of Philom degree in ZOOlOQ‘V and Environmental Toxicology 05%,? “[4441 / Major WW Mew MSU is an Affirmative Action/Equal Opportunity Institution 0- 12771 PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE 0g gggE l I A 1 11/00 emIRCIDatoDuopeS-pu Instrumental and Bioanalytical Measures of Endocrine Disruptors in Water By Shane Allen Snyder A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Zoology and Institute for Environmental Toxicology 2000 ABSTRACT Instrumental and Bioanalytical Measures of Endocrine Disruptors in Water By Shane Allen Snyder A number of compounds released into the environment by human activities can modulate endogenous hormone activities and have been termed endocrine-disrupting compounds (EDCs). It has been hypothesized that such compounds may elicit a variety of adverse effects in both humans and wildlife, including promotion of hormone-dependent cancers and reduction in reproductive fitness. Concern over EDCs has created a need to monitor for these compounds in the environment. This need is underscored by recent legislation mandating that chemicals and formulations be screened for potential estrogenic activity before they are manufactured or used in certain processes (Safe Drinking Water Act Amendments of 1995 - Bill Number S1316; Food Quality Protection Act of 1996 - Bill Number PL. 104-170). Several methods for the sensitive detection and quantitation of EDCs from water and tissue samples have been developed. Nonylphenol (NP), a minor transient degradation product of the widely used nonylphenol polyethoxylate (NPE) non-ionic surfactants, was tested for bioaccumulation in fathead minnows. After a 42 d continuous exposure, fish tissues were analyzed. The resulting bioconcentration factors (BCFs) ranged from 203 — 268. While this method was appropriate for fathead minnows weighing less than 2 9, it was not applicable for larger tissue sample sizes. Therefore, a more reliable method was desired. Extractive steam-distillation was employed to extract NP and lower oligomer NPEs from fish tissue samples. Optimization of this method resulted in consistent recoveries greater than 70% and detection limits less than 20 ng/g. The method was tested using carp captured in the Las Vegas Bay of Lake Mead, Nevada. The average concentration of NP detected in the carp tissue was 184 ng/g wet weight. Wastewater effluents and surface waters from Michigan and Nevada were screened for estrogenic and anti-estrogenic compounds using a novel method which combines analytical chemistry and in vitm bioassays. Bioassay-directed chemical fractionation and instrumental analyses indicated that all observed estrogenicity was caused by the fraction of greatest polarity. Results indicate that the natural steroid 17B-estradiol and the synthetic steroid ethynylestradiol were most likely responsible for observed bioactivity. A follow-up study was initiated at Lake Mead, Nevada. Water samples of 100 L volume were extracted and analyzed using various instrumental and bioanalytical methods. Concentrations of priority pollutants were found to be low relative to other sites in the US. However. several new contaminants were identified. These included pharmaceuticals, flame-retardants, and personal care products. Many of these compounds have not been reported previously in the surface waters of North America. Copyright by Shane Allen Snyder 2000 ACKNOWLEDGMENTS The reseach contained herein would not have been possible without the guidance and support of many people and organizations. I would like to thank Dr. John Giesy for serving as my advisor and for his years of support and friendship. I would also like to thank Dr. Frank D’ltri, Dr. Robert Huggett, and Dr. Matthew Zabik for serving as members to my graduate committee and for their time and efforts in helping me achieve my research and professional goals. I would like to express my sincere appreciation to my friends and colleagues, without their support this research would not have been possible. Acknowledgments for specific components of my research are made in the following chapters. The MSU Aquatic Toxicology Laboratory has provided me with a breadth of experience. This was in no small part due to interactions with peers of various expertise and I would like to express my appreciation for their help and support. I would like to specifically thank Erin Snyder, Dan Villenueve, Tim Keith, Kevin Henry, Dave Verbrugge, K. Kannan, and Pete Horvath. The organizations which provided funding and material and technical assistance are acknowledged in the appropriate chapters of this dissertation. However, I would like to specifically thank those who have inspired and guided my research, namely: Dr. Carter Naylor of Huntsman Corporation; Dr. Kevin Kelly, Margaret Lake, and Dr. James LaBounty of US. Department of the Interior - Bureau of Reclamation; Peggy Roefer, Kim Zikmund, and Kay Brothers of the Southern Nevada Water Authority; Doug Karafa and Bill Shepherd of the Clark County Sanitation District; and Bill Burke, Fred DeSiIva, and Roy Irwin of the National Park Service. vi TABLE OF CONTENTS LIST OF TABLES ............................................................................................. . ..... x LIST OF FIGURES ............................................................................................... xii INTRODUCTION ................................................................................................... 1 CHAPTER 1. OVERVIEW OF INSTRUMENTAL AND BIOANALYTICAL MEASURES OF ENDOCRINE DISRUPTERS IN WATER ................................... 6 INTRODUCTION ......................................................................................... 6 IN VITRO BIOASSAYS ............................................................................. 11 IN VIVO VERSUS IN VITRO BIOASSAYS FOR TESTING EDCs ........... 16 USE OF FISH TO TEST FOR EDCs ........................................................ 16 Selection of test species ................................................................. 18 End points and biomarkers used to assess effects of EDCs .......... 18 Field Studies ................................................................................... 19 Laboratory Studies ......................................................................... 20 MONITORING OF BUTYL TIN COMPOUNDS IN WATER ............... . ...... 26 Ecological impacts .......................................................................... 28 Instrumental analysis and biomoniton'ng ........................................ 29 BlOASSAY-DIRECTED CHEMICAL FRACTIONATION .......................... 32 CONCLUSIONS ........................................................................................ 35 REFERENCES .......................................................................................... 38 CHAPTER 2. BIOCONCENTRATION OF NONYLPHENOL IN FATHEAD MINNOWS (PIMEPHALES PROMELAS) ............................................................ 50 INTRODUCTION .................................... ' ...... . ............................................ 5 0 EXPERIMENTAL SECTION ..................................................................... 53 Standards and reagents ............ . .................................................... 53 Fish exposure .............................. . .................................................. 54 Water extraction ................................ - ............................................ 55 Fish extraction and clean-up .......................................................... 56 Quantitation of compounds ............................................................ 57 Recovery and precision ............... . .................................................. 57 RESULTS .................................................................................................. 58 DISCUSSION ............................................................................................ 59 ACKNOWLEDGMENTS ............................................................................ 60 REFERENCES .......................................................................................... 60 vii CHAPTER 3. A METHOD FOR DETERMINING CONCENTRATIONS OF NONYLPHENOL AND LOWER OLIGOMER NONYLPHENOL ETHOXYLATES IN FISH TISSUES ................................................................................................ 69 ABSTRACT ............................................ . .................................................. 69 INTRODUCTION ....................................................................................... 70 EXPERIMENTAL SECTION ..................................................................... 72 Standards and reagents ................................................................. 72 Sample collection and preservation ............................................... 72 Extraction ....................................................................................... 73 NonnaI-phase HPLC fractionation .................................................. 74 Identification and Quantitation ........................................................ 75 Recovery and Detection Limits ....................................................... 75 RESULTS AND DISCUSSION .................................................................. 76 ACKNOWLEDGMENTS ................................................................ . ........... 77 REFERENCES .......................................................................................... 77 CHAPTER 4. ANALYTICAL METHODS FOR DETECTION OF SELECTED ESTROGENIC COMPOUNDS IN AQUEOUS MIXTURES ................................. 84 ABSTRACT .................................................. . ........................................... 84 INTRODUCTION ....................................................................................... 85 EXPERIMENTAL SECTION ..................................................................... 88 Standards and reagents ................................................................. 88 Sample collection and preservation ............................................... 90 Extraction ....................................................................................... 92 Normal-phase HPLC fractionation .................................................. 92 Quantitation of compounds ............................................................ 93 Radioimmunoassay (RIA) .............................................................. 94 Recovery and Precision ................................................................. 95 Detection Limits ................................................. .......................... 97 RESULTS .................................................................................................. 97 DISCUSSION ............................................................................................ 99 ACKNOWLEDGMENTS .......................................................................... 1 03 REFERENCES ........................................................................................ 104 CHAPTER 5. TOXICITY IDENTIFICATION AND EVALUATION OF ESTROGENIC AND DIOXIN-LIKE COMPOUNDS IN WASTEWATER EFFLUENTS ...................................................................................................... 118 ABSTRACT ............................................ .. ................................................ 118 INTRODUCTION ..................................................................................... 119 MATERIALS AND METHODS ................................................................ 122 Sample collection and fractionation .............................................. 122 viii Cell culture and bioassay ............................................................. 122 RESULTS AND DISCUSSION ................................................................ 126 Dioxin-Iike activity .............................. . .......................................... 126 Estrogen-like activity .................................................................... 127 SUMMARY .............................................................................................. 1 32 REFERENCES ........................................................................................ 133 CHAPTER 6. BIOACTIVE ORGANIC COMPOUNDS IN THE WATERS OF LAKE MEAD, NEVADA, USA ............................................................................ 154 ABSTRACT ............................................................................................. 154 INTRODUCTION ..................................................................................... 155 EXPERIMENTAL SECTION ................................................................... 158 Standards and reagents ............................................................... 158 Sample collection ........................................................................ 159 Extraction .................................... , ................................................ 160 Silica gel fractionation ............................................... . .................. 162 Identification and quantitation ....................................................... 163 Recovery and precision ................................................................ 165 Detection Limits ............................................................................ 165 RESULTS AND DISCUSSION ................................................................ 166 Analytical considerations .............................................................. 166 Organochlon'ne pesticides and PCBs ........................................... 167 Polycyclic aromatic hydrocarbons ................................................ 168 Alkylphenols and EPA .................................................................. 169 Pharmaceuticals .......................... . ................................................ 1 70 Other compounds ......................................................................... 1 71 High-resolution mass spectrometry .............................................. 172 SUMMARY .............................................................................................. 172 ACKNOWLEDGMENTS .......................................................................... 172 REFERENCES ........................................................................................ 173 LIST OF TABLES CHAPTER 1 Table 1. End points used to assess reproductive impacts of EDCs in fish ............................................................................................................. 37 CHAPTER 2 Table 1. Detection Limits for Compounds of Interest ............................... 65 Table 2. Bioconcentration of NP in Fathead Minnows (mean concentrations) .......................................................................................... 66 CHAPTER 3 Table 1. Ions Monitored and Recovery Data ........................................... 80 Table 2. Detection Limits for Compounds of Interest ............................... 81 Table 3. Average Concentrations of NP and NPEs in Lake Mead Carp..82 CHAPTER 4 Table 1. Recoveries through the analytical procedure (n=3) (% Recovered i SD) ..................................................................................... 108 Table 2. Dection Limits for Selected Xenoestrogens ............................. 109 Table 3. Concentrations of Selected Xenoestrogens in the Trenton Channel ................................................................................................... 110 Table 4. Concentrations of Selected Xenoestrogens at WWTPs ................................................................................................... 111 Table 5. Concentrations of Selected Xenoestrogens in Lake Mead ...... 112 CHAPTER 5 Table 1. Extract Concentrations ............................................................ 138 Table 2. E2-EQs and Predicted Responses .......................................... 139 CHAPTER 6 Table 1. F1 and F2 Compounds Detected, Estimated Method Detection Limits (nglL), and Ions Monitored ............................................................ 176 Table 2. Pharmaceuticals Compounds (F3) Detected, Estimated Method Detection Limits (nglL), and Ions Monitored ........................................... 177 Table 3. F3 Compounds Detected, Estimated Method Detection Limits (nglL), and Ions Monitored — Part I ......................................................... 178 Table 4. F3 Compounds Detected, Estimated Method Detection Limits (nglL), and Ions Monitored — Part II ........................................................ 179 Table 5. Concentrations of HCHs in Lake Mead (ng/L) ......................... 180 Table 6. Concentrations of DDTs Detected in Lake Mead (nglL) .......... 181 Table 7. Concentrations of Alkylphenols and Bisphenol A in Lake Mead (ng/L) ....................................................................................................... 182 Table 8. Concentrations of Pharmaceuticals in Lake Mead (ng/L), Part I ........................................................................................................ 183 Table 9. Concentrations of Pharmaceuticals in Lake Mead (ng/L), Part II ....................................................................................................... 184 Table 10. Concentrations of Pharmaceuticals in Lake Mead (ng/L), Part III ...................................................................................................... 185 Table 11. Concentrations of Other Compounds in Lake Mead (ng/L)...186 Table 12. Compounds Identified in Las Vegas Bay by HRMS .............. 187 xi LIST OF FIGURES CHAPTER 1 Figure 1. Mechanism for activation of the estrogen receptor (ER) in MVLN cells ................................................................................................ 49 CHAPTER 2 Figure 1. Structures of target compounds ............................................... 67 Figure 2. Chromatograms of target compounds in fish tissues: (A) standards (B) fish tissue extract from 3.4 pg NP/L exposure (C) fish tissue extract from 0.4 pg NP/L exposure ................................................. 68 CHAPTER 3 Figure 1. Structures of compounds of interest ........................................ 83 CHAPTER 4 Figure 1. Structures of target compounds ............................................. 113 Figure 2. Flow diagram of analytical method ......................................... 114 Figure 3. Reverse-phase HPLC of F2: (A) spiked standards; (B) Trenton Channel - Black Lagoon 8/30/97; (C) WWI'P - BV effluent 10l8l97 ....... 115 Figure 4. Reverse-phase HPLC of F3: (A) spiked standards; (B) Trenton- Channel — Black Lagoon 8/30/97; (C) WWTP — BV effluent 10l8l97 ...... 116 Figure 5. Normal-phase HPLC of NPE oligomer distribution: (A) NPE standard; (B) extracted NPE ................................................................... 117 CHAPTER 5 Figure 1. Flow diagram of analytical method ......................................... 140 Figure 2. Reverse-phase HPLC fine fractionation ................................. 141 Figure 3. Dioxin-Iike bioactivity (various sites): Significance bars represent 3x SD of the background sign ................................................. 142 Figure 4. Estrogen-like bioactivity — Michigan WWTPs (October 1997): Significance bars represent 3x SD of the background signal .................. 143 xii Figure 5. Estrogen-like bioactivity — Trenton Channel (August 1997): Significance bars represent 3x SD of the background signal .................. 144 Figure 6. Estrogen-like bioactivity - Lake Mead (April 1997): Significance bars represent 3x SD of the background signal ...................................... 145 Figure 7. Estrogen-like bioactivity — Lake Mead (September 1997): Significance bars represent 3x SD of the background signal .................. 146 Figure 8. Fine fractionation and corresponding estrogen-like bioactivity of WWTP BV-effluent F2 extract ................................................................. 147 Figure 9. Fine fractionation and corresponding estrogen-like bioactivity of Black Lagoon, Trenton Channel, F2 extract ............................................ 148 Figure 10. Fine fractionation and corresponding estrogen-like bioactivity of LV Wash, Lake Mead (April 1997), F3 extract .................................... 149 Figure 11. Fine fractionation and corresponding estrogen-like bioactivity of LV Bay, Lake Mead (April 1997), F3 extract ....................................... 150 Figure 12. Further fractionation and corresponding estrogen-like bioactivity of LV Wash, Lake Mead (April 1997), F3 extract ................... 151 Figure 13. Fine fractionation and corresponding estrogen-like bioactivity of WWTP BV-effluent, F3 extract ............................................................ 152 Figure 14. Fine fractionation and corresponding estrogen-like bioactivity of Black Lagoon, Trenton Channel, F3 extract ........................................ 153 CHAPTER 6 Figure 1. Map of the Boulder Basin study area of Lake Mead, Nevada .................................................................................................... 188 Figure 2. Flow diagram of analytical method ........................ . ................. 189 Figure 3. Mass spectra of phenytoin (Dilantin): (A) authentic standard; (B). LV Bay water extract ........................................................................ 190 Figure 4. Mass spectral scan of phenobarbital: (A) authentic standard; (B) LV Bay water extract ........................ . ................................................ 191 Figure 5. Mass spectral SIM Chromatograms I: (A) authentic standards; (B) LV Bay water extract ......................................................................... 192 xiii Figure 6. Mass spectral SIM Chromatograms II: (A) standards; (B) LV Bay water extract. Peak # 1. DEET; 2. Guafenesin; 3. Acetaminophen; 4. Meprobamate; 5. Caffeine; 6. Oxybenzone; 7. Bisphenol A; 8. A8 THC; 9. A9 THC; 10. Primidone; 11. Carbamazepine; 12. Hydrocodone ............. 193 Figure 7. Mass spectral scan of DEET: (A) authentic standard; (B) LV Bay water extract .................................................................................... 194 Figure 8. Mass spectral scan of tris(1 ,3-dichIoroisopropyl)phosphate: (A) NIST database spectra; (B) LV Bay water extract .................................. 195 xiv INTRODUCTION The threat of modulation of animal endocrine systems by xenobiotic compounds has become a major issue facing society today. These compounds are varied in form and mechanism of action. This poses unique challenges in the identification and evaluation of these compounds from environmental matrixes. The aquatic environment is exceptionally susceptible to xenobiotic insult. Water can be polluted by a multitude of sources and acts as a sink for many types of pollution. This chapter outlines several methods for the detection and quantitation of endocrine disrupting compounds (EDCs) in the aquatic environment. However, no single method alone can predict or detect all EDCs present in an environmental sample, nor can all the biological mechanisms of action be accounted for in one simple test. Therefore, comprehensive screening for EDCs must combine several types of analyses including in vivo and in vitro bioassays, and analytical chemistry. This dissertation will discuss several of these types of analyses and demonstrate advantages and disadvantages of each. Each chapter of this dissertation is in journal format and publication status is provided. Chapter one of this dissertation is an overview of the research conducted at the Aquatic Toxicology Laboratory (ATL) at Michigan State University (MSU) related to endocrine disruption in the aqautic environment. The role of various types of in vitro and in vivo bioassays for the detection of enodcrine disrupting chemicals will be discussed. Specific examples are provided as well as advantages and disadvantages of each type of analysis. A discussion of butyltins is provided as an example of a unique type of endocrine disrupting chemical (EDC) which has been shown to cause adverse effects in the environment. The final section in this chapter deals with toxicity identification and evaluation of EDCs. Here we integrate results from in vitro bioassays with instrumental analyses. By using bioassay-directed fractionation and subsequent instrumental analyses, we are able to determine the compounds of greatest potency from complex mixtures. In conclusion, we discuss the need to use an integrated approach combining in vitro and in vivo bioassays with advanced instrumental techniques to determine what compounds in the aquatic environment may have the potential to impact the endocrine systems of aquatic organisms. Chapter two discusses a controlled laboratory study to determine the bioconcentration of p-nonylphenol (NP) in fathead minnows. NP is one of several transient intermediate degradation products of the widely used nonylphenol polyethoxylate (NPE) nonionic surfactants. NP is a ubiquitous contaminant of wastewater treatment plant (WWT P) effluents. In both in vitro and in vivo bioassays, NP has been shown to be estrogenic. NP is fairly lipophilic and persistent, therefore, it was desirable to determine bioconcentrations factors (BCFs) in an aquatic organism. Methods for determining NP concentrations in water and tissue were developed. BCFs ranging from 203 to 268 were determined. Chapter three expands upon the methodology developed in chapter two. A monitoring study for NP in the tissues of fish was desired. However, no reliable and sensitive methods for the identification and quantitation of NP in tissue samples were available. The methodology presented in chapter two did not provide the necessary sensitivity for a monitoring study. Several methods were investigated. Also, the methodology was to be readily transferable to other laboratories by using common and/or inexpensive equipment. Extract steam- distillation followed by gas chromatography with mass selective detection (GC/MS) was developed. This method provided method detection limits (MDLs) ranging from 2 — 5 ng/g, wet weight, for NP and nonylphenol mono— and di- ethoxylate. Themethod was also highly reproducible with coefficients of varation (CVs) generally less than 10% among subsamples. The method was tested using carp captured in the Las Vegas Bay of Lake Mead, Nevada. Average tissue concentrations of NP and nonylphenol monoethoxylate (NPE1) were determined to be 184 and 242 ng/g, respectively. The fourth chapter presents a novel method to determine concentrations of certain estrogenic compounds in aqueous mixtures. It provides the first report in North America of natural and sythetic estrogens in surface waters. Octylphenol (OP), NP, and NPE were also identified and quantified. These compounds were extracted in situ from 5 L water samples using solid-phase extraction (SPE) disks. The resulting extracts were fractionated based on polarity. Various instrumental techniques were used to identify and quantify compounds of interest. The study locations include four WWl'Ps in central Michigan; the Trenton Channel of the Detront River, Michigan; and Lake Mead, Nevada. Concentrations of NP and OP ranged from less than the MDL to 37 and 0.7 pg/L, respectively. NPE concentrations ranged from less than the MDL to 332 pg/L. Concentrations of 17B-estradiol (endogenous estrogen) and ethynylestradiol (synthetic estrogen) ranged from less than the MDLs to 3.7 ng/L and 0.8 ng/L, respectively. The fifth chapter describes a toxicity identification and evaluation (TIE) method for estrogen- and dioxin-like compounds in complex aqueous mixtures. Extracts from the method presented in chapter three were tested using in vitro bioassays. Whole extracts and corresponding fractions were tested for estrogen- and dioxin-like activity using MVLN and H4IIE-Iuc in vitro bioassays, respectively. No significant dioxin-like activity was observed. Estrogenic activity was quantitated by comparing the magnitude of induction elicited by the extract or fraction to the maximum induction caused by the endogenous estrogen, 17B- estradiol (E2). Several extracts and fractions displayed significant estrogenic activity. In each case, the most polar fraction (F3) was associated with the greatest estrogenicity. Instrumental analyses and further fractionation were used to identify compounds associated with estrogenic responses. Mass balance calculations suggested that E2 and the synthetic estrogen, ethynylestradiol (EE2), were the most likely cause of the responses observed. Alkylphenols (APs) did not appear to contribute significantly to the observed estrogenicity. The final chapter of this dissertation (chapter 6) expands upon the methods developed in the previous chapters to determine the concentrations of several classes of bioactive compounds in the waters of Lake Mead, Nevada. Several recent reports have indicated a flux of organic compounds flowing into the Boulder Basin of Lake Mead, Nevada from the Las Vegas Wash. The primary objective of this study was to screen for a wide range of organic contaminants in Lake Mead. Mass spectrometry was utilized to screen for previously unreported compounds. Samples of 100 L of filtered water were extracted using large-scale SPE. Following a series of elution and extraction procedures, resulting extracts were fractionated into 3 polarity classes. Each fraction was analyzed by gas chromatography with mass selective detection (GC/MS). DDT, DDE, and DDD (collectively, DDTs) and four isomers of hexachlorocyclohexane (HCH) were detected at