| i 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 DATE DUE 5/08 K:lProj/Acc&Pres/CIRC/DaIeDue.indd REPRODUCTIVE EFFECTS OF GESTATIONAL AND LACTATIONAL EXPOSURE TO ESTROGENIC ENDOCRINE MODULATORS IN MICE BY Jiyou Han A DISSERTATION Submitted to Michigan State University In partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Animal Science - Environmental Toxicology 2008 ABSTRACT REPRODUCTIVE EFFECTS OF GESTATIONAL AND LACTATIONAL EXPOSURE TO ESTROGENIC ENDOCRINE MODULATORS IN MICE By Jiyou Han Our previous studies demonstrated decreased sperm production and decreased in vitro sperm fertilizing ability in male offspring (F-1) after in utero and lactational exposure to 1.0 and 10 pg diethylstilbestrol (DES)/kg maternal body weight/day (bw/d). In contrast, fertilizing ability of sperm was increased exposure to 0.1 pg DES/kg maternal bw. Genistein (GEN, an isoflavonoid) increased after sperm production and fertilizing ability at a maternal dose as low as 10 mg/kg bw. The aims of the present study were to examine the effects of maternal exposure to 17a-ethynyl estradiol (EE2, a component of oral contraceptives) on sperm production, sperm quality, and sperm fertilizing ability in male Offspring (F-1) and effects on superovulation and egg fertilizing ability in female offspring (F-1) maternally exposed to EE2, DES or GEN. Pregnant C57BL/6 mice (F-O), bred with DEA/2 males, were gavaged daily with DES (0.1, 1.0, or 10 ug/kg maternal bw), EE2 (0.1, 1.0, or 10 ug/kg maternal bw) or GEN (0.1, 0.5, 2.5, or 10 mg/kg maternal bw) from gestational day 12 to postnatal day (PND) 20. Sperm production and quality were examined by using computer- assisted sperm analysis (CASA). Fertilizing ability of sperm and eggs was assessed using an in vitro fertilization (IVF) assay. Our results demonstrated that in utero and lactational exposure to a high dose of EE2 (10 ug/kg maternal bw) could decrease sperm production without Changing fertilizing ability and sperm motion parameters in 45- to 48-week-old male offspring. On the other hand, a significant increase in the number of ovulated eggs was observed as a result of in utero and lactational exposure to 0.1 pg DES/kg maternal bw without altering fertilizing ability of eggs. Fertilizing ability Of eggs was significantly decreased in the 10 pg EE2/kg maternal BW animals at 3 to 6 weeks of age, whereas GEN had no effect on the number of ovulated eggs and fertilizing ability of eggs. ACKNOWLEDGEMENTS I grateful acknowledge Professor Karen Chou for encouraging and providing me to learn science. She has contributed her time and hands in helping me perform the research described in this dissertation. I would like to express the deepest appreciation to my committee members, especially Professor Steve Bursian and Professor Dale Rozeboom for their valuable input. In addition, I thank Dr. Peter Saama and Professor Robert Tempelman, who helped me analyze data. I also thank Raquel and Barb for their persistent help. Many thanks go out to my family; Kun-Mo Han, Young-Hee Kim, Yongga Chae, Hyongwoo Ham, Kyuhi Han, Anrakyugo, Kyong-A Han, Jinho Kim, Dong Hun Han, and Sunjoo Lee. Especially, I thank my husband, Hyeongpil Ham, for giving me endless support and love. I would like to thank my 11-month-old son, Juwon Ham, for giving me strength to complete my doctoral degree and for inspiring my sprits. Finally, I thank God who is always guiding me and loving me. TABLE OF CONTENTS LIST OF TABLES ................................................................................. viii LIST OF FIGURES ................................................................................. x ABBREVIATIONS ................................................................................. xi CHAPTER 1 1.1 Rationale ................................................................................ 1 1.2 Hypothesis .............................................................................. 3 1.3 Objectives ............................................................................... 3 CHAPTER 2- Literature review I 2.1 Estrogen synthesis ................................................................... 4 2.1.1 Estrogen synthesis in females ............................................. 8 2.1.2 Estrogen synthesis in males .............................................. 12 2.4 Estrogen receptors .................................................................. 13 2.5 Modes of action of estrogen ...................................................... 16 2.5.1 Ligand-dependent mode .................................................... 16 2.5.2 Ligand-independent mode ................................................. 17 2.5.3 Estrogen response element-independent mode......... .........18 2.5.4 Nongenomic mode ........................................................... 20 2.6 Effects of estrogen on ovarian development, oogenesis, and Folliculogenesis .................................................................... 21 2.6.1 Estrogen in ovarian development ........................................ 21 2.6.2 Estrogen in oogenesis and folliculogenesis ........................... 22 2.7 Effects of estrogen on testicular development and spennatogenesis.26 2.7.1 Estrogen in testicular development ..................................... 26 2.7.2 Estrogen in sperrnatogenesis ............................................. 31 2.8 Effects of estrogen on fertilization and implantation ........................ 38 2.9 Effects of estrogen on sex ratio of offspring .................................. 40 2.10 Effects of estrogen on anogenital distance .................................. 44 2.11 Effects of estrogen on puberty ................................................. 47 2.12 Effects of estrogen on body weight ........................................... 48 2.13 Bibliography ......................................................................... 52 CHAPTER 3- Literature review II 3.1 Definition of estrogenic endocrine modulators ............................... 77 3.2 Sources and exposure to EEMs ................................................. 78 3.3 Modes of action of EEMs ......................................................... 81 3.3.1 Altered sex steroid hormone synthesis ................................. 81 3.3.2 Altered sex steroid hormone storage and/or release...............83 3.3.3 Altered sex steroid hormone transport and Clearance...... ........83 3.3.4 Altered sex steroid hormone receptor recognition/binding........86 3.3.5 Altered sex steroid hormone postreceptor activation ............... 87 3.3.6 Altered gene expression without changing DNA sequence ....... 88 3.4 Phytoestrogen and mycotoxin ................................................... 91 3.4.1 Genistein ....................................................................... 91 3.4.2 Zearalenone .................................................................. 93 3.5 Pharmaceuticals .................................................................... 96 3.5.1 Diethylstilbestrol ............................................................. 96 3.5.2 Tamoxifen ..................................................................... 98 3.5.3 17o-ethynylestradiol ...................................................... 1 00 3.6 Pesticides ........................................................................... 103 3.6.1 Methoxychlor .............................................................. 103 3.6.2 Atrazine ..................................................................... 104 3.7 Industrial Chemicals ............................................................... 106 3.7.1 Bisphenol A ................................................................. 106 3.7.2 Phthalates .................................................................. 109 3.8 Environmental pollutants ........................................................ 112 3.8.1 Polychlorinated biphenyls .............................................. 112 3.8.2 Benzo[a]pyrene ........................................................... 114 3.9 Bibliography ......................................................................... 1 16 CHAPTER 4 Effects of In utero and Lactational Exposure to Diethylstilbestrol, 17a-Ethynyl estradiol, and Genistein on Ovulation and In Vitro Fertilizing Ability of Eggs in Female Mouse Offspring 4.1 Abstract ............................................................................... 146 4.2 Introduction .......................................................................... 147 4.3 Materials and methods ........................................................... 150 4.4 Results ............................................................................... 155 4.5 Discussion .......................................................................... 165 4.6 Bibliography ......................................................................... 171 CHAPTER 5 Effects of In utero and Lactational Exposure to 17a—Ethynyl Estradiol on Sperm Quality and In Vitro Fertilizing Ability in Male Mouse Offspring 5.1 Abstract ............................................................................... 178 5.2 Introduction .......................................................................... 1 79 5.3 Materials and methods ........................................................... 182 5.4 Results ............................................................................... 188 5.5 Discussion ........................................................................... 196 5.6 Bibliography ......................................................................... 203 CHAPTER 6 Effects of Gestational and Lactational Exposure of Female Mice to 17a- Ethynyl Estradiol on Reproduction 6.1 Abstract ............................................................................... 212 6.2 Introduction .......................................................................... 213 6.3 Materials and methods ........................................................... 214 vi 6.4 Results ............................................................................... 217 6.5 Discussion .......................................................................... 224 6.6 Bibliography ........................................................................ 228 CHAPTER 7 7.1 Conclusion ............................................................................ 232 vii LIST OF TABLES CHAPTER 2 Table 2-1. Enzymes involved in steroidogenesis .......................................... 6 Table 2-2. Production rates and serum concentrations of estradiol during the menstrual cycle in normal women ............................................ 