some sites, with concentrations ranging from less than detection to 28 nglL. Octylphenol, nonylphenol, nonylphenol monoethoxylate, and nonylphenol diethoxylate were detected in some samples, with concentrations ranging from less than detection to 1500 nglL. Caffeine, nicotine, oxybenzone, N,N-diethyl-meta-toluamide (DEET), phosphate-based flame-retardants, and several pharmaceuticals were detected in some samples with concentrations ranging from less than detection to 360 nglL. The identities of several of these previously unreported compounds were verified using high-resolution mass spectrometry. PAHs were detected at each site and did not vary significantly among the sites in concentration or in pattern. Total PAH concentrations ranged from 5 to 78 nglL. Polychlorinated biphenyls, triazine herbicides, and steroids were not detected in any of the samples. Chapter 1 OVERVIEW OF INSTRUMENTAL AND BIOANALYTICAL MEASURES OF ENDOCRINE DISRUPTERS IN WATER (Published as a chapter in the American Chemical Society Book: “Analysis of Environmental Endocrine Disruptors” 2000) A number of compounds released into the environment by human activities can modulate endogenous hormone activities and have been termed endocrine-disrupting compounds (EDCs) (1, 2). Environmental EDCs have been defined as exogenous agents which interfere with the "synthesis, secretion, transport, binding, action, or elimination of natural hormones in the body that are responsible for the maintenance of homeostasis, reproduction, development, and/or behavior" (3). It has been hypothesized that such compounds may elicit a variety of adverse effects in both humans and wildlife, including promotion of hormone-dependent cancers, reproductive tract disorders, and reduction in reproductive fitness (4-10). Endocrine "disruption" by environmental contaminants has become a cause for concern (11) and as a result there is an urgent need to develop methods for monitoring EDCs in the environment. This need is underscored by recent legislation mandating that chemicals and formulations be screened for potential estrogenic activity before they are manufactured or used in certain processes (Safe Drinking Water Act Amendments of 1995 - Bill Number 8.1316; Food Quality Protection Act of 1996 - Bill Number PL. 104-170). EDCs act through a number of mechanisms including receptor-mediated hormonal responses such as hormone mimics (1). These include xenobiotic effects through thyroid hormone receptor, epidermal growth factor (EGF) receptor, aryl hydrocarbon receptor (AhR), estrogen receptor (ER) and androgen receptor (AR) (12, 13). Of all the EDCs, those that are direct-acting estrogen receptor (ER) agonists (xenoestrogens or estrogen mimics), direct acting ER antagonists (antiestrogens), or androgen antagonists have received the greatest attention, due in part to their importance in embryonic development. The “estrogen hypothesis” stemmed from the observation that some of the effects observed in oviparous wildlife exposed to persistent and bioaccumulative chemicals were similar to those that could be caused by injecting estrogen into eggs (2). This hypothesis was supported by the fact that naturally occurring exoestrogens, such as phytoestrogens, could cause reproductive dysfunction in animals. Further support came from the observation that some xenobiotics that can bind the ER are weak estrogen agonists or antagonists in in vitro expression assays (12). Because the major initial concern over endocrine modulating compounds has been the estrogen mimics (xenoestrogens), we will restrict our discussion to “estrogenic” and “antiestrogenic” compounds, but the techniques presented can be applied to any receptor-mediated process for which a suitable in vitro cell system can be developed (12). There are a wide variety of ER-active compounds in the environment (14). These include both natural products (11) and synthetic compounds (10). The synthetic compounds include both chlorinated and non-chlorinated compounds that can either mimic or antagonize the effects of endogenous estrogen (12). Some of the compounds found in the surface waters that have been reported to be estrogenic include nonylphenol (NP), octylphenol (OP), 17B-estradiol (E2) (produced in the body) and the synthetic estrogen ethynylestradiol (EE2) (used in oral birth control medication) (15). Several types of in vitro assays are available for measuring the estrogenic activity of single compounds or complex mixtures. The range of responses includes everything from simple receptor binding, to expression of endogenous or exogenous genes, to cell proliferation and differentiation. In vitro systems are attractive as screening tools because they are rapid, inexpensive, and fairly reproducible. For these reasons, precise estimates of the relative potency of a great many samples or compounds can be obtained in a rather short period of time. Expression assays examine induction or suppression of proteins encoded by genes whose transcription is thought to be modulated through an ER- mediated mechanism. Increases or decreases in the activity of the protein of interest upon exposure to a single compound or complex mixture, such as an environmental extract, suggest the presence of one or more ligands with the potential to modulate a broad range of genomically controlled estrogenic responses. Although in vitro assays are an attractive option for screening and mechanistic studies, they may miss effects that would take place only in whole organisms. Some toxicants require metabolic activation through one or more pathways that occur in vivo, but not in vitro. Homeostatic controls and bioaccumulation generally are not simulated by in vitro testing systems. Toxicant effects can occur by different mechanisms in multiple tissues simultaneously. In addition, the same chemical exposure can result in very different responses in an animal depending upon its life stage, sex, and reproductive state. Fish are useful in vivo models for testing for effects of endocrine disrupting chemicals in water. Some species are extremely sensitive to such effects, and fish can act as integrators of responses to mixtures of toxicants that occur in the environment. In addition, effects related to growth and reproduction in fish are more easily related to population-level and ecological effects than are effects in in vitro systems. Fish can also be used in conjunction with in vitro testing systems to investigate mechanisms of toxicant action and to calibrate in vitro to in vivo responses. The aquatic environment is especially susceptible to the effects of contaminants. Effluents from municipal and industrial wastewater treatment facilities, storm and agricultural run-off, and boating add many exogenous compounds to the aquatic ecosystem. Of the most frequently studied EDCs, butyltin compounds are next in abundance to alkylphenols in wastewater effluents. These compounds are also present in surface waters from many locations. The impact of tributyltin on the marine environment has received considerable attention during recent years because of its high toxicity to aquatic organisms (16-20). The use of TBT as an antifouling agent in paints applied on ships results in direct contact with aquatic environments. However, studies on the contamination of surface water (lakes and rivers) by butyltin compounds originating from the disposal of municipal wastewater are limited. In the aquatic environment, instrumental analytical techniques are used to identify and quantify EDCs in sediments, tissues, and water. In some cases the toxicant is already known. In other cases, screening can be used to identify new toxicants based on bioassay-directed fractionation coupled with subsequent instrumental analyses. This is part of a toxicity identification and evaluation (TIE). By using bioassay-directed fractionation, compounds with certain mechanisms of action can be isolated and identified. A mass-balance can then be performed to see if all observed bioactivity can be accounted for by the compounds identified (21). This comprehensive approach allows for screening of various types of EDCs and may identify yet unknown compounds with significant bioactivity. The issue of environmental endocrine disruption is extremely complex. No single method will identify or explain all types of EDCs in the aquatic environment. Integrated approaches that combine in vitro and in vivo bioassays with analytical chemistry techniques are needed to screen for and assess the effects of EDCs in the environment. This introduction should serve as an overview of work completed at the Aquatic Toxicology Laboratory (ATL) at Michigan State University relative to instrumental and bioanalytical techniques used for the determination of endocrine disrupters in water. 10 IN VITRO BIOASSAYS In vitro bioassays have an important role to play in an integrated approach to the study of endocrine disrupting compounds in the environment (12). In vivo studies in both the field and laboratory are critical for linking exposure to biologically relevant effects. They are, however, impractical for routine, high throughput screening, and characterization of individual compounds or environmental samples. Procedurally, in vitro bioassays are typically performed more quickly and at significantly less cost than in vivo studies. Use of simplified biological systems circumvents much of the inter-individual, seasonal, and temporal variability, which can confound interpretation of in vivo responses. Additionally, in vitro bioassays avoid many of the complex socio—political and ethical issues associated with whole animal studies. Thus, in vitro bioassays are more amenable systems for routine use than in vivo assays. In vitro bioassays use simplified biological or biochemical systems to measure responses elicited by exposure to individual compounds or environmental samples. The responses measured are generally highly sensitive, integrated, and mechanistically-based. These properties allow in vitro bioassays to overcome many of the limitations of in vivo studies and instrumental analyses. While instrumental analyses are essential for the identification and quantitation of compounds, they provide no information regarding the efficacy and potency of these compounds to modulate endocrine function. In vitro bioassays measure mechanistically-based biological responses. In vitro 11 bioassays provide an integrated measure of the combined potency of all compounds in a sample. This is a practical advantage, because instrumental analysis of complex mixtures can be very expensive, difficult, and time consuming. Additionally, in vitro bioassays can detect compounds for which there are no analytical methods available and can be useful tools to measure potential additive and non-additive interactions between compounds. When the biological response being measured is highly sensitive, in vitro bioassays can also account for compounds that can exert a biological effect at concentrations less than analytical detection limits. 17B-estradiol and ethynylestradiol, for instance, can elicit significant responses in MVLN cells at concentrations less than 50 pM (a 15 ppt) (12), while sensitive instrumental analyses, such as HPLC fluorescence which has a detection limit of approximately 100 ng/mL (15) (z 350 nM) for these compounds. As a result, in vitro bioassays provide information which can complement instrumental analyses. In vitm bioassays have been used widely for the study of potentially estrogenic compounds (12). Competitive receptor binding assays (12, 22-25), cell proliferation assays (26-30) and in vitro gene expression assays (12, 24, 29, 31-35) are among the most common in vitro approaches employed in the study of estrogenic and other endocrine disrupting compounds. The mechanistic foundation, advantages, and disadvantages of these approaches have been described elsewhere (12, 22, 36). Work in our laboratory has primarily involved the use of the MVLN (MCF- 7-Iuc) in vitro gene expression assay. MVLN cells are human breast carcinoma 12 cells stably transfected with a Iuciferase reporter gene under control of estrogen responsive elements (EREs) of the Xenopus vitellogenin A2 gene (37, 38). When cells are exposed to a single compound or environmental sample, ER ligands can enter cells and bind the ER (Fig. 1). In MVLN cells, binding also upregulates expression of an exogenous Iuciferase reporter gene (Fig. 1). Upon addition of the appropriate substrate, Iuciferase catalyzes a light producing reaction. As a result, luciferase activity can be measured conveniently and with great sensitivity using a 96 well plate-reading Iuminometer. The measured Iuciferase activity is proportional to the sample’s ability to modulate ERE- mediated gene expression. Although gene expression in MVLN and other estrogen responsive cells is generally regarded to be ligand induced, there are also ligand-independent pathways that can modulate the transcriptional activity of the ER through activation of protein kinases (12). For example, treatment of cells with growth factors or agents that activate protein kinases in general, can cause the ER to be activated (without ligand binding) to bind to EREs and activate transcription. Complex interactions within mixtures are also possible. Investigations have included mixtures of estrogenic and anti-estrogenic compounds (39) and mixtures of weak estrogens (12). Use of the MVLN assay by our laboratory provides examples of useful applications of in vitro bioassays as part of an integrated approach to the study of endocrine disrupters. Screening with the MVLN bioassay has identified both individual compounds and complex mixtures extracted from environmental 13 samples that were able to induce ERE-mediated gene expression (12, 15, 40- 42). Active samples have been compared in terms of either their relative efficacy (the magnitude of response elicited; (15, 40, 41)) or relative potency (the mass, volume, or concentration of sample or compound needed to elicit some defined level of response; 1). Environmental samples from a diverse array of sources including surface waters (15, 43), wastewaters (15), sediments (40), and commercial fish diets (42) have been evaluated successfully. MVLN—based screening and relative potency or efficacy comparison has been used to focus analytical efforts on samples that show the greatest potential to exert estrogenic effects, identify field sites that warrant more extensive study, and profile the distribution of potentially estrogenic agents in relation to point sources, land use, and wastewater treatment processes. In cases where chemical fractionation has been coupled with MVLN screening, the results have facilitated the formulation and testing of hypotheses regarding which specific compounds or groups of compounds in an environmental sample which may be responsible for the in vitro responses observed (40). MVLN responses to individual compounds have been used to characterize the relative “estrogenic" potencies of compounds (12). Comparison of responses elicited by a compound in several in vitro bioassays based on different mechanistic assumptions can provide lnfonnation regarding that compound’s mechanism of action. Thus, in vitro bioassays can provide a great deal of useful information to aid in the assessment of endocrine disrupting compounds. 14 As with other approaches, in vitro bioassay responses must be interpreted with a clear understanding of their limitations. They can detect and provide an integrated measure of the ability of a compound or sample to elicit a mechanism- specific biological response. The relevance of that response for modeling or predicting effects in vivo is dependent on the relevance of the mechanistic principles and assumptions upon which the assay is based. The MVLN assay, for example, would not detect compounds that exert estrogenic responses in vivo by acting as an anti-androgen or by altering endogenous estradiol levels through effects on the hypothalamus or pituitary. Thus, unless a general and fairly universal mechanism of action for a specific type of in vivo effect can be defined, in vitro bioassays provide a rather narrow field of view for detecting compounds which may alter endocrine function on a whole-organism level. Furthermore, because they use simplified systems, in vitro assays fail to account for a number of important factors which may modulate responses via a given mechanism of action in vivo. Such factors include toxicokinetics, cross-talk between biological pathways, and environmental factors (12, 22). In order to be useful for predicting effects at the whole-animal or population level, in vitro bioassays must be rigorously calibrated to in vivo responses (44, 45). The greatest limitation of in vitro bioassays for either screening or monitoring is the need for a priori knowledge about the mechanisms of action of a compound or mixture for which the assay is designed to screen. Thus, although in vitro bioassays have a role to play in the evaluation of endocrine disrupting compounds, they are just one of the tools needed to address the issue. 15 IN VIVO VERSUS IN VITRO TESTING FOR EDCs Despite public pressure to use in vitro testing whenever possible to spare animal lives and minimize the cost and time involved, whole animal studies are crucial for the development of biologically relevant methods to screen for and characterize EDCs in water. In vitro bioassays are mechanism specific and cannot account for the broad spectrum of potential mechanisms of effects that may be relevant in vivo (22). Whole animals can integrate and account for effects of chemicals in a mixture that may act through different mechanisms, at different tissues, either directly or indirectly to modulate overall endocrine function as well as specific endpoints. Additionally, effects of some compounds may be affected by metabolic transformation. Such transformations are generally not accounted for by in vitro bioassays (22). Furthermore, whole animal studies account for bioaccumulation and homeostatic controls that affect the potency or efficacy of compounds in vivo. Although not necessarily amenable for routine monitoring and screening, in vivo studies are critical for relating exposure to population level-effects and ecologically relevant responses as well as the development and validation of relevant in vitro screening assays and biomarkers. 16 USE OF FISH TO TEST FOR EDCs. Fish have long been used as biosentinel models of water quality, particularly because of the concerns associated with the increased production, use, and disposal of chemicals in the environment (13, 46). Among these chemicals are the EDCs, which are thought to interfere (directly or indirectly) with the processes governing reproduction, behavior, and other aspects used to evaluate population health. Many factors must be taken into account when using fish to test for effects of EDCs. It is well known that environmental factors play an important role in fish reproduction. Temperature, photoperiod, diet, availability of spawning substrates, proximity and reproductive readiness of potential mates (47), and water quality all affect reproductive status, ovulation, and spawning in fish (48). Stress, including that from capture, handling, and confinement, can have a rapid and marked effect on plasma sex hormone levels (49, 50). Seasonal and dailly changes in sex hormone levels must be considered in sampling schemes (49). If fish are fed during a study, diet must be selected carefully since some animal diets can contain hormonally active components or contaminants (30, 51, 52). Fish exhibit a variety of reproductive strategies. Some are ovovivlparous and some are oviparous. Some, like the fathead minnow (Pimephales promelas), demonstrate parental care of eggs or offspring, while others, like the goldfish (Carassius auratus), ignore or even consume their eggs. Some spawn only once per breeding season, while others spawn repeatedly. This last point is 17 particularly important to consider, since hormone levels and other end points can change rapidly and dramatically around the time of spawning. SELECTION OF TEST SPECIES Different species of fish have been used to test for effects of EDCs, and each has its advantages and disadvantages. Desirable species should be easy to culture and handle in the laboratory, readily available year-round, widespread, and economically or aesthetically important. It is also important to choose a species for which there is a great deal of knowledge about its reproductive biology. It is difficult to determine whether an EDC has caused a departure from the normal condition if the normal condition is not known. Easily recognized sexual dimorphism is important for sexing fish, and in some cases, reduction of prominent secondary sex characteristics is used as evidence of endocrine disruption (44, 45). Some species that are currently used include trout and salmon, particularly rainbow trout (Oncorhynchus mykiss), carp (Cyprinus carpio), goldfish, white sucker (Catostomus commersonr), Iargemouth bass (Micmpterus salmoides), fathead minnow, Japanese medaka (Oryzias Iatipes), mummichog (Fundulus heteroclitus), and mosquitofish (Gambusia affinis). 18 END POINTS AND BIOMARKERS USED TO ASSESS EFFECTS OF EDCs Fish have been used in both laboratory and field studies to test individual chemicals or mixtures, effluents, and natural waters impacted by human activity for potential to alter reproductive structure and function. Some end points used to assess the effects of EDCs on fish and some studies that employed these end points are listed (Table 1). This table does not list all end points that could be used or all of the studies in which they have been used. For a review of methods to assess effects of EDCs on fish, see (53). Plasma vitellogenin (Vtg) induction is a popular marker of exposure of fish to chemicals that bind to the estrogen receptor. Vtg is an egg-yolk precursor synthesized in the liver of oviparous animals in response to estrogen (54). Vtg is released from the liver to the bloodstream where it is carried to the ovary and sequestered into developing oocytes. Vtg usually is not detected in significant amounts in the plasma of male or immature fish, but they can be stimulated by exposure to natural or synthetic estrogenic chemicals to produce Vtg (55). Thus, Vtg is a useful tool to detect estrogenic activity of test chemicals or environmental waters potentially impacted by estrogenic EDCs. However, it is important to keep in mind that Vtg induction has not been shown to have any adverse effects in fish and is a functional measure of exposure useful only for monitoring for estrogenic compounds (55). 19 FIELD STUDIES Other researchers in the United States, the United Kingdom, and Canada have conducted field studies to determine whether EDCs in the environment have the ability to alter the reproductive structure and function of feral or wild (44, 56—60) and caged fish (61-63). Representative examples of these studies are those conducted with adult fish at the Aquatic Toxicology Laboratory at Michigan State University. In situ exposure of caged fathead minnows to municipal wastewater treatment plant (VVWT P) effluents was conducted. Fathead minnows were caged in rivers directly in the outfalls of municipal WWI'Ps in mid-Michigan (64). Plasma Vtg, E2, testosterone (T), gonad histology, and male secondary sex characteristics were examined. Adult male and female minnows were caged for 3 weeks. No changes in any of these end points could be attributed to EDCs in WWTP effluents. In situ exposure of caged common goldfish to municipal waste water treatment plant effluents was conducted. This study was similar to the fathead minnow study previously described. Adult male and female goldfish were caged for 6 weeks at the same sites with the fathead minnows (65). Plasma Vtg, E2, and T were measured by enzyme immunoassay. Gonad histology was examined, and gonad tissue was removed and incubated in vitro to evaluate gonadal steroidogenic activity. Interpretation of the data is in progress. 20 LABORATORY STUDIES The Aquatic Toxicology Laboratory at Michigan State University has conducted both field and controlled laboratory studies. All of the laboratory studies were conducted with adult fathead minnows exposed to test compounds in a proportional flow-through diluter system. Fathead minnows were exposed for 19 days to water borne E2 at measured concentrations ranging from 3.5 to 15 ng/L in a proportional flow- through diluter (66). Fish were exposed in duplicate groups of six, with three males and three females per exposure tank. Interestingly, E2 was detected even in control and solvent control tanks, indicating that the fish were a source for some of the estrogen in the exposure tanks. Egg production, plasma Vtg, plasma E2, and gonad histology were examined. The E050 (concentration expected to cause 50% effect) for inhibition of egg production was 120 nglL. and the E050 for Vtg induction in males was 251 ng/L. Vtg was measured indirectly as alkaline labile phosphorus. In male fish, plasma E2 increased in a dose-dependent manner with concentrations of water borne E2. Plasma E2 levels in females were not affected by water borne E2 exposure. This difference in response may be due to sex-related differences in homeostatic control or metabolism of estrogen. Histopathological changes of the gonad were noted in a companion study (45) (see next section). The fathead minnow was used as a test subject in part because of its prominent sexual dimorphism. Males have a fatpad (for which the fish is named) on the dorsal surface of the head. Males also develop nuptial tubercles, rows of 21 small, hardened projections on the rostrum. The fatpad and nuptial tubercles are easily seen with the naked eye, and are absent in females. The female is generally lighter in color and smaller than the male, which usually develops a darker color pattern when sexually mature. In addition, the female has a more tapered head shape. Sexually mature fathead minnows were exposed to water borne E2 as described in the previous section (66). In males, E2 exposure caused atrophy of secondary sex characteristics and histologic lesions of the testes. Lesions of the testes included proliferation of Sertoli cells, degenerative spermatozoa, and enlarged phagolysosomes containing necrotic spermatozoa and other cellular debris. Ovaries of females exposed to E2 contained more primary follicles and fewer secondary and Graafian follicles than ovaries of control fish (45), indicating that ovarian development in preparation for spawning had been retarded. Adult fathead minnows were exposed to water borne 4-nonylphenol (NP) in a proportional flow-through diluter at concentrations ranging from <0.01 to 3.4 [lg/I. for 42 days (67). The study was conducted first early in the natural breeding season (Experiment I, or EX l) and again later in the breeding season (Experiment II, or EX ll). Measured exposure concentrations for the two experiments were similar (EX I: 0.05, 0.16, 0.4, 1.1, and 3.4 pg NPIL ; EX II: <0.01, 0.10, 0.33, 0.93, and 2.4 pg NPIL ). Examination of dose-response relationships between NP and egg production per female and between NP and plasma E2 in females revealed curves indicative of "Inverted-U" type responses, with lesser doses resulting in an increased response and greater doses resulting 22 in lesser responses. For males and females in EX l, plasma E2 levels were greater than controls for all of the NP treatment groups except the greatest (3.4 pg NPIL), which was not different from the controls. Plasma E2 levels for both sexes ranged roughly from 2 to 25 ng/mL. For females in EX ll, two intermediate exposure concentrations (0.1 and 0.33 pg NP/L) produced plasma E2 concentrations statistically greater than those in both controls groups. The plasma E2 concentrations for females ranged from about 2 to 8 ng/mL. In EX II males, none of the treatment levels produced plasma E2 levels statistically greater than both controls, but the sample sizes were small, and the shape of the curve indicated a trend similar to that seen in the females. For females in EX l, the dose-response curve for NP versus plasma Vtg suggests that NP caused a decrease in plasma Vtg levels, with the two greatest NP concentrations resulting in the least plasma Vtg levels. For females in EX II, the dose-response curve for NP versus plasma Vtg suggests an Inverted-U response, with the least NP exposure level producing a statistically significant increase over the controls, and greater exposure levels producing statistically lesser Vtg levels that are still significantly greater than the control Vtg levels. For males in both EX | and EX II, there was no significant increase in plasma Vtg in any of the NP exposure concentrations. In EX I, NP caused no statistically significant effect on egg production per female. However, the sample sizes used were small, and the shape of the dose-response curve suggests the same inverted-U type response, with increased egg production at the least treatment level (0.05 pg NPIL) and a decrease in greater treatment levels. In EX ll, there was an increase in egg 23 production per female for the greatest treatment level (2.4 pg NPIL) and for one of the intermediate treatment levels (0.01 pg NPIL), with two treatment levels between exhibiting no difference from the controls. The mechanism of NP action in the alteration of fathead minnow reproductive end points is unclear, but may be related to positive and negative feedback on endogenous estrogen production. The differences in response over the two different experiments may have resulted from differences in the reproductive state of the fish. This underscores the importance of timing of the study relative to the reproductive state in fish. Furthermore, while NP has been reported to be a weak estrogen agonist, it is likely that the mechanism of action is not due to the binding of NP to the ER. Adult fathead minnows were exposed to an industrial mixture of NPEO for 42 days in a proportional flow-through diluter. Three males and three females were placed in each exposure tank. NPEO exposure concentrations ranged from 0 to 10 pg/L. Plasma Vtg, E2, and T were measured by enzyme immunoassay. Egg production per female (fecundity) was determined. No statistically significant concentration-dependent relationship was demonstrated between fecundity and NPEO exposure. However, the shape of the curve indicated a possible inverted- U response. NPEO was not observed to have any statistically significant effect on any of the other end points examined. The mechanisms playing a role in the disruption of endocrine, and related systems, in fish need a better understanding (68). Considering that testing of EDCs in feral populations, or even representative captive adults in laboratory 24 studies represent an economical, practical, and even technological obstacle, attention has been shifted to possible alternatives, not just of surrogate species, but also of exposure methodologies. The medaka (Oryzias Iatipes), a small fish frequently used as adults and/or early life stages (embryos and/or larvae) in aquatic toxicity studies (69-71) has proven useful as a tool in those alternative studies. The attributes that make this species attractive for in vivo experimentation have been discussed elsewhere (46); however, only until recently has micromanipulation technology started to take advantage of a more novel exposure approach: in ovo nanoinjection of chemicals into fertilized medaka eggs (72, 73). While egg microinjecting techniques have been developed in several fish (mainly salmonids) over ten years ago (74-76), few studies have been attempted in species with small eggs. Recently, Villalobos et al. (73) reported success in using this medaka assay to test the toxicity of several technical mixtures of polychlorinated napthalenes (Halowax). The authors injected 0.5 nl of Halowax 1014, Halowax 1013, or Halowax 1051 in triolein as a carrier solvent. Mortality for the injected triolein controls was below 10%, and the Halowax-induced responses suggested involvement of the arylhydrocarbon- (Ah-) receptor. The morphological effects of each of the Halowaxes was easily observed in the embryos and could be associated with structure-activity dependent effects (responses varied according to the chloronaphthalene speciation, chlorine localization within the molecule, and total mass contribution). Furthermore, the effects were consistently reproduced after repeated tests. However, these 25 compounds did not appear to have elicited a disrupting effect in the medaka endocrine system. These studies, done in collaboration with the US. Geological Survey (USGS) Columbia Environmental Research Center (Columbia, MO), demonstrate the feasibility of nanoinjection of compounds into the small eggs of medaka. MONITORING OF BUTYLTIN COMPOUNDS IN WATER Recently, widespread industrial and agricultural applications of organotin compounds, particularly butyltins, have led to increasing concerns about environmental contamination and biological effects caused by these compounds. Di- (DBT) and mono-butyltins (MBT) have been widely used as heat and light stabilizers for polyvinyl chloride (PVC) plastics and catalysts in the production of polyurethane and silicone. TBT has been used as antifouling agents, biocides, fungicides, molluscicides, insecticides and wood preservatives (77). The use of butyltin compounds in a variety of consumer products and household items (including sanitary napkins, diaper, cellophane wrap, sponges used in washing dishes, butter parchments, and gloves) (78, 79) and the leachates from PVC pipes have led to their occurrence in municipal wastewater and sewage sludge (80, 81). In addition, TBT is used as a disinfectant in waxes, polishes, sprays and laundry washes, which may allow contamination of municipal wastewater (82). Industrial discharges of TBT used as a slimicide in the paper industry and 26 for textile and lumber treatment, and in cooling water treatment are further sources of contamination. Input of DBT and TBT into surface waters from applications other than antifouling paint on vessels (i.e., use of TBT as a slimicide in the cooling water of a thermoelectric power plant in Italy) was first reported in the late 19805 (83). Concentrations of butyltins in raw wastewater in Zurich, Switzerland, on six sampling days ranged from 136 to 564, 127 to 1026 and 64 to 217 ngIL, respectively (84). Approximately 90% of each butyltin species was associated with suspended particles, and butyltins are removed at a rate of 62, 80 and 66% for MBT, DBT and TBT, respectively, by sedimentation (clarifier) in the sewage treatment plant. After secondary clarification, which usually represents the outflow from treatment plants, MBT, DBT and TBT ranged from 7 to 47 ng/L. MBT, DBT and TBT concentrations in the range of 01-097, 0.41-1.24 and 0.28- 1.51 png (dry wt) were found in sewage sludge. Similarly, MBT was found in all the 36 influent and effluent samples from VWVTPs in five Canadian cities. Concentrations of MBT were in the range of 28-31 pglL for influent and 1-22 ngL for effluent. DBT and TBT were found only infrequently at a few pglL (85). Concentrations of MBT, DBT and TBT in sewage sludge were as great as 650, 600, and 675 pg/g, dry wt, respectively (85). In Bordeaux, France, concentrations of MBT and DBT in influents from a WWTP were 18 and 12 ng/L, respectively (86), whereas those in effluent samples were 15 and 8 ng/L, respectively. TBT was not detected in these samples. Effluents from a WWTP in Mainz, Germany, contained MBT and DBT concentrations in the range of 13-28 27 and 6-96 ng/L, respectively (87). Total butyltin concentration in sewage sludge from Patna, India, was 340 ng/g, dry wt (88). Studies describing the occurrence of butyltins in wastewater effluents in the US. are not available. Nevertheless, results from the above studies suggest that butyltins are widespread contaminants in wastewater. Even in those WWI'Ps with exceptionally modern systems (84), the concentrations of TBT remaining in effluents were greater than the environmental quality standard of 2 ngIL (89). Occurrence of butyltin compounds in unnavigable areas in certain rivers (90), as well as in aquatic organisms of certain unnavigable rivers, suggest the release of these compounds from wastewater (91). ECOLOGICAL IMPACTS OF BUTYLTINS Butyltin compounds, particularly TBT has been implicated in the development of “imposex” (development of male sex characteristics in females) in about 50 species of gastropods worldwide (92). This effect was attributed to inhibition by TBT of cytochrome P450-dependent aromatase, the enzyme responsible for the conversion of testosterone to 178-estradiol (17). Because cytochrome P450 systems control the conversion of cholesterol into a variety of hormones including testosterone and estradiol, effects of TBT on this enzyme system can result in hormonal imbalance in animals. Although the concentrations of TBT in wastewater effluents were greater than those known to cause imposex in several sensitive gastropods, discharge of effluents into 28 surface waters could result in dilution. Nevertheless, aquatic animals concentrate TBT in tissues, particularly in liver or hepatopancreas (20, 93, 94). Because lipovitellin (provides protein and lipid for the developing embryo) is produced in the hepatopancreas or liver, accumulation of butyltins in these organs can interfere with vitellogenesis (95). TBT can also interact with cytochrome P450 and modulate the toxic effects of contaminants such as polychlorinated biphenyls (96). Although MBT and DBT were not shown to induce imposex in gastropods, they have been reported to be toxic to rainbow trout yolk sac fry (97). No observed effect concentrations (NOEC) for DBT and TBT in water were 40 ng/mL and 40 ngmL, respectively, for the mortality of rainbow trout yolk sac fry (97). Similarly, DBT has been reported to produce a wide variety of toxic effects such as immunosuppression in exposed fish (97, 98). INSTRUMENTAL ANALYSIS AND BIOMONITORING OF BUTYLTINS The need to identify and determine concentrations of different butyltin species at trace levels was driven by their great toxicity. There are now a variety of instrumental methods available for the routine determination of butyltins in different matrices, but it should be noted that these analytical techniques are still somewhat laborious. The modern techniques are generally sensitive enough to measure butyltin species at sub nglL levels, but quality assurance and quality control efforts should be implemented on a regular basis to assess the accuracy and precision of analytical results. Some of the recent instrumental analytical 29 techniques for butyltins are discussed elsewhere (18). The most sensitive methods for analysis of butyltins involve the conversion to either alkyl derivatives or volatile hydrides followed by chromatographic separation and determination with specific detectors. The gas chromatography (GC) techniques used in butyltin analyses include: GC-MS (mass spectrometery), GC-GFAA (graphite furnace atomic spectrometry), and GC-FPD (flame photometric detector). Methods based on G0 are widely used due to its high resolution and detector versatility. Due to the potential for TBT to Induce imposex in gastropods at water concentrations of a few ng/L, studies have suggested the application of field bioassays using transplanted bivalves to assess TBT contamination in aquatic systems (17, 92). The intensity of expression of imposex in Nucella IapiIIus can generally be related to the water concentration of TBT. The appearance of a small penis and the partial development of a vas deferens first occurs at TBT concentrations below 0.5 ng/L (as tin), although reproduction is unaffected at this level (89). At 1-2 nglL (as tin) penis size is markedly increased and in some females proliferation of vas deferens tissue overgrows the genital papilla, thus sterilizing the animal. At slightly greater concentrations all females become sterile and a concentration of 10 ng tin/L caused suppression of oogenesis and initiated sperrnatogenesis (89). For monitoring purposes, the intensity of imposex is most simply described by the relative penis size (RPS) index in which the bulk of the female penis [(female length)3/(male length)3 x 100] is expressed as a percentage of that of the male. Measurement of the length of 30 a penis in species such as Nugefla is a simple procedure since the penis can be expressed without dissection by placing the animal (minus shell) dorsal side up and drawing back the flap of tissue forming the rook of the mantle cavity. Under a binocular microscope and using 1 mm graduated graph paper, the length of the penis can be measured to the nearest 0.1 mm from its tip to its base. Bifurcate or trifurcate penises are occasionally found, particularly on females, and in these cases the longer or the longest structure is measured. A female lacking any measurable penile outgrowth is registered as zero and this value is included in the calculation of the mean. Whilst length is the most convenient parameter of penis size to measure, it does not convey a true impression of the difference in mass between, for example, a penis of 1 mm and another of 3 mm. However, the weight of the penis is related to the cube of its length (89) and thus the RPS calculation involves the cube of the length. An RPS of 50% indicates that the mean penis size of the female is half the bulk of that of the male. RPS is a reliable measure in all areas except those where contamination is severe. In these areas the male penis is often deformed and consequently the RPS value is lowered and imposex thereby underestimated. Because RPS does not positively identify the presence of sterilized females and thus a second index, the vas deferens sequence (VDS) was devised (89). This index is based on the division of imposex into six stages ranging from early development to the final stage of sterilization. Calculation of the mean VDS stage provides an index by which to compare imposex in different populations. Any population with an index above 4 contains sterilized females and thus has a reduced reproductive capacity. 31 Further details on this biomonitoring technique are given in Bryan et al., 1988. A positive relationship between the incidence of imposex and body residues of TBT has been shown (99). Although the induction of imposex tends to be regarded as a specific response to TBT, field bioassays can be confounded by several environmental factors and, therefore, their applicability to quantitatively monitor butyltin contamination is in question. In vitro bioassays have been used to study the effects of butyltin compounds on EROD activity. Due to high cytotoxic potential of butyltin compounds, use of in vitro studies to elucidate the mechanism of toxic effects of butyltin compounds has been hindered. This indicates the need for a combination of several techniques to identify the sources and effects of butyltins in the environment. BIOASSAY-DIRECTED CHEMICAL FRACTIONATION Unique challenges face the analytical chemist when trying to develop methods for EDCs. The primary difficulty is determining what compounds are EDCs. While various groups and agencies have published lists of EDCs, the question of new or unknown EDCs in the environment remains. By the use of in vitro bioassays like those previously discussed as an analytical tool, the chemist may begin to develop a more comprehensive screening program. When a bioactive sample is discovered, subsequent chemical fractionation and in vitro bioassays may lead to compounds or classes of compounds likely responsible for the observed effect. Once candidate compounds have been identified, a 32 predicted activity can be estimated by applying relative potency factors derived from pure compounds. This predicted activity may be compared to the actual activity measured in the bioassay to determine if a mass-balance exists. A reliable method for sensitive detection of certain EDCs in complex aqueous mixtures has been developed using solid-phase extraction (SPE) and in vitro bioassays (15). Dissolved and particulate phase organics were simultaneously extracted from 5 L water samples at the field site by pumping the samples sequentially through a 90 mm glass fiber filter and a 90 mm Empore styrenedivinylbenzene (SDB) SPE disk. The SDB matrix permits the extraction of both non-polar and semi-polar organics from water. Flow rates and volumes were measured with an in-Iine electronic flow meter/totalizer. Vacuum filtration was used at the laboratory to extract the SPE disks sequentially with acetone, dichloromethane and hexane. All extracts were dried over sodium sulfate and concentrated to near dryness. HPLC with a preparative silica column was used to generate three fractions of the extract based on polarity. The least polar fraction contained halogenated aromatic hydrocarbons (HAHs) and polycyclic aromatic hydrocarbons (PAHs), the moderately polar fraction contained alkylphenols and phthalates, and the fraction of greatest polarity contained steroids, bisphenol A, and alkylphenol polyethoxylate surfactants. Both total and fractionated extracts were assayed for estrogenic/antiestrogenic (ERE-mediated) activity or dioxin-like (AhR-mediated) activity using cell lines transfected with Iuciferase reporter genes under control of estrogen- or dioxin- responsive enhancer sequences, respectively. Additional fractionation by reverse phase 33 HPLC and in vitro testing were sometimes used to more narrowly isolate bioactive compounds. Compounds identified using this strategy included: alkyphenols, steroids, PAHs, and HAHs. Instrumental analyses performed included: HPLC/fluorescence, GCIMS and GCIECD. ELISA was used to quantify synthetic and endogenous estrogen in some bioactive extracts. Sites screened included several wastewater treatment plants in south central Michigan, effluents entering the Trenton Channel of the Detroit River, and areas of Lake Mead in Nevada. The degree of induction over background was used as a measure of the activity of unknown samples to direct further fractionation and instrumental analyses. Most AhR-mediated activity occurred in the most non-polar fraction, while the greatest ERE-mediated activity occurred in the most polar of the three fractions. These polar fractions were further separated by reverse-phase HPLC into 9 fractions. In every case, the estrogenic activity occurred in the fractions containing E2 and EE2. These compounds were determined to be the most likely causes of observed in vitro estrogenicity. Further confirmation was completed using radioimmunoassays (RIAs) for E2 and EE2. Water concentrations of E2 and EE2 ranged from less than detection to 3.7 ng/L and 0.8 nglL, respectively. AhR-active compounds were detected by bioassay in only one sample, indicating that the levels of “dioxin-like” compounds were less than the method detection limits for most of the water samples. This is not suprising as only 5 L of water was extracted. 34 Although NP and OP, known environmental estrogens, were present in many water samples, there was no correlation between these alkylphenols and observed estrogenicity. Not only did the fraction containing these compounds show no bioactivity, the mass balance also would not predict a significant response based on the concentrations in the resulting extracts. Fine fractionation also confirmed that the lack of estrogenicity in the alkylphenol fraction was not due to antagonistic effects. The concentrations of NP and OP ranged from less than detection to 37 and 0.7 pglL, respectively. The in vitro bioassay systems were found to be effective tools for the screening of estrogen and dioxin-like activity in extracts of surface waters and for directing further fractionation and instrumental analyses. The results of the assay can also be compared to the predicted activity calculated from an additive model of estrogenic activity. In the second method, the concentrations of all of the putative estrogenic compounds in a mixture are multiplied by their relative potency factors (determined for individual compounds) and the total activity is predicted by summing the products of these two values. This method allows a mass balance of activity to be calculated. By comparing the response of the cells with whole extracts and various fractions, it is possible to determine whether all of the likely estrogenic compounds have been identified and to investigate nonadditive interactions among compounds. 35 CONLUSIONS Instrumental and bioanalytical methods for the determination and measurement of EDCs in the aquatic environment have been presented. There are many tools available to aid in the identification and characterization of EDCs. In vitro bioassays have great potential for routine use in characterizing EDCs. They become particularly useful tools when used in conjunction with instrumental analyses. They are, however, only relevant as the mechanisms the are based on. Calibration to in vivo responses are crucial for proper application and interpretation of in vitro bioassay results. In vivo bioassays offer an integrated measure of endocrine disruption as the whole animal is taken into account. In vivo testing suffers from the complexity of sorting out mechanisms of toxicity. Analytical chemistry techniques are important in the identification and quantitation of EDCs in the environment and in controlled laboratory studies. The issue of endocrine disruption is extremely complex. Only by combining various investigative techniques can the risks, if any, of EDCs be better understood. EDCs have a wide variety of toxicity mechanisms, while also having a great range of physical properties. An integrated approach is required to better detect, quantify, and understand EDCs in the environment. 36 TABLE 1: End points used to assess reproductive impacts of EDCs in Fish End point vitellogenin (alkaline labile phosphorus, immunoassay, liver mRNA) gonadosomatic index (GSI) plasma sex hormone levels (T, 11-KT, E2 and other estrogens) steroidogenesis enzyme activities and intermediates production of hormones during in vitro organ incubation (gonad, pituitary) gonadotropins and gonadotropin releasing hormone gonad structure and histology (intersex, gonad duct development) activity of liver enzymes involved in steroid metabolism (mixed function oxygenase, or MFO, activity) fertility and fecundity secondary sex characteristics reproductive behavior 37 Selected references (56, 62) (100-103) (104,105) (105) (103,105) (103, 104) (107-109) (57,110) (57,102) (44, 45) (44,111) (1) (2) (3) (4) (5) (6) (7) (8) (9) (10) (11) REFERENCES Kendall, R. J.; Dickerson, R. L.; Giesy, J. P.; Suk, W. A. Principles and Processes for Evaluating Endocrine Disruptors in Wildlife; SETAC Press: Pensacola, FL, 1998. Feyk, L. L.; Giesy, J. P. In Principles and Processes for Evaluating Endocrine Disruptors in Wildlife; Kendall, R. J., Dickerson, R. L., Giesy, J. P., Suk, W. A., Eds.; SETAC Press: Pensacola, FL, 1998, pp 121-140. US. EPA. 1997, Special Report on Environmental Endocrine Disruption: Effects Assesment and Analysis. Office of Research and Development, Washington DC. EPA/630/R-96/012. Safe, S. H. Pharrnacol. Ther. 1995, 67, 247-281. Ahlborg, U. 6.; al., e. Critical Reviews in Toxicology 1995, 25, 463-531. Sharpe, R. M.; Skakkebaek, N. E. Lancet 1993, 341, 1392-1395. Carlsen, E.; Giwercman, A.; Keiding, N.; Skakkebaek, N. E. Environmental Health Perspectives 1995, 130, 137-139. Horwitz, K. B.; Jackson, T. A.; Rain, D. L.; Richer, J. K; Takimoto, G. S.; Tung, L. Mol. Endocrinol. 1996, 10, 1167-1177. Katzenellenbogen, J. A. Environmental Health Perspectives 1995, 103, 99-101. Gillesby, B. E.; Zacharewski, T. R Environmental Toxicology and Chemistry 1998, 17, 3-14. Colborn, T. Environmental Toxicology and Chemistry 1998, 17, 1-2. 38 (12) (13) (14) (15) (16) (17) (18) (19) (20) Villeneuve, D. L.; Blankenship, A. L.; Giesy, J. P. In Toxicant-Receptor Interactions and Modulation of Gene Expression; Denison, M. S., Helferich, W. G., Eds.; Lippincott-Raven Publishers: Philadelphia, PA, 1997, pp 69-99. Giesy, J. P.; Snyder, E. M. In Principles and Processes for Evaluating Endocrine Disruptors in Wildlife; Kendall, R. J., Dickerson, R. L., Giesy, J. P., Suk, W. A., Eds.; SETAC Press: Pensacola, FL, 1998, pp 155-237. Kendall, R. J.; Brouwer, A.; Giesy, J. P. In Principles and Processes for Evaluating Endocrine Disruptors in Wildlife; Kendall, R. J., Dickerson, R. L., Giesy, J. P., Suk, W. A., Eds.; SETAC Press: Pensacola, FL, 1998, pp 1-16. Snyder, S. A.; Keith, T. L.; Verbrugge, D. A.; Snyder, E. M.; Gross, T. S.; Kannan, K.; Giesy, J. P. Environ. Sci. Technol. 1999 (in press). Alzieu, C. Mar. Environ. Res. 1991, 32, 7-17. Spooner, N.; Gibbs, P. E.; Bryan, G. W.; Goad, L. J. Mar. Environ. Res. 1991, 32, 37-39. Fent, K. Crit. Rev. Toxicol. 1996, 26, 1-117. Kannan, K.; Senthilkumar, K; Loganahthan, B. G.; Takahashi, S.; Odell, D. K.; Tanabe, S. Environ. Sci. Technol. 1997, 31, 296-301. Kannan, K.; Guruge, K. S.; Thomas, N. J.; Tanabe, S.; Giesy, J. P. Environ. Sci. Technol. 1998, 32, 1169-1175. 39 (21) (22) (23) (24) (25) (26) (27) (28) (29) (30) Sanderson, J. T.; Giesy, J. P. In Encyclopedia of Environmental Analyses and Remediation; Meyers, R. A., Ed.; John Wiley and Sons, Inc.: New York, NY, 1998, pp 5272-5297. Zacharewski, T. Environmental Science & Technology 1997, 31, 613—623. Kelce, W. R.; CR, 8.; 8.0., L.; LE, 6.; J.A., K.; E.M., W. Nature 1995, 375, 581 -585. White, R.; Jobling, S.; Hoare, A.; Sumpter, J. R; Parker, M. G. Endocrinology 1994, 135, 175-182. Martin, P. M.; Honrvitz, K. B.; Ryan, D. S.; McGuire, W. L. Endocrinology 1978, 103, 1860-1867. Soto, A. M.; C., S. Biochemical and Biophysical Research Communications 1984, 122, 1097-1103. Kelce, W. R.; E., M.; M.P., G.; S.C., L.; L.E., G. Toxicology and Applied Pharmacology 1994, 126, 276-285. Soto, A. M.; Chung, K. L.; Sonnenschein, C. Environmental Health Perspectives 1994, 102, 380-383. Olea, N.; Pulgar, R.; Pilar, P.; Clea-Serrano, F.; Rivas, A.; NovilIo-Fertrell, A.; Pedraza, V.; Soto, A. M.; Sonnenshein, C. Environmental Health Perspectives 1996, 104, 298-305. Welshons, W. V.; Rottinghaus, G. E.; Nonneman, D. J.; Dolan-Timpe, M.; Ross, P. F. Joumal of Veterinary Diagnostic Investigations 1990, 2, 268- 273. 40 (31) (32) (33) (34) (35) (36) (37) (33) (39) (40) (41) Fluoriot, G.; Vaillant, C.; Salbert, G.; Pelissero, C.; Guirard, J. M.; Valotaire Joumal of Cell Science 1993, 105, 407-416. Anderson, M. J.; Miller, M. R.; Hinton, D. E. Aquatic Toxicology 1996, 34, 327-350. Ramamoorthy, K.; Wang, F.; Chen, l. C.; Norris, J. D.; McDonnell, D. P.; Leonard, L. S.; Gaido, K. W.; Bocchinfuso, W. P.; Korach, K. 3.; Safe, S. Endocrinology 1997, 138, 1520-1527. Miksicek, R. J. Molecular Pharmacology 1993, 44, 37-43. Kramer, V. J.; Helferich, W. G.; Bergman, A.; Klasson-Wehler, E.; Giesy, J. P. Toxicology and Applied Pharmacology 1997, 144, 363-376. Zacharewski, T. Environmental Health Perspectives Supplements 1998, 106, 577-582. Demirpence, E.; Duchesne, M. J.; Badia, E.; Gagne, D.; Pons, M. Journal of Steroid Biochemistry and Molecular Biology 1993, 46, 355-363. Pons, M.; Gagne, D.; Nicolas, J. C.; Mehtali, M. Biotechniques 1990, 9, 450-459. Blankenship, A.; Villeneuve, D.; Giesy, J. P. Organohallogen Compounds 1997, 34, 412-416. Khim, J. S.; Villeneuve, D. L.; Kannan, K.; Lee, K. T.; Snyder, S. A.; Koh, C. H.; Giesy, J. P. Submitted to: Environ. Toxicol. Chem. 1999. Koistinen, J.; Soimasuo, M.; Tukia, K.; Oikari, A.; Blankenship, A.; Giesy, J. P. Environ. Toxicol. Chem. 1998, 17, 1499-1507. 41 (42) (43) (44) (45) (46) (47) (48) (49) (50) (51) (52) Kemler, K.; Duda, C. A.; Blankenship, A. L.; Kannan, K.; Giesy, J. P. , SETAC Annual meeting, Charlotte, NC 1998. Lee, K. T.; Villeneuve, D.; Kannan, K.; Giesy, J. P. , SETAC Annual meeting, Charlotte, NC 1998. Bortone, S. A.; Davis, W. P. BioScience 1994, 44, 165-172. Miles-Richardson, S. R.; Kramer, V. J.; Fitzgerald, S. D.; Render, J. A.; Yamini, B.; Barbee, S. J.; Giesy, J. P. Aquatic Toxicology 1998, Submitted. Villalobos, S. A.; Anderson, M. J.; Denison, M. S.; Hinton, D. E.; Tullis, K.; Kennedy, l. M.; Jones, A. 0.; Chang, D. P. Y.; Yang, G.; Kelly, P. Envir. Health Perspect. 1996, 104, 735-743. Dulka, J. G.; Stacey, N. E.; Sorensen, P. W.; Van Der Kraak, G. J. Nature (London) 1987, 325, 251-253. Stacey, N. E.; Cook, A. F.; Peter, R. E. Journal of Fish Biology 1979, 15, 349-361. McMaster, M. E.; Munkittrick, K. R.; Van Der Kraak, G. J. Canadian Technical Report of Fisheries and Aquatic Sciences 1992, 1836, 1-29. Pankhurst, N. W.; Van Der Kraak, 6.; Peter, R. E. Gen. Comp. Endocrinol 1995, 99, 249-257. Pelissero, C.; Le Menn, F.; Kaushick, S. General and Comparative Endocrinology 1 991, 83, 447-457. Murphy, P. A.; Farrnakalidis, E.; Johnson, L. D. Food Chem. Toxicol. 1982, 20, 315-317. 42 (53) (54) (55) (55) (57) (58) (59) (50) (51) Donaldson, E. M.; Scherer, E. In Methods for Assessing the Effects of Chemicals on Reproductive Functions; Vouk, V. B., Sheehan, P. J., Eds; John Wiley & Sons: New York, NY, 1983, pp 365-404. Mommsen, T. P.; Walsh, P. J. In Fish Physiology, CRC: Ann Arbor, 1988; Vol. XIA, pp 348-406. Sumpter, J. P.; Jobling, S. Environ. Health Perspect. 1995, 103, 173-178. Folmar, L. C.; Denslow, N. D.; Rao, V.; Chow, M.; Crain, D. A.; Enblom, J.; Marcino, J.; Guillette, L. J., Jr. Environ. Health Perspect. 1996, 104, 1096- 1101. McMaster, M. E.; Van Der Kraak, G. J.; Portt, C. B.; Munkittrick, K. R.; Sibley, P. K.; Smith, I. R.; Dixon, D. G. Aquat. Toxicol. 1991, 21, 199-218. Goodbred, S. L.; Gilliom, R. J.; Gross, T. S.; Denslow, N. P.; Bryant, W. B.; Schoeb, T. R. ; United States Department of the Interior. United States Geological Survey.: Sacramento, California, 1997, pp 48. Bevans, H. E.; Goodbred, S. L.; Miesner, J. F.; Watkins, S. A.; Gross, T. S.; Denslow, N. D.; Schoeb, T. ; United States Department of the Interior. United States Geological Survey.: Carson City, Nevada, 1996, pp 47. Lye, C. M.; Frid, C. L. J.; Gill, M. E.; McCormick, D. Mar. Pollut. Bull. 1997, 34, 34-41. Harries, J. E.; Sheahan, D. A.; Jobling, S.; Matthiessen, P.; Neall, P.; Routledge, E. J.; Rycroft, R.; Sumpter, J. P.; Tylor, T. Environmental Toxicology and Chemistry 1996, 15, 1993-2002. 43 (52) (53) (54) (55) (56) (57) (53) (59) (70) (71) (72) Harries, J. E.; Sheahan, D. A.; Jobling, S.; Matthiessen, P.; Neall, P.; Sumpter, J. P.; Tylor, T.; Zaman, N. Environmental Toxicology and Chemistry 1997, 16, 534-542. Purdom, C. E.; Hardiman, P. A.; Bye, V. J.; Eno, N. 0.; Tyler, C. R.; Sumpter, J. P. Chemistry and Ecology 1994, 8, 275-285. Nichols, K. M.; Miles-Richardson, S. R.; Snyder, E. M.; Giesy, J. P. Environmental Toxicology and Chemistry 1999, Submitted. Snyder, E. M.; Nichols, K. M.; Pierens, S. L.; Snyder, S. A.; Giesy, J. P. , 1999. Kramer, V. J.; Miles-Richardson, S.; Pierens, S. L.; Giesy, J. P. Aquatic Toxicology 1998, 40, 335-360. Giesy, J. P.; Pierens, S. L.; Snyder, E. M.; Miles-Richardson, S.; Kramer, V. J.; Snyder, S. A.; Nichols, K. M.; Villeneuve, D. A. Environmental Toxicology and Chemistry 1999, Submitted. Arcand-Hoy, L. D.; Benson, W. H. Environ. Toxicol. Chem. 1998, 17, 49- 57. Vlfrsk, J. D.; Cooper, K. R. Environ. Toxixol. Chem. 1990, 1159-1169. Marty, G. D.; Wetzlich, S.; Nunez, J. M.; Craigmill, A.; Hinton, D. E. Aquat. Toxicol. 