11 Table 2-3. ERs and aromatase distribution in the rodent fetal testis ................ 29 Table 2-4. ERs and aromatase distribution in postnatal immature rodent testis .................................................................................. 29 Table 2-5. ERs and aromatase distribution in the adult rodent testis ............... 29 Table 2-6. Reproductive phenotypes of ERK0 and ArKO male mouse models ................................................................................ 32 CHAPTER 4 Table 4-1. Body weight of female offspring Of dams treated with DES, EE2, and GEN from gestational day 12 through lactation ............................ 158 Table 4-2. Anogenital distance of female offspring Of dams treated with DES, EE2, and GEN from gestational day 12 through lactation ............... 160 Table 4-3. Number of ovulated eggs of female offspring of dams treated with DES, EE2, and GEN from gestational day 12 through lactation ...... 161 Table 4-4. In Vitro fertilizing ability of eggs from female offspring of dams treated with DES, EE2, and GEN from gestational day 12 through lactation .............................................................................. 162 Table 4-4a. Relative percentage of in Vitro fertilizing ability of eggs from female offspring of dams treated with DES, EE2, and GEN from gestational day 12 through lactation .......................................................... 163 Table 4-5. Percent of degenerated eggs from female offspring of dams treated with DES, EE2, and GEN from gestational day 12 through lactation .............................................................................. 164 viii CHAPTER 5 Table 5-1. Organ weights of male offspring of dams treated with EE2 from gestational day 12 through lactation ...................................... 190 Table 5-2. Anogenital distance of male offspring of dams treated with EE2 from gestational day 12 through lactation ......................................... 192 Table 5-3. Epididymal sperm quality of male Offspring of dams treated with EE2 from gestational day 12 through lactation ................................... 193 Table 5-4. In Vitro fertilizing ability of epididymal sperm from male offspring of dams treated with EE2 from gestational day 12 through lactation...194 Table 5-4a. Relative percentage of in Vitro fertilizing ability of epididymal sperm from male offspring of dams treated with EE2 from gestational day 12 through lactation .............................................................................. 195 CHAPTER 6 Table 6-1. Reproductive performance of F-0 females gavaged with 17a-ethynyl estradiol from gestational day 12 to lactation (postnatal day 20)....220 Table 6-2. Reproductive performance of F-O females gavaged with diethylstilbestrol from gestational day 12 to lactation (postnatal day 20) .................................................................................... 222 Table 6-3. Reproductive performance of F-0 females gavaged with genistein from gestational day 12 to lactation (postnatal day 20) ................. 223 LIST OF FIGURES CHAPTER 2 Figure 2-1. Schematic presentation of steroidogenesis .................................. 5 Figure 2-2. Ovarian steroidogenesis ......................................................... 10 Figure 2-3. Schematic drawing of the mean serum levels of E1 and E2 in relation to progesterone, luteinizing hormone, and follicular stimulating hormone during pre- and postmenopause .................. 11 Figure 2-4. Schematic representative of the structural and functional domains of ERor and ERB ...................................................................... 15 Figure 2-5. Schematic representation of modes Of action of estrogen .............. 19 Figure 2-6. Schematic representation of hormonal regulation of oogenesis in the ovary .................................................................................. 25 Figure 2-7. Schematic representation of a cross-sectional view of the testis and the seminiferous tubules ......................................................... 27 Figure 2-8. Schematic representation of hormonal regulation of sperrnatogenesis ............................................................................... 35 CHAPTER 4 Figure 4-1. Schedule of maternal treatments and parameters measured in their female offspring (F-1) ........................................................... 152 CHAPTER 5 Figure 5-1. Schedule of maternal treatments and parameters measured in their male offspring (F-1) .............................................................. 184 CHAPTER 6 Figure 6-1. Schedule of treatments and parameters measured in females ...... 216 ABBREVIATIONS ADI... ..Acceptable daily intake AF-1 .....Activation function 1 AF-2. . . ..Activation function 2 AhR. . . ..Ary hydrocarbon receptor AGD. . . .. Anogenital distance ALH.....Amplitude of lateral head displacement ANOVA ..... Analysis Of variance AP-1 .. . ..Activator protein-1 AR.....Androgen receptor ArKO. . . ..Aromatase knockout mouse ATP... ..Adenosine tri-phosphate B[a]P ..... Benzo[a]pyrene BBP. . . ..Butyl benzyl phthalate BG-1.....A human ovarian adenocarcinoma cell line BSA... ..Bovine serum albumin BPA ..... Bisphenol A BW ..... Body weight cAMP. . . ..Cyclic adenosine monophosphate CASA.....Computer assisted sperm analysis CCA. . . ..Cervical clear cell adenocarcinoma CCI4. . . ..Carbon tetrachloride C/EBP. . . ..CCAAT/enhancer binding protein xi CREB.....cAMP response element binding protein CYP 19 ..... Cytochrome P450 19 (aromatase) Cyp11a.....P450 side chain cleavage enzyme DBCP. . . ..Dibromochloropropane DBD. . . ..DNA binding domain DBP ..... Dibutyl phthalate DDE. . . ..Dichlorodiphenyl dichloroethene DDT. . . ..1 , 1, 1-trichloro—2, 2,-bis (p-Chlorophenyl)ethane DE2. . . ..(+)-Diol—epoxide-2 DES. . . ..Diethylstilbestrol DEHP ..... Di-(2-ethylhexyl)-phthalate DHD. . . ..(-)-Dihydrodiol DHEA. . . ..And rostenedione DH EAS. . . ..Dehyd roepiand rosterone sulfate DHT. . . ..Dihyd rotestosterone DINP. . . ..Diisononyl phthalate DNA. . . ..Deoxyribonucleic acid E1 .....Estrone E2... ..17B-estradiol E3... ..Estradiol ECSCF ..... The European Commission’s Scientific Committee on Food EE2. . . ..17or-ethynyl estradiol EEMs. . . ..Estrogenic endocrine modulators xii EGF. . . ..Epiderrnal growth factor ER... ..Estrogenic receptor ERor. . . ..Estrogen receptor alpha ERB. .. ..Estrogen receptor beta ERE. . . ..Estrogen response element ERKO. . . ..Estrogen receptor knockout mouse orERKO.....Estrogen receptor alpha knockout mouse BERKO.....Estrogen receptor beta knockout mouse EU.....The European Union FDA.....US Food and Drug Administration ERR1 ..... Estrogen receptor-related receptor 1 FISH... ..Fluorescence in situ hybridization FSH. . . ..Follicle stimulating hormone GD.....Gestationa| day GEN.....Genistein GLUTs. . . ..Glucose transporter proteins GnRH. . . ..Gonadotropin releasing hormone HCB ..... Hexachlorobenzene hCG ..... Human Chorionic gonadotropin hERa ..... Human estrogen receptor a HDL ..... High density Iipoprotein Hoxa.....10 Homeobox A 10 HPTE. . . ..2,2-Bis(p—hydroxyphenyl)-1 ,1 ,1 -trichloroethane . xiii HSD. . . ..Hydroxy-steroid dehyd rogenase 3B-HSD ..... 3B-hydorxysteroid dehydrogenase HSP70.....Heat shock protein 70 kDa HSP90.....Heat shock protein 90 kDa 17B-HSR.. . ..17B-hydroxysteroid red uctase lCl 164, 384 ..... Estra-1,3,5 (10)—triene-7-undecanamide IGF-1 .....lnsulin-like growth factor-1 IL-1 .....lnterieukin-1 IVF. . . ..In Vitro fertilization LBD ..... Ligand binding domain LDso ..... Lethal dose to 50% of the experimental animals LDL ..... Low density Iipoprotein LH. . . ..Luteinizing hormone LOAEL. . . ..The lowest Observed adverse effect level LS-mean. . . ..Least squares mean MAPK.....Mitogen activated protein kinase mBP. . . ..Monobutyl phthalate mBzP. . . ..Monobenzyl phthalate MEHP ..... Mono-(2-ethylhexyl)-phthalate mEP. . . ..Monoethyl phthalate mEHP. . . ..Monoethylhexyl phthalate mRNA ..... Messenger ribonucleic acid MTD. . . ..Maximum tolerated dose xiv MXC. . . ..Methoxychlor NADP.....Nicotinamide adenine dinucleotides phosphate NF-xB.....Nuclear factor kappa B NHANES.....The National Health and Nutrition Examination Survey NGF.....Naturally occurring growth factor NOAEL.....No observable adverse effect level NOEL... ..No Observed effect levels NR. . . ..Nuclear receptor o,p-DDE. . . ..Ortho, para-dichlorodiphenyl dichloroethene P ..... progesterone p300/CBP.....A transcriptional CO-activating protein involved in G protein signaling P450scc ..... P450 side Chain cleavage enzyme PCB... ..Polychlorinated blphenyl PMSG ..... Pregnant mare’s serum gonadotropin PND. . . ..Postnatal day PPAR. . . ..Peroxisome proliferator-activated receptor ppm.....Part per million PR... ..Progesterone receptor PVC... ..Polyvinyl chloride RNA ..... Ribonucleic acid SD... ..Standard deviation SE.....Standard error XV SF-1 .....Steroidogenic factor 1 SHBG. . . ..Sex-steroid hormone-binding globulin SMRT.....Silencing mediator for retinoids and thyroid hormone receptors SOX—9.....SRY-box containing gene 9 SR-B1 .....Scavenger receptor B1 SRC-1.....Steroid receptor coactivator 1 StAR.....SterOidogenic acute regulatory protein TCDD. . . ..2,3,7,8-Tetrachlordibenzo-p-dioxin TDl ..... Tolerable daily intake TEBG. . . ..Testosterone-estrogen-binding globulin TGF. . . ..Transfonning growth factor US. . . ..The United States US EPA.....US Environmental Protection Agency UK... ..The United Kingdom Wnt-4.....Wingless-type MMTV integration site family, member 4 Wnt-7a ..... Wingless-type MMTV integration site family, member 7a ZEA. . . ..Zearalenone xvi CHAPTER 1 RATIONALE, HYPOTHESIS AND OBJECTIVES 1.1 RATIONALE Reproductive health is the most important fundamental factor for . preservation of the species. The reproductive system is delicately maintained by endogenous hormones such as the sex steroid hormones, estrogen and androgen. Estrogen and androgen are mainly produced by female and male gonads. They stimulate the development of female and male sex Characteristics during organ developmental and puberty and the production of female and male gametes by sexually matured gonads. Disruption of homeostasis by an exogenous substance, which can act as antagonist or agonist of endogenous hormones during the critical stage of development, may result in potential adverse effects on reproductive health. . Estrogen exerts its biological activities via binding to estrogen receptors (ER) in estrogen target tissues. Laboratory in Vitro assays and in vivo models