1991, 329-340. Harris, G. E.; Metcalfe, T. L.; Metcalfe, C. D. Environ. Toxicol. Chem. 1994, 13, 1393-1403. Papoulias, D. M.; Nolte, D.; Tillitt, D. Submitted to: Aquat. Toxicol. 1999. 44 (73) (74) (75) (75) (77) (73) (79) (80) (81) (82) (33) (84) (85) Villalobos, S. A.; Papoulias, D. M.; Meadows, J.; Blankenship, A. L.; Pastva, S. D.; Kannan, K.; Hinton, D. E.; Tillitt, D. E.; Giesy, J. P. Submitted to: Environ. Toxicol. Chem. 1 999. Black, J. J.; Maccubbin, A. E.; Schiffert, M. J. Natl. Cancer Inst. 1985, 1123-1128. Metcalfe, C. D.; Cairns, V. W.; Fitzsimons, J. D. Aquat. Toxicol. 1988, 347- 356. Norrgren, L.; Andersson, T.; Bjork, M. Aquat. Toxicol. 1993, 301-316. Piver, W. T. Environ. Health Perspect. 1973, 4, 61-79. Yamada, S.; Fujii, Y.; Mikami, E.; Kawamura, N.; Hayakawa, J.; Aoki, K.; Fukaya, M.; Terao, C. J. AOAC Int. 1993, 76, 436-441. Takahashi, S.; Mukai, H.; Tanabe, S.; Sakayama, K.; Miyazaki, T.; Masuno, H. Submitted to: Environ. Pollut. 1999. Wu, W.; Roberts, R. S.; Chung, Y.-C.; Ernst, W. R.; Havlicek, S. C. Arch. Environ. Contam. Toxicol. 1989, 18, 839-843. Quevauviller, P. H.; Bruchet, A.; Donard, O. F. X. Appl. Organomet. Chem. 1991, 5, 125-129. Maguire, R. J. Water Poll. Res. J. Canada 1991, 26, 243-360. Bacci, E.; Gaggi, C. Mar. Pollut. Bull. 1989, 20, 290-292. Fent, K.; Muller, M. D. Environ. Sci. Technol. 1991, 25, 489-493. Chau, Y. K.; Zhang, S.; Maguire, R. J. Sci. Total Environ. 1992, 121, 271- 281. 45 (35) (87) (83) (39) (90) (91) (92) (93) (94) (95) (95) (97) Donard, O. F. X.; Quevauviller, P. H.; Bruchet, A. Wat. Res. 1993, 27, 1085-1089. Schebek, L.; Andreae, M. 0.; Tobschall, H. J. Environ. Sci. Technol. 1991, 871-878. Kannan, K.; Senthilkumar, K.; Sinha, R. K. Appl. Organomet. Chem. 1997, 11, 223-230. Bryan, G. W.; Gibbs, P. E.; Burt, G. R. J. Mar. Biol. Ass. UK. 1988, 68, 733-744. Maguire, R. J.; Chau, Y. K.; Bengert, G. A.; Hale, E. J.; Wong, P. T. S.; Kramar, O. Environ. Sci. Technol. 1982, 16, 698-702. Kannan, K.; Grove, R. J.; Senthilkumar, K.; Henny, C. J.; Giesy, J. P. Arch. Environ. Contam. Toxicol. (in press) 1999. Gibbs, P. E.; Bryan, G. W.; Pascoe, P. L. Mar. Environ. Res. 1991, 32, 79- 87. Kannan, K.; Yasunaga, Y.; lwata, H.; Ichihashi, H.; Tanabe, S.; Tatsukawa, R. Arch. Environ. Contam. Toxicol. 1995, 28, 40-47. Kannan, K.; Senthilkumar, K.; Elliott, J. E.; Feyk, L. A.; Giesy, J. P. Arch. Environ. Contam. Toxicol. 1998, 35, 64—69. Lee, R. F. Mar. Environ. Res. 1993, 35, 181-187. Kannan, K.; Villeneuve, D. L.; Blankenship, A. L.; Giesy, J. P. J. Toxicol. Environ. Health 1998, 55, 373-384. deVries, H.; Penninks, A. H.; Snoeij, N. J.; Seinen, W. Sci. Total Environ. 1991, 103, 229-243. 46 (93) (99) (100) (101) (102) (103) Boyer, l. J. Toxicol. 1989, 55, 253-298. Horiguchi, T.; Shiraishi, H.; Shimizu, M.; Yamazaki, S.; Morita, M. Mar. Pollut. Bull. 1995, 13, 402-405. Pereira, J. J.; Mercaldo-Allen, R.; Kuropat, C.; Luedke, D.; Sennefelder, G. Archives of Environmental Contamination and Toxicology 1993, 24, 427- 431. Jobling, S.; Sheahan, D.; Osborne, J. A.; Matthiessen, P.; Sumpter, J. P. Environmental Toxicology and Chemistry 1996, 15, 194-202. Monosson, E.; Fleming, W. J.; Sullivan, C. V. Aquatic Toxicology 1994, 29, 1-19. Singh, P. B.; Kime, D. E.; Epler, P.; Chyb, J. Journal of Fish Biology 1994, 44, 195-204. (104) Van Der Kraak, G. J.; Munkittrick, K. R.; McMaster, M. E.; Portt, C. B.; (105) (106) (107) (108) Chang, J. P. Toxicology and Applied Pharmacology 1992, 115, 224-233. MacLatchy, D. L.; Van Der Kraak, G. J. Toxicology and Applied Pharmacology 1995, 134, 305-312. MacLatchy, D.; Peters, L.; Nickle, J.; Van Der Kraak, G. Environmental Toxicology and Chemistry 1997, 16, 1895-1904. Gimeno, S.; Komen, H.; Venderbosch, P. W. M.; Bowmer, T. Environmental Science & Technology 1997, 31, 2884-2890. Jobling, S.; Nolan, M.; Tyler, C. R.; Brighty, G.; Sumpter, J. P. Environmental Science & Technology 1998, 32, 2498-2506. 47 (109) Gray, M. A.; Metcalfe, C. D. Environmental Toxicology and Chemistry 1997, 16, 1082-1086. (110) Johnson, L.; Casillas, E.; Sol, S.; Collier, T.; Stein, J.; Varanasi, U. Marine Environmental Research 1993, 35, 165-170. (111) Kindler, P. M.; Bahr, J. M.; Philipp, D. P. Hormones and Behavior 1991, 25, 410-423. 48 Estrogen or xenoestrogen 777V fITI é ITI :04 l ‘-—---0’ ERE-Luc . , \ gene \LUleel‘ase III \ \ m NA ’I’I ’ ’ ‘ - o luiferin . . ' xy lucrferrn Luciferase Figure 1. Mechanism for activation of the estrogen receptor (ER) in MVLN cells 49 Chapter 2 BIOCONCENTRATION OF NONYLPHENOL IN FATHEAD MINNOWS (PIMEPHALES PROMELAS) (Submitted to Chemosphere 2000) ABSTRACT Bioaccumulation of p-nonylphenol (NP) by fathead minnows was determined under laboratory conditions. Fish were exposed continuously for 5 wk to 3.4, 1.6, 0.4, 0.16 and 0.05 pg NPIL in a flow-through system. NP was Soxhlet extracted from whole fish homogenates with dichloromethane (DCM). The resulting extract was concentrated and bulk lipids removed by gel permeation and silica gel chromatography. Compounds were identified and quantified by reverse-phase high-pressure liquid chromatography (RP-HPLC) with fluorescence detection. Mass spectrometry was used for verification of peak assignments. Bioconcentration factors ranged from 203 to 268. INTRODUCTION Alkylphenol polyethoxylates (APEs) are non-ionic surfactants that are used in various industrial, institutional, agricultural, and household applications (T almage, 1994). APEs are among the most widely used non-ionic surfactants, with approximately 250 million kg per year used in the United States (US) 50 (Chemical Market Reporter, Oct. 18, 1999). The molecular structure of APEs consist of two moieties, an alkyl-substituted phenol and an ethoxylate oligomer chain. The alkylphenol (AP) portion of the surfactant is fairly non-polar and allows the APEs to dissolve grease and other materials that have small water solubilities. The ethoxylate portion of the surfactant is water-soluble and aids in the transfer of materials to the aqueous phase. APEs are produced commercially by the base-catalyzed ethoxylation of alkylphenols (Cahn & Lynn Jr., 1978). Greater than 90% of the APs used in APE production are of the para substituted isomer. Nonylphenol polyethoxylates (NPEs) that contain a 9 carbon branched isomeric alkyl group (CgI‘I1g) (Figure 1) represent approximately 80% of production (Lye et al., 1999; Naylor et al., 1992; Talmage, 1994). The remaining production is comprised almost entirely of octylphenol polyethoxylates (OPEs). NPEs and the less widely used OPEs degrade during wastewater treatment or in the environment to form transient intermediates such as alkylphenol ethoxycarboxylates (APECs), shorter-chain oligomers of APEs, and alkylphenols (Bennie et al., 1997; Field & Reed, 1996; Naylor et al., 1992; Rudel et al., 1998; Tanghe, Dhooge & Verstraete, 1999). The type and efficiency of wastewater treatment affects the total removal rate of NPEs and the type and proportion of the transient intermediates. Primary treatment removes only 25 - 78% of the total NPE entering wastewater treatment plants (WWTPs). With primary treatment alone, the hydraulic retention time is short and degradation is minimal. Primary effluents contain approximately 82% NPE3 - 20, 12% NPEm, 3% nonylphenol monoethoxycarboxylate and nonylphenol diethoxycarboxylate 51 (NPECm), and 3% nonylphenol (NP) from the initial NPE loading (Ahel, Giger & Koch, 1994). In the United States (US), WWTPs with secondary or tertiary treatment achieve average removal rates in excess of 97% for NPE. Effluents from these types of WWTPs contain approximately 28% NPE3 .. 20, 22% NPEm, 46% NPECm, and 4% NP from the initial NPE loading (Ahel et al., 1994). Although NP is a minor degradation component of NPE, significant amounts of this slightly lipophilic, moderately toxic compound enter the environment. NP has a log K0W of 4.84 (Ahel & Giger, 1993b), water solubility of 5.4 mg/L (Ahel & Giger, 1993a), and a log Koc of 3.58 (Naylor et al., 1992). This physico-chemical profile indicates that NP may bioaccumulate in aquatic organisms. This has been documented in several species of fish from natural waters and from controlled laboratory exposures (T almage, 1994). NP bioconcentration factors (BCFs) for the fathead minnow (Pimephales promelas) exposed to 5 and 25 pg NP/L for 20-d was determined to be 270 and 350, respectively (Naylor, 1995). Half-times for depuration have been determined to be 1.4 and 1.2 (1, respectively, following exposure to 5 and 25 pg NPIL (Naylor, 1995). Nonylphenol has been shown to be weakly estrogenic in a variety of both in vitro (Desbrow et al., 1998; Jobling & Sumpter, 1993; Mueller & Kim, 1978; Soto et al., 1992; White et al., 1994) and in vivo bioassays (Giesy et al., 2000; Jobling et al., 1996; Nimrod & Benson, 1996). L050 values for acute toxicity to fish range from 0.135 to 0.3 mg NPIL for fathead minnows (Pimephales promelas) (Dwyer et al., 1995) and from 0.167 to 0.221 mg NPIL for three 52 species of trout (Naylor, 1995). Reports of APEs and their degradation products in the environment in the US. are scarce. The most comprehensive survey reports concentrations from 30 US. rivers that are influenced by municipal or industrial wastewater effluents (Naylor et al., 1992). That study found that 60- 75% of samples had no detectable levels of NP, NPE1, and NPEZ. NP concentrations observed in that study ranged from <0.11 to 0.64 pglL (Naylor et al., 1992). Concentrations of NP in effluents and surface waters ranging from not detectable to 37 pglL have also been reported (Snyder et al., 1999). The objectives of this study were to develop a suitable method for extraction, clean-up, and quantification of NP in fish tissues and to use this method to determine the BCF for partitioning of NP into fathead minnows during a controlled laboratory exposure. EXPERIMENTAL SECTION Standards and Reagents: All standards and reagents used were of the highest purity commercially available. High purity standards of p-nonylphenol (NP), p-tert octylphenol (OP), and p-tert butylphenol (BP) were obtained from Schenectady International (Freeport, TX). Pesticide residue grade acetonitrile (ACN), hexane, dichloromethane (DCM), and acetone were obtained from Burdick and Jackson (Muskegon, MI). Reagent water was first purified by reverse osmosis (RO) followed by NanopureTM (Barnstead, Dubuque, IO) treatment. Glass wool (8 Elm, Corning Glass Works, Corning, NY) was Soxhlet extracted overnight with DCM, 53 then dried and stored in glass containers until needed. Glass fiber filters, fine grade (GF/F) of 0.7 pm nominal pore size were heated at 450 °C for 4 h in aluminum foil prior to use. Anhydrous granular sodium sulfate (EM Science, Gibbstown, NJ) was heated at 450 °C overnight then stored at 125 °C until day of use. Fish Exposure. Fathead minnows (Pimephales promelas) were cultured in the Michigan State University Aquatic Toxicology Laboratory (MSU-ATL) under a 16:8 lightzdark cycle in continuously flowing, well water at 17-19 °C. Fish were fed a 1:1 (v:v) mixture of Tetramin® (Tetrawerke, Melle Germany) and ground Sterling Silver Cup (2-4 mm pellet size) (Murray, UT) once daily at a rate of approximately 0.5% of fresh body weight. The dry food was supplemented with Artemia (brine shrimp) nauplii. Freeze-dried Spirulina sp algae was fed to fry up to 2.5 cm length. Reproductively mature fish, 12 to 18 mo of age, were transferred to randomly assigned 19 L glass aquaria in a circulating water- jacketed bath (Frigid Units Living Stream, Toledo, OH). Each tank received 3 Uh treated well water through a solenoid-controlled proportional-flow diluter apparatus (Ace Glass, Vineland, NJ). Each tank received vigorous aeration, and glass lids prevented escape of aerosols from the tanks. The temperature of the water jacket was maintained at 26 °C. Fish were randomly assigned to 20 groups, each containing 2 males and 2 females and placed into 19L aquaria. Treatments were in triplicate with the exception of the solvent control group, which due to the restriction of 54 space in the water bath, contained only two aquaria. Fish were allowed to acclimate in control water for 7 d then exposed to NP for a total of 42 d. The aquaria were randomly assigned to receive treatment concentrations of NP or control solutions delivered by a proportional-flow diluter. Treatments included control, which received only water, and a solvent control, which received the same concentration of solvent (0.0001% ethanol) as the NP treatments. Fathead minnows were exposed to target (nominal) concentrations of 0.1, 0.3, 1.0, 3.0 and 10 pg NPIL. The actual, measured concentrations were 0.05, 0.16, 0.4, 1.1 and 3.4 pg NPIL (Table 2). Three fish from each exposure level were analyzed for NP. Water Extraction. Water samples were collected 3 times during the exposure to determine actual NP concentrations. Analytical methods were adapted from those published previously (Snyder et al., 1999). One L samples were spiked with 1.0 pg BP in methanol as a surrogate standard. The fortified samples were vacuum filtered and extracted using a 47 mm glass fiber filters (GF/F) directly above a 47 mm styrenedivinylbenzene (SDB) EmporeTM solid-phase extraction (SPE) disk. The SPE disk was frozen until extraction. The SPE disk was solvent extracted with 15 mL of acetone followed by 25 mL DCM. Moisture was removed by passing the extract through 30 g of anhydrous sodium sulfate. The dried extract was concentrated to 1 mL in iso-octane. A 2 g gravimetric silica column (60 - 100 mesh, Aldrich Chemical Company, Milwaukee, WI) column was washed with 100 mL of 20% DCM in hexane and the extract loaded. 55 Alkylphenols were eluted with 100 mL of DCM. This fraction was concentrated to 1 mL in acetonitrile (ACN) for RP-HPLC analysis. Fish Extraction and Clean-up. Individual, whole fathead minnows were weighed then homogenized with 30 g of anhydrous sodium sulfate. The homogenate was spiked with 0.50 pg of OP as a surrogate standard and Soxhlet extracted for 12 h using 300 mL DCM. The resulting extract was concentrated at 30 °C by rotary evaporation followed by a gentle stream of nitrogen to a final volume of 2 mL. Gel permeation chromatography (GPC) was used to remove bulk lipids from the extracts. The GPC system consisted of a Rheodyne 7725i injector (Cotati, CA) with a 5 mL sample loop, a quaternary high-pressure liquid chromatography (HPLC) pump (Perkin Elmer, Series 410, Norwalk, CT), and an electronic fraction collector (ISCO Foxy 200, Lincoln, NE). Two Phenomenex (Torrance, CA) GPC columns were connected in tandem; a PhenogelTM 5 pm 500 A (300 x 21.20 mm) followed by an Envirosep-ABC (350 x 21 .20 mm). Both GPC columns were preceded by an Envirosep-ABC (60 x 21.20 mm) guard column (Phenomenex, Torrance, CA). The pump was operated isocratically at 3 mL/min with DCM as the mobile-phase solvent. A fraction containing OP and NP was collected from 20-35 min. This fraction was concentrated and cleaned-up in the same manner as the water extracts. 56 Quantitation of Compounds. Extracts were analyzed by RP-HPLC as described previously (Snyder et al., 1999). The reverse-phase HPLC system included a Perkin Elmer (PE) (Norwalk, CT) series 200 autosampler, a PE series 200 binary pump, a Hewlett Packard (HP) 1046A fluorescence detector, and PE TurboChrome 4.0 data software package. A Phenomenex ProdigyTM 5 pm octadecylsilica (ODS) 100 A (250 mm x 4.6 mm, Torrance, CA) preceded by a 30 mm x 4.6 mm guard column of the same packing material was used. For all compounds of interest, the fluorescence detector settings were: 229 nm excitation, 310 nm emission, PMT gain of 12, lamp time of -1, response time of 2 sec, stop time at 27 min, gate and delay at zero. The elution profile was a 20 min gradient (curve = -2) from 50% water in ACN to 2% water in ACN followed by a 10 min ACN purge. Each sequence began with one blank and one standard. The injection volume of standards and samples was 25 pL. Peak areas were determined by electronic integration, and sample concentrations were determined using Microsoft Excel version 7.0. Chromatography and retention times for the compounds of interest are given (Figure 2). All calibration curves were linear (r‘2 > 0.99). An HP 5890 series ll plus GC with a 30 m DB5-MS capillary column (J&W Scientific, Folsom, CA) and a HP 5972 series mass selective detector (MSD) were used for confirmation of peak assignments. Recovery and Precision. Instrumental detection limits (lDLs) and limits of quantitation (LOQs) were determined by analyzing dilute standards (near the 57 estimated IDL) and calculating the signal-to-noise ratio [peak height (mV) divided by baseline noise] for each standard concentration. Linear regression of the signal to noise ratios against concentrations was then used to determine the IDL (signal to noise ratio = 3) and LOQ (signal to noise ratio = 10). The method detection limit (MDL) was determined by spiking seven 1.5 g homogenized fathead minnow tissue samples with dilute concentrations of NP and OP (at the estimated MDL) and analyzing as described previously. The MDL was calculated by multiplying the standard deviation (SD) of the resulting concentrations by a t—value of 3.1 (for n=7). RESULTS Recoveries for each of the compounds of interest were in excess of 70%. For both water and tissue analyses, data were adjusted for surrogate recovery. The lDLs, LOQs, and MDLs for these compounds in water and tissue are shown (Table 1). The clean-up steps for this method did not remove all lipids present in the fish tissue extracts, resulting in an increased baseline and elevated operating pressures. Therefore, after approximately 5 injections of fish tissue extracts, the analytical column was purged with successively less polar solvents to remove residual lipids. With the exception of the controls, NP was detected in the water in each of the exposure tanks (Table 2). For each exposure level, measured concentrations were approximately 1/3 of nominal concentrations. 58 Concentrations were fairly constant throughout the exposure period. Nonylphenol was not detectable in fish tissues from the lowest (measured) exposure concentrations, 0.16 and 0.05 pg NPIL (Table 2). Therefore, bioconcentration factors (BCFs) were calculated only for the 0.4, 1.6, and 3.4 pg NPIL (measured) exposure levels (Table 2). The coefficient of variation (CV) for triplicate water samples was less than 26%. These fish were homogenized, then 1 g aliquots were removed for analysis. The CV for triplicate measurements of NP in fish tissue was less than 29%. BCFs were calculated by dividing the average tissue concentration by the average measured exposure concentration (Table 2). DISCUSSION The BCFs determined here are similar to previously reported values ranging from 100 -300 (Liber et al., 1999; Naylor, 1995; Staples et al., 1998). The analytical method described here works well for tissue samples with a mass of less than 1.5 9. For environmentally exposed fish, it would be necessary to assure that other residues that can fluoresce (for example, polycyclic aromatic hydrocarbons) present in the tissue do not interfere with the HPLC analysis. Concentrations of NP in surface waters are often less than 0.16 pg NPIL (Naylor et al., 1992). Fish exposed to 0.16 pg NPIL would, assuming a BCF of 300, contain a concentration of 48 pg NPlkg. Thus, the MDL for tissues presented 59 here may be insufficient to assess accurately the degree of accumulation of NP in fish from surface waters containing small concentrations of NP. ACKNOWLEDGMENTS This work was supported by the Chlorine Chemical Council of the Chemical Manufacturer’s Association, the National Institute of Environmental Health Sciences (NIEHS) Superfund Basic Research Program (E80491), and the US. Environmental Protection Agency (EPA) Office of Water Quality (CR 822983-01-0). REFERENCES Ahel, M. & Giger, W. (1993a). Aqueous solubility of alkylphenols and alkylphenol polyethoxylates. Chemosphere 26, 1461-1470. Ahel, M. & Giger, W. (1993b). Partitioning of alkylphenols and alkylphenol polyethoxylates between water and organic solvents. Chemosphere 26, 1471- 1478. Ahel, M., Giger, W. & Koch, M. (1994). Behaviour of alkylphenol polyethoxylate surfactants in the aquatic environment - ll. Occurence and transformation in sewage treatment. Water Research.28, 1131 -1 142. 60 Bennie, D. T., Sullivan, C. A., Lee, H. B., Peart, T. E. & Maguire, R. J. (1997). Occurence of alkylphenols and alkylphenol mono- and diethoxylates in natural waters of the Laurentian Great Lakes basin and the upper St. Lawrence River. The Science of the Total Environment 193, 263-275. Cahn, A. & Lynn Jr., J. L. (1978). Surfactants and Detersive Systems. In “Kirk-Othmer Encyclopedia of Chemical Technology”, vol. 22, pp. 365. J. Wiley & Sons, Inc., New York. Desbrow, C., Routledge, E. J., Brighty, G. C., Sumpter, J. P. & Waldock, M. (1998). Identification of Estrogenic Chemicals in STW Eflleunt. 1. Chemical Fractionation and in vitro Biological Screening. Environmental Science & Technology 32, 1549-1558. Dwyer, F. J., Sappington, L. C., Buckler, D. R. & Jones, S. B. (1995). “Use of surrogate species in assessing contaminant risk to endangered and threatened species”, pp. 71. National Technical lnforrnation Service, Springfield, VA. Field, J. A. & Reed, R. L. (1996). Nonylphenol Polyethoxy Carboxylate Metabolites of Nonionic Surfactants in US. Paper Mill Effluents, Municipal Sewage Treatment Plant Effluents, and River Waters. Environmental Science & Technology 30, 3544-3550. 61 Giesy, J. P., Pierens, S. L., Snyder, E. M., Miles-Richardson, S., Kramer, V. J., Snyder, S. A., Nichols, K. M. & Villeneuve, D. A. (2000). Effects of 4-Nonyl Phenol on Fecundity and Biomarkers of Estrogenicity in Fathead Minnows (Pimephales promelas). Environmental Toxicology and Chemistry (in press). Jobling, S., Sheahan, D., Osborne, J. A., Matthiessen, P. & Sumpter, J. P. (1996). Inhibition of testicular growth in rainbow trout (Oncorhynchus mykiss) exposed to estrogenic alkylphenolic chemicals. Environmental Toxicology and Chemistry 1 5, 194-202. Jobling, S. & Sumpter, J. P. (1993). Detergent components in sewage effluent are weakly oestrogenic to fish: an in vitro study using rainbow trout (Oncorhynchus mykiss) hepatocytes. Aquatic Toxicology 27, 361 -372. Liber, K., Gangl, J. A., Corry, T. D., Heinis, L. J. & Stay, F. S. (1999). Lethality and Bioaccumulation of 4-Nonylphenol in Bluegill Sunfish in Littoral Enclosures. Environmental Toxicology and Chemistry 18, 394-400. Lye, C. M., Frid, C. L. J., Gill, M. E., Cooper, D. W. & Jones, D. M. (1999). Estrogenic Alklphenols in Fish Tissues, Sediments, and Waters from the UK. Tyne and Tees Estuaries. Environmental Science & Technology 33, 1009-1014. 62 Mueller, G. C. & Kim, U. H. (1978). Displacement of estradiol from estrogen receptors by simple alkylphenols. Endocrinology 102, 1429-1435. Naylor, C. G. (1995). Environmental fate and safety of nonylphenol ethoxylates. Text. Chem. Color. 27, 29-33. Naylor, C. G., Mieure, J. R, Adams, W. J., Weeks, J. A., Castaldi, F. J., Ogle, L. D. & Romano, R. R. (1992). Alkylphenol ethoxylates in the environment. JAOCS 69, 695-703. Nimrod, A. C. & Benson, W. H. (1996). Environmental Estrogenic Effects of Alkylphenol Ethoxylates. Critical Reviews in Toxicology 26, 335-364. Rudel, R. A., Melly, S. J., Geno, P. W., Sun, G. & Brody, J. G. (1998). Identification of Alkylphenols and Other Estrogenic Phenolic Compounds in Wastewater, Septage, and Groundwater on Cape Cod, Massachusetts. Environmental Science & Technology 32, 861-869. Snyder, S. A., Keith, T. L., Verbrugge, D. A., Snyder, E. M., Gross, T. S., Kannan, K. & Giesy, J. P. (1999). analytical methods for detection of selected estrogenic compounds in aqueous mixtures. Environmental Science & Technology 33, 2814-2820. 63 Soto, A. M., Tien-Min, L., Honorato, J., Silvia, R. M. & Sonnenschein, C. (1992). An "in culture" bioassay to assess the estrogencity of xenobiotics (e- screen). In “Chemically-induced alterations in sexual and functional development: The wildlife/human connection”, vol. 21 (ed. M. A. Mehlman). Princeton Scientific Publishing Co.,lnc, Princeton,NJ. Staples, C. A., Weeks, J., Hall, J. & Naylor, C. (1998). Evaluation of Aquatic Toxicity and Bioaccumulation of C8- and 09- Alkylphenol Ethoxylates. Environmental Toxicology and Chemistry 17, 2470-2480. Talmage, S. S. (1994). “Environmental and Human Safety of Major Surfactants: Alcohol Ethoxylates and Alklphenol Ethoxylates”. Lewis Publishers, Boca Raton. Tanghe, T., Dhooge, W. & Verstraete, W. (1999). Isolation of a bacterial strain able to degrade branched nonylphenol. Applied and Environmental Microbiology 65, 746-751. White, R., Jobling, S., Hoare, S. A., Sumpter, J. P. & Parker, M. G. (1994). Environmentally Persistent Alkylphenolic Compounds Are Estrogenic. Endocrinology 135, 175-1 82. 64 TABLE 1. Detection Limits for Compounds of Interest Detection Limit BP OP NP IDL (nglml) 2 2 7 L00 (nglml) 7 8 23 MDL (ng/L Water) 23 16 45 MDL (nglg Tissue) NA 39 67 NA = not applicable BP = butylphenol OP = octylphenol NP = nonylphenol IDL = instrumental detection limit LOQ = limit of quantitation MDL = method detection limit 65 TABLE 2. Bioconcentration of NP in fathead minnows (mean concentrations) Nominal Measured Tissue BCF Concentration Concentration Concentration (HQ/LI (nglL) (91kg) 0.1 0.05 0.9) across the entire calibration range. Recovery and Detection Limits. Instrument detection limits (lLDs) were defined as the minimum mass of analyte required to yield a signal to noise ratio of 3. Seven replicates of homogenized goldfish tissues were spiked with NP and NPE(1-3) at the estimated method detection limits (MDLs) to determine recovery and precision (Table 1). MDLs were calculated by multiplying the standard deviation (SD) of the recovered concentrations by a t-value of 3.1427 (for n=7 replicates). The limits of quantitation (LOQs) were calculated by adding 5 80s to the MDL. 75 RESULTS AND DISCUSSION It was determined that two successive distillations with fresh iso-octane were required to achieve acceptable analyte recovery. It was also observed that two successive extractions of 1.5 h each were more efficient than one 3 h distillation. Recoveries of the compounds of interest using two distillations were consistently greater than 70% with the exception of NPE3 (Table 2). The coefficient of variation (CV) in recoveries were less than 20% (T able 2). The lesser recovery of NPE3 is a function of its lesser volatility and lipophillicity. Initial method development with steam-distillation resulted in extreme foaming that resulted in contamination of the iso-octane extract. While silicon-based anti- foaming agents successfully reduced foaming, recoveries were reduced and inconsistent. The addition of concentrated sulfuric acid reduced foaming and did not negatively effect recovery or precision. Although cooling the distillation condensers resulted in slightly greater recovery, the improvement was small. NP and NPE1 were detected in the tissue from the carp captured in Las Vegas Bay (Table 3). The variability in concentrations of NP and NPE1 among the subsamples was small (Table 3). These concentrations observed are reasonable, based upon reported water concentrations (18) and published bioconcentration factors (BCFs) of 100 to 300 (8, 24, 25). Although concentrations of NP and NPE in sediments of Las Vegas Bay are unknown, it is likely that sediment bound NP and NPE effected the carp tissue concentrations, as carp are bottom-feeders. 76 The method presented here was successful for sensitive and reproducible measurements of concentrations of NP and NPE“ a. 2) in fish tissues. The resulting detection limits are sufficient to determine toxicologicalIy-relevant concentrations of the compounds of interest in fish tissues. This method also utilizes laboratory equipment that is common or inexpensive, which makes the method ammenable for use in most environmental laboratories. ACKNOWLEDGMENTS Funding for this project was provided by the Alkylphenol Exthoxylate Research Coucil (APEC). Carp were provided by the Nevada Division of Wildlife, Las Vegas, Nevada. The authors would like to thank Erin M. Snyder for critically reviewing this manuscript. We would also like to thank Bob Fensterhelm of the APERC for technical guidance of this project. REFERENCES (1) Talmage, S. S. Environmental and Human Safety of Major Surfactants: Alcohol Ethoxylates and Alklphenol Ethoxylates; Lewis Publishers: Boca Raton. 1994. (2) Naylor, C. G.; Mieure, J. R; Adams, W. J.; Weeks, J. A.; Castaldi, F. J.; Ogle, L. D.; Romano, R. R. JAOCS1992, 69, 695-703. (3) Field, J. A.; Reed, R. L. Environ. Sci. Technol. 1996, 30, 3544-3550. 77 (4) (5) (6) (7) (8) (9) (10) (11) (12) (13) (14) Rudel, R. A.; Melly, S. J.; Geno, P. W.; Sun, 6.; Brody, J. G. Environ. Sci. Technol. 1998, 32, 861-869. Bennie, D. T.; Sullivan, C. A.; Lee, H. B.; Peart, T. E.; Maguire, R. J. Sci. Total Environ. 1997, 193, 263-275. Naylor, C. G. Text. Chem. Color. 1995, 27, 29-33. Weeks, J. A.; Adams, W. J.; Guiney, P. D.; Hall, J. F.; Naylor, C. G. The Alkyphenols and Alkylphenol Ethoxylates Review 1998, 1, 64-74. Staples, C. A.; Weeks, J.; Hall, J.; Naylor, C. Environ. Toxicol. Chem. 1998, 17, 2470-2480. White, R.; Jobling, S.; Hoare, S. A.; Sumpter, J. R; Parker, M. G. Endo. 1994, 135, 175-182. Mueller, G. C.; Kim, U. H. Endo. 1978, 102, 1429-1435. Soto, A. M.; Tien-Min, L.; Honorato, J.; Silvia, R. M.; Sonnenschein, C. In Chemically-induced alterations in sexual and functional development: The wildlife/human connection; Mehlman, M. A., Ed.; Princeton Scientific Publishing Co.,lnc: Princeton,NJ, 1992; Vol. 21. Desbrow, C.; Routledge, E. J.; Brighty, G. C.; Sumpter, J. P.; Waldock, M. Environ. Sci. Technol. 1998, 32, 1549-1558. Jobling, S.; Noylan, M.; Tyler, C. R.; Brighty, G.; Sumpter, J. P. Environ. Sci. Technol. 