demonstrated that selected xenobiotics and natural substances can bind to ER and exert estrogenic responses. Thus, chemicals are called estrogenic endocrine modulators (EEMs). There has been a great deal of attention and debate regarding the hypothesis that exposure, particularly in utero exposure, to EEMs might be capable of causing a spectrum of adverse reproductive and developmental effects in human and animals. EEMs can alter development of the endocrine system and of the organs that respond to endocrine signals in organisms indirectly exposed during prenatal and/or early postnatal life. The effects of exposure during development may be permanent and irreversible. Although the exact modes of action of EEMs remain unclear, evidence supporting the hypothesis that EEMs impact the endocrine system include: 1) reproductive tract abnormalities and Clear cell adenocarcinoma (CCA) of the vagina and cervix of human females exposed to the potent estrogenic drug diethylstilbestrol in utero, 2) decreased sperm production and altered sperm quality in laboratory animals exposed to EEMs, and 3) decreased fertility and reproductive tract abnormalities in wildlife populations exposed to EEMs at high concentration. Therefore, to assess the risk of exposure to EEMs on reproductive and developmental health in humans, long-term health consequences of exposure to EEMs during developmental periods need to be investigated. Additionally, it is important to compare the potential effects and modes of action between synthetic estrogenic compounds (e.g. diethylstilbestrol or DES and 17a-ethynyl estradiol or EE2) and naturally occurring phytoestrogens (e.g. genistein or GEN) for better assessing health risk. Therefore, in this study, EE2 (a major estrogenic component of oral contraceptives) and GEN (a phytoestrogen formed in soybeans) were selected to examine the potential adverse effects of EEMs on reproductive health. DES was selected as estrogenic positive control since its estrogenic effects on reproductive health have been well documented. To examine sperm production and quality, computer assisted semen analysis (CASA) was used. Egg and sperm fertilizing abilities were investigated by using an in-vitro fertilization assay. 1.2 HYPOTHESIS Gestational and lactational exposure Of adult female mice to estrogenic endocrine disruptors cause long-term alterations in ovulation and egg fertilizing ability in female offspring and alterations in testicular development and sperm quality in male offspring. 1.3 OBJECTIVES . Determine the effects of weak (GEN) and potent (DES and EE2) EEMs on murine ovulation and egg fertilizing ability following gestational and lactational exposure. 0 Determine the effects of weak (GEN) and potent (DES and EE2) EEMs on murine testicular development and sperm quality following gestational and lactational exposure. CHAPTER 2 LITERATURE REVIEW I. 2.1 ESTROGEN SYNTHESIS There are three endogenous estrogens, estrone (E1), 178-estradiol (E2), and estriol (E3). Estrogens are mainly synthesized in the gonad and also produced in many other tissues such as the adrenal cortex, adipose tissue, brain, and skin (1). Figure 2-1 illustrates the general scheme for synthesis of estrogens. The synthesis of estrogens requires Cholesterol and they can be synthesized de novo from intra—cellular lipid stores acquired from serum lipoproteins through the scavenger receptor B1 (SR-B1). Steroidogenesis is the passage of Cholesterol from the outer to the inner mitochondrial membrane, which is facilitated by steroidogenic acute regulatory protein (StAR). Within the inner mitochondrial membrane, cholesterol is cleaved into pregnenolone by the action of the side- Chain-Cleavage enzyme (P450scc). Pregnenolone enters the smooth endoplasmic reticulum (ER) where the remaining steroidogenic enzymes responsible for testosterone synthesis are located (2). Aromatase (CYP19) converts androstenedione or testosterone to E1 or E2. Steroid hormone biosynthesis is catalyzed by two major types of enzymes: cytochrome P450 (CYPs) and hydroxysteroid dehydrogenase (e.g. 3B—HSD and 17B—HSD) (3). Table 2-1 presents the major catalytic enzymes that are involved in steroidogenesis. Thus, biosynthesis of specific steroids is depended on quantitative expression of these two steroidogenic enzymes and their activity in specific cells comprising to the specific endocrine tissue. Furthermore, these two .Nm. .o E 9 95.90909 .o 05.09.906.20 9.02.00 8E>Ov 0090Eo.< 09000. 0.0 0.09.930 9.990909 .2 0.9.9.0000. 00E>Nc0 0.como.0.o.90 9.....0E0. 9: 0.9.3 Em. E23030. 0.Emm_aouc0 £095 9.. 0.9...0 9.0.8059... .Aoomomvav 9.5.9.0 wag—020-509.0003 9.. Co c0300 9: >0 05.9.0390 9:. 0050.0 0. 6.98.9.0 0:05:05. .m_.ucoc09.E .09.. 9: :.£.>> .Efiwv E90... .5530. 9:00 0.580.990 .3 00.9...09 .9.m.nE0E .m..9.9.09.E .09.. 9.. 9 .9:o 9.. E0... 8.93.9.0 .0 03009. 9.. 0. 0.39.09.06.90 .0 090 95...... .90. 9.... .Cmiwv Fm .9800. 09.0500 9: 59.0.... 9:90.09... E200 E9. 09.300... 00.90 0.0.. 8.2.0009... E2. o>oc 0.0 03.00550 9. :00 0.... 9.0 6.900.050 09.3.00. 00.990 .0 0.00550 9.... 0.8582990 .0 9.0.9509... 0..0E0...0m TN 0.39“. o fl 0 u 0 fl 0 fl o a“ nu fl 0 fl 0 fl 0 D D U n "v u U a U o 0 o u 0 fl 0 fl 0 “v 0 fl 9 fl 0 “v o n 0 fl 0 D a ”U a 0019 r a o .o.00..wm 0c0..mm. _ m m 00m: .. a a m 8 Hi0 .. a Owflhw a a a m _ 0:90.08005535 m. a g a m 0:0.0.mo.0c0.000.0>c00 N wax/0 oémm o o ... fl . 0:0.OE0E w on H mm LHOOEmU 0 fl 0 fl 0 fl 0 U a “- 0 fl 0 fl o "v 0 “v 0 fl 0 a” 0 U 0 fl 0 fl 0 fl 0 "v 0 flaw 0 fl 0 U . AU 0 U 0m 0 .w G o o n m o 0:0.OE0E (00.2006. Edam IIIIiIIlIlv 6.0.00.0ch m 0on Beccozooeu. fr 923 . 6.900.050 L 0:0.OE0E =00 Table 2-1. Enzymes involved in steroidogenesis Enzyme Gene Reaction P450 type Cholesterol side chain cleavage P450800 CYP1 1A (3 succesive monooxygenations) C-11 hydroxylation P450c11 CYP1 1 B1 018 hydroxylation C-18 hydroxylation P450018 CYP11BZ C-18 oxidation 1 7a-hyd roxylation P450017 CYP1 7 C17-C20 bond Cleavage P450c21 CYP21 C-21 hydroxylation Steroid ring-A aromatization P450arom CYP19 (3 succesive monooxygenations) Dehydrogenase type _ 3B-hydroxy-steroid dehydrogenation 3B HSD H3038 5-ene-4—ene isomerization 17B-HSD HSD17B 17B-hydroxy-steroid dehydrogenation (1 7-reductase) 1 7-keto-steroid reduction Catalyzes testosterone to sa-reductase SRD5A2 dihydrotestosterone enzymes are expressed not only in reproductive tissues (e.g. gonads and reproductive tract) but also in many other tissues (e.g., adipose tissue, liver, brain etc) that have the ability to convert Circulating precursors (e.g. androstenedione) into active hormones (e.g. testosterone or E2) (4-6). Variations in a certain enzyme activities are generally dependent on gene expression of an enzyme with three Characteristics (1) basal expression of the enzyme in the cell; (2) hormonal signal regulated expression; (3) tissue- and cell-specific expression. Cytochrome P450 constitutes a large superfamily of membrane bound oxygenases. P450 families 1, 2, 3 and 4 have function in detoxification of xenobiotic Chemicals by modifying substrates to become more polar forms prior to excretion and the metabolism of natural endogenous steroids. They are not substrate specific. Cytochrome P450 families 11, 17, 19, 21 and 27 are involved in the synthesis of steroid hormones and they are substrate specific. Among these P450 families, P450arom coded in Cyp19 gene irreversibly converts testosterone or androstenedione into E2. P450arom is a microsomal enzymatic complex composed of two proteins: a ubiquitous NADPH-cytochrome p450 reductase and a cytochrome P450 aromatase, which contains the heme and the steroid-binding site. Animal and epidemiological studies indicate that estrogens play important roles in development and function of several target tissues involved in cardiovascular performance, bone maintenance, homeostasis, and behavior by affecting cell proliferation and cell differentiation (7-11). Specifically, numerous studies and reviews have evaluated the effect Of estrogens on proper development and function of human and animal reproductive tracts (12-15). This chapter will review the general biological role of estrogen in reproduction. More specifically, estrogen synthesis in females and males, estrogen receptors, and modes of activation of estrogen will be discussed. Then, the effects of estrogen on normal ovarian and testicular development and function, spermatogenesis, oogenesis, fertilization, implantation, sex ratio, anogenital distance (AGD), puberty, and body weight will be reviewed. 2.1.1 ESTROGEN SYNTHESIS IN FEMALES In females, estrogens are mainly produced in the ovary and thus have been thought of as female sex steroid hormones. Around the ninth week of gestation in the human, estrogen synthesis occurs in the primitive ovary of the female fetus (16). The ovarian steroidogenesis pathway is presented in Figure 2- 2. Estradiol is mainly synthesized by ovarian granulosa cells Of the growing follicle (17). In the theca cells of the preovulatory follicle Of the ovaries, luteinizing hormone (LH) stimulates the theca cell via the adenylyl cyclase pathway, and low density lipoprotein (LDL) and high density lipoprotein (HDL) across the cell membrane of theca cells due to an increase in the expression of LDL and HDL receptors (SR-B1) on the cell membrane. With facilitation of StAR protein, intracellular cholesterol passes from the outer to the inner mitochondrial membrane. Cytochrome P450scc enzyme cleaves Cholesterol, which is transported within the inner mitochondrial membrane, into pregnenolone. Thus, the stimulated theca cell increases its synthesis of androstenedione. The androstenedione synthesized in the theca cells freely diffuses to the granulosa cells, whose aromatase activity has been stimulated by follicular stimulating hormone (FSH). FSH, also acting via the adenylyl cyclase pathway, stimulates the granulosa cell to prOduce aromatase. Aromatase converts androstenedione to E1 and then 17B-hydroxy steroid dehydrogenase (17B-HSD) converts E1 to E2 (18, 19). Production rates and serum concentrations of E2 vary dowsing the different stages of the menstrual cycle. During the last five days before ovulation in humans, E2 concentration rapidly increases from 30 to 40 up to 300 to 400 pg/ml and then decreases to approximately 100 pg/ml by the sixth day after ovulation (20). The E2 concentration increases to 200 pg/ml during the luteal phase, corresponding to the increase in progesterone, but it is not as high as during the follicular phase (21). During the menstrual cycle, E2 production varies cyclically, with the highest production rates and serum concentrations in the preovulatory phase (Table 2-2) (22). In addition to ovarian production of E1, E2 is converted to E1 by 17B-HSD in the liver and E1 sulfate by aromatase in the adrenal gland. During early pregnancy, maternal estrogen is synthesized in the maternal ovaries, but by seven weeks of gestation, the placenta produces estrogen (23, 24). With progression of pregnancy, maternal serum concentrations of E1 and E2 increase from 50 to 100 pg/ml up to 30,000 pg/ml and serum concentrations of E3 increase from 1.3 to 3.2 ng/ml at 20 weeks of gestation to 6.0 to 19.5 ng/ml at 42 weeks of gestation (20). In perimenopausal or menopausal women (the mean age of menopause is 51 years in Europe), the serum concentrations of E2 are frequently below 20 pg/ml since depletion of ovarian follicles leads to a steady decline in ovarian E2 production (25) (Figure 2-3). Because the negative feedback effects of E2 on pituitary gonadotropin secretion are lost, semm FSH and LH concentrations are significantly increased. Peripheral aromatization increases 3- to 4-fold and becomes the main source of estrogen in menopausal women (26). In several studies, aromatase activity has also been detected in adipose-containing tissues ..omm. ”00.30w 0000.50 60.00.93. ...... 