1998, 32, 2498-2506. Jobling, S.; Sheahan, D.; Osborne, J. A.; Matthiessen, P.; Sumpter, J. P. Environ. Toxicol. Chem. 1996, 15, 194-202. 78 (15) (15) (17) (13) (19) (20) (21) (22) (23) (24) (25) Nimrod, A. C.; Benson, W. H. Critical Reviews in Toxicology 1996, 26, 335-364. Jobling, S.; Sumpter, J. P. Aquat. Toxicol. 1993, 27, 361-372. Routledge, E. J.; Sumpter, J. P. Joumal of Biological Chemistry 1997, 272, 3280-3288. Snyder, S. A.; Keith, T. L.; Verbrugge, D. A.; Snyder, E. M.; Gross, T. S.; Kannan, K.; Giesy, J. P. Environ. Sci. Technol. 1999, 33, 2814-2820. Lye, C. M.; Frid, C. L. J.; Gill, M. E.; Cooper, D. W.; Jones, D. M. Environ. Sci. Technol. 1999, 33, 1009-1014. Kubeck, E.; Naylor, C. G. JAOCS 1990, 67, 400-405. Fowler, B. R.; Haviland, L.; Kennedy, S. SETAC Presentation 1998. Ahel, M.; McEvoy, J.; Giger, W. Environmental Pollution 1993, 79, 243- 248. Veith, G. D.; Kiwus, L. M. Bull. Environ. Contam. Toxicol. 1977, 17, 631- 636. Liber, K.; Gangl, J.; Corry, T.; Heinis, L.; Stay, F. Environ. Toxicol. Chem. 1999, 18, 394-400. Snyder, S. A.; Keith, T. L.; Pierens, S. L.; Snyder, E. M; Giesy, J. P. Chemosphere, 2000 (submitted). 79 TABLE 1. lens Monitored and Recovery Data Compound Ions (mlz) Spike (nglg) % Recovered NP 107, 135, 149 15.4 78.1 x 9.21 NPE1 135, 179, 193 74.8 76.1 i 9.84 NPE; 135, 223, 237 67.2 69.4 i 13.0 NPE3 135, 267, 281 40.0 17.0 i 20.1 TBC 149, 164, 121 NA NA NA = not applicable NP = nonylphenol NPE1 = nonylphenol monoethoxylate NPE2 = nonylphenol diethoxylate NPE3 = nonylphenol triethoxylate TBC = tert-butyl orthocresol 80 TABLE 2. Detection Limits for Compounds of Interest Compound IDL(ng) MDL(ngIg) LOQ(ngIg) NP 5.12 3.30 4.82 NPE1 15.5 16.8 18.5 NPE; 17.3 18.2 20.6 NPE3 112 20.6 28.9 NP = nonylphenol NPE1 = nonylphenol monoethoxylate NPE; = nonylphenol diethoxylate NPE3 = nonylphenol triethoxylate 81 TABLE 3. Average Concentrations of NP and NPEs in Lake Mead Carp Compound nglg wet wt. %CV NP 184 4.4 NPE1 242 9.3 NPEz ND NA NPE3 ND NA ND = not detectable at MDL NA = not applicable NP = nonylphenol NPE1 = nonylphenol monoethoxylate NPE; = nonylphenol diethoxylate NPE3 = nonylphenol triethoxylate 82 Ho. Ho. p-tert-butyl orthocresol p-Cumylphenol H0 (C9H19) p-Nonylphenol H— (o— c—cy; o—.—(c,H,,) Nonylphenol ethoxylate1 4 Figure 1. Structures of Compounds of Interest. 83 Chapter 4 ANALYTICAL METHODS FOR DETECTION OF SELECTED ESTROGENIC COMPOUNDS IN AQUEOUS MIXTURES (Published in Environmental Science and Technology, 1999) ABSTRACT Both natural estrogens and synthetic compounds that mimic estrogen can reach the aquatic environment through wastewater discharges. Because nonylphenol (NP), octylphenol (OP), nonylphenol polyethoxylates (NPE), 17B- estradiol (E2), and ethynylestradiol (EE2) have previously been found to be estrogenic and occur in wastewater effluents, they were the primary analytes for which the method was developed. Water samples were extracted in situ with solid-phase extraction disks. Analytes were separated by high-pressure liquid chromatography (HPLC) and detected by fluorescence or competitive radioimmunoassay (RIA). Method detection limits (MDLs) using HPLC with fluorescence detection were 11, 2, and 52 nglL of water for NP, DP, and NPE, respectively. The RIA MDLs for E2 and EE2 were 107 and 53 pglL, respectively. Samples were collected from 4 municipal wastewater treatment plants (WWTPs) in south central Michigan, 8 locations on the Trenton Channel of the Detroit River, Michigan, and 5 locations in Lake Mead, Nevada. Concentrations of NP and OP ranged from less than the MDL to 37 and 0.7 pglL, respectively. NPE 84 concentrations ranged from less than the MDL to 332 pglL. Concentrations of E2 and EE2 ranged from less than the MDLs to 3.7 nglL and 0.8 nglL, respectively. INTRODUCTION The potential effects of chemicals that alter the normal endocrine function and physiological status of animals have been an increasing concern in recent years (1). Reports of apparent increases in hormone-dependent cancers and decreases in sperm quantity and quality in humans have raised questions about the role of natural and synthetic chemicals in these trends (2-7). A number of pollutants including pesticides, certain polychlorinated biphenyls (PCBs), dioxins, furans, alkylphenols, synthetic steroids and natural products such as phytoestrogens have been reported to disrupt normal hormonal pathways in animals and collectively have been referred to as endocrine disrupters or endocrine disrupting chemicals (EDCs) (2, 8). While EDCs can operate through a number of both direct and indirect mechanisms of action, of particular concern are those compounds that mimic endogenous estrogens. The Safe Drinking Water Act Amendments of 1995 (bill number S1316) and the Food Quality Protection Act of 1996 (bill number PL. 104-170), which mandate comprehensive screening for estrogenic and anti-estrogenic chemicals, are examples of the increasing public concern regarding endocrine disruption. While it is known that many natural and synthetic chemicals are estrogenic, it is unclear whether or not the concentrations of estrogenic agents present in the 85 environment are sufficient to cause adverse physiological effects. One aspect of conducting human or wildlife risk assessments is an exposure assessment. This suggests the need for assays and techniques to monitor the quantity and effects of endocrine disrupters. WWTPs receive natural and synthetic EDCs from urban and industrial dischargers. WWTPs use a variety of treatment processes of varying efficiency such that in some cases compounds are not completely removed by the treatment processes and are ultimately discharged into surface waters. Fish caged below WWTP outfalls have been reported to have abnormal ratios of sex steroid hormones as well as to exhibit histological changes in their reproductive organs (9-11). Fish living in waters influenced by WWTP effluents have been observed to express similar effects attributed to unknown or unspecified EDCs (12-14). Extracts of some WWTP effluents in the United Kingdom have been shown by use of in vitro assays, to be estrogenic due to the presence of alkylphenols and natural and synthetic estrogens (15). Alkylphenol polyethoxylates (APEs) are non-ionic surfactants widely used in various industrial (55% of total demand), institutional (30% of total demand) and household applications (15% of total demand) (16). APEs are among the most widely used non-ionic surfactants, with 1998 US. use of approximately 204 million kg per year (16). Most of the APEs used are of the nonylphenol polyethoxylate (NPE) types that contain a 9 carbon branched isomeric alkyl group (16, 17). NPEs and the less widely used octylphenol polyethoxylates (OPEs) degrade during wastewater treatment or in the 86 environment to alkylphenol ethoxycarboxylates (APECs), lower oligomer APEs such as NPE1-3, and alkylphenols such as octylphenol (OP) and nonylphenol (NP) (Figure 1) (17-20). NP and OP have been shown to be estrogenic in a variety of both in vitro (9, 15, 21-23) and in vivo bioassays (24-26). Some oligomers of NPE have also been found to be estrogenic in vitro (9, 23, 24, 27) and in vivo (24). Reports of APEs and their corresponding degradation products from the US. are scarce. The most comprehensive survey reports concentrations from 30 US. rivers that are influenced by municipal or industrial wastewater effluents (17). That study found that 60-75% of samples had no detectable levels of NP, NPE1, and NPEz (17). The endogenous hormone 178-estradiol (E2) and the synthetic hormone ethynylestradiol (EE2) have been detected in several WWTP effluents (Figure 1) (15, 28-31). Synthetic hormones are generally more stable in water than are natural hormones and have greater potency (31, 32). EE2 was found to be unchanged after 120 h in activated sludge treatment, indicating possible release into surface waters after waste water treatment (29). EE2 is a widely used pharmaceutical with daily oral doses ranging from 30 - 50 pg (33). Estrogens are excreted by humans in concentrations as great as 2.7 mg/L on a daily basis (34). Conjugated estrogens used in the treatment of cancer, hormonal imbalance, osteoporosis, and other ailments are the second most prescribed drug in the US. and are administered orally in doses ranging from 0.3 — 2.5 mg (33). Estrogens are excreted by mammals as water-soluble conjugates in urine or as “free” estrogens in feces (30, 35, 36). Conjugated forms of these estrogens 87 can be deconjugated during wastewater treatment and In the environment to generate the more potent “free” estrogen (15, 31). Environmental concentrations of E2 and EE2 range from less than detection to greater than 140 ng/L and less than detection to 15 nglL, respectively (15, 28, 29, 32). Fish exposed to E2 and EE2 exhibit changes in biomarkers for estrogenicity at concentrations as low as 0.5 nglL (10, 15, 37, 38). In this report, we describe a rapid and sensitive method for detecting and quantifying EDCs in surface waters. The method was designed to isolate and concentrate a broad range of organic compounds from surface waters so that it could be used as an analytical tool for toxicity identification and evaluation (TIE). An in situ extraction technique using 90 mm styrenedivinylbenzene (SDB) EmporeTM solid-phase extraction disks was developed. This technique permits the extraction of greater volumes of water more rapidly and efficiently than the typical vacuum filtration methods (39). This method also provides detection limits less than the reported effects concentrations for these compounds in the aquatic environment. The SDM matrix is effective for extracting polar organics, which is necessary has the compounds of interest all have polar hydroxyl functional groups. Previously EmporeTM disks have been used for the extraction of natural water sucessfully (18, 39-41). However, these compounds of interest were not investigated. APs and certain steroids are extracted by this method without requiring the transport of large volumes of water, thus reducing losses due to adsorption to transport containers and due to microbial degradation (Figure 2). To demonstrate the 88 utility of this method, samples of effluents and surface waters were collected from several locations in the Trenton Channel of the Detroit River, Michigan; below the outfalls of municipal WWTPs in south central Michigan; and in the Las Vegas (LV) Wash and in Lake Mead, Nevada. EXPERIMENTAL SECTION Standards and Reagents. All standards and reagents used were of the highest purity commercially available. High purity standards (98% pure) of p-nonylphenol (NP), p-tert butylphenol (BP), and p-tert octylphenol (OP) were obtained from Schenectady International (Freeport, TX). Standards of nonylphenol monoethoxylate (NPE1) and nonylphenol polyethoxylates (SurfonicTM N-95) were obtained from Huntsman Corporation (Austin, TX). Standards of E2 and EE2 of 98% purity were obtained from Sigma Chemical Company (St. Louis, MO). High purity, pesticide residue grade ACN, methanol (MeOH), hexane, dichloromethane (MeClz), iso-octane, 2-propanol, and acetone were obtained from Burdick and Jackson (Muskegon, MI) or Sigma-Aldrich (Milwaukee, WI). Solvents were also tested for interferences by examining 150-fold concentrated solvents using high-pressure liquid chromatography (HPLC) with fluorescence detection and gas chromatography/mass spectrometery (GCIMS). Reagent water was first purified by reverse osmosis (RO) followed by NanopureTM (Barnstead, Dubuque, IA) treatment. Glass fiber filters, fine grade (GF/F) of 0.7 pm nominal pore size and coarse grade (GF/C) of 1.2 pm nominal pore size 89 (\Nhatman, Maidstone, England), were heated at 450 °C for 4 h in aluminum foil prior to use. Anhydrous granular sodium sulfate (EM Science, Gibbstown, NJ) was heated at 550 °C overnight then stored at 125 °C until day of use. Sample Collection and Preservation. Water samples were collected from 8 sites in the Trenton Channel of the Detroit River, MI; 4 WWTPs in south central Michigan, and 5 sites in Lake Mead, NV. To minimize effects due to dilution, effluent samples were collected as close to the source as possible. Samples from the Trenton Channel and Lake Mead were collected at 50% of the maximum depth or 3 m, whichever was less. Samples were collected between April and October 1997. All samples were filtered and extracted at the sample location (Figure 2). Water samples were pumped through fluoropolymer (PFA) tubing (3l8” o.d.) by a Fluid Metering (Oyster Bay, NY) DC pump (model QB) fitted with a ceramic lined stainless steel pump head (model Q1-CSC) to a 90 mm stainless steel pressure filtration holder (Millipore, Bedford, MA). Flow rate was maintained at 100 mL/min until 5 L total volume had been extracted or until the pressure exceeded 100 psi. Stainless steel screen (250 pm mesh, Aquatic Ecosystems, Apopka, FL) was used to prevent large particles from entering the inlet tubing. A glass fiber filter was placed on each side of a 90 mm poly (styrenedivinylbenzene) (SDB-XC) EmporeTM (3M Corporation, St. Paul, MN). The bottom filter (90 mm GF/C filter) was used to improve flow characteristics, and the top filter (90 mm GF/F) was used to filter particulates from the sample. 90 Fifteen mL each of acetone, MeClz, and MeOH, and 20 mL reagent water were injected sequentially through a custom Luer lock fitting on the relief valve of the filtration holder with a 2 min residence period to condition the EmporeTM disk. A McMillan (Georgetown, TX) electronic flow meter (model 102-6TP), calibrated to i 1% accuracy with National Institute of Standards and Technology (NIST) traceable standards, was installed downstream of the filtration holder. This digital rate meter and totalizer was used to monitor flow rate and to record the total volume. Once the desired sample volume had been reached, a volume of 20 mL of air was pushed through the filter holder to move headspace water through the holder. The filter holder then was disassembled and the GF/F and EmporeTM disk were stored separately in clean aluminum foil and kept on ice for transport to the laboratory. The GF/C filter was discarded. At each sampling site, prior to collecting the samples, the apparatus was purged with sample for 10 min by bypassing the filter holder. At sites with concentration gradients, samples were taken sequentially from the least to greatest predicted concentrations. Flow meter calibration was verified prior to each sampling trip by comparing the reported totalizer output to the measured volume of water exiting the meter. The sampling apparatus was cleaned with water and solvents before and after each sample. Field blanks were taken daily by passing 5 L of NanopureTM water through the system as previously described. Laboratory blanks were performed on each day of extractions and instrument blanks were conducted daily. No blank had detectable concentrations of any of the compounds of interest. None of the 91 compounds of interest were detected in the blanks. Breakthrough tests were performed by placing two EmporeTM disks in series and analyzing each disk separately. Breakthrough was detected only at the greatest concentrations spiked and the breakthrough was determined to be less than 11%. Extraction. EmporeTM disks were eluted on a 90-mm vacuum manifold (3M Corporation, St. Paul, MN). Fifteen mL of acetone, 25 mL of MeClz, and 10 mL of hexane were added sequentially to the reservoir and collected in a 50-mL centrifuge tube. Slight vacuum was applied to initiate solvent transfer and the remainder was permitted to flow without vacuum. Vacuum was applied once all solvents had passed through the disk to remove residual solvents from the disk. Moisture was removed from the solvent extract by passing it through 30 g anhydrous sodium sulfate placed in a custom glass funnel containing a stopcock at 1 drop/sec. The eluate was collected in a flat bottom flask and rotary evaporated at 30 °C to approximately 5 mL. The sample was concentrated and solvent-exchanged to 1 mL iso-octane under a gentle stream of nitrogen at 30 °C. One mL of 40% hexane in MeClz was added to the test tube and vortexed for 5 sec. The sample was collected in a 2.5 mL Hamilton syringe (Reno, NV) and injected into a normal phase HPLC system for fractionation. Normal-Phase HPLC Fractionation. The HPLC fractionation system consisted of a Rheodyne 7725i injector (Cotati, CA) with a 5 mL sample loop, a quaternary pump (Perkin Elmer, Series 410, Nonrvalk, CT), and an electronic fraction 92 collector (ISCO Foxy 200, Lincoln, NE). A Phenomenex Luna 5 pm silica column (250 mm x 4.6 mm, Torrance, CA) was used for nonnal-phase separations with 3-step isocratic elution. The mobile-phase solvent profile was 30% MeClz in hexane for 15 min, MeCIz for 20 min, and MeOH for 20 min, each at 1 mUmin with no gradient curves. The column was returned to initial conditions by passing MeClz for 10 min followed by 30% MeClz in hexane for 35 min at 1mUmin. Fractions were collected from 0-20 min (F 1), 20-45 min (F2), and 45-70 min (F3) with no delay. F1 contained the most non-polar compounds such as PAHs, PCBs, organochlorine (OC) pesticides, and fluorescent compounds leached from the EmporeTM SDB-XC disk. F2 contained BP, NP, and OP. F3 contained the most polar compounds, including E2, EE2, and NPEs. Each fraction was concentrated by rotary evaporation at 30 °C to approximately 5 mL. The sample was evaporated and solvent-exhanged to 1 mL ACN under a gentle stream of nitrogen. No internal or surrogate standards were added to the samples since these could interfere with the in vitro bioassays used to screen for total estrogenic and dioxin-like activity. Quantitation of Compounds. F2 and F3 were analyzed by reverse-phase HPLC (Figures 3 and 4). The reverse-phase HPLC system included a Perkin Elmer (PE) (Nomalk, CT) series 200 autosampler, a PE series 200 binary pump, a Hewlett Packard (HP) 1046A fluorescence detector, and PE TurboChrome 4.0 data software package. The column used was a Phenomenex Prodigy 5 pm octadecylsilica (ODS) 100 A (250 mm x 4.6 mm, Torrance, CA) preceded by a 30 93 mm x 4.6 mm guard column of the same packing material. For all compounds of interest, thefluorescence detector settings were: 229 nm excitation, 310 nm emission, PMT gain of 12, lamp time of -1, response time of 2 sec, stop time at 27 min, gate and delay at zero. Elution solvents for gradient elution were reagent water and ACN delivered at a constant flow rate of 1 mL/min. The elution profile was a 20 min gradient (curve = -2) from 50% water and 50% ACN to 2% water and 98% ACN followed by a 10 min isocratic ACN purge. Each sequence began with one blank and one standard. The injection volume of standards and samples was 10 pL. Peak areas were determined by electronic integration, and sample concentrations were determined using Microsoft Excel version 7.0. Chromatography and retention times for the compounds of interest are shown in Figures 3 and 4. All calibration curves were linear (r2 > 0.99) across the entire calibration range. An HP 5890 series ll plus GC with a 30 m DB5-MS capillary column (J&W Scientific, Folsom, CA) and a HP 5972 series mass selective detector (MSD) were used for confirmation of peak assignments. Radioimmunoassay (RIA). Competitive radioimmunoassay (RIA) was used for detection of E2 and EE2 following a procedure previously described for measurement of these compounds in plasma and adapted for environmental samples (14). The most polar fraction (F3) from each of the HPLC fractionation procedures was analyzed in duplicate for both E2 and EE2. A 20 pL aliquot of sample was evaporated to dryness in a 5 mL glass test tube by use of a vortex 94 evaporator (Labconco, Kansas City, MO) and nitrogen gas. Each sample was reconstituted in 0.5 M sodium phosphate buffered saline buffer (PBSGA). Standard curves were prepared in PBSGA. Accuracy and precision of the assay were determined in several ways. Cross-reactivities of the E2 antiserum with other steroids are 11.2 % for estrone; 1.7% for estriol; <1.0 % for 178-estradiol and androstenedione; and <0.1% for EE2, DES, and all other steroids examined. Cross-reactivities of the EE2 antiserum with other steroids were 0.3% for E2; <0.1% for norethindrone, estrone, 178-estradiol, diethylstilbestrol, hexoestrol and dienoestrol; and <0.01% for all other steroids and compounds tested. To determine recovery during reverse-phase HPLC fractionation, known amounts (1, 2, 5, 10, 25, 50, 100, 250, and 500 pg) of E2 or EE2 were fractionated by HPLC and the masses of the recovered materials determined. No significant losses occurred during fractionation. Parallelism between the dose-response relationships for standards and samples were evaluated by assaying dilutions of HPLC fractions (2.5, 5, 10, 15, and 20 pL HPLC fraction volumes). The standard curves were determined to be parallel to the dilution curves for HPLC fractions. Recovery and Precision. Spike/recovery experiments were performed using reagent water or untreated ground water to determine the accuracy and precision of the method. Nineteen-L glass carboys were spiked with NP, BP, OP, and EE2 and mixed for at least 1 h. Three 4-L samples were sequentially extracted from the 19-L mixture using the procedure described as above. Three spikes were 95 also performed to determine the effect of larger volumes by spiking 19 L and extracting 17 L. Flow rate was optimized by spiking 4 L of water with the compounds of interest and loading the EmporeTM disks at different rates. For NPE, quantitation was based on a sum of all oligomers and reported as total NPE (NPE). The instrument was calibrated using SurfonicTM N-95. Therefore, all results relative to NPE concentrations are semi-quantitative in relation to the defined conditions. To assure NPE oligomer distribution integrity, oligomer distribution of extracted NPE was compared to that of an NPE (as Surfonicm N-95) standard. The NPE oligomer distribution was determined by normal phase HPLC using a Phenomenex Phenosphere 5 pm cyanopropyl column (250 mm x 4.6 mm). The PE binary HPLC instrument and fluorescence detector have been described previously. The mobile phase solvents consisted of 35% iso-propanol in MeOH (A) and hexane (B). The elution profile was 3% A and 97% B for 7 min at a flow rate of 0.5 mUmin isocratic followed by a 20 min gradient to 95% A and 5% B at a curve of -5 and flow rate of 1 mUmin. The column was purged a 1 mUmin with 3% A and 97% B to return the column to the initial conditions. It was determined that the oligomer distribution remained unchanged throughout the procedure (Figure 5). Oligomer-spec'rfic distributions were not determined for environmental samples. To determine losses of EE2, OP, NP, and BP during evaporation procedure, these compounds were spiked into a 250 mL flat bottom flasks containing the correct amounts and ratios of solvents used for the EmporeTM 96 extraction. Solvents were rotary evaporated and the volume was adjusted as shown earlier. Detection Limits. The instrumental detection limits (IDLs) and limits of quantitation (LOQs) for NP, OP, BP, NPE, and EE2 by reverse phase HPLC with fluorescence detection were determined by injecting 10 pL of a 50 ngImL mixture of each of these compounds 5 times. The IDLs and LOQs are defined as 3 times and 10 times the standard deviation (SD) of the quantified peak for each compound in the mixture, respectively. The method detection limits (MDLs) using reverse phase HPLC with fluorescence detection (10 pL injections) were estimated from the mass of analyte at the LOQ divided by 5 L sample size plus 25% to account for a 75% average recovery. The IDLs for E2 and EE2 using RIA were determined using lmmunoFitTM EIAIRIA Data Analysis Software (Beckman, Fullerton, CA). The minimum concentration distinguishable from zero for E2 and EE2 was 427 and 211 pg/mL extract, respectively. The MDLs for E2 and EE2 using RIA detection were calculated in the same manner as for HPLC with fluorescence detection. RESULTS Recoveries for these compounds at various concentrations are shown (Table 1). It was determined that recovery did not vary significantly between untreated ground water and reagent water. The compounds of interested added 97 to a natural water sample from Lake Mead were recovered within the range determined by laboratory spike-recovery experiments. Flow rates greater than 500 mLImin resulted in poor recoveries (<50%) possibly due to breakthrough. The greatest average recovery of > 80% for all analytes was achieved using a flow rate of 50 mLImin; however, this resulted in extraction times > 1.5 h. A flow rate of 100 mLImin resulted in consistent recoveries of the compounds of interest with an acceptable average recovery >70% (Table 1). BP was the only compound with poor recoveries (average 60%). BP was tested for use as a possible surrogate standard in future studies because it has been used previously as an internal standard (42). BP has a greater volatility than the other compounds of interest, which is the likely cause for its lesser recoveries (Table 1). If rotary and nitrogen evaporation temperatures and final volumes were not carefully monitored, losses of up to 50% were observed. By maintaining the evaporation temperatures at 30 °C and avoiding over-concentration, evaporation losses could be kept consistent and less than 10%. It was determined that EE2 could function as a surrogate for E2. Recoveries for E2 and EE2 were virtually the same for each experiment performed. Additionally, E2 required fresh standards weekly, while EE2 was stable and remained in calibration for at least 6 mo. NPE was also spiked and recovered 4 times with an average recovery of 71% i 12. In environmental samples, oligomers greater than NPE4 rarely were observed (qualitatively) by GC/MS. 98 The IDLs, LOQs, and MDLs for the compounds of interest for each method of detection are presented in Table 2. The coefficient of variation (CV) for duplicate RIA analyses was less than 12% in each case. The compounds of interest were detected in some samples (Tables 3, 4, & 5). NP was detected in 17 of the 23 samples collected (not including replicate samples) with concentrations ranging from less than detection to 37 pglL. OP was detected in 13 of the 23 samples analyzed (not including replicate samples) with concentrations ranging from less than the MDL to 0.67 pglL. NPE was detected in 16 of the 23 samples analyzed (not including replicate samples) with concentrations ranging from less than the MDL to 332 pglL. E2 was detected in 16 of the 22 samples analyzed (not including replicate samples) with concentrations ranging from less than the MDL to 3.7 nglL. EE2 was detected in 8 of the 22 samples analyzed (not including replicate samples) with concentrations ranging from less than the MDL to 0.76 ng/L. DISCUSSION The Trenton Channel of the Detroit River, Michigan, is in an industrialized and urbanized area with point sources of industrial and municipal waste water effluents (20, 43, 44). Concentrations of NP and OP at Grosse lle, an island that forms the Trenton Channel, have been reported to be 0.12 and 0.045 pglL, respectively, in June, 1994 (20). These concentrations are similar to those observed in our studies. The Trenton Channel is influenced by these compounds 99 not only in the head waters but by point sources entering the Channel. Monguagan Creek appears to provide the greatest loading of NP, OP, and NPE to the Trenton Channel (Table 3). This Creek was previously reported to be a source for unique tert-alkylphenols such as 4-tert-pentylphenol (45). Many of these unique alkylphenols were also detected during GCIMS screening in this study. However, no concentrations are reported here. No trend was observed from the concentrations of E2 and EE2 detected in the Trenton Channel. The greatest concentration of E2 and the only detectable concentration of EE2 were at the Black Lagoon site. Concentrations of NP, OP, NPE, E2, and EE2 were determined in 4 WWTP effluents in south central Michigan. Samples were also analyzed upstream from 3 of the WWTP discharges. Concentrations of these compounds varied greatly among locations (Table 4). NP and E2 were detected in each effluent sample analyzed. No upstream site contained detectable concentrations of NP, OP, NPE, or EE2. The only upstream location with a detectable concentration of E2 was the BV site. This site is located in an agricultural area with many livestock farms, which may have contributed to the detection of E2, but we do not have enough data to draw conclusions on the reason for this occurrence. It should be noted, however, that the BV site uses a lagoon system for wastewater treatment. Operators of that plant during the October sampling event indicated that only primary treatment was in operation at that time. This effect can be seen in the F3 chromatogram (diluted 50x) of the sample collected during that time period (Figure 4). The NPE oligomer distribution is different from 100 that of the other sites (Figure 4). This is an indication of inefficient treatment. Removal rates for NP during wastewater treatment have been shown to be 95- 99% (46). Concentrations of NP in municipal wastewater effluents in the US. have been previously demonstrated to range from less than detection to 4.9 pglL (46). These concentrations are similar to the concentrations observed in our studies. In US. rivers, NP has been reported to be undetectable (MDL=0.11 pglL) in 60-75% of samples collected and averaged 0.12 pglL in samples where it was detected (17). Concentrations of analytes varied greatly among locations within Lake Mead (Table 5). The LV Wash site was located at the confluence of VWVTP effluents from the city of Las Vegas and Clark County Santitation District, Nevada. This combined effluent stream represents an average of approximately 460 million Uday of tertiary treated wastewater. The LV Bay site is located approximately 8 km downstream of the LV Wash site and is located in Lake Mead near the entry point of the LV Wash. The LV Bay site would be expected to have some mixing of the effluent with Lake Mead water and some dilution from non-point source ground water entry. Concentrations of NP, OP, and NPE decreased by nearly 50% from LV Wash to LV Bay, while E2 and EE2 concentrations decreased and increased, respectively. The increase in EE2 could be due to the deconjugation of EE2 conjugates (15, 31). The LV Marina site lies approximately 2.3 km downstream of the LV Bay site. While only E2 was detected at this site, it should be noted that previous research by the US. Bureau of Reclamation has shown an interflow of the dense waste water extending from 101 the Las Vegas Wash to the Hoover Dam (47). This interflow lies on top of the hypolimnion and its depth changes seasonally. It is likely that our samples collected at 3 m of depth do not represent the influence of the wastewater effluent at the LV Marina and Saddle Island sites, thus, offering a possible explanation of the non-detectable concentrations of the compounds of interest. Both of these sites lie in the path of the wastewater interflow as it moves towards the Hoover Dam. Samples collected in September followed a rain event that resulted in a three-fold increase of the LV Wash average daily flow (personal communication from Clark County Sanitation District). The addition of large amounts of storm water in the LV Wash could explain the lesser concentrations of the compounds of interest from this sampling event. To our knowledge this paper offers the first reported data for these compounds in the waters of Lake Mead, Nevada. This method permits the sensitive detection of NP, OP, NPE, E2 and EE2 without the transportation of large volumes of water. This equates to savings in time and costs while achieving biologically relevant detection limits. While HPLC fluorescence provides sensitive detection of E2 and EE2, polar interferences were encountered in all environmental samples. RIA allowed a very sensitive detection of E2 and EE2 without a rigorous clean-up. However, RIA is also sensitive to compounds that are structurally similar to the analyte. Future efforts will include greater sample volumes and derivatizations to achieve structural confirmations of E2 and EE2 by GCIMS. 102 Although the sites studied represent only one point in time and were chosen to demonstrate the utility of the method, rather than the dynamics of the target compounds, it can be concluded that estrogenic compounds are widespread contaminants of WWTP effluents. A monitoring program would be required to better describe the flux and fate of these compounds in the environment. While the concentrations of these compounds determined in this study are in the range of those predicted to cause physiological responses in certain fish (10, 15, 37, 38), no direct link currently exists between the sites evaluated and a direct cause-effect relationship. ACKNOWLEDGMENTS This work was supported by the Chlorine Chemical Council (CCC) of the Chemical Manufacturer’s Association, the National Institute of Environmental Health Sciences (NIEHS) Superfund Basic Research Program (ES0491), and the US. Environmental Protection Agency (EPA) Office of Water Quality (CR 822983-01-0). Technical and material support was provided by Carter Naylor of Huntsman Corp., Austin TX; Eric Hulsey of Phenomenex Corp.; and Oliver Bell of 3M Corp. We would also like to thank Dr. Matthew Zabik of Michigan State University for his technical assistance throughout this project. Logistical support for Lake Mead, NV samples was provided by the Southern Nevada Water Authority, US. National Park Service, and the US. Bureau of Reclamation. 103 REFERENCES (1) (2) (3) (0 (Q (5) (7) (8) (9) (10) (11) (12) Colborn, T. Environ. Toxicol. Chem. 1998, 17, 1-2. Gillesby, B. E.; Zacharewski, T. R. Environ. Toxicol. Chem. 1998, 17, 3- 14. Katzenellenbogen, J. A. Environ. Health Perspect. 1995, 103, 99-101. Honrvitz, K. B.; Jackson, T. A.; Rain, D. L.; Richer, J. K.; Takimoto, G. S.; Tung, L. Mol. Endocrinol. 1996, 10, 1167-1177. Carlsen, E.; Giwercman, A.; Keiding, N.; Skakkebaek, N. E. Environ. Health Perspect. 1995, 130, 137-139. Sharpe, R. M.; Skakkebaek, N. E. Lancet 1993, 341, 1392-1395. Ahlborg, U. G.; Critical Reviews in Toxicology 1995, 25, 463-531. Safe, S. H.; Gaido, K. Environ. Toxicol. Chem. 1998, 17, 119-126. White, R.; Jobling, S.; Hoare, S. A.; Sumpter, J. R; Parker, M. G. Endo. 1994, 135, 175-182. Purdom, C. E.; Hardiman, P. A.; Bye, V. J.; Eno, N. 0; Tyler, C. R.; Sumpter, J. P. Chem. Ecol. 1994, 8, 275-285. Harries, J. E.; Sheahan, D. A.; Jobling, S.; Matthiessen, P.; Neall, P.; Sumpter, J. P.; Tylor, T.; Zaman, N. Environ. Toxicol. Chem. 1997, 16, 534-542. Jobling, S.; Noylan, M.; Tyler, C. R.; Brighty, G.; Sumpter, J. P. Environ. Sci. Technol. 1998, 32, 2498-2506. 104 (13) (14) (15) (16) (17) (18) (19) (20) (21) (22) Folmar, L. C.; Denslow, N. D.; Rao, V.; Chow, M.; Crain, D. A.; Enblom, J.; Marcino, J.; Guillette Jr., L. J. Environ. Health Perspect. 1996, 104, 1096- 1101. Bevans, H. E.; Goodbred, S. L.; Miesner, J. F.; Watkins, S. A.; Gross, T. S.; Denslow, N. D.; Schoeb, T. Water-Resources Investigations Report 96—4266 1996. Desbrow, C.; Routledge, E. J.; Brighty, G. C.; Sumpter, J. P.; Waldock, M. Environ. Sci. Technol. 1998, 32, 1549-1558. Talmage, S. S. Environmental and Human Safety of Major Surfactants: Alcohol Ethoxylates and Alklphenol Ethoxylates; Lewis Publishers: Boca Raton, 1994. Naylor, C. G.; Mieure, J. R; Adams, W. J.; Weeks, J. A.; Castaldi, F. J.; Ogle, L. D.; Romano, R. R. JAOCS1992, 69, 695-703. Field, J. A.; Reed, R. L. Environ. Sci. Technol. 1996, 30, 3544-3550. Rudel, R. A.; Melly, S. J.; Geno, P. W.; Sun, 6.; Brody, J. G. Environ. Sci. Technol. 1998, 32, 861-869. Bennie, D. T.; Sullivan, C. A.; Lee, H. B.; Peart, T. E.; Maguire, R. J. Sci. Total Environ. 1997, 193, 263-275. Mueller, G. C.; Kim, U. H. Endo. 1978, 102, 1429-1435. Soto, A. M.; Tien-Min, L.; Honorato, J.; Silvia, R. M.; Sonnenschein, C. In “Chemically-induced alterations in sexual and functional development: The wildlife/human connection”; Mehlman, M. A., Ed.; Princeton Scientific Publishing Co.,lnc: Princeton,NJ, 1992; Vol. 21. 105 (23) (24) (25) (26) (27) (28) (29) (30) (31) (32) (33) (34) (35) (35) Jobling, S.; Sumpter, J. P. Aquat. Toxicol. 1993, 27, 361-372. Jobling, S.; Sheahan, D.; Osborne, J. A.; Matthiessen, P.; Sumpter, J. P. Environ. Toxicol. Chem. 1996, 15, 194-202. Giesy, J. P.; Pierens, S. L.; Snyder, E. M.; Miles-Richardson, S.; Kramer, V. J.; Snyder, S. A.; Nichols, K. M.; Villeneuve, D. A. Environ. Toxicol. Chem. 2000 (in press). Nimrod, A. C.; Benson, W. H. Critical Reviews in Toxicology 1996, 26, 335-364. Routledge, E. J.; Sumpter, J. P. Joumal of Biological Chemistry 1997, 272, 3280-3288. Shore, L. S.; Gurevitz, M.; Shemesh, M. Bull. Environ. Contam. Toxicol. 1993, 51, 361-366. Aherne, G. W.; Briggs, R. J. Pharrn. Phamiacol. 1989, 41, 735-736. Knight, W. M. Estrogens in the Environment; Elsevier North Holland: Amsterdam, 1980. Tabak, H. H.; Bloomhuff, R. N.; Bunch, R. L. Dev. Ind. Microbiol. 1981, 22, 497-519. Aherne, G. W.; English, J.; Marks, V. Ecotoxicology and Environmental Safety 1985, 9, 79-83. Unknown In World Wide Web, 1998; Vol. www.rxlist.comltop200.htm. F ostis, T. J. Stet. Biochem. 1987, 28, 21 5-226. Shore, L. S.; Shemesh, M.; Cohen, R. Aust. Vet. J. 1988, 65, 67. Velle, W. General and Comparative Endocrinology 1963, 3, 621-635. 106 (37) (33) (39) (40) (41) (42) (43) (44) (45) (45) (47) Kramer, V. J.; Miles-Richardson, S.; Pierens, S. L.; Giesy, J. P. Aquat. Toxicol. 1998, 40, 335-360. Routledge, E. J.; Sheahan, D.; Desbrow, C.; Brighty, G. C.; Waldock, M.; Sumpter, J. P. Environ. Toxicol. Chem. 1998, 32, 1559-1565. Durand, G.; Chiron, S.; Bouvot, V.; Barcelo, D. International Journal of Environmental Analytical Chemistry 1992, 49, 31 -42. Barcelo, D.; Durand, G.; Bouvot, V.; Nielen, M. Environ. Sci. Technol. 1993, 27, 271-277. Evans, 0.; Jacobs, B. J.; Cohen, A. L. Analyst 1991, 116, 15-19. Blackburn, M. A.; Waldock, M. J. Water Res. 1995, 29, 1623-1629. Froese, K. L.; Verbrugge, D. A.; Snyder, S. A.; Tilton, F.; Tuchman, M.; Ostaszewski, A.; Giesy, J. P. J. Great Lakes Res. 1997, 23, 440-449. Carter, D. S.; Hites, R. A. Environ. Sci. Technol. 1992, 26, 1333-1341. Shiraishi, H.; Carter, D. S.; Hites, R. A. Biomedical and Environmental Mass Spectrometry 1989, 18, 478-483. Naylor, C. G. Text. Chem. Color. 1995, 27, 29-33. LaBounty, J. F.; Horn, M. J. Journal of Lake and Reservoir Management 1997, 13, 95-108. 107 TABLE 1. Recoveries through the analytical procedure (n=3) (% Recovered : SD) Spike cone (nglL) Volume EE2 BP OP NP 240 5 78:6 65:7 76:4 77:7 1128 5 78:4 62:3 72:3 74:5 2374 5 76:5 61:8 74:4 76:5 5934 5 74:3 65:6 73:6 72:8 23737 5 72:7 46:4 68:9 69:4 10761 17 63:7 48:5 67:10 67:9 EE2 = ethynylestradiol BP = butylphenol OP = octylphenol NP = nonylphenol 108 TABLE 2. Dection Limits for Selected Xenoestrogens Method E2 EE2 BP OP NP NPE HPLC/ IDL (nglml) NM 5 4 3 13 62 Fluorescence “ LOQ (nglml) NM 16 13 9 42 208 “ MDL (nglL) NM 4 3 2 11 52 RIA IDL (pg/ml) 427 21 1 NA NA NA NA “ LOQ (pg/ml) NA NA NA NA NA NA “ MDL (pg/L) 1 07 53 NA NA NA NA NM = not measured NA = not applicable IDL = instrumental detection limit LOQ = limit of quantitation MDL = method detection limit RIA = radioimmunoassay 109 TABLE 3. Concentrations of Selected Xenoestrogens in the Trenton Channel Location Date NP OP NPE E2 EE2 (no/L) (nglL) (nglL) (poll-I (pg/Ll Lower Transect 8/30/97 993 26 6970 ND ND Trenton WWTP 8/30/97 479 5 5390 1070 ND Chemical Plant 8/30/97 862 15 7310 91 1 ND Power Plant 8/30I97 721 17 8600 ND ND Elizabeth Park 8/30/97 269 ND 1910 435 ND Black Lagoon 8/30/97 936 66 8680 1290 359 Monguagan 8/30/97 1 190 81 17800 1060 ND Creek Upper Transect 8/30/97 665 23 8330 1280 ND ND = not detectable NM = not measured NP = nonylphenol OP = octylphenol NPE = nonylphenolethoxylate E2 = 178-estradiol EE2 = ethynylestradiol 110 TABLE 4. Concentrations of Selected Xenoestrogens at WWTPs Location Date NP OP NPE E2 EE2 (nglL) (HQ/L) (nglL) (Pg/L) (Pg/L) ER-effluent 5/16/97 806 N D 9020 NM NM EL-effluent 5/16/97 1020 72 9310 656 248 BV-effluent 5/16/97 22800 249 21800 3230 242 BV-upstream 10l8l97 ND ND ND 711 ND BV-effluent 10l8l97 37000 673 332000 3660 759 MA-upstream 10l8l97 ND ND ND ND ND MA-effluent 10/8/97 590 16 4850 905 357 ER-upstream 10l8l97 ND ND ND ND ND ER-effluent 10l8l97 171 ND ND 477 ND ND = not detectable NM = not measured NP = nonylphenol OP = octylphenol NPE = nonylphenolethoxylate E2 = 178-estradiol EE2 = ethynylestradiol 111 TABLE 5. Concentrations of Selected Xenoestrogens in Lake Mead Location Date NP OP NPE E2 EE2 (HI/L) (nglL) (nglL) (Pg/L) (Pg/L) LV Wash 4/30/97 1 140 43 8990 2670 480 LV Bay 4/30/97 750 27 4850 2210 520 9/5/97 160 ND 3180 188 253 LV Marina 9/5/97 ND ND ND 270 ND Saddle 4/30/97 ND ND ND ND ND Island Callville Bay 9/5/97 ND ND ND ND ND ND = not detectable NM = not measured NP = nonylphenol OP = octylphenol NPE = nonylphenolethoxylate E2 = 178-estradiol EE2 = ethynylestradiol 112 .0 Q p-t-Octylphenol HO Ethinylestradiol Ho—@—(CQH19) p-Nonylphenol 17B-Estradiol Figure 1. Structures of target compounds 113 5L in situ SPE extraction l Vacuum/Solvent Extraction l Concentrate to 1 mL l Normal Phase Chromatography Fractionation i F1 v F2 V F3 Non-polar Semi-Polar Polar HAHs and PAHs OP, NP NPE, E2, EE2 Reverse Phase Radioimmunoassay Fluorescence E2, EE2 Figure 2. Flow diagram of analytical method OP, NP, NPE 114 BP OP A. NP B. C Figure 3. Reverse-phase HPLC of F2: (A) spiked standards; (B) Trenton Channel — Black Lagoon 8/30/97; (C) WWTP - BV effluent 10l8l97 115 EE2 B. r—I—II r 1 I I _.'_. C. r—I—I—w . r fiT I w—F—r—w—r—u Figure 4. Reverse-phase HPLC of F3: (A) spiked standards; (B) Trenton- Channel — Black Lagoon 8/30/97; (C) WWTP — BV effluent 10l8l97 116 Figure 5. Normal-phase HPLC of NPE oligomer distribution: (A) NPE standard; (B) extracted NPE 117 Chapter 5 TOXICITY IDENTIFICATION AND EVALUATION OF ESTROGENIC AND DIOXIN-LIKE COMPOUNDS IN WASTEWATER EFFLUENTS (To be submitted to Environmental Science and Technology) ABSTRACT Many xenobiotics enter the aquatic environment from wastewater discharges. Recent reports have shown that some compounds in wastewater effluent are estrogenic while others are dioxin-like. The project described here was designed to screen extractable organic compounds dissolved in water samples for in vitro estrogen and dioxin-like activity. Samples from 3 municipal wastewater treatment plants (WWTPs) in south central Michigan (upstream and effluent); 4 point source locations on the Trenton Channel of the Detroit River, Michigan; and 5 locations in Lake Mead, Nevada were analyzed. Organic compounds were extracted from 5 L water samples using solid-phase extraction disks. Extracts were further fractionated based on polarity. Whole extracts and fractions were tested for estrogen- and dioxin-like activity using MVLN and H4IIE- Iuc in vitro bioassays, respectively. No significant dioxin-like activity was observed. Estrogenic activity was quantitated by comparing the magnitude of induction elicited by the extract or fraction to the maximum induction caused by 178-estradiol (E2). Several extracts and fractions displayed significant 118 estrogenic activity. In each case, the most polar fraction (F3) was associated with the greatest estrogenicity. Instrumental analyses and further fractionation were used to identify compounds associated with estrogenic responses. Results suggested that the endogenous estrogen, E2, and the synthetic estrogen, ethynylestradiol (EE2), were the most likely cause of the responses observed. Alkylphenols (APs) did not appear to contribute significantly to the observed estrogenicity. INTRODUCTION A number of compounds released into the environment by human activities can modulate endogenous hormone activities and have been termed endocrine-disrupting compounds (EDCs) (1, 2). EDCs have been defined as exogenous agents which interfere with the "synthesis, secretion, transport, binding, action, or elimination of natural hormones in the body that are responsible for the maintenance of homeostasis, reproduction, development, and/or behavior" (3). It has been hypothesized that such compounds may elicit a variety of adverse effects in both humans and wildlife, including promotion of hormone-dependent cancers, reproductive tract disorders, and reduction in reproductive fitness (1, 4-10). Endocrine disruption by environmental contaminants has become a cause for concern (11), and as a result there is an urgent need to develop methods for monitoring EDCs in the environment (12). This need is underscored by recent legislation mandating that chemicals and 119 formulations be screened for potential estrogenic activity before they are manufactured or used in certain processes (Safe Drinking Water Act Amendments of 1995 - Bill Number S.1316; Food Quality Protection Act of 1996 - Bill Number PL. 104-170). Estrogenic and anti-estrogenic compounds are of particular concern. These compounds have the ability to mimic or block the functions of endogenous estrogen. Estrogenic effects have been documented in fish exposed to municipal wastewater treatment plant effluents (13-18). Some marine gastropods exhibited decreased reproductive success and developed imposex (females with male genitalia) when exposed to tributyltins (19, 20). Nonylphenol (NP), nonylphenol polyethoxylates (NPEs), octylphenol (OP), and synthetic and natural steroids are estrogenic organic constituents of many wastewater effluents (13, 21-24). These compounds were targeted in this investigation. Halogenated aromatic hydrocarbons (HAHs) and polycyclic aromatic hydrocarbons (PAHs) are classes of compounds that have been shown to exert adverse effects on aquatic organisms. Toxic effects associated with HAHs and PAHs include mortality, wasting syndrome, hepatotoxicity, immunotoxiclty, reproductive impairment, and carcinogenicity (25-27). Many of these effects are the result of an aryl hydrocarbon receptor (AhR)-mediated mechanism (25). Additionally, HAHs and PAHs also modulate estrogen receptor (ER)-mediated activities. HAHs such as polychlorinated dibenzo-p-dioxins (PCDDs), polychlorinated dibenzofurans (PCDFs), and some polychlorinated biphenyls (PCBs), have been reported to cause anti-estrogenic effects in vitro 120 (28, 29). PAHs have been associated with both estrogenic and anti-estrogenic responses in vitro (30, 31). As a result, both AhR- and ER-mediated effects of HAHs and PAHs are of concern when screening surface water samples for potential (anti)estrogenic compounds. Although instrumental analyses can be used to detect and quantify known (anti)estrogenic compounds associated with wastewater treatment plant (WWT P) effluents, in vitro bioassays provide useful information that can complement instrumental analyses to provide a more comprehensive characterization of a sample’s potential to modulate estrogenic responses. In vitro bioassays measure the ability of a sample to alter a mechanism-specific biochemical or physiological process. As a result, they provide an indication of the biological relevance of a sample. Furthermore, this is accomplished in a manner that integrates the overall potency of complex mixtures. Thus, in vitro bioassays can account for both unknown compounds and potential non-additive interactions among compounds. This study involved use of in vitro bioassays to screen samples for compounds able to modulate ER- or AhR-mediated gene expression. Successive iterations of sample fractionation coupled with bioassay and instrumental analysis were employed to identify specific compounds associated with in vitro responses. Furthermore, bioassay-derived estimates of estrogenic activity were compared to estimates based on analytical concentrations of known estrogen agonists and their relative potencies (REPs) in a mass balance analysis (32, 33). Mass balance analysis was used to generate hypotheses regarding whether the known composition of a sample could account 121 for the response observed or whether unknown compounds or non-additive interactions between components of the sample influenced the activity. Thus, instrumental analyses and in vitro bioassays were applied, in a complementary manner, to screen wastewater outfalls for compounds able to elicit estrogenic or dioxin-like biological responses and generate hypotheses regarding the probable cause of the activity observed. MATERIALS AND METHODS Sample Collection and Fractionation. A detailed description of the analytical methodology used in this study was published previously (34). Briefly, 5 L water samples were extracted at each field site using solid-phase extraction (SPE) EmporeTM disks. Organic extracts from these SPE disks were separated into 3 fractions based on polarity using normal-phase high-pressure liquid chromatography (NP-HPLC) (Figure 1). In some cases, EDCs of interest were isolated further from F2 and F3 by fractionating these again with reverse-phase HPLC (RP-HPLC), with fractions collected approximately every 3 min (Figure 2). Chromatographic conditions for RP-HPLC fractionation were described previously (34). NP-HPLC and RP-HPLC separations were accomplished using silica and Cu; analytical columns, respectively. Cell Culture and Bioassay. An MCF-7 human breast carcinoma cell line, stably transfected with an estrogen receptor-controlled luciferase reporter gene 122 construct (MVLN or MCF-7-luc cells), was developed and characterized by Dr. M.D. Pons, Institut National de la Sante et de la Recherche Medicale (35). MVLN cells were cultured in 75-cm2 disposable polyethylene tissue culture flasks (Corning, Corning NY, USA) containing 20-25 mL of Dulbecco’s Modified Eagle Medium (DMEM) with Hams F-12 nutrient mixture (Sigma D-2906; St Louis, MO, USA) supplemented with 10% defined fetal bovine serum (Hyclone, Logan UT, USA), 27.3 I.U. insulin (Sigma I-1882) IL, and 1.0 mM sodium pyruvate (Sigma). H4llE-Iuc cells are rat hepatoma cells containing a Iuciferase reporter gene under control of dioxin responsive enhancers (DREs) (36). They were cultured in 100- mm disposable petri plates (Corning) containing 12 mL Dulbecco’s Modified Eagle Medium (DMEM; Sigma D2902). Cells were incubated at 37 °C in a humidified 95:5 air:COz atmosphere and were passaged when plates became confluent (generally every 5 d). New cultures were started from frozen stocks after 30 passages. In preparation for bioassay, cells were trypsinized from flasks or plates containing 80-100% confluent cell growths. The number of cells per mL was determined microscopically by use of a hemacytometer. MVLN cells were diluted in stripped media [DMEM with Hams F-12 nutrient mixture, supplemented with 10% dextran-coated charcoal filtered fetal bovine serum (Hyclone), 27.3 I.U. insulin (Sigma I-1882) IL, and 1.0 mM sodium pyruvate (Sigma)] to a concentration of approximately 1.5 x 105 cells/mL. H4llE-luc cells were diluted to the same concentration in DMEM. Cells were seeded into the 60 interior wells of 96-well flat bottom microplates (Packard Instruments 6005181; Meriden CT, 123 USA) at 125 pL per well (15-20,000 cells per well) using a repeating pipettor. To assure homogeneity, the cell solution was vortexed continuously during seeding. The 36 exterior wells of each microplate were filled with 125 pL of media. Cells were dosed after an overnight incubation to allow for cell attachment. Extracts or fractions were dissolved in the appropriate culture media (stripped media for MVLN, DMEM for H4llE-luc) to yield a final concentration of 1.0% extract. A 3- fold dilution of each extract or fraction was also prepared yielding a concentration of 0.33% extract. Test wells were dosed with 125 pL 1.0% or 0.33% extract in media to yield final in-well concentrations of 0.50% and 0.165% extract. Solvent control wells were dosed with 125 pL of media spiked with 1.0% of the appropriate solvent to yield a final in-well concentration of 0.50% solvent. Blank wells received 125 pL of the appropriate media. Each plate tested included a minimum of three solvent control wells, three blank wells, and three replicates of each fraction tested (at both 0.50% and 0.165% levels). Dosed cells were exposed for 72 h at standard incubation conditions. Each test plate was inspected visually and differences in cell numbers and condition relative to control wells and conditions normally observed during routine culturing were noted for each well. Culture medium was then removed and each well was rinsed twice with phosphate buffered saline (PBS) supplemented with 1.0 mM Ca2+ and Mg2+ using an eight channel vacuum manifold. Plates were inspected for cell loss during washing. Following inspection, 75 pL PBS supplemented with Ca2+ and Mg2+ was added to each well, followed by 75 pL Luc-liteTM reagent (Packard Instruments). Each plate was 124 incubated for 10 min at 30 °C then scanned with an ML 3000 microplate reading luminometer (Dynatech Laboratories, Chantilly, VA, USA). Following the luminomter scan, 125 pL 1.08 mM fluorescamine (Sigma) in acetonitrile (ACN) was added to each well and plates were assayed for protein after a 15 min incubation at room temperature (37). Plates were scanned using a Cytofluor 2300 (excitation 400 nm, emission 460 nm), and responses were compared to a standard curve consisting of 6 concentrations of bovine serum albumin (BSA) (Sigma) ranging from 50-1.5 pg per well. All data were collected electronically and imported into a spreadsheet (Excel 7.0, Microsoft Inc., Seattle, WA) for data analysis. Protein content per well was calculated by regression against the BSA standard curve. Protein data were used as an index of cell number to detect outliers that were not apparent by visual inspection. Relative luminescence units (RLU) were not adjusted for protein. Sample responses in RLU were expressed as a percentage of the mean maximum response observed for E2 or 2,3,7,8-tetrachlorodibenzo-p- dioxin (T CDD) standard curves developed on the same day (% TCDD-max and % E2-max, respectively). The greatest response of the two extract dilutions was reported. However, for each significant response, the greatest response came from the greater extract concentration (0.5% in the well). In addition, for some samples, the dose-response relationship was used to determine the total estrogenic or dioxin-like activity of the samples. Activities derived from the bioassay were designated E2-EQ and TCDD-EQ for estrogenic and dioxin-like activities, respectively. 125 In addition to determining the total activity of extracts by the above- mentioned bioanalytical techniques, for some samples the total activity was estimated by correcting the concentrations of individual compounds present in the extract for their relative potencies. This was accomplished by multiplying the concentration of each compound by the corresponding toxic equivalency factor (TEF) or relative potency factor (REP) and summing the products. The calculated concentrations were referred to as EEQ and TEQ, respectively. A mass balance of activities determined by bioanalytical techniques and those calculated from the measured concentrations of individual compounds could be calculated. This was done to determine whether there were unidentified compounds contributing to the total activities measured in the bioassays. To make an accurate comparison, the potential for antagonistic and synergistic interactions needed to be addressed. This was done by the fine fractionation of samples, by which compounds known to be antagonistic to the measurement of EEO were separated from the active compounds. RESULTS AND DISCUSSION Dioxin-Iike Activity. None of the extracts or fractions tested in this study induced a significant response in the H4llE-luc bioassay (Figure 3). This was not surprising, because water concentrations of dioxin-like compounds were expected to be very small. No PCBs or PCDDs/DFs were detected in any of the extracts analyzed in this study. PAHs were detected at each site; however, 126 summed concentrations did not exceed 40 ng/L at any site. Of the OC pesticides analyzed, only hexachlorocyclohexanes (HCHs) were observed and only in the Las Vegas Bay sites. HCH concentrations were less than 20 nglL in all samples that contained detectable concentrations. The method detection limit (MDL) for the H4llE-luc bioassay was around 0.2 frnol TCDD-EQlwell (32). Assuming there were no antagonistic interactions among components of the sample, the H4IIE- luc results suggest that the water samples analyzed contained less than 10 pg TCDD-EQ/L. Estrogen-like Activity. Whole extracts and corresponding fractions were screened for estrogenic activity using the MVLN in vitro bioassay. Non-polar compounds such as PAHs, PCBs, and most organochlorine (OC) pesticides, if present, would have been contained in F1 (Figure 1) (34). Certain OC pesticides and PAHs such as chrysene, benz[a]anthracene, and benzo[a]pyrene have been reported to cause weak estrogenic responses in vitro (31, 38). None of the F1 samples; however, induced a significant response in the MVLN assay (Figures 4 - 7). Based on the MDL for E2 in the MVLN assay, concentrations of estrogenic compounds present in F1 contributed less than 110 pg E2-EQIL. These results support the conclusion that concentrations of non-polar estrogenic compounds in the surface waters and effluents examined were small. Weak estrogen agonists such as NP and OP were present in F2 (Figure 1). Concentrations of E2-EQ in F2 were less than the MDL, despite the confirmed presence of NP and OP (Figures 4 — 7). These results suggest that 127 the compounds present in F2 contributed less than 110 pg EEQ/L. Estrogenic potencies of NP and OP, relative to E2, for luciferase induction in MVLN cells have been reported to be 1.25 x 10'5 and 1.9 x 10'5 for NP and OP, respectively (33). Concentrations of NP and OP present in the sample extracts (Tables 1 & 2) were used to calculate EEQ. NP and OP were estimated to contribute less than 15 pg E2-EQ/L for fifteen of the sixteen samples tested. Thus, the lack of significant induction of the MVLN cells was consistent with the known concentrations of estrogenic alkylphenols present in the F2 samples. The BV- effluent sample extract contained approximately 380 pg EEQ/L, which would correspond to a dose of approximately 8.5 fmol E2-EQ/well in the MVLN bioassay. Based on regression against an E2 standard curve, this dose could have elicited a response as great as 53% E2-max. The F2 extract of the BV- effluent; however, failed to induce a significant MVLN response (Figure 4). This suggests that F2 of the BV—effluent sample might have contained interfering compounds that suppressed the estrogenic activity of NP and OP. Therefore, the F2 extract of the BV-effluent was further fractionated and E2-EQ measured (Figure 8). No significant estrogen-like responses were observed in this fine fractionation. The same fine fractionation was applied to the F2 extract from Black Lagoon (Figure 5). Once again, no significant estrogen-like activity was observed. In general, however, concentrations of E2-EQ in F2 extracts were in agreement with the EEO based on the known concentrations and relative potencies of these compounds. 128 F3 extracts were the most active of the fractions tested. Six of the sixteen F3 samples elicited significant estrogenic activity in the MVLN bioassay (Figures 4 — 7). The greatest magnitudes of response for F3 extracts, approximately 80% E2-max, were observed for samples collected from the LV Wash and LV Bay in April 1997 (Figure 6). However, samples from these sites collected in September 1997 did not contain significant concentrations of E2-EQ (Figure 7). The samples collected in September 1997 were after a large storm event, which diluted the wastewater entering the LV Wash and LV Bay (34). The estrogenic potency of EE2 relative to E2 for Iuciferase induction in the MVLN assay was previously reported to be approximately 0.1 (33). Based on the concentrations of E2 and EE2, and their corresponding relative potencies, samples collected from LV Wash and LV Bay in April 1997 were estimated to contain 2180 and 1810 pg E2/EE2-derived EEQIL, respectively. These concentrations should have yielded doses of approximately 50 and 41 frnol E2- EQ/well in the MVLN bioassay. Based on regression against an E2 standard curve, such doses would be expected to yield responses of approximately 92% and 88% E2-max, respectively. Given the potential range of uncertainty associated with the bioassay responses and the relative potency of EE2, the responses observed for the samples collected from LV Wash and LV Bay in April 1997 were not significantly different from predicted responses. Thus, the known E2 and EE2 composition of F3 of the samples collected from LV Wash and LV Bay appeared to account for all the estrogenic activity observed. Further fractionation of the F3 extracts from LV Wash and LV Bay revealed that all of the 129 estrogenicity was associated with the fine fractions (F Fs) 3 and 4, which equates roughly to the retention times of E2 and EE2 (Figures 10 & 11). FFs 3 and 4 from the F3 extract of the LV Wash sample were collected, combined, and fractionated again by RP-HPLC using a slower flow rate and solvent gradient to separate E2 and EE2 (Figure 12). The estrogen-like bioactivity was isolated to the fractions where E2 and EE2 elute, and the magnitude of induction was similar to that predicted from the E2-EQs measured in the MVLN bioassay (Tables 1 & 2). The greater estrogenic activity of the water extracts from LV Wash and LV Bay was most likely due to the fact that the WWTPs discharging into the Las Vegas Wash represent a larger population of humans. Significant estrogenic activity also was associated with F3 extracts of water from three locations on the Trenton Channel of the Detroit River (B. Lagoon, Chem, and WWTP) (Figure 5), and BV—effluent (Figure 4). Concentrations of E2-EQ in extracts from B. Lagoon, Chem., WWTP, and BV- effluent contained 1060, 730, 850, and 2980 pg E2/EE2-derived EEQ/L, respectively. Based on regression against an E2 standard curve, these concentrations of E2-EQ were predicted to yield responses of 76%, 67%, 71%, and 99% E2-max, respectively. Observed MVLN responses for these F3 samples were; however, less than predicted (Figures 4 & 5). Further fractionation and bioanalysis of F3 extracts from BV-effluent and B. Lagoon indicated that all observed estrogenicity is contained in FFs 3 and 4. However, the magnitude of induction of the FFs was significantly different from that of the corresponding total F3 extract. This suggests that interfering and/or anti- 130 estrogenic compounds present in F3 might have modulated the activity of the known estrogen agonists. The potential presence of interfering and/or anti-estrogenic (estrogen antagonists) compounds also was suggested by the lack of significant response for several samples. Based on concentrations of E2 and EE2, six additional F3 samples should have elicited significant responses in the MVLN bioassay. Concentrations of EEQs calculated from concentrations of EE2 and E2 in samples collected from LV Bay (Sept. 1997), LV Marina, M. Creek, BV-upstream, MA-effluent, and ER-effluent were estimated to range from 170-850 pg EEQIL. Regression against an E2 standard curve would indicate, predicted responses of 35-71% E2-max in the MVLN assay for these samples. Thus, the responses were less than predicted for these samples. The reason for this observation is unknown at this time. Concentrations of EEO in unfractionated (total) extracts were similar to those for F3 fractions. Samples for which F3 elicited a significant response also elicited a significant whole extract response (Figures 4 - 6). In five of the six cases, the whole extract response was slightly lesser than the response elicited by F3. This suggests that F1 and F2 may have contained some interfering or anti-estrogenic substances that modulated the activity of the known ER agonists in the samples. However, no significant anti-estrogenic responses were observed (Figures 4 — 6). Because the decreases were slight; however, the results suggest that the bulk of potential interfering (antagonistic) compounds were present in F3. The extract of BV-effluent was the only sample for which the 131 whole-extract response was greater than the corresponding F3 response. It was also the only sample for which the concentrations of NP and OP in F2 were predicted to yield significant estrogenic activity. NP and OP accounted for 11% of the total E2-EQ calculated for the BV-effluent extract. Thus, although F2 of the BV-effluent sample failed to elicit a significant response, NP and OP may have contributed to the response of the whole extract, such that the total extract response was greater than the F3 response. In general, however, NP and OP accounted for less than 1% of the total concentrations of sample E2-EQs present in samples. SUMMARY The mass balance calculations based on instrumental analyses and bioassay-directed fractionation support the conclusion that E2 and EE2 were the dominant environmental estrogens in the samples. Also, interfering or anti- estrogenic compounds present in samples (predominantly in F3) may have acted to mask or dampen the activity of the known ER agonists in the MVLN bioassay. All observed responses in the MVLN bioassay were either less than or approximately equal to responses predicted based on the concentrations and relative potencies of known ER agonists measured by instrumental techniques. NP and OP generally contributed less than 1% of the total E2-EQs. Furthermore, sample fractions containing NP and OP did not elicit significant activity. For most samples, fractions containing E2 and EE2 elicited responses slightly greater than 132 the responses of the corresponding whole extracts. Thus, among the ER agonists detected in the samples, E2 and EE2 appear to be responsible for the bulk of the activity. The fact that observed responses were generally lower than predicted suggests the presence of interfering compounds. Concentrations of E2-EQ in whole extracts were only slightly less than those from F3 extracts. This suggests that the interfering compounds may have been present. Because there were few instances where the concentrations of E2-EQ were greater than the concentrations of EEO in any of the fractions or in the subsequent fine fractions, the known composition can account for the magnitude of response observed. It is unlikely that there were additional ER-active compounds of significant concentration that were not identified. There are insufficient data to explain differences in bioactivity among the locations investigated. It should be noted that samples from the Trenton Channel of the Detroit River in Michigan received less effluent from municipal WWl'Ps relative to the volume of the receiving water compared to the other sites. Also, the population served by the WWI'Ps varied greatly among sites. Further investigations would be necessary to determine the actual loading of bioactive compounds as a function of population density. Without knowing the available fractions and bioaccumulation potential of the various compounds and dose- response relationships for target species, it is not possible to predict the potential effects of the observed concentrations of environmental estrogens on biota. REFERENCES 133 (1) (2) (3) (4) (5) (6) (7) (8) (9) Kendall, R. J.; Dickerson, R. L.; Giesy, J. P.; Suk, W. A. Principles and Processes for Evaluating Endocrine Disruptors in Wildlife; SETAC Press: Pensacola, FL, 1998. Ankley, G.; Mihaich, E.; Stahl, R.; Tillitt, D.; Colborn, T.; McMaster, S.; Miller, R.; Bantle, J.; Campbell, P.; Denslow, N.; Dickerson, R.; Folmar, L.; Fry, M.; Giesy, J.; Gray, E.; Guiney, P.; Hutchinson, T.; Kennedy, 8.; Kramer, V.; LeBlanc, G.; Mayes, M.; Nimrod, A.; Patino, R.; Peterson, R.; Purdy, R.; Ringer, R.; Thomas, P.; Touart, L.; Van Der Kraak, G.; Zacharewski, T. Environ. Toxicol. Chem. 1998, 17, 68-87. US. EPA, Special report on environmental endocrine disruption: an effects assessment and analysis; Office of Research and Development. Washington, DC. February 1997. EPA/630/R-96I012 1997. Safe, S. H. Pharrnacol. Ther. 1995, 67, 247-281. Ahlborg, U. G.; Lipworth, L.; Titus-Ernustoff, L.; Hsieh, C.-C.; Hanberg, A.; Baron, J.; Trichopoulos, D.; Adami, H. 0. Crit. Rev. Toxicol. 1995, 25, 463-531. Sharpe, R. M.; Skakkebaek, N. E. Lancet 1993, 341, 1392-1395. Carlsen, E.; Giwercman, A.; Keiding, N.; Skakkebaek, N. E. Environ. Health Perspect. 1995, 130, 137-139. Horwitz, K. B.; Jackson, T. A.; Rain, D. L.; Richer, J. K.; Takimoto, G. S.; Tung, L. Mol. Endocrinol. 1996, 10, 1167-1177. Katzenellenbogen, J. A. Environ. Health Perspect. 1995, 103, 99-101. 134 (10) (11) (12) (13) (14) (15) (15) (17) (18) (19) Gillesby, B. E.; Zacharewski, T. R. Environ. Toxicol. Chem. 1998, 17, 3- 14. Colborn, T. Environ. Toxicol. Chem. 1998, 17, 1-2. Kavlock, R. J.; Daston, G. P.; DeRosa, C.; Fenner—Crisp, P.; Gray, L. E.; Kaattari, S.; Lucier, G.; Luster, M.; Mac, M. J.; Maczka, C.; Miller, R.; Moore, J.; Rolland, R.; Scott, G.; Sheehan, D. M.; Sinks, T.; Tilson, H. A. Environ. Health Perspect. 1996, 104, 715-740. White, R.; Jobling, S.; Hoare, S. A.; Sumpter, J. R; Parker, M. G. Endo. 1994, 135, 175-182. Purdom, C. E.; Hardiman, P. A.; Bye, V. J.; Eno, N. C.; Tyler, C. R.; Sumpter, J. P. Chem. Ecol. 1994, 8, 275-285. Harries, J. E.; Sheahan, D. A.; Jobling, S.; Matthiessen, P.; Neall, P.; Sumpter, J. P.; Tylor, T.; Zaman, N. Environ. Toxicol. Chem. 1997, 16, 534-542. Jobling, S.; Noylan, M.; Tyler, C. R.; Brighty, G.; Sumpter, J. P. Environ. Sci. Technol. 1998, 32, 2498-2506. Folmar, L. C.; Denslow, N. D.; Rao, V.; Chow, M.; Crain, D. A.; Enblom, J.; Marcino, J.; Guillette Jr., L. J. Environ. Health Perspect. 1996, 104, 1096- 1101. Bevans, H. E.; Goodbred, S. L.; Miesner, J. F.; Watkins, S. A.; Gross, T. S.; Denslow, N. D.; Schoeb, T. Water-Resources Investigations Report 96-4266, 1996. Gibbs, P. E.; Bryan, G. W.; Pascoe, P. L. Mar. Environ. Res. 1991, 79-87. 135 (20) (21) (22) (23) (24) (25) (25) (27) (23) (29) (30) (31) Bryan, G. W.; Gibbs, P. E.; Burt, G. R. J. Mar. Biol. Ass. UK 1988, 733- 744. Snyder, S. A.; Snyder, E.; Villeneuve, D.; Kurunthachalam, K.; Villalobos, A.; Blankenship, A.; Giesy, J. In Analysis of Environmental Endocrine Dismptors; Keith, L. H., Jones-Lepp, T. L., Needham, L. L., Eds.; American Chemical Society: Washington,DC, 2000, pp 73-95. Belfroid, A. C.; Van der Horst, A.; Vethaak, A. D.; Schafer, A. J.; Rijs, G. B. J.; Wegener, J.; Cofino, W. P. Sci. Total Environ. 1999, 225, 101-108. Bennie, D. T. Water Qual. Res. J. Canada 1999, 34, 79-122. Naylor, C. G.; Mieure, J. R; Adams, W. J.; Weeks, J. A.; Castaldi, F. J.; Ogle, L. D.; Romano, R. R. JAOCS 1992, 69, 695-703. Poland, A.; Knutson, J. C. Ann. Rev. Phamacol. Toxicol. 1982, 22, 517- 554. Neff, J. M. Polycyclic Aromatic Hydrocarbons in the Aquatic Environment: Sources, Fates, and Biological Effects; Applied Science: London, 1979. Schecter, A. Dioxins and Health; Plenum Press: New York, 1994, pp 710. Krishnan, V.; Safe, S. Toxicol. Appl. Phannacol. 1993, 120, 55-61. Anderson, M. J.; Miller, M. R.; Hinton, D. E. Aquat. Toxicol. 1996, 34, 327- 350. Tran, D. Q.; lde, C. F.; McLachlan, J. A.; Arnold, S. F. Biophys. Res. Commun. 1996, 229, 102-108. Clemons, J. H.; Allan, L. M.; Marvin, C. H.; Wu, Z.; McCarry, B. E.; Zacharewski, T. R. Environ. Sci. Technol. 1998, 32, 1853-1860. 136 (32) (33) (34) (35) (36) (37) (38) Sanderson, J. T.; Giesy, J. P. In Encyclopedia of Environmental Analysis and Remediation; Meyers, R. A., Ed.; John Wiley and Sons: New York, 1998, pp 5272-5297. Villeneuve, D. L.; Blankenship, A. L.; Giesy, J. P. In Toxicant-Receptor Interactions; Denison, M. S., Helferich, W. G., Eds.; Taylor & Francis, 1998, pp 69-97. Snyder, S. A.; Keith, T. L.; Verbrugge, D. A.; Snyder, E. M.; Gross, T. S.; Kannan, K.; Giesy, J. P. Environ. Sci. Technol. 1999, 33, 2814-2820. Pons, M.; Gagne, 0.; Nicolas, J. C.; Mehtali, M. Biotechniques 1990, 9, 450-459. Sanderson, J. T.; Aarts, J. M.; Brouwer, A.; Froese, K. L.; Denison, M. S.; Giesy, J. P. Toxicol. Appl. Pharmacol. 1996, 137, 316-325. Kennedy, S. W.; Jones, S. P. Anal. Biochem. 1994, 222, 217-223. Soto, A. M.; Chung, K. L.; Sonnenschein, C. Environ. Health Perspect. 1994, 102, 380-383. 137 Table 1. Extract Concentrations Location Date NP Lake Mead LV Wash 4/30/97 4560 LV Bay 4/30/97 3000 9/5/97 640 LV Marina 9/5/97 ND Saddle Island 4/30/97 ND Callville Bay 9/5/97 ND Trenton Channel WWTP 8l30/97 1916 Chem. 8l30/97 3450 B. Lagoon 8l30/97 3740 M. Creek 8l30/97 4740 WWTPs BV-upstream 1 0/8/97 ND BV-effluent 10l8l97 148000 MA-upstream 1 0/8/97 N D MA-effluent 1 0/8/97 2065 ER-upstream 1 0/8/97 ND ER-effluent 1 0/8/97 680 OP 172 108 ND ND ND ND 20 60 264 324 ND 2350 ND 54 ND ND NPE 35960 19400 12710 ND ND ND 21600 29200 34700 71260 ND 1 160000 ND 19400 ND ND ND = not detectable NA = not applicable NP = nonylphenol OP = octylphenol NPE = nonylphenolethoxylate E2 = 178-estradiol EE2 = ethynylestradiol 138 E2 1 0700 8840 752 1080 ND ND 4260 3640 51 80 4244 2500 14600 ND 3620 ND 1 900 EE2 (ng/mL) (ng/mL) (ng/mL) (pg/mL) (pg/mL) 1920 2080 1012 ND ND ND ND ND 1440 ND ND 3036 ND 1430 ND ND Table 2. E2-EQs and Predicted Responses Location Date NPIOP- Predicted E2IEE2- Predicted E2-EQa Responseb E2-EQ° Responsed (PS/L) (0%) Lake Mead LV Wash 4/30/97 12. 1 0 2180 92 LV Bay 4/30/97 7.91 0 1810 88 9/5/97 1.60 0 171 35 LV Marina 9/5/97 NA NA 216 41 Saddle Island 4/30/97 NA NA NA NA Callville Bay 9/5/97 NA NA NA NA Trenton Channel WWTP 8/30/97 4.87 0 852. 71 Chem. 8l30/97 8.85 0 728 68 B. Lagoon 8130/97 10.4 0 1 060 76 M. Creek 8l30/97 13.1 0 849 71 WWTPs BV-effluent 1 0/8/97 379 53 2980 99 BV-upstream 1 0/8/97 NA NA 500 59 MA-upstream 1 0/8/97 NA NA NA NA MA-effluent 1 0/8/97 5.41 0 753 68 ER-upstream 1 0/8/97 NA NA NA NA ER-effluent 1 0I8/97 1 .70 0 380 53 NA = not applicable 3 Nonylphenol and octylphenol-derived 17-B-estradiol equivalents. NPIOP-EZ-EQ = (NPNW. potency (REP)x Npmnmum) '1' (OPREP X opooncentratlonIo NPREP = 1.25 X 10.5. OPREP = 1.9 X 10.5. A REP estimate was not available for NPE, therefore it was not considered when deriving E2-EQ estimates. Values presented were back-calculated to water concentrations (pg E2-EQI L H20). b MVLN bioassay response magnitudes predicted based on regression of NPIOP-derived E2-EQ against a 17-B-estradiol standard curve. Units are %-E2-max. ° Estradiol and ethynylestradiol-derived 17-B-estradiol equivalents. E2IEE2-E2-EQ = (EZREp x EZWO") + (EEZREP X EEanmmjon). EZREP = 1.0. EEZREP = 0..10 values presented were back-calculated to water concentrations (pg E2-EQ/ L H20). d MVLN bioassay response magnitudes predicted based on regression of E2IEE2-derived E2- EQ against a 17-B-estradiol standard curve. Units are %-E2.-max. Note: Total E2-EQ = NPIOP-EZ-EQ + E2IEE2-E2-EQ. Predicted bioassay response magnitudes are not additive. 139 5L in situ SPE extraction l Vacuum/Solvent Extraction l Concentrate to 1 mL l Normal-phase Fractionation in F2 t Non-polar PAHs and HAHs Semi-Polar OP and NP F3 Polar NPE, E2, and EE2 l Reverse-phase Fractionation Figure 1. Flow diagram of analytical method 140 1 2 3 4 5 6 7 8 9 10 OP F2 NP 5152 F3 52 NPE1 I17 3. .. .1,o .fi .. ..20.. .. - Tlme (min) Figure 2. Reverse-phase HPLC fine fractionation 141 % TCDD Max 100. I Total 80.0 _ . F1 50.0 CI F2 _ 5 F3 40.0 20.0 -20. MA LV-Bay B. Lagoon Blank Site Figure 3. Dioxin-Iike bioactivity (various sites): Significance bars represent 3x SD of the background signal 142 80.0 I roan a I F1 60.0 CI F2 _ I F3 40.0 20.0 0.0 -20.0 MA-up MA BV-up ev ER-up ER Blank Site Figure 4. Estrogen-like bioactivity — Michigan WWTPs (October 1997): Significance bars represent 3x SD of the background signal 143 % E2 Max 1 00.0 80.0 I roar — fl F1 60.0 CI F2 F— m F3 40.0 20.0 0.0 -20.0 B.Lagoon Chem. M.Creek WWTP Blank Site Figure 5. Estrogen-like bioactivity — Trenton Channel (August 1997): Significance bars represent 3x SD of the background signal 144 % E2 Max 100.0 80.0 60.0 40.0 20.0 0.0 -20.0 LV LV Saddle Blank Wash Bay Island * Site Figure 6. Estrogen-like bioactivity — Lake Mead (April 1997): Significance bars represent 3x SD of the background signal 145 80.0 I Total ’— n F1 50.0 5 F2 _ 5 F3 40.0 LV LV Callville Blank Bay Marina Bay Site Figure 7. Estrogen-like bioactivity - Lake Mead (September 1997): Significance bars represent 3x SD of the background signal 146 %E2Max d e 8 3 S 8 8 iv a Figure 8. Fine fractionation and corresponding estrogen-like bioactivity of WWTP BV-effluent F2 extract 147 °/o E2 Max nb OP 0” 90 N15 00 r'o c Figure 9. Fine fractionation and corresponding estrogen-like bioactivity of Black Lagoon, Trenton Channel, F2 extract 148 % E2 Max Figure 10. Fine fractionation and corresponding estrogen-like bioactivity of LV Wash, Lake Mead (April 1997), F3 extract 149 % E2 Max NPE — 10 100 40 20 Figure 11. Fine fractionation and corresponding estrogen-like bioactivity l i of LV Bay, Lake Mead (April 1997), F3 extract 150 % E2 Max EE2 E2 100 I N-tsoaco Booooo Figure 12. Further fractionation and corresponding estrogen-like bioactivity of LV Wash, Lake Mead (April 1997), F3 extract 151 °/o E2 Max Figure 13. Fine fractionation and corresponding estrogen-like bioactivity of VWVTP BV-effluent, F3 extract 152 % E2 Max Figure 14. Fine fractionation and corresponding estrogen-like bioactivity of Black Lagoon, Trenton Channel, F3 extract 153 Chapter 6 BIOACTIVE ORGANIC COMPOUNDS IN THE WATERS OF LAKE MEAD, NEVADA, USA (To be submitted to Environmental Science and Technology) ABSTRACT Several recent reports have indicated a flux of organic compounds flowing into the Boulder Basin of Lake Mead, Nevada from the Las Vegas Wash. The primary objective of this study was to screen for a wide range of organic contaminants from 5 locations in Lake Mead between May 1998 and June 1999. Certain pesticides, polychlorinated biphenyls (PCBs), alkylphenols, polycyclic aromatic hydrocarbons (PAHs), and steroids were target compounds. Mass spectrometry was utilized to screen for previously unreported compounds. Samples of 100 L of filtered water were collected in Teflon-lined containers and were extracted using large-scale solid-phase extraction. Following a series of elution and extraction procedures, resulting extracts were fractionated into 3 polarity classes. Each fraction was analyzed by gas chromatography with mass selective detection (GCIMS). DDT, DDE, and DDD (collectively, DDTs) and four isomers of hexachlorocyclohexane (HCH) were detected at some sites, with concentrations ranging from less than detection to 28 nglL. Octylphenol, nonylphenol, nonylphenol monoethoxylate, and nonylphenol diethoxylate were 154 detected in some samples, with concentrations ranging from less than detection to 1500 ngIL. Caffeine, nicotine, oxybenzone, N,N-diethyl-meta-toluamide (DEET), phosphate-based flame-retardants, and several pharmaceuticals were detected in some samples with concentrations ranging from less than detection to 360 nglL. The identities of several of these previously unreported compounds were verified using high-resolution mass spectrometry. PAHs were detected at each site and did not vary significantly among the sites in concentration or in pattern. Total PAH concentrations ranged from 5 to 78 ng/L. Polychlorinated biphenyls, triazine herbicides, and steroids were not detected in any of the samples. INTRODUCTION With approximately 36.7 x 109 m3 of storage capacity, Lake Mead is the largest reservoir on the Colorado River system and was formed with the completion of Hoover Dam in 1935. The lake encompasses approximately 593 km2 of surface area and extends 185 km east from Hoover Dam, up the old course of the Colorado River. The majority of flow into Lake Mead (97%) comes from the Colorado River. Approximately 1.5% of the flow originates from the Virgin and Muddy Rivers, which discharge into the Overton Arm of Lake Mead, and 1.5% comes from the Las Vegas Wash, which discharges into the Boulder Basin (Figure 1). Approximately 22 million people depend on water from Lake Mead and the lower Colorado River for domestic and agricultural usage (1). 155 Las Vegas Wash is a 19 km channel that drains the entire 4100 km2 Las Vegas Valley Hydrographic Basin. Sources of flow to the wash include storm water, urban runoff, shallow groundwater, and tertiary treated wastewater. The average daily flow in Las Vegas Wash is approximately 580 million L per day (excluding storm water). The drinking water intakes for Southern Nevada lie approximately 10 km down stream of the Las Vegas Wash confluence. The Las Vegas Wash provides significant nutrient and particulate loading into the Las Vegas Bay (1-3). This increased nutrient loading results in frequent eutrophication of the Las Vegas Bay. Secchi readings of less than 0.5 m and chlorophyll a concentrations of greater than 300 mg/m3 have been reported (1). Water from the Las Vegas Wash is warmer and has greater conductivity, turbidity, and suspended and dissolved solids than main body Lake Mead water. Despite being warmer, Las Vegas Wash water has a greater density, which results in an underflow that extends until density equilibrium is met. This intrusion then can form an interflow above the hypolimnion. At certain times of the year, the intrusion of the Las Vegas Wash can be detected at various depths over the 16 km to Hoover Dam (1). In 1996, the US. Geological Survey (USGS) reported endocrine disruptive type effects in feral carp from the Las Vegas Wash and Bay. These carp had significantly different sex steroid and plasma vitellogenin levels than carp collected from Callville Bay (reference site) in Lake Mead (4). Furthermore, concentrations of some synthetic organic chemicals were found to be greater in water, sediment, and fish tissues from the Las Vegas Wash and Bay as 156 compared to Callville Bay (4). The presence of alkylphenols and natural and synthetic estrogen also has been reported in the Las Vegas Wash and Bay (5). In addition, in vitro bioassays were used to screen extracts of Lake Mead water samples for estrogenicity. This study indicated that natural and synthetic estrogen were the most potent xenoestrogens present in Las Vegas Wash and Bay water extracts (6). These reports indicate an urgent need to identify organic compounds present in the Las Vegas Bay that may have an impact on the aquatic organisms which reside there. In this report we describe a novel method for the sensitive detection of several classes of organic compounds. The basic methodology is a modified version of that reported previously (5). Organic compounds from a 100 L sample of water were concentrated using solid-phase extraction (Figure 2). Compounds reported previously and known to induce endocrine disruptive type effects in certain aquatic organisms were targeted. These include organochlorine (OC) pesticides, polycyclic aromatic hydrocarbons (PAHs), polychlorinated biphenyls (PCBs), atrazine, simazine, alkylphenols (APs), 178-estradiol (E2), and ethynylestradiol (EE2). Mass spectrometry was used to quantify and identify these target compounds and to screen for previously unreported organic compounds. Gas chromatography with high-resolution mass spectrometry (GCIHRMS) was used for structural verification for some compounds never before reported in US. waters. Mass Peak Profiling from Selected Ion Recording Data (MPPSIRD) and a Profile Generation Model (PGM) were used in conjunction with GC/HRMS to identify of trace concentrations of organic 157 compounds, including pharmaceuticals, in these complex extracts. The data presented for novel compounds is semi-quantitative due to the methodologies applied. EXPERIMENTAL SECTION Standards and Reagents. All standards and reagents used were of the highest purity commercially available. Standards (approximately 98% pure) of p- nonylphenol (NP) and p-tert octylphenol (OP) were obtained from Schenectady International (Freeport, TX). Standards of nonylphenol monoethoxylate (NPE1) and nonylphenol diethoxylate (NPEZ) were obtained from Huntsman Corporation (Austin, TX). Organochlorine (OC) pesticides, polycyclic aromatic hydrocarbons (PAHs), polychlorinated biphenyls (PCBs), and triazine herbicide standards were obtained from AccuStandard Inc. (New Haven, CT). All pharmaceutical and steroid standards were obtained from Sigma Chemical Company (St. Louis, MO) with the exception of fluoexetine (Prozacm), which was obtained from US. Pharrnacopeia (Rockville, MD). Bisphenol A (BPA), DEET, and 2-hydroxy-4- methoxybenzophenone (oxybenzone) were obtained from Aldrich (Milwaukee, WI). Pesticide residue grade methanol (MeOH), hexane, dichloromethane (DCM), iso-octane, and acetone were obtained from Burdick and Jackson (Muskegon, MI) or Sigma-Aldrich (Milwaukee, WI). Solvents were tested for interfering compounds by examining 150-fold concentrated solvents using gas chromatography with mass selective detection (GCIMS). Reagent water was first 158 purified by reverse osmosis (RO) followed by Nanopurem (Barnstead, Dubuque, IA) treatment. Glass fiber filters, fine grade (GF/F) with 0.7 pm nominal pore size (293 mm, Whatman, Hillsboro, OR), were heated at 450 °C for 4 h in aluminum foil prior to use. A 25% salt solution was made by dissolving 250 g sodium chloride (Baker, Phillipsburg, NJ) in 1 L reagent grade water and liquid-liquid extracted 3x with DCM to remove contaminants. Anhydrous granular sodium sulfate (EM Science, Gibbstown, NJ) was heated at 550 °C, overnight then stored at 125 °C until day of use. Sample Collection. Water samples were collected between May 1998 and June 1999 from 5 locations in Lake Mead, Nevada (Figure 1). Samples were taken from 3 m below the surface except at the LV1 site where samples also were collected at the zone of greatest conductivity as determined using an H20 SondeTM unit connected to a Surveyor 3TM data recorder (Hydrolab Corp., Austin, TX). Samples were pumped through one to five 30 cm glass fiber filters (GF/Fs) in a 293 mm stainless steel parallel filtration system (Pentaplate) using fluoropolymer (PFA) tubing (3I8” o.d.) and a peristaltic pump fitted with platinum- cured silicone peristaltic tubing at the field site. The Pentaplate was cleaned with water and solvents before and after each sample was filtered. At each sampling site, prior to collecting the samples, the apparatus was purged with sample water for 10 min by bypassing the filter holder. At sites with predicted concentration gradients, samples were taken sequentially from the least to greatest 159 concentrations. The initial 4 L portion of filtered sample was discarded, and the remaining sample was collected in a custom-built clean TeflonTM bag (Berghof America, Concorde, CA) lining a 113 L high-density polyethylene tank. Once the TeflonTM bag was filled, the tank was transported to the laboratory. Field blanks were taken with each sampling event by passing 100 L of NanopureTM water through the system as previously described. A column blank was processed with each batch of columns extracted by extracting a prepared SPE column through which no sample or blank water had passed. Laboratory blanks were performed on each day of extractions, and instrument blanks were conducted daily. Flow meter calibrations were verified prior to each sampling trip by comparing the reported totalizer output to the measured volume of water exiting the meter. Extraction. Immediately upon arrival in the laboratory, solid-phase extraction (SPE) began. Samples were extracted by pumping the samples through a stainless steel preparative high pressure liquid chromatography (HPLC) column (250 x 21.2 mm, Phenomenex, Torrance, CA). The HPLC column was dry- packed with approximately 55 g of pre-cleaned Bond Elut ENVTM (125 pm particle size, Varian, Harbor City, CA) SPE resin. These assembled SPE columns were additionally cleaned by pumping sequentially, for one hour each, acetone, 50% DCM in hexane, methanol, and water through the column at 5 mUmin. Columns were stored at 4 °C until use. 160 Samples were pumped through the SPE columns using a metering pump (Fluid Metering, Syosset, NY) at a flow rate of 0.1 lein until 100 L total volume had been extracted (approximately 16.7 h). An electronic flow meter (model 102- 6TP, McMillan, Georgetown, TX), calibrated to i 1% accuracy with National Institute of Standards and Technology (NIST) traceable standards, was installed downstream of the SPE column. This digital rate meter and totalizer was used to monitor flow rate and to record the total volume. Once the desired sample volume had been reached, the column was removed, capped, and stored at 4 °C until elution. The SPE column was eluted using a quaternary HPLC pump (Perkin Elmer, Series 410, Nonrvalk, CT) at 5 mUmin sequentially for 10 min with acetone, 40 min with 50% DCM in hexane, and an additional 10 min with acetone. The effluent of the SPE column was collected in a 1 L separatory funnel containing 100 mL 25% sodium chloride solution, and the resulting mixture was liquid-liquid extracted for 5 min, and the aqueous layer (bottom) was collected into a 1 L separatory funnel. The original aqueous layer was then extracted twice more with 50 mL DCM and the organic layer (bottom) combined with the original organic layer. The combined organic layers were then passed at a rate of 1 drop/sec through a funnel containing 100 g anhydrous sodium sulfate and collected in a RapidVapTM tube (Labconco, Kansas City, MO). Extracts were then concentrated to approximately 3 mL in a RapidVapTM N2 evaporation system (Labconco, Kansas City, M0) at 35 °C and vortex speed of 30. The sample was quantitatively transferred to a 15 mL centrifuge tube and concentrated to 1 mL 161 under a gentle stream of nitrogen at 30 °C. The extract was diluted to 4 mL with iso-octane, vortexed, and concentrated to 2.0 mL under nitrogen. Silica Gel Fractionation. One mL of iso-octane extract was fractionated using a gravimetric silica gel column prepared by placing a small plug of DCM extracted glass wool in a glass chromatography column followed by 10 g of acid-activated granular copper (10 - 40 mesh, Aldrich, Milwaukee, WI), 1 cm of sodium sulfate, 3 g of silica gel (60 — 100 mesh, Aldrich, Milwaukee, WI) in hexane, and an additional 1 cm of sodium sulfate. This column was rinsed with 25 mL of hexane. The sample was then loaded onto the column and the column drained until the sample was adsorbed into the top sodium sulfate layer. The first fraction (F 1) was eluted with 100 mL of 20% DCM in hexane at 1 drop/sec. A second and third fraction (F2 and F3) were eluted with 100 mL of 75% DCM in hexane and 100 mL 50% DCM in MeOH, respectively, at 1 drop/sec. F1 contained the least polar compounds, such as PAHs, PCBs, most organochlorine (OC) pesticides, and contaminant compounds leached from the SDB resin (Figure 2). F2 contained nonylphenol (NP), octylphenol (OP), and greater polarity OC pesticides (Figure 2). F3 contained the most polar compounds, including triazine herbicides, pharmaceuticals, DEET, oxybenzone, flame-retardants, steroids, and NPE1-2 (Figure 2). Each fraction was collected in a N2 Rapid VapTM tube (Labconco, Kansas City, MO) and concentrated using a N2 RapidVapTM (Labconco, Kansas City, MO) as previously described. The fractions were further concentrated under a gentle stream of nitrogen and solvent-exchanged to 162 1 mL iso-octane (F1 and F2) or MeOH (F3). No internal or surrogate standards were added to the samples because these could interfere with bioassays to be used for estrogenic and dioxin-like bioactivity. Identification and Quantitation. All compounds were identified and quantified using an HP 5890 series Il GC with an HP 5972 mass selective detector (MSD). A 30 m DB—5MS capillary column (0.25 mm l.D., 0.25 urn film, J&W Scientific, Folsom, CA) was used for PAH, OC pesticides, atrazine, simazine, and PCB analyses. An injection volume of 3 pL was used for these compounds. Temperature programming was optimized based on that provided in the J&W catalog (1998, J&W Scientific, Folsom, CA) for these analytes with the DB-5MS column. NP and OP were analyzed using the same GCIMS system, but using a 30 m DB-17MS capillary column (0.25 mm l.D., 0.25 um film, J&W Scientific, Folsom, CA). NPE122, pharmaceuticals, steroids, personal care products, and BPA were analyzed using the same GCIMS system described previously. These compounds were analyzed using the DB-5MS and DB-17MS described above, plus a 30 m Rtx-50 capillary column (0.25 mm ID, 0.25 pm film, Restek, Bellefonte, PA). Injection volumes of 2 uL were used for the analyses of these compounds. For all compounds quantitated, the MSD was operated in selected ion monitoring (SIM) mode with 2 -3 ions per compound (Tables 1 - 4). Identification was determined by proper retention time and ion ratio. Quantitation was performed using HP ChemStation software. All calibration curves were linear (r2 > 0.9). All screening was performed with the MSD operating in scan 163 mode, and structural matching was accomplished using the NlST/EPAINIH mass spectral database (May 1992 release, NIST, Gaithersburg, MD) (Tables 1 - 4). Complete instrumental parameters are available as supplemental materials upon request. Further confirmation of previously unreported compounds from F3 extracts was accomplished using GC/HRMS. A CTC Analytics Model A2208 autosampler (Zwingen, Switzerland) was used to make 1 uL splitless injections onto a 30 m DB-5 capillary GC column (0.25 mm ID, 0.25 pm film, J&W Scientific, Folsom, CA) located within the oven of an HP 6890 GC. A Finnigan MAT 900 S-trap hybrid mass spectrometer (Bremen, Germany) was used for analysis of the column effluent. Only the double focusing MS stage was used in these studies. Full scan low resolution (1000) mass spectra over mass ranges of 50-300 Da and 100-400 Da were acquired at 1.8 sec/cycle and 1.1 sec/cycle, respectively. Elemental compositions of specific ions observed in background- subtracted mass spectra were determined using MPPSIRD to acquire data and the PGM to automatically interpret the data. Multiple mlz ratios were monitored in selected ion recording mode across three to five calibration and analyte mass peak profiles with a constant magnetic current and jumps in the accelerating and electrostatic sector potentials. Each mlz ratio was monitored for 20 msec to provide 1 sec/cycle for 31 monitored mlz ratios. Five mlz ratios each were used to monitor 40% of the mass range of the lock mass and calibration mass profiles which bracketed the analyte ion profiles investigated. The remaining 21 mlz ratios were used to monitor a 2000 parts per million (ppm) wide mass range 164 centered on an estimated mass from a low resolution mass spectrum using 3000 resolution, a single analyte ion with 10 ppm mass increments between mlz ratios using 10,000 resolution, or 60% of the mass range of three partial analyte profiles using 25,000 resolution. This system was used only for identification purposes and no quantitation was attempted. Recovery and Precision. Spike recovery experiments were performed using laboratory and Lake Mead water to determine the accuracy and precision of the method. Target compounds were spiked into laboratory water at various concentrations several times. During the initial sample collection at Lake Mead, samples were collected in duplicate at two sites. One sample was spiked with the target compounds while the other was not. To facilitate sample homogenization, the standards were spiked into approximately 50 L of water and mixed thoroughly. An additional 50 L of water was added vigorously and mixed thoroughly. The analysis procedure was the same as previously described for water samples. Detection Limits. Instrumental detection limits (IDLs) were based on the minimum concentration necessary to achieve a signal 3x the baseline noise. The method detection limits (MDLs) were estimated based on lDLs and the sample volume extracted (Tables 1 - 4). Furthermore, signals were not quantified in sample extracts unless the signal to noise ratio in that sample was greater than 165 10. Detection limits could not be calculated for compounds detected by use of NIST structural database matching or MPPSIRD and PGM. RESULTS AND DISCUSSION Analytical Considerations. Recoveries for the target compounds were consistent in both laboratory and matrix spikes and ranged from 73 — 107%. Therefore, no recovery adjustments were made to the data. Some compounds of interest were detected in trace quantities in field blanks. These concentrations were consistent and therefore subtracted from reported data. Contamination of samples was most likely due to the Pentaplate filtration system because it had great surface area and was difficult to clean thoroughly. Although the SPE columns should have been able to achieve good recoveries with much faster flow rates, back-pressure as a result of the small particle size and pressure limitations of the metering pump (approximately 60 psi) limited the flow rate to 100 mUmin. F3 extracts caused rapid deterioration of the GCIMS. Therefore, a 20 m guard column of deactivated fused silica (0.32 mm I.D., J&W Scientific, Folsom, CA) was added, and 1 m was removed when chromatographic resolution deteriorated. It also was observed that contamination of the injector liner caused the greatest impact on chromatography. The use of a cyclo double gooseneck liner (4 mm, Restek, Bellefonte, PA) allowed more injections of F3 extracts without replacement. In order to determine if chromatographic resolution had deteriorated, a set of standards was analyzed after every 5 sample 166 injections. If the resulting quantitation varied by greater than 20%, the guard column was trimmed of 1 to 2 m and the injector liner replaced. Even with these measures, some polar F3 pharmaceuticals were not particularly amenable to GC separation, and at times the resulting quantitation varied by as much as 50%. Also, it was unknown whether these compounds were stable over time. To reduce sample weathering, all extracts remained cooled at --15 °C until analysis. Organochlorine Pesticides and PCBs. Concentrations of OC pesticides in the waters of Lake Mead were generally low (Tables 5 & 6). Concentrations of HCHs and DDTs ranged from 0.12 to 25.9 nglL and 0.16 to 28.2 nglL, respectively (Tables 5 & 6). One water blank that was collected following sampling in Las Vegas Bay contained 4.8 ng DDT/L. Data were not adjusted for this contamination. The observed patterns of relative concentrations of HCHs and DDTs indicate leaching from a source that has been under anaerobic conditions (such as burial). Ground water seepage entering Las Vegas Wash has been shown contain HCHs with the same isomer pattern observed in this study (1999 Southern Nevada Water Authority, unpublished data). Historical manufacturing of OC pesticides in the Las Vegas Valley is the most likely source of these pesticides in the Las Vegas Bay (Southern Nevada Water Authority communication). Concentrations of OC pesticides are extremely low in other areas of Lake Mead. The US. EPA fresh water chronic exposure to aquatic life protection value for DDT is 0.001 pglL. The EPA values for bioconcentration to 167 predators (including humans) and human health cancer risk for DDT are 0.024 and 0.59 ng/L, respectively. Therefore, concentrations of DDT in the Las Vegas Bay may be sufficient to cause adverse affects in certain aquatic organisms (Federal Register 45-FR-79331). The chronic aquatic toxicity values for all HCH isomers is 0.08 pglL. For the protection of predatory species (through biomagnification) is 0.0163, 0.0186, and 0.0123 ug/L for a-, [3-, and y- HCH, respectively (Federal Register 45-FR-79335). Therefore, Las Vegas Bay water concentrations for B-HCH would have exceeded the recommended limits on two occasions (Tables 5 & 6). This indicates that HCHs may pose a threat to predator species in Lake Mead. PCBs were not detected in any water samples from Lake Mead. This is not surprising since these compounds are extremely lipophilic and very insoluble in water. The chronic toxicity value for fresh water and water quality criteria for aquatic life and predators such as humans (including safety factors and bioaccumulation factors) are 0.065 and 0.00079 ug/L, respectively. Therefore, it is unlikely that total PCB concentrations in Lake Mead waters pose a significant ecological threat. Polycyclic Aromatic Hydrocarbons. PAHs were detected at every site in Lake Mead. PAHs were also detectable in all water and column blanks. Summed concentrations of the 16 PAHs investigated did not vary significantly among the sites and ranged from 14 to 78 ng/L. Patterns of relative concentrations of PAHs did not vary significantly among locations with the exception of naphthalene, 168 concentrations of which were greater in the Las Vegas Bay. It is possible that motors from the sample collection vessels may have influenced PAH results since fuel from the motors occasionally was visible in the water. Ubiquitous detection of PAHs in Lake Mead is consistent with the great usage of recreational vehicles on the Lake. The value of the protection of water and organisms (including biomagnification potential and is based primarily on human health) for total PAHs is 0.0028 jig/L. Therefore, most sites tested at Lake Mead exceed this threshold and may have adverse affects on aquatic organisms. Alkylphenols and Bisphenol A. NP, OP, NPE1&2, and BPA were detected at some sites in Lake Mead (I' able 7). Detectable concentrations of NP and OP ranged from 5.0 to 308 nglL and 1.4 to 23.2 ng/L, respectively, and were consistently greater in the Las Vegas Bay. NPE1 and NPE2 concentrations ranged from 40.1 to 1540 ng/L and 10.0 to 702 nglL, respectively. Concentrations in the Las Vegas Bay were always much greater than concentrations at the reference sites. BPA was detected in only one sample from the Las Vegas Bay at 2.4 ng/L. Acute and chronic toxicity values for NP range from approximately 17 to 3,000 and 4 to 900 pglL for a wide range of aquatic organisms (7). Concentrations of NP observed in Lake Mead are far below those likely to cause acute or chronic toxicity (Table 7). Although NP has been shown to be weakly estrogenic in a variety of both in vitro (8-12) and in vivo bioassays (13-15), 169 concentrations to elicit effects are generally greater than those found in the waters of Lake Mead. Pharmaceuticals. Several pharmaceuticals were detected in Las Vegas Bay water samples (T ables 8 - 10). Initially, pharmaceuticals were detected using the MSD in scan mode. Mass spectra of the suspected pharmaceuticals were compared to the spectra of authentic standards (Figures 3 & 4). Because these matches were extremely good, the samples and standards were also analyzed in SIM mode (Figures 5 & 6). Concentrations of pharmaceuticals detected using the MSD in SIM mode and calibrated with authenticated standards ranged from less than the MDLs to 260 ng/L. These data are presented semi-quantitatively. These concentrations of pharmaceuticals are extremely small. Because there are no water quality criteria for these compounds, the toxicological significance needs to be placed into proper perspective. Furthermore, because persons using these compounds are exposed to pham'iacologically appropriate doses, it is inappropriate to compare exposures to maximum tolerated or toxic doses. Instead, the relevance of the dose that would be received by persons consuming untreated water was assesses by calculating the volume of water that would need to be consumed to obtain a single daily dose. For instance, a typical human dose of phenytoin is 50 mglday. If one were to drink the water from the location in Las Vegas Bay with the greatest concentration of phenytoin (260 nglL), it would require drinking approximately 190,000 L of this water to achieve a therapeutic dosage. The average daily 170 water intake of humans is 2 Ud. Thus, if the entire volume of water consumed contained phenytoin at the maximum concentration observed in surface waters of Las Vegas Bay, a person would receive 0.001% of a therapeutic dose. While these calculations do not represent a rigorous risk assessment, it does indicate that the concentrations of pharmaceuticals in the waters of Lake Mead probably do not present a significant risk to humans. However, the potential of the observed concentrations of pharmaceuticals directly on wildlife is unknown and can not be accurately assessed due to a lack of information of the effects of these compounds on wildlife species. A thorough review of pharmaceuticals in the environment has been published (16). Other Compounds. Several other classes of novel compounds were detected in Lake Mead water samples (Tables 1 & 6). Initial validation of these compounds was performed using the MSD in scan mode using authenticated standards or the NIST mass spectral database (Figures 7 & 8). Concentrations of compounds analyzed by GCIMS SIM for which authentic standards were available are presented (Table 11). The resolution and reproducibility of DEET and oxybenzone were such that 2 significant figures are provided. Whereas, due to poorer analytical resolution, concentrations of nicotine and caffeine are reported to only one significant figure. Compounds identified only by spectral matching with the NIST mass spectral database are presented (Tables 1 - 4). These compounds were observed only in water samples from the Las Vegas Bay. 171 High-resolution Mass Spectrometry. Analytical tools have been developed recently that utilize GC/HRMS for identification of unknown organic compounds in environmental samples. For example, several isomers of acrylonitrile-styrene, which are byproducts of a polymerization process, were identified in water from a municipal well near Toms River, NJ, where an increased incidence of childhood cancer was observed (17). Several novel compounds were verified and identified using MPPSIRD and PGM (Table 12). These data are considered to be qualitative only and were detected only Las Vegas Bay. Summary. This paper provides information on a methodology that used large volume water sampling with solid-phase extraction to screen for a broad range of classes of organic compounds. Several classes of organic compounds were determined to be entering Lake Mead via the Las Vegas Wash. However, the Virgin, Muddy, and Colorado Rivers entering Lake Mead were not tested. Concentrations of OC pesticides and halogenated aromatic hydrocarbons were relatively small. A number of previously unreported compounds including pharmaceuticals, health care products, and flame-retardants were identified. This study may serve as a basis for further research that could validate and better quantitate these novel compounds. ACKNOWLEDGMENTS 172 This work was supported by grants and donations from the Southern Nevada Water Authority, US. Department of the Interior - Bureau of Reclamation, Clark County Sanitation District, and the National Park Service. Technical and instrumental support was provided by US. Environmental Protection Agency - National Environmental Research Laboratory, Las Vegas, NV. The authors would like to thank Fred DeSiIva and the rest of the National Park Service Volunteers who operated and organized boats for this project. REFERENCES (1) LaBounty, J. F.; Horn, M. J. J. Lake Reser. Manag. 1997, 13, 95-108. (2) Prentki, R. T.; Paulson, L. J. In Aquatic Resources Management of the Colorado River Ecosystem; Adams, V. D., Lamarra, V. A., Eds.; Ann Arbor Science: Ann Arbor,Ml, 1983, pp 105-123. (3) Paulson, L. J.; Baker, J. R. In Proceedings of the Symposium on Surface Water lmpoundments; Stefan, H. G., Ed.; American Society of Civil Engineering: New York, 1981, pp 1647-1658. (4) Bevans, H. E.; Goodbred, S. L.; Miesner, J. F.; Watkins, S. A.; Gross, T. S.; Denslow, N. D.; Schoeb, T. Water-Resources Investigations Report 96-4266 1996. (5) Snyder, S. A.; Keith, T. L.; Verbrugge, D. A.; Snyder, E. M.; Gross, T. S.; Kannan, K.; Giesy, J. P. Environ. Sci. Technol. 1999, 33, 2814-2820. 173 (6) (7) (8) (9) (10) (11) (12) (13) (14) (15) Snyder, S. A.; Snyder, E.; Villeneuve, D.; Kurunthachalam, K.; Villalobos, A.; Blankenship, A.; Giesy, J. In Analysis of Environmental Endocrine Disruptors; Keith, L. H., Jones-Lepp, T. L., Needham, L. L., Eds.; American Chemical Society: Washington,DC, 2000, pp 73-95. Staples, C. A.; Weeks, J.; Hall, J.; Naylor, C. Environ. Toxicol. Chem. 1998, 17, 2470-2480. Mueller, G. C.; Kim, U. H. Endo. 1978, 102, 1429-1435. Soto, A. M.; Tien-Min, L.; Honorato, J.; Silvia, R. M.; Sonnenschein, C. In Chemically-induced alterations in sexual and functional development: The wildlife/human connection; Mehlman, M. A., Ed.; Princeton Scientific Publishing Co.,lnc: Princeton,NJ, 1992; Vol. 21. Desbrow, C.; Routledge, E. J.; Brighty, G. C.; Sumpter, J. P.; Waldock, M. Environ. Sci. Technol. 1998, 32, 1549-1558. Jobling, S.; Sumpter, J. P. Aquat. Toxicol. 1993, 27, 361-372. White, R.; Jobling, S.; Hoare, S. A.; Sumpter, J. R; Parker, M. G. Endo. 1994, 135, 175-182. Jobling, S.; Sheahan, D.; Osborne, J. A.; Matthiessen, P.; Sumpter, J. P. Environ. Toxicol. Chem. 1996, 15, 194-202. Giesy, J. P.; Pierens, S. L.; Snyder, E. M.; Miles-Richardson, S.; Kramer, V. J.; Snyder, S. A.; Nichols, K. M.; Villeneuve, D. A. Environ. Toxicol. Chem. 2000, (in press). Nimrod, A. C.; Benson, W. H. Crit. Rev. Toxicol. 1996, 26, 335-364. 174 (16) Daughton, C. G.; Ternes, T. A. Environ. Health Perspect. 1999, 107, 907- 938. (17) Grange, A. H.; Sovocool, G. W. Rapid Commun. Mass Spectrom. 1998, 1161-1169. 175 TABLE 1. F1 and F2 Compounds Detected, Estimated Method Detection Limits (nglL), and Ions Monitored MDL Ions Usage/Source E1 HCHs' 0.10 Various Insect control DDT,DDE, and DDD 0.15 Various Insect Control PAHs“ 4.0 Various Petroleum-based products 3 NP 5.0 107, 135 Degradation product of NPEs OP 1.0 107, 135 Degradation product of OPEs 176 TABLE 2. Pharmaceuticals Compounds (F3) Detected, Estimated Method Detection Limits (nglL), and Ions Monitored \ Compound MDL Ions UsagaISource Carbamazepine 10 165, 193, 236 Pharmaceutical — anti-seizure Primidone 20 1 17, 146, 190 Pharmaceutical - anti-seizure Phenytoin 50 180, 223, 209 Pharmaceutical — anti-seizure Phenobarbital 10 204, 117 Pharmaceutical — anti-seizure Diazapam 1 256, 283, 284 Pharmaceutical - anxiety disorders Meprobamate NA Scan Pharmaceutical - anxiety disorders Guaifenesin 20 109, 124, 198 Pharmaceutical - expectorant Codeine 10 162, 229, 299 Pharmaceutical — pain relief Hydrocodone 4 185, 242, 299 Pharmaceutical — pain relief Pentoxifylline 5 180, 193, 221 Pharmaceutical — blood thinner Carisoprodol NA Scan Pharmaceutical - muscle spasms Methocarbamol NA Scan Pharmaceutical — muscle spasms Mequinol NA Scan Pharmaceutical - pigmentation NA = not applicable 177 TABLE 3. F3 Compounds Detected, Estimated Method Detection Limits (nglL), and Ions Monitored - Part I Compound MDL Ions Usage/Source NPE1 30 219, 223, 228 Surfactants NPE2 30 107, 223, 237 Surfactants Nicotine 4 84, 133, 161 Tobacco products Caffeine 7 67, 109, 194 Coffee, tea, etc. DEET 2 91, 1 19, 190 Insect repellant BPA 2 213, 219, 228 Plasticizer Oxybenzone 10 151 , 227, 228 Sunscreen Homomenthyl salicylate NA Scan Sunscreen o-Methoxyacetophenone NA Scan Fragrance Vanillin NA Scan Fragrance Terbuthylazine NA Scan Weed control NA = not applicable 178 TABLE 4. F3 Compounds Detected, Estimated Method Detection Limits (nglL), and Ions Monitored - Part II Compound MDL Ions Usage/Source p-Chloroaniline NA Scan Dye Acridine NA Scan Dye Triclosan NA Scan Anti-fungal F ulvicin NA Scan Anti-fungal Tris(2-chloroethyl)phosphate NA Scan F lame-retardant I Tris(2-chloropropyl)phosphate NA Scan Flame-retardant Tris(1,3-dichIoroisopropyl)phosphate NA Scan Flame-retardant Triphenylphosphate NA Scan Industrial chemical Triethylphosphate NA Scan Industrial chemical Ethyl citrate NA Scan Food additive Mequinol NA Scan Anti-pigmentation Cholesterol NA Scan Biomolecule NA = not applicable 179 TABLE 5. Concentrations of HCHs in Lake Mead (nglL) Location Date a-HCH B-HCH S-HCH y-HCH LV Wash 6/23/98 7.94 25.91 23.53 7.43 3/26/99 1.43 3.18 2.26 1.95 5/05/99 1.88 5.70 3.34 2.86 6I09/99 2.96 8.07 5.20 4.17 LV-0.5 3/26/99 1 .06 2.23 1 .45 1.58 5/05/99 1.15 2.61 1.75 1.66 6/09/99 1.21 0.80 2.11 1.32 LV-1 (plume) 8/03l98 3.25 13.3 10.8 4.88 3/26/99 6.88 20.3 12.3 10.6 5/05/99 2.45 6.84 4.56 3.49 LV-1 (surface) 6/23/98 1 .49 4.56 3.32 2.33 8/03/98 0.42 2.40 0.85 ND Water Barge 6/22I98 ND ND ND ND 8/03/98 ND ND ND ND 3/26/99 ND ND ND ND 5/05/99 ND 0.26 ND 0.21 6I09/99 ND ND ND ND Moon Cove 6/22/98 0.12 0.38 0.31 ND 3/26/99 ND 0.47 ND 0.27 5/05/99 ND 0.26 ND 0.24 6/09/99 ND 0.24 ND 0.23 ND = not detectable 180 TABLE 6. Concentrations of DDTs Detected in Lake Mead (nglL) Location Date DDE DDD DDT LV Wash 6/23/98 ND 0.39 1.09 3/26/99 ND 0.24 0.48 5/05/99 ND 0.42 4.26 6/09/99 ND 0.52 9.61 LV-0.5 3/26/99 ND 0.47 12.0 5105/99 ND 0.28 ND 6/09/99 ND ND ND LV-1 (plume) 8/03/98 0.18 1.15 19.5 3/26/99 ND 0.36 28.2 5/05/99 0.29 0.44 25.7 LV-1 (surface) 6/23/98 ND 0.16 1.81 8l03l98 ND 0.46 4.62 Water Barge 6/22/98 ND ND ND 8/03/98 ND ND ND 3/26/99 ND ND 0.31 5/05/99 ND ND ND 6/09/99 ND ND ND Moon Cove 6122/98 ND ND ND 3/26/99 ND ND 0.46 5/05/99 ND ND 0.45 6/09/99 ND ND 0.50 ND = not detectable 181 TABLE 7. Concentrations of Alkylphenols and Bisphenol A in Lake Mead (nglL) Location Date OP NP NPE. NPE2 BPA LV Wash 6/23/98 22.0 308 1000 663 ND 3/26/99 3.7 32.3 193 138 ND 5I05/99 ND ND 192 94 ND 6/09/99 7.1 61 .9 496 350 ND LV-0.5 3/26/99 1 .4 13.6 129 33.0 ND 5/05/99 4.0 40.2 198 58 2.4 6/09/99 1.8 18.3 117 57 ND LV-1 (plume) 8l03l98 15.1 172 398 222 ND 3/26/99 23.2 1 96 1 540 702 ND 5I05/99 20.7 232 978 468 ND LV-1 (surface) 6/23/98 ND 78.7 677 489 ND 8/03/98 ND 10.1 47 ND ND Water Barge 6/22/98 ND ND ND ND ND 8l03l98 ND ND ND ND ND 3/26/99 ND ND ND ND ND 5I05/99 ND ND ND ND ND 6/09/99 ND ND ND ND ND Moon Cove 6/22/98 ND 5.6 40.1 10.0 ND 3/26/99 ND 5.0 ND ND ND 5/05/99 ND ND ND ND ND 6/09/99 ND ND ND 29.3 ND ND = not detectable 182 TABLE 8. Concentrations of Pharmaceuticals in Lake Mead (nglL), Part I Location Date Phenytoin Codeine Primidone LV Wash 6/23/98 100 20 80 3l26l99 70 ND 40 5/05/99 80 30 40 6/09/99 260 ND 80 LV-0.5 3l26l99 50 20 20 5I05I99 ND 20 40 6/09/99 70 30 40 LV-1 (plume) 8l03l98 250 100 80 3l26l99 230 120 50 5/05/99 250 ND 1 30 LV-1 (surface) 6/23/98 ND ND 10 8/03/98 ND ND 20 Water Barge 6/22/98 ND ND ND 8/03/98 ND ND ND 3/26/99 ND ND ND 5/05/99 ND ND ND 6I09/99 ND ND ND Moon Cove 6/22/98 ND ND ND 3l26l99 ND ND ND 5I05/99 ND ND ND 6/09/99 ND ND ND ND = not detectable 183 TABLE 9. Concentrations of Pharmaceuticals in Lake Mead (nglL), Part II Location Date Pentoxifylline Carbamazepine. Guaifenisen. LV Wash 6/23/98 10 30 40 3/26/99 1 0 30 40 5/05/99 ND 20 30 6/09/99 10 30 50 LV-0.5 3/26/99 ND ND ND 5I05/99 ND 40 20 6/09I99 1 0 30 30 LV-1 (plume) 8/03/98 ND 40 40 3l26l99 ND ND ND 5/05/99 ND ND ND LV-1 (surface) 6/23/98 ND 20 ND 8/03/98 ND 10 ND Water Barge 6/22/98 ND ND ND 8/03/98 ND ND ND 3l26l99 ND ND ND 5/05/99 ND ND ND 6/09l99 ND ND ND Moon Cove 6/22/98 ND ND ND 3/26/99 ND ND ND 5/05/99 ND ND ND 6/09/99 ND ND ND ND = not detectable 184 TABLE 10. Concentrations of Pharmaceuticals in Lake Mead (nglL), Part III Location Date Phenobarbital Hydrocodone Diazepam LV Wash 6/23/98 1 0 ND ND 3l26l99 ND ND 50 5/05/99 ND ND 60 6/09/99 ND 10 ND LV—0.5 3l26l99 ND ND ND 5/05/99 ND ND ND 6/09/99 ND 10 ND LV-1 (plume) 8/03/98 30 10 ND 3l26l99 40 ND ND 5I05/99 40 10 10 LV-1 (surface) 6l23/98 ND ND ND 8/03/98 ND ND ND Water Barge 6/22/98 ND ND ND 8/03/98 ND ND ND 3/26/99 ND ND ND 5/05/99 ND ND ND 6/09/99 ND ND ND Moon Cove 6/22/98 ND ND ND 3l26l99 ND ND ND 5/05/99 ND ND ND 6I09I99 ND ND ND ND = not detectable 185 TABLE 11. Concentrations of Other Compounds in Lake Mead (nglL) Location Date DEET Oxybenzone Caffeine Nicotine LV Wash 6/23/98 270 200 10 1 0 3l26l99 20 56 30 1 0 5/05/99 24 1 50 30 1 0 6I09I99 87 82 30 1 0 LV-0.5 3l26l99 12 140 20 ND 5I05l99 1 1 120 20 10 6I09I99 45 210 20 10 LV-1 (plume) 8/03/98 164 220 30 10 3/26/99 51 120 30 ND 5I05l99 67 58 50 20 LV-1 (surface) 6/23/98 57 360 10 ND 8l03l98 17 200 ND ND Water Barge 6/22/98 ND 73 ND ND 8/03/98 4 79 ND ND 3l26l99 N D 38 ND ND 5/05/99 ND 53 1 0 ND 6/09/99 3 78 ND ND Moon Cove 6/22l98 3 38 ND ND 3l26l99 ND 180 ND ND 5I05l99 ND 30 ND ND 6/09/99 ND 59 ND ND ND = not detectable 186 TABLE 12. Compounds Identified in Las Vegas Bay by HRMS Phenobarbital Phenytoin Oxybenzone Trichlorophenol Ethyl citrate Tris(2-chloroethyl)phosphate Tris(2-chloropropyl)phosphate Tris(1 ,3-dichIoroisopropyl)phosphate Tertbutylazine 187 l c r-, ) \_"' .§ Water Barge .2' 7 -l Cove ‘- k, ,1‘\_,' ‘ I I Government Wash . ‘ r t. ,u 7 95‘. 2t, Lx I ‘l ‘ ‘ ‘\ ya 0 ’I _’ ~ e I m LV-t -- 1, ml (061° ‘ (x ' ' ‘ ‘ n ‘ we»... , » ,\ Q \ \ l f‘ "I \ \ ‘ .. ‘ d 5 $6 ‘ of.“ I I D 'P‘ ' ’ I I \\ ' I \ ' ls " -- o 34 I a 09 ‘ 9, ‘ ye. . I 'Las Vegas Bay ' . 4, , :~ ' Boat Harbor . Boulder .' r' ’1 "“ Moon ' la Fish Hatchery“ ' v Cove Basrn ' ' Pumping :‘\ Saddle ." '_ (" Statlons 4\ Island (S, . ‘ _ \ t Boulder Boat Harbor ‘ \‘ ‘h‘ ‘1 ‘ \ Boulder ‘~ 2 3? : Beach ‘-~, I, ’ 3’ \\"‘~ I ’1’) \ Figure 1. Map of the Boulder Basin study area of Lake Mead, Nevada 188 130 L filtered in situ I 100 L solid-phase extracted (SPE) I SPE eluted, liquid/liquid extracted, and concentrated to 2 mL I 1 mL silica gel fractionated iF1 vF2 lF3 Non—polar Semi-Polar Polar I I I PAHs NP NPE122 OC Pesticides A OP Atrazine PCBs OC Pesticides Simazine Estrone 1 78-estradiol Ethynylestradiol Progesterone Pharmaceuticals DEET Nicotine Caffeine Oxybenzone Bisphenol A Figure 2. Flow diagram of analytical method 189 180 A. 77 10“ e 209 223 l 165 l 252 147 l I J . u L I. .1. l l ,7 l 104 Be 77 223 209 I65 252 193 147 236 mlz Figure 3. Mass spectra of phenytoin (Dilantin): (A) authentic standard; (B). LV Bay water extract 190 204 O O A N N . T 117 O 77 “’4 161 91 146 II 174 i1 232 II "I In I I I 204 B. 117 104 77 91 146 161 174 i 232 All A. - . J] I. m/z Figure 4. Mass spectral scan of phenobarbital: (A) authentic standard; (B) LV Bay water extract 191 DEET Oxybenzone Phcnacetin A Guaifenesin Bromphenlramine O Chlorcyclizinc Pentoxifylline Phenytoin r l I I A f B . I I I T '1 _ I 18 21 24 27 30 33 36 RT (min) Figure 5. Mass spectral SIM chromatograms I: (A) authentic standards; (B) LV Bay water extract 192 f I 1 I 9 12 15 18 21 24 RT (min) Figure 6. Mass spectral SIM chromatograms II: (A) standards; (B) LV Bay water extract. Peak # 1. DEET; 2. Guafenesin; 3. Acetominophen; 4. Meprobamate; 5. Caffeine; 6. Oxybenzone; 7. Bisphenol A; 8. A8 THC; 9. A9 THC; 10. Primidone; 11. Carbamazepine; 12. Hydrocodone 193 119 <“"\ A. 91 s b3 i— =— [:_—— 119 It .I .---I . .I Figure 7. Mass spectral scan of DEET: (A) authentic standard; (B) LV Bay water extract 194 CI 75 CI Cl Cl 99 \__2 O S—/ II 0— 1'2— 0 A. 191 o \CC| 155 209 C' 381 u 243 321 1. 1. 75 99 B- .9. 155 209 m I L 321 1. . L .. m/z Figure 8. Mass spectral scan of tris(1,3-dichloroisopropyl)phosphate: (A) NIST database spectra; (B) LV Bay water extract 1 195