0.0.0.000: 5.0000 .50. ...n... 00000690000 0.0.0.0085? .Qw... 009050 60..0.3E..0 0.0...0. .Iwn. 900000000000. 00.000000 2.9.0 .0526 90000000... 00.000000 .n....< 000.05 330000 .O< .Nm. 0. .m 000.000 690.005 00000690300 0.0.0.0 90.0.3-0: 000. 000 :m. o. 000.005.00.000 0...0>000 000.9005 00 .r .000.0E0.0 00300.0 9:00 000.3006 9.. 090.3800 $030.00 000.0:0 ......0000 00. 0.> 60:00 00.0 .19. .Iwu. >0 00.0.3E..0 0000 000 >..>..00 000.0E0.0 00003 .250 000.3006 00. 0. 003.00 000.. 0. 0:00 009.. 00. 0. 000000.00 000.005.00.000 9.. 000300 ..00 0000. 00. 0. 00900.90 .0 0.00.. 002000909000 .0 0.000.000 0.. 00000.00. ..00 0000. 00. 090.3600 030 .. 0005000690 0.0. 00900.06 .0..000000..E .000. 00. 0.0....) 00.50009. 0. 00.0.5 5.900.000 00>00.0 000.000 000...va 00 0. 00900.08 .0..000000..0. .000. 00. 0. .930 00. E0.. 0630.0. 000000 5.900.000 .0.3..000..0. 0.0.0.0 02$ .0 00..0....00. 0.05 .0090E0E ..00 00. 00 Swim. 0.0.0000. ..o... 000 ..n... .0 5.00900 00000.00. 0. 030 0:00 0000. .0 00900.00 ..00 00. 0.0. 000.0 00... 000 ..n... 000 $030.00 000.05 ...:0000 00. 0.> ..00 0000. 00. 00.0.3E..0 I.. 00005 00. .0 0.2.5. 00.0.3590 00. .0 0:00 0000. 00. 0. .0.0000600.0.90 00..0>O .N-N 0.36.“. Imu- . =00 .000.-3:000 i, jil :00 0005.0 i _0_Uw._u0m_ iilil Il\ 00.0.0u00u00... M0 \\\.Q / DWI: 000.0050?! fi\\ .J@. 0000:: P \ /«\\. \i/ 0:0.u0w \\ / U< . 0C0..0u00u00.r (Hibii/ / N \\ 0C0..00C0u00..uc< A.\ 005“ [V 009.000.0010. CWT—I \ \\ 00 //Al\i 8020 F \\ a ““5303... u \ . \ 0:00.0«000000. / l\\.. 0:0.UOC/0u000UC< kl IOIGSQENI r ,i . .i A/ QF__ 0.001 pmol/I of genistein for 30 min. In the same study, sperm fertilizing ability was significantly increased in the 10 pmollL E2 treatment, compared with untreated control sperm (76 vs. 36%, respectively). Furthermore, the influence exerted by estrogen on maturation of sperm has been investigated in ERKO mice and was found to decrease motility and fertilizing potential (101 ). Estrogen has an important role in the implantation of the blastocysts in the uterus and its action is mediated by the progesterone receptor (PR) and growth factors at the local sites. In the wild-type mouse uterus, where ERor predominates estradiol treatment induces both mRNA and protein levels of the progesterone receptor in the stroma, and down-regulates PR expression in the uterine epithelium. In the aERKO females, up-regulation of the PR in the uterine stroma 38 occurred (159). This suggests that estrogen has important roles in implantation by regulating the progesterone receptor in the uterus. The successful growth and development of the fetus are dependent on the balance of endocrinological events after implantation. Approximately 8 to 12% of all pregnancy losses in humans are the result of endocrine factors (160). Prior to four weeks of gestation, the main source of estrogen in maternal Circulation is the maternal ovaries but around seven weeks of gestation, the main source of both maternal and fetal estrogen is the placenta (23). During pregnancy in humans and primates, maternal serum estrogen concentration increase from 50 to 100 pg/ml to 30,000 pg/ml at birth (20). The major role of estrogen is to maintains the endometrium by enhancing uteroplacental blood flow and to regulate the production of progesterone throughout the pregnancy by stimulating receptor- mediated uptake of LDL increase the activity of placental mitochondrial Cholesterol side-chain Cleavage enzyme (P450scc) (161-163). A study demonstrated that estrogen administration (5 mglanimal by interamuscular injection on gestational day 9 and 10), before the conceptus secretes estrogen on gestational day 12, alters the pattern of IGF gene expression through the nuclear factor kappa B (NF-KB) system desynchronizing the uterine environment for conceptus implantation resulting in later embryonic loss in pigs (164). These studies indicate that estrogen serves as the signal for maternal recognition and maintenance of pregnancy but at the same time, timing of endometrial exposure to estrogen is critical to embryonic survival. 39 2.9 EFFECTS OF ESTROGEN ON SEX RATIO OF OFFSPRING It has been hypothesized that the mammalian sex ratio at birth (male births/total births) is influenced by biological factors (e.g., high level of parental sex steroid hormones at the time Of conception, excision of accessory sex glands in rodents) and environmental factors (e.g., exposure to dioxin) (165-169). Review studies by James indicated that high concentrations of LH at the time of conception may result in more female Offspring, while high levels of estrogen, testosterone and progesterone may be associated with male offspring (168, 170- 172). Furthermore, he suggested that the same hormones may affect the sex ratio in opposite directions in different species and more male offspring are produced as a result of suboptimal health of the parents (173). Based on James’s hypothesis, EEMs may have potential effects in altering the sex ratio in both humans and wildlife species. However, there are no Clear answers for why different hormone concentrations cause variations of the sex ratio and the possible mechanisms underlying his hypothesis (174). In addition to biological factors, epidemiologic studies demonstrated that some environmental contaminants such as 2,3,7,8-tetraChlorochlibenzo-p-dioxin (TCDD), hexachlorobenzene (HCB), dibromochloropropane (DBCP) and Clomiphene Citrate may be associated with the decline in the sex ratio in humans that has been observed in some western countries (166, 169, 175-177). It is known that these compounds have estrogenic and antiestrogenic properties, thus altering the sex ratio via Changing parental biological factors. For example, high serum concentrations of TCDD in fathers exposed during the Seveso accident in 40 1976, were significantly associated with a decline in sex ratio up to eight years after the incident in two areas having the highest levels of contamination (Meda and Seveso) (P = 0.008) (178). The effect of TCDD on the sex ratio was detected at serum concentration lower than 20 ng/kg body weight in the fathers who were exposed to TCDD (166). Furthermore, studies suggest that exposure to high concentration of TCDD may decrease male offspring for up to 10 years, but this effect diminished over time. On the other hand, some studies related to dioxin-like Chemicals conducted in Taiwan and in European countries did not find any significant association between dioxin-like Chemicals and altered sex ratio or high concentration of pesticide or industrial pollution and the proportion of male births (177, 179, 180). Hexachlorobenzene is a fungicide used to control Tilletia tritiCi, a wheat based fungus. It is resistant to metabolic degradation and has an undetermined haIf-life in humans because of its intense lipophilicity (181). An epidemiological study in Turkey reported that women exposed to high concentrations of HCB between 1955 and 1957 had a significantly lower proportion of males than those exposed before 1950 or later 1970 (181). Based on in Vitro cytochrome P4501A induction, the potency of HCB is approximately 10,000 times less than dioxin. Furthermore, HCB binds to the aryl hydrocarbon receptor (AhR) with 10,000 times less affinity compared to dioxin (182). This suggests that HCB may act via the same mechanism Of action as TCDD to decrease the sex ratio. However, the mechanisms of TCDD and HCB in altering sex ratio need further investigation. 41 Dibromochloropropane, a pesticide, was used to control worms that cause damage to crops and other plants. It was banned in the US in 1977 because it was found to lower the sex ratio in DBCP-exposed male workers. For example, an epidemiological study in Israel reported that the decreased sex ratio was significantly associated with paternal DBCP exposure (P < 0.025)(183). Studies confirmed that DBCP causes infertility (e.g., azosperrnia), spontaneous abortion, and it has potential to cause tumors in the breast, lung and other organs in laboratory animals (184-187). Rats subcutaneously injected with DBCP had decreased testes weights and increased infertility with degenerated seminiferous tubules. Dibromochloropropane significantly increased gonadotrophin levels in DBCP-exposed agricultural workers who had azosperrnia and in infertile DBCP- treated laboratory rats (188, 189). However, serum testosterone concentrations were not different in either exposed humans or laboratory species, compared to control groups (183, 189). Corresponding to James’s hypothesis, high concentrations of gonadotrophin induced by exposure to DBCP may cause a decreased sex ratio in humans and laboratory rats (174). However, the mechanism of this phenomenon is also unknown. A potential mechanism of action of EEMs in altering sex-ratio could be through an alteration of the uptake of glucose in the female reproduction tract or in sperm after fertilization has occurred. It has been postulated that high concentrations of glucose in the reproductive tissues of both parents may increase the sex ratio. For example, the presence of glucose (5.56 mmol/l) in the culture medium when bovine embryos were cultured for 24 to 48 h after 42 insemination, lead to a higher percentage of Cleavage among the male embryos (190). In addition to increased male embryo Cleavage, glucose (5.56 mmOl/l) in the culture medium increased the number of acrosome reactions and sperm motility after 18 hours of capacitation of human sperm, compared to media containing no glucose (191). Furthermore, sperm motility in fertilization medium can be sustained by least 2.78 mM of glucose. Chemically induced diabetes in pregnant female mice resulted in maternal hyperglycemia and a significantly decreased sex ratio in their live offspring (192). Corresponding to these results, a meta-analysis study demonstrated that excess glucose in the maternal circulation around conception favors the development of male blastocysts during early embryo cell division (193). The other possible mode of EEMs in decreasing sex ratio may be via altering the maturation of the oocytes. A study investigated the effects of an in Vitro maturation culture period for bovine oocytes on the sex ratio of in Vitro produced blastocysts (194). An increase in the proportion of male blastocysts with an increase in maturation culture period (34h vs. 16 and 22h) Of cumulus- oocyte complexes was Observed (194). Studies demonstrated that the heads Of Y-bearing spermatozoa and their length were significantly smaller and their necks and tails were shorter than those of X-bearing ones (195). Therefore, differential migration of the Y- and X-bearing spermatozoa was suggested (196). However, published studies addressing possible association or effects of EEMs on mammalian sex ratios have still produced contradictory results because of unknown mechanisms. Further studies are needed to confirm the effects of 43 hormones on the sex-selection and to investigate biological mechanisms of the sex-selection. 2.10 EFFECTS OF ESTROGEN ON ANOGENITAL DISTANCE (AGD) Anogenital distance (AGD) is the distance between the anus and the base Of the external genitals. The AGD in males is approximately twice as long compared to females because testosterone produced by the gonads increases the length of the perineum separating the anus from the testicles via stimulation of cell proliferation (197, 198). The use of AGD has been explored as an indicator of prior androgen exposure due to intrauterine position (199). Corresponding to this, recent studies demonstrated that AGD is a sensitive indicator of demasculinization of male genitalia exposed to exogenous estrogen or antiandrogenic Chemicals during the prenatal developmental period (197, 200- 202). The reason is that the reproductive system is Changing rapidly at both the organ and molecular levels via hormonal stimulation during the prenatal period. Therefore, the consequences of hormonal effects are established for the duration of the individual’s life span. For example, testicular testosterone synthesis in humans begins approximately 65 days after fertilization and influences differentiation of the Wolffian duct system into the epididymis, vas deferens, and seminal vesicles (203). In the male rat, the growth rate of AGD in male rats is increased around gestational day 15, when testosterone synthesis begins (204). Clark et al. (205) confirmed that the growth rate of the AGD was controlled by dihydroxytestosterone rather than by testosterone. Consequently, EEMs that 44 interfere with the synthesis of dihydroxytestosterone or decrease its bioactivity can decrease the growth rate of AGD. Evan et al. (206) reported that GEN inhibits the activity of Soc-reductase in genital skin fibroblasts. A number of studies demonstrated that prenatal and postnatal exposure to EEM decreases the male AGD (12, 207-210). For example, a study demonstrated that estrogen inhibited human fetal penile smooth muscle cell proliferation in a dose-dependent manner from 10'12 mol/l, whereas tamoxifen acted as an antiestrogen to reverse the estrogen effect, in an in Vitro cellular model (211). Another well-known example of EEMs that decrease male AGD in laboratory animals and humans is phthalate esters, which are among the most extensively produced industrial Chemicals and used principally as plasticizers. to improve flexibility and durability in soft vinyl plastic toys, shampoos, soaps, nail polish, and vinyl flooring. The first reported evidence of their antiandrogenic effects on male AGD was Sohoni and Sumpter’s paper (212). In their study, rats were orally administered high doses Of phthalate esters (250, 500, and 750 mglkg bw/d) throughout gestation to postnatal day 20. Further epidemiologic studies confirmed that certain phthalates are significantly associated with reduced AGD in human male infants (207, 213-215). Among these epidemiologic sttidies, a study in the United State reported that the estimated median and 95th percentile of daily exposure to phthalates and its metabolites [di-n-butyl phthalate (DnBP), diethyl phthalate (DEP), butylbenzyl phthalate (BBzP), diisobutyl phthalate (DiBP), di-2-ethylhexyl phthalate (DEHP)] are approximately 9.45 and 126.77 pg/kg bw/d (213). These values are substantially 45 lower than the US EPA reference doses of 100 pg/kg bw/d for DBP, 800 pg/kg bw/d for DEP, 200 pg/kg bw/d for BBzP, and 20 pg/kg bw/d for DEHP. It suggests that phthalates have antiandrogenic effects on the AGD at low doses during the prenatal developmental period and that further risk assessments for these Chemicals are needed. Conversely, a known natural mycotoxin EEM, zearalenone, increases AGD in both male and female rat offspring maternally exposed to zearalenone (oral daily doses of 1 mg zearalenone/kg bw/d from gestational day 6 to 19) (216). This suggests that zearalenone has androgenic effects during prenatal development or has specific effects on the AGD. Additionally, bisphenol A, used to produce polycarbonate plastic and epoxy resins, is a known EEM in both in vivo and in Vitro assays. However, AGDs in female and male rat offspring exposed to bisphenol A (4 to 40 mglkg bw/d gestational day 6 to postnatal day 20) were not different, compared to the control groups (217). Our previous study also demonstrated that gestational and lactational exposure to the phytoestrogen, genistein, did not Change AGD in male and female mouse Offspring on postnatal day 21 (218) (female data shown in Chapter 4). Overall, these results suggest how complex the hormonal balance in developing animal fetuses is. Taken together, the AGD can be used as a sensitive biomarker of effects of EEMs altering the intrauterine androgen environment but several confounding factors should be considered including species differences, doses of EEMs, and the timing of exposure. 46 2.11 EFFECTS OF ESTROGEN ON PUBERTY Puberty is initiated by the activity of the hypothalamic pulse. In infancy and Childhood, the activity of the hypothalamic pulse doest not occur, and it increases slowly as reflected by the increase of the number and amplitude of LH and FSH pulses between the sixth and ninth year of life (219). The serum levels of estrogen in humans remain less than 12 pg/ml until the onset of puberty (approximately 7 years Of age), and increase up to 392 pg/ml in girls and 86 pg/ml in boys (20, 220). The activity Of the hypothalamic pulse during puberty is initiated by unknown mechanisms (221). Estrogen has been thought of as an activator of the GnRH pulse generator but studies demonstrated that estrogens are not activators Of the GnRH pulse generator but rather have modulating effects on gonadotropin release (222, 223). As a result of increased GnRH secretion from the hypothalamus, FSH from anterior pituitary increases intracellular CAMP concentrations via a G-protein-coupled receptor and it leads to an induction of the aromatase for estrogen synthesis (220). Under the influence of increased estrogen from gonads, ovaries, uterus, the fallopian tube, vagina, and breast glands grow and mature in females. The first menstrual bleeding, called menarche, is initiated by a temporary slight drop in estrogen concentrations, which leads to an estrogen withdrawal hemorrhage. In the pubertal boy, estrogen is produced in testicular tissue, and increased estrogen concentrations are much higher than those found before puberty (12 vs. 86 pg/ml) (20). Epidemiological studies reported that estrogen 47 deficiency due to mutations in the aromatase gene and estrogen resistance due to dismptive mutations in the estrogen receptor gene have no effect on normal male sexual maturation at puberty (224). There has been concern about precocious puberty. Even though there are several causes of this, including improved nutrition, exposure to EEMs could partly be associated with precocious puberty. Relationships between pubertal development and perinatal and postnatal exposure to EEMs (i.e., polychlorinated biphenyls, 1,1,1-trichloro-2,2-bis(p-Chlorophenyl)ethane, phthalate esters, furans and the pesticide endosulfan) have been reported (225). However, further study is needed to understand the mechanisms that link pubertal development and EEMs. 2.12 EFFECTS OF ESTROGEN ON BODY WEIGHT The increasing prevalence of obesity is a growing concern woridwide. The incidence Of Obesity has significantly increased in the US over the past two to three decades (226). According to the National Health and Nutrition Examination Survey (NHANES)1 in 1999 to 2002, 31% of adults aged at least 20 years were Obese and 5% were extremely obese with respect to children aged 6 through 19 years, 16% were overweight (227, 228). Since overweight adolescents have a 70% chance of becoming overweight or obese adults (80% if ‘ The National Health and Nutrition Examination Survey (NHANES) have been monitoring the national prevalence Of overweight and Obesity. Since 1960, NHANES data on measured height and weight have been used to determine obesity levels in the US. Body mass index (BMI) is calculated as weight in kilograms divided by the square of height in meters. 48 one parent is obese or overweight), the prognosis for the future health of Americans is declining (229). Based on dramatically increased obesity after the 19803 and an important role of estrogen in natural weight-control mechanisms, a hypothesis has been suggested that EEMs alter the body's natural weight-control mechanisms resulting in interference of the normal developmental and homeostatic controls over adipogenesis and energy balance. Epidemiological evidences has indicated that EEMs have a positive association with increased body weight in humans and laboratory animals (230, 231). The ERa (aERKO) and ERB (BERKO) knockout mice as well as the double ER knockout (orBERKO) mouse and the aromatase knockout (ArKO) mouse, have shown that lipid and carbohydrate phenotypes involve increased adiposity with insulin resistance, hyperiipidemia and hyperleptinemia (232-235). ERa knockout mice have a significantly increased body fat content with a 10-fold increase in E2 (235, 236). This suggests that EEMs, which can increase signaling ERB or block signaling ERa, may contribute to obesity in ERor knockout mice. A cross-sectional epidemiological study of two single-nucleotide polymorphisms (a T to C (Pvull) and an A to G (Xbal)) in the first intron of the ERor gene showed that there is no association between the ERor gene polymorphisms and body fat distribution in men, but women with the Xbal polymorphism may be predisposed to the development of upper-body obesity in middle-aged (40 to 59 years) individuals. However, older aged (68 to 69 years) 49 women with the Xbal polymorphism had decreased whole-body and abdominal fat tissue (237). These results indicate that the ERor has an active role in female upper-body obesity in an age-related manner. Generally, EEMs at high levels of exposure decrease body weight of laboratory animals but at much lower levels of exposure, they may increase body weight with no adverse effects (238). Low levels of EEMs have been widely measured since they have been used as pharmaceuticals ingredients (e.g., EE2), organochlorine pesticides (e.g., DDT), plasticizers (e.g., phthalate), and growth promoting hormones (e.g. zearalenone) in humans and livestock (239-242). Laboratory animal studies demonstrated that concentrations as low as 0.5 pg E2 caused approximately an approximate doubling of uterine wet weight in mice (243). Therefore, it is important to consider increased body weight as a low-dose effect of EEMs, which can be a potential contributor of Obesity. Most EEMs have lipophilic properties. Therefore, they can be bioaccumulated in body fat in relatively high concentrations and transfer across the fetal-matemal blood barrier. Consequently, the current obesity epidemic may be significantly associated with the level of EEM exposure in early life and in utero (226, 238). Suggested mechanisms of altering the natural body weight- control in early life and in utero are: (1) disruption of the major weight controlling hormones, such as thyroid hormones, estrogen, testosterone, corticosteroids, insulin, growth hormone, and leptin (2) alteration of the levels of, and sensitivity to, neurotransmitters (in particular dopamine, noradrenaline, and serotonin) (3) interference with many metabolic processes (4) induction of widespread damage 50 to body tissues (nerve and muscle tissue in particular), often at concentrations that human beings are currently exposed to. However, the extent to which each individual is affected could also be significantly related to a given individual’s genetic ability to deal with exposure to EEMs. Furthermore, it is known that decreased endogenous estrogen concentrations in postmenopausal women and in ovariectomized laboratory rodents cause increased adipose tissues in females (244, 245). Additionally, studies demonstrated that estrogen has direct effects on adipocytes to inhibit lipogenesis and on food consumption and energy expenditure that contribute to its inhibitory effect on adipose deposition in both females and males during the developmental period and adulthood (246, 247). Taken together, further research is needed to investigate which of the environmental estrogenic compounds is probably causing the greatest damage to the natural weight- Control system. 51 2.1 3 BIBLIOGRAPHY 1. 10. Nelson LR, Bultm SE 2001 Estrogen production and action. J Am Acad Derrnatol 45:81 16-124 Fisher JS 2004 Are all EDC effects mediated via steroid hormone receptors? Toxicology 205:33—41 Sanderson JT 2006 The steroid hormone biosynthesis pathway as a target for endocrine-disrupting Chemicals. Toxicol Sci 94:3—21 Meseguer A, Puche C, Cabero A 2002 Sex steroid biosynthesis in white adipose tissue. Horm Metab Res 34:731-736 Kershaw EE, Flier JS 2004 Adipose tissue as an endocrine organ. J Clin Endocrinol Metab 89:2548-2556 Roselli CE, Resko JA, Stormshak F 2003 Estrogen synthesis in fetal sheep brain: effect of maternal treatment with an aromatase inhibitor. Biol Reprod 682370-374 Dubey RK, Tofovic SP, and Jackson EK. 2004 Cardiovascular pharmacology of estradiol metabolites. J Phannacol Exp Ther 308:403- 409. Hess RA, Bunick D, Lee KH, Bahr J, Taylor JA, Korach KS, and Lubahn DB. 1997 A role for oestrogens in the male reproductive system. Nature 390:509-512. Jones ME, Thorbum AW, Britt KL, Hewitt KN, Wreford NG, Proietto J, Oz OK, Leury BJ, Robertson KM, Yao S, and Simpson ER. 2000 Aromatase-deficient (ArKO) mice have a phenotype Of increased adiposity. PrOC Natl Acad Sci U S A 97:12735-12740. Jones ME, Thorbum AW, Britt KL, Hewitt KN, Misso ML, Wreford NG, Proietto J, Oz OK, Leury BJ, Robertson KM, Yao S, and Simpson ER. 2001 Aromatase-deficient (ArKO) mice accumulate excess adipose tissue. J Steroid Biochem Mol Biol 79:39 52 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Migliaccio S, Newbold RR, Bullock BC, Jefferson WJ, Sutton FG Jr, McLachIan JA, and Korach KS. 1996 Alterations of maternal estrogen levels during gestation affect the skeleton of female offspring. Endocrinology 137:21 18-2125. Mylchreest E, Cattley RC, Foster PM 1998 Male reproductive tract maiforrnations in rats following gestational and lactational exposure to Di(n-butyl) phthalate: an antiandrogenic mechanism? Toxicol Sci 43:47-60 Carreau S, Genissel C, Bilinska B, Levallet J 1999 Sources of oestrogen in the testis and reproductive tract of the male. Int J Androl 22:211-223. Hess RA, Bunick D, Bahr J 2001 Oestrogen, its receptors and function in the male reproductive tract - a review. Mol Cell Endocrinol 178229-38. Yin Y, Ma L 2005 Development of the mammalian female reproductive tract. J Biochem (Tokyo) 137:677-683 Jones EJ, DeChemey AH 2003 The female reproductive system. In: Boron WF, Boulpaep EL eds. Medical physiology. Philadelphia: Saunders; 1141-1208 Dubey RK, Tofovic SP, Jackson EK 2004 Cardiovascular pharmacology of estradiol metabolites. J Pharmacol Exp Ther 308:403-409. Hillier SG, Whitelaw PF, Smyth CD 1994 Follicular oestrogen synthesis: the 'two-cell, two-gonadotrophin' model revisited. Mol Cell Endocrinol 100251-54 Erickson GF 1978 Normal ovarian function. Clin Obstet Gynecol 21 :31-52 Dotsch J, Dorr HG, Wildt L 2001 Exposure to endogenous estrogens during lifetime. In: Hutzinger Oe, Metzler M eds. Endocrine disruptors part I. New York: Springer-Verlag Berlin Heidelberg New York; 83-98 Lenton EA 1988 Pituitary and ovarian hormones in implantation and early pregnancy. In: Chapman M, Grudzinskas G, Chard T eds. Implantation. Heidelberg: Springer; 17 53 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. Baird DT, Fraser IS 1974 Blood production and ovarian secretion rates Of estradiol-17 beta and estrone in women throughout the menstrual cycle. J Clin Endocrinol Metab 38: 1 009—1 01 7 Siiteri PK, MacDonald PC 1966 Placental estrogen biosynthesis during human pregnancy. J Clin Endocrinol Metab 26:751-761 MacDonald PC, Siiter PK 1965 Origin of estrogen in women pregnant withan anencephalic fetus. J Clin Invest 44:465-474 Dotsch J, Dorr HG, Wildt L 2001 Exposure to endogenous estrogens during lifetime. In: Metzler M ed. Endocrine disruptors part I. New York: Springer; 81-99 Brodie AM 1979 Recent advances in studies on estrogen biosynthesis. J Endocrinol Invest 2:445-460 Grow DR 2002 Metabolism of endogenous and exogenous reproductive hormones. Obstet Gynecol Clin North Am 29:425-436 Simpson ER, Zhao Y, Agarwal VR, Michael MD, Bulun SE, Hinshelwood MM, Graham-Lorence S, Sun T, Fisher CR, Qin K, Mendelson CR 1997 Aromatase expression in health and disease. Recent Prog Horm Res 52:185-213. Simpson ER 2004 Aromatase: biologic relevance of tissue-specific expression. Semin Reprod Med 22:11-23. Jakob F, Homann D, Seufert J, Schneider D, Kohrle J 1995 Expression and regulation of aromatase cytochrome P450 in THP 1 human myeloid leukaemia cells. Mol Cell Endocrinol 110:27-33 Naftolin F, Ryan KJ, Davies IJ, Petro Z, Kuhn M 1975 The formation and metabolism of estrogens in brain tissues. Adv Biosci 15:105-121 Simpson ER 2003 Sources of estrogen and their importance. J Steroid Biochem Mol Biol 86:225-230 54 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. Simpson ER 2003 Sources of estrogen and their importance. J Steroid Biochem Mol Biol 862225-230 Miller WR 1991 Aromatase activity in breast tissue. J Steroid Biochem Mol Biol 39:783—790 Matsumine H, Hirato K, Yanaihara T, Tamada T, Yoshida M 1986 Aromatization by skeletal muscle. J Clin Endocrinol Metab 632717-720 Miller WR 1991 Aromatase activity in breast tissue. J Steroid Biochem Mol Biol 392783-790 Schlinger BA, Arnold AP 1991 Brain is the major site of estrogen synthesis in a male songbird. Proc Natl Acad Sci 88:4191-4194 Patchev AV, Gotz F, Rohde W 2004 Differential role of estrogen receptor isoforrns in sex-specific brain organization. The FASEB Journal Express Article Kula K, Walczak-Jedrzejowska R, SIowikowska-Hilczer J, Wranicz JK, Kula P, Oszukowska E, Marchlewska K 2005 Important functions of estrogens in mew-breakthrough in contemporary medicine. Przegl Lek 622908-915 Payne AH, Kelch RP, Muslch SS, Halpem ME 1976 lntratesticular site of aromatization in the human. J Clin Endocrinol Metab 42:1081-1087 Carreau S 1996 Paracrine control of human Leydig cell and Sertoli cell functions. Folia Histochem Cytobiol 34:111-119 Pomerantz DK 1979 Effects of in vivo gonadotropin treatment on estrogen levels in the testis Of the immature rat. Biol Reprod 2121247-1255 Hess RA, Bunick D, Lee KH, Bahr J, Taylor JA, Korach KS, Lubahn DB 1997 A role for oestrogens in the male reproductive system. Nature 390:509-512 55 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. Hess RA 2003 Estrogen in the adult male reproductive tract: A review. Reprod Biol Endocrinol 1252. Pepe GJ, Albrecht ED 1995 Actions of placental and fetal adrenal steroid hormones in primate pregnancy. Endocr Rev 162608-648 Janulis L, Bahr JM, Hess RA, Janssen S, Osawa Y, Bunick D 1998 Rat testicular germ cells and epididymal sperm contain active P450 aromatase. J Androl 19:65-71. Lambard S, Carreau S 2005 Aromatase and oestrogens in human male germ cells. Int J Androl 282254-259 Weniger JP 1993 Estrogen production by fetal rat gonads. J Steroid Biochem Mol Biol 44:459-462 Delbes G, Levacher C, Habert R 2006 Estrogen effects on fetal and neonatal testicular development. Reproduction 132:527-538 Gennari L, Nuti R, Bilezikian JP 2004 Aromatase activity and bone homeostasis in men. J Clin Endocrinol Metab 89:5898-5907 Levine AC, Kirschenbaum A, Gabrilove JL 1997 The role of sex steroids in the pathogenesis and maintenance of benign prostatiC hyperplasia. Mt Sinai J Med 64220-25 Dorrington JH, Fritz IB, Armstrong DT 1978 Control of testicular estrogen synthesis. Biol Reprod 18:55-64 Free MJ, Jaffe RA 1979 Collection of rete testis fluid from rats without previous efferent duct ligation. Biol Reprod 20:269-278 Brewster ME, Anderson WR, Pop E 1997 Effect of sustained estradiol release in the intact male rat: correlation of estradiol serum levels with actions on body weight, serum testosterone, and peripheral androgen- dependent tissues. Physiol Behav 61 :225-229 56 55. 56. 57. 58. 59. 60. 61. 62. 63. Nagata C, Takatsuka N, Shimizu H, Hayashi H, Akamatsu T, Murase K 2001 Effect of soymilk consumption on serum estrogen and androgen concentrations in Japanese men. Cancer Epidemiol Biomarkers Prev 10:179-184 Weinstein RL, Kelch RP, Jenner MR, Kaplan SL, Grumbach MM 1974 Secretion of unconjugated androgens and estrogens by the normal and abnormal human testis before and after human Chorionic gonadotropin. J Clin Invest 53:1-6 Akingbemi BT 2005 Estrogen regulation of testicular function. Reprod Biol Endocrinol 3:51 Klinge CM 2000 Estrogen receptor interaction with co-activators and CO- repressors. Steroids 652227-251 Pettersson K, Grandien K, Kuiper GG, Gustafsson JA 1997 Mouse estrogen receptor beta forms estrogen response element-binding heterodimers with estrogen receptor alpha. Mol Endocrinol 1121486-1496 Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA 1996 Cloning of a novel estrogen receptor expressed in rat prostate and ovary. Proc Natl Acad Sci 93:5925-2930 Kurebayashi J, Otsuki T, Kunisue H, Tanaka K, Yamamoto S, Sonoo H 2000 Expression levels of estrogen receptor-alpha, estrogen receptor- beta, coactivators, and corepressors in breast cancer. Clin Cancer Res 6:512-518 Glaros S, Atanaskova N, Zhao C, Skafar DF, Reddy KB 2006 Activation Function -1 domain Of estrogen receptor regulates the agonistic and antagonistic actions of Tamoxifen. MOI Endocrinolepub ahead of print Kobayashi Y, Kitamoto T, Masuhiro Y, Watanabe M, Kase T, Metzger D, Yanagisawa J, Kato S 2000 p300 mediates functional synergism between AF-1 and AF-2 of estrogen receptor alpha and beta by interacting directly with the N-terminal A/B domains. J Biol Chem 275:15645-15651 57 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. Osborne CK, Zhao H, Fuqua SA 2000 Selective estrogen receptor modulators: structure, function, and Clinical use. J Clin Oncol 1823172- 3186 Enmark E, Gustafsson JA 1999 Oestrogen receptors - an overview. J lntem Med 246:133-138 Pettersson K, Gustafsson JA 2001 Role of estrogen receptor beta in estrogen action. Annu Rev Physiol 63:165—192 Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS, Smithies O 1993 Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci U S A 90:11162-11166 Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, Sar M, Korach KS, Gustafsson JA, Smithies O 1998 Generation and reproductive phenotypes Of mice lacking estrogen receptor beta. Proc Natl Acad Sci U S A 95:15677-15682 Dupont S, Krust A, Gansmuller A, Dierich A, Chambon P, Mark M 2000 Effect of single and compound knockouts of estrogen receptors alpha (ERaIpha) and beta (ERbeta) on mouse reproductive phenotypes. Development 127:4277—4291 Pelletier G 2000 Localization of androgen and estrogen receptors in rat and primate tissues. Histol Histopathol 15:1261-1270 Fitzpatrick SL, Funkhouser JM, Sindoni DM, Stevis PE, Deecher DC, Bapat AR, Merchenthaler I, Frail DE 1999 Expression of estrogen receptor-beta protein in rodent ovary. Endocrinology 140:2581-2591 Hall J, McDonnell DP 1999 The estrogen receptor beta-isoforrn (ERbeta) of the human estrogen receptor modulates ERalpha transcriptional activity and is a key regulator of the cellular response tO estrogens and antiestrogens. Endocrinology 140:5566-5578. Beato M, Klug J 2000 Steroid hormone receptors: an update. Hum Reprod Update 6:225-236 58 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. Suen CS, Berrodin TJ, Mastroeni R, Cheskis RJ, Lyttle R, Frail DE 1998 A transcriptional coactivator, steroid receptor coactivator-3, selectively augments steroid receptor transcriptional activity. The Journal Of Biological Chemistry 273:27645—27653 Smith CL 1998 Cross-talk between peptide growth factor and estrogen receptor signaling pathways. Biol Reprod 58:627—632 Hall JM, Couse JF, Korach KS 2001 The multifaceted mechanisms of estradiol and estrogen receptor signaling. J Biol Chem 276:36869-36872 Ohmichi M, Tasaka K, Kurachi H, Murata Y 2005 Molecular mechanism of action of selective estrogen receptor modulator in target tissues. Endocr J 52:161-167 Kato S 2001 Estrogen receptor-mediated cross-talk with growth factor signaling pathways. Breast Cancer 8:3—9 Tremblay A, Tremblay GB, Labrie F, Giguere V 1999 Ligand- independent recruitment of SRC—1 to estrogen receptor beta through phosphorylation Of activation function AF-1. Mol Cell 3:51 3-519 Dutertre M, Smith CL 2003 Ligand-independent interactions Of p160/steroid receptor coactivators and CREB-binding protein (CBP) with estrogen receptor-alpha: regulation by phosphorylation sites in the NB region depends on other receptor domains. Mol Endocrinol 17:1296-1314 Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, Scanlan TS 1997 Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science 277:1508-1510 Cheung E, Acevedo ML, Cole PA, Kraus WL 2005 Altered pharmacology and distinct coactivator usage for estrogen receptor- dependent transcription through activating protein-1. Proc Natl Acad Sci U S A 102:559-564 Matthews J, Wihlen B, Tujague M, Wan J, Strom A, Gustafsson JA 2005 Estrogen receptor (ER) beta modulates ERalpha-mediated transcriptional activation by altering the recruitment of C-Fos and C-Jun to estrogen-responsive promoters. Mol Endocrinol 20:534-543 59 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. Krebs CJ, Jarvis ED, Pfaff DW 1999 The 70-kDa heat shock cognate protein (Hsc73) gene is enhanced by ovarian hormones in the ventromedial hypothalamus. Proc Natl Acad Sci U S A 96:1686—1691 Voss MR, Stallone JN, Li M, Comelussen RN, Knuefermann P, Knowlton AA 2003 Gender differences in the expression of heat shock proteins: the effect of estrogen. Am J Physiol Heart Circ Physiol 285:H687-692 Nelson J, Clarke R, Murphy RF 1986 The unoccupied estrogen receptor: some comments on localization. Steroids 482121-124 Falkenstein E, Wehling M 2000 Nongenomically initiated steroid actions. Eur J Clin Invest 30:51-54 Falkenstein E, Tillmann HC, Christ M, Feuring M, Wehling M 2000 Multiple actions of steroid hormones—a focus on rapid, nongenomic effects. Pharmacol Rev 522513-556 Falkenstein E, Norman AW, Wehling M 2000 Mannheim Classification of nongenomically initiated (rapid) steroid action(s). J Clin Endocrinol Metab 85:2072-2075 Revelli A, Massobrio M, Tesarik J 1998 Nongenomic actions of steroid hormones in reproductive tissues. Endocr Rev 1923-17 Luconi M, Francavilla F, Porazzi I, Macerola B, Forti G, Baldi E 2004 Human spermatozoa as a model for studying membrane receptors mediating rapid nongenomic effects of progesterone and estrogens. Steroids 692553-559 Moats RKn, Ramirez VD 1998 Rapid uptake and binding of estradiol- 17beta-6-(O-carboxymethyl)oxime:125l-labeled BSA by female rat liver. Biol Reprod 582531-538 Boulware MI, Mermelstein PG 2005 The influence of estradiol on nervous system function. Drug News Perspect 182631 -637 60 94. 95. 96. 97. 98. 99. 100. 101. 102. Pepe GJ, Billiar RB, Leavltt MG, Zachos NC, Gustafsson JA, Albrecht ED 2002 Expression of estrogen receptors alpha and beta in the baboon fetal ovary. Biol Reprod 66:1054-1060 Vaskivuo TE, Maentausta M, Torn S, Oduwole O, Lonnberg A, Herva R, Isomaa V, Tapanainen JS 2005 Estrogen receptors and estrogen- metabolizing enzymes in human ovaries during fetal development. J Clin Endocrinol Metab 90:3752-3756 Brandenberger AW, Tee MK, Jaffe RB 1998 Estrogen receptor alpha (ER-alpha) and beta (ER-beta) mRNAs in normal ovary, ovarian serous cystadenocarcinoma and ovarian cancer cell lines: down-regulation of ER- beta in neoplastic tissues. J Clin Endocrinol Metab 83:1025-1028 Okada A, Ohta Y, Buchanan DL, Sato T, Inoue S, Hiroi H, Muramatsu M, Iguchi T 2002 Changes in ontogenetic expression of estrogen receptor alpha and not of estrogen receptor beta in the female rat reproductive tract. J Mol Endocrinol 28:87-97 Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS, Smithies O 1993 Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. PrOC Natl Acad Sci USA 90:11162-11166 Couse JF, Hewitt SC, Bunch DO, Sar M, Walker VR, Davis BJ, Korach KS 1999 Postnatal sex reversal of the ovaries in mice lacking estrogen receptors alpha and beta. Science 286:2328-2331 Britt KL, Findlay JK 2002 Estrogen actions in the ovary revisited. J Endocrinol 175:269-276 Couse JF, Korach KS 1999 Estrogen receptor null mice: what have we Ieamed and where will they lead us? Endocr Rev 20:358—41 7 Byskov AG 1994 Embryology of mammalian gonads and ducts. In: Knobil E, Neill JD eds. The physiology of reproduction. New York: Raven Press; 487-540 61 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. Gosden RG, Bownes M 1995 Cellular and molecular aspects of oocyte development. In: Grudzinskas JG, YoviCh JL eds. The oocyte. Cambridge: Cambridge reviews in human reproduction; 25-53 Couse JF, Yates MM, Deroo BJ, Korach KS 2005 Estrogen receptor- beta is critical to granulosa cell differentiation and the ovulatory response to gonadotropins. Endocrinology 146:3247-3262 Drummond AE 2006 The role Of steroids in follicular growth. Reprod Biol Endocrinol 4216 Huynh K, Jones G, Thouas G, Britt KL, Simpson ER, Jones ME 2004 Estrogen is not directly required for oocyte developmental competence. Biol Reprod 70:1263—1269. de Kretser DM, Kerr JB 1988 The cytology of the testis. In: Knobil E NJ ed. The Physiology of Reproduction. New York: Raven Press Ltd; 837-932 Byskov AG 1986 Differentiation of mammalian embryonic gonad. Physiol Rev 66:71-117 Winter JSD, Faiman C, Reyes Fl 1977 Sex steroid production by the human fetus: its role in morphogenesis and control by gonadotropins. In: Blandau RJ, Bergsma D eds. Morphogenesis and Malformation of the Genital System. New York, NY: AR Liss; 41 -58 Sar M, Welsch F 2000 Oestrogen receptor alpha and beta in rat prostate and epididymis. Andrologia 32:295-301 Fisher JS, Millar MR, Majdic G, Saunders PT, Fraser HM, Sharpe RM 1997 lmmunolocalisation of oestrogen receptor-alpha within the testis and excurrent ducts of the rat and marmoset monkey from perinatal life to adulthood. J Endocrinol 153:485-495 Yamashita S 2004 Localization of estrogen and androgen receptors in male reproductive tissues of mice and rats. Anat Rec A Discov Mol Cell Evol Biol 279:768-778 62 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. Hughes lA, Coleman N, Faisal Ahmed S, Ng KL, Cheng A, Lim HN, Hawkins JR 1999 Sexual dimorphism in the neonatal gonad. Acta Paediatr Suppl 88:23-30 Gustafson ML, Donahoe PK 1994 Male sex determination: current concepts of male sexual differentiation. Annu Rev Med 45:505—524 Greco TL, Furlow JD, Duello TM, Gorski J 1992 lmmunodetection of estrogen receptors in fetal and neonatal male mouse reproductive tracts. Endocrinology 1 30:421-429 Carreau S, Lambard S, Delalande C, Denis-Galeraud l, Bilinska B, Bourguiba S 2003 Aromatase expression and role of estrogens in male gonad : a review. Reprod Biol Endocrinol 11:35. O'Donnell L, Robertson KM, Jones ME, Simpson ER 2001 Estrogen and sperrnatogenesis. Endocr Rev 222289-318 Nielsen M, Bjomsdottir S, Hoyer PE, Byskov AG 2000 Ontogeny of oestrogen receptor alpha in gonads and sex ducts of fetal and newborn mice. J Reprod Fertil 1 18:195-204 Majdic G, Millar MR, Saunders PT 1995 lmmunolocalisation of androgen receptor to interstitial cells in fetal rat testes and to mesenchymal and epithelial cells of associated ducts. J Endocrinol 147:285-293 Cooke PS, Young P, Hess RA, Cunha GR 1991 Estrogen receptor expression in developing epididymis, efferent ductules, and other male reproductive organs. Endocrinology 128:2874-2879 Shughrue PJ, Lane MV, Scrimo PJ, Merchenthaler I 1998 Comparative distribution of estrogen receptor-alpha (ER-alpha) and beta (ER-beta) mRNA in the rat pituitary, gonad, and reproductive tract. Steroids 63:498- 504 Pelletier G, Labrie C, Labrie F 2000 Localization of oestrogen receptor alpha, oestrogen receptor beta and androgen receptors in the rat reproductive organs. J Endocrinol 165:359-370 63 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. van Pelt AM, do Rooij DG, van der Burg B, van der Saag PT, Gustafsson JA, Kuiper GG 1999 Ontogeny of estrogen receptor-beta expression in rat testis. Endocrinology 140:478-483 Weniger JP, Zeis A 1987 Oestrogen synthesis by the foetal rat testis in organ culture. J Steroid Biochem 28:307-310 Lambard S, Silandre D, Delalande C, Denis-Galeraud I, Bourguiba S, Carreau S 2005 Aromatase in testis: expression and role in male reproduction. J Steroid Biochem Mol Biol 95:63-69 Janulis L, Bahr JM, Hess RA, Bunick D 1996 P450 aromatase messenger ribonucleic acid expression in male rat germ cells: detection by reverse transcription-polymerase chain reaction amplification. J Androl 17:651-658. Eddy EM, Washbum TF, Bunch DO, Goulding EH, Gladen BC, Lubahn DB, Korach KS 1996 Targeted disruption of the estrogen receptor gene in male mice causes alteration of sperrnatogenesis and infertility. Endocrinology 137:4796-4805 Robertson KM, O'Donnell L, Jones ME, Meachem SJ, Boon WC, Fisher CR, Graves KH, McLachIan RI, Simpson ER 1999 Impairment of sperrnatogenesis in mice lacking a functional aromatase (cyp 19) gene. Proc Natl Acad Sci U S A 96:7986-7991. Derelanko MJ 2000 Toxicologists's pocket handbook. In. Boca Raton, Florida: CRC Press; 85-122 Jones EE, DeChemey AH 2003 The male reproductive system. In: Boron WF, Boulpaep EL eds. Medical physiology. Philadelphia: Saunders; 1122- 1 140 Fisher CR, Graves KH, Parlow AF, Simpson ER 1998 Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the cyp19 gene. Proc Natl Acad Sci U S A 95:6965-6970 Robertson KM, O'Donnell L, Jones ME, Meachem SJ, Boon WC, Fisher CR, Graves KH, McLachIan RI, Simpson ER 1999 Impairment of 64 133. 134. 135. 136. 137. 138. 139. 140. 141. sperrnatogenesis in mice lacking a functional aromatase (cyp 19) gene. Proc Natl Acad Sci U S A 96:7986-7991 Nitta H, Bunick D, Hess RA, Janulis L, Newton SC, Millette CF, Osawa Y, Shizuta Y, Toda K, Bahr JM 1993 Germ cells of the mouse testis express P450 aromatase. Endocrinology 132:1396-1401 Sharpe RM, Skakkebaek NE 1993 Are oestrogens involved in falling sperm counts and disorders of the male reproductive tract? Lancet 341 :1392-1395 Macomber D, Sanders M 1929 The spermatozoa count: Its value in the diagnosis, prognosis, and treatment of sterility. N Engl J Med 200:981-984 Eskenazi B, Wyrobek AJ, Sloter E, Kidd SA, Moore L, Young S, Moore D 2003 The association of age and semen quality in healthy men. Hum Reprod 18:447-454 Fisch H, Goluboff ET, Olson JH, Feldshuh J, Broder SJ, Barad DH 1996 Semen analyses in 1,283 men from the United States over a 25-year period: no decline in quality. Fertil Steril 65:1009-1014 Giwercman A, Carlsen E, Keiding N, Skakkebaek NE 1993 Evidence for increasing incidence of abnormalities of the human testis: a review. Environ Health Perspect 101 :65-71 Toppari J, Skakkebaek NE 1998 Sexual differentiation and environmental endocrine disrupters. Baillieres Clin Endocrinol Metab 12:143-156 Kula K 1988 Induction of precocious maturation of sperrnatogenesis in infant rats by human menopausal gonadotropin and inhibition by simultaneous administration of gonadotropins and testosterone. Endocrinology 122:34-39 Li H, Papadopoulos V, Vidic B, Dym M, Culty M 1997 Regulation of rat testis gonocyte proliferation by platelet-derived growth factor and estradiol: identification of signaling mechanisms involved. Endocrinology 138:1289- 1298 65 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. Mullaney BP, Skinner MK 1991 Growth factors as mediators of testicular cell-cell interactions. Baillieres Clin Endocrinol Metab 52771-790 Fijak M, Meinhardt A 2006 The testis in immune privilege. Immunol Rev 213:66-81 Jefferson WN, Couse JF, Banks EP, Korach KS, Newbold RR 2000 Expression of estrogen receptor ?is developmentally regulated in reproductive tissues of male and female mice. Biol Reprod 622310-317 Saunders PT, Fisher JS, Sharpe RM, Millar MR 1998 Expression of oestrogen receptor beta (ER beta) occurs in multiple cell types, including some germ cells, in the rat testis. J Endocrinol 156:R13-17 Pentikainen V, Erkkila K, Suomalainen L, Parvinen M, Dunkel L 2000 Estradiol acts as a germ cell survival factor in the human testis in vitro. J Clin Endocrinol Metab 85:2057-2067 Shetty G, Krishnamurthy H, Krishnamurthy HN, Bhatnagar S, Moudgal RN 1997 Effect of estrogen deprivation on the reproductive physiology of male and female primates. J Steroid Biochem Mol Biol 61:157-166 Tsutsumi l, Fugimori K, Nakamura RM, Mather JP, Ono T, diZerega GS 1987 Disruption of seminiferous epithelial function in the rat by ovarian protein. Biol Reprod 362451-461 ' Zhou Q, Clarke L, Nie R, Carnes K, Lai LW, Lien YH, Verkman A, Lubahn D, Fisher JS, Katzenellenbogen BS, and Hess RA. 2001 Estrogen action and male fertility: roles of the sodium/hydrogen exchanger-3 and fluid reabsorption in reproductive tract function. Proc Natl Acad Sci U S A 98:14132-14137. Hess RA, Zhou Q, Nie R, Oliveira C, Cho H, Nakaia M, Carnes K 2001 Estrogens and epididymal function. Reprod Fertil Dev 13:273-283. Rago V, Bilinska B, Palma A, Ando S, Carpino A 2003 Evidence of aromatase localization in cytoplasmic droplet of human immature ejaculated spermatozoa. Folia Histochem Cytobiol 41:23-27 66 152. 153. 154. 155. 156. 157. 158. 159. 160. 161. Aquila S, Sisci D, Gentile M, Middea E, Siciliano L, Ando S 2002 Human ejaculated spermatozoa contain active P450 aromatase. J Clin Endocrinol Metab 87:3385-3390 Sharpe RM, Atanassova N, McKinneII C, Parte P, Turner KJ, Fisher JS, Kerr JB, Groome NP, Macpherson S, Millar MR, Saunders PT 1998 Abnormalities in functional development of the Sertoli cells in rats treated neonatally with diethylstilbestrol: a possible role for estrogens in Sertoli cell development. Biol Reprod 59:1084-1094. Kelly MJ, Levin ER 2001 Rapid actions of plasma membrane estrogen receptors. Trends Endocrinol Metab 122152-156 Luconi M, Muratori M, Forti G, Baldi E 1999 Identification and characterization of a novel functional estrogen receptor on human sperm membrane that interferes with progesterone effects. J Clin Endocrinol Metab 84:1670-1678 Fraser LR, Beyret E, Milligan SR, Adeoya-Osiguwa SA 2006 Effects of estrogenic xenobiotics on human and mouse spermatozoa. Hum Reprod 21:1184—1193 Furuya S, Endo Y, Oba M, Nozawa S, Suzuki S 1992 Effects of modulators of protein kinases and phosphatases on mouse sperm capacitation. J Assist Reprod Genet 9:391-399 Adeoya-Osiguwa SA, Markoulaki S, Pocock V, Milligan SR, Fraser LR 2003 17beta-Estradiol and environmental estrogens significantly affect mammalian sperm function. Hum Reprod 182100-107 Emmen JM, Korach KS 2003 Estrogen receptor knockout mice: phenotypes in the female reproductive tract. Gynecol Endocrinol 17:169- 176 Arredondo F, Noble LS 2006 Endocrinology of recurrent pregnancy loss. Semin Reprod Med 24:33-39 Babischkin JS, Pepe GJ, Albrecht ED 1989 Regulation of progesterone biosynthesis by estrogen during baboon pregnancy: placental mitochondrial cholesterol side-chain cleavage activity in antiestrogen 67 162. 163. 164. .165. 166. 167. 168. 169. 170. (ethamoxytriphetol, MER—25)—treated baboons. Endocrinology 124:1638- 1645 Babischkin JS, Pepe GJ, Albrecht ED 1997 Estrogen regulation of placental P-450 cholesterol side-chain cleavage enzyme messenger ribonucleic acid levels and activity during baboon pregnancy. Endocrinology 138:452-459 Baggia S, Albrecht ED, Pepe GJ 1990 Regulation of 11 beta- hydroxysteroid dehydrogenase activity in the baboon placenta by estrogen. Endocrinology 126:2742-2748 Geisert RD, Ross JW, Ashworth MD, White FJ, Johnson GA, DeSiIva U 2006 Maternal recognition of pregnancy signal or endocrine disruptor. the two faces of oestrogen during establishment of pregnancy in the pig. Reprod Suppl 62:131-145 James WH 1996 Evidence that mammalian sex ratios at birth are partially controlled by parental hormone levels at the time of conception. J Theor Biol 1802271 -286 Mocarelli P, Gerthoux PM, Ferrari E, Patterson DGJ, Kieszak SM, Brambilla P, Vincoli N, Signorini S, Tramacere P, Carreri V, Sampson EJ, Turner WE, Needham LL 2000 Paternal concentrations of dioxin and sex ratio of offspring. Lancet 355:1858-1863 Pergament E, Todydemir PB, Fiddler M 2002 Sex ratio: a biological perspective of 'Sex and the City'. Reprod Biomed OnIine 5:43-46 James WH 1976 Timing of fertilization and sex ratio of offspring—a review. Ann Hum Biol 32549-556 Davis DL, Gottlieb MB, Stampnitzky JR 1998 Reduced ratio of male to female births in several industrial countries: a sentinel health indicator? JAMA 279:1018-1023 James WH 1980 Time of fertilisation and sex of infants. Lancet 121124- 1126 68 171. 172. 173. 174. 175. 176. . 177. 178. 179. 180. 181. James WH 1985 Sex ratio, dominance status and maternal hormone levels at the time of conception. J Theor Biol 1 142505-510 James WH 1986 Hormonal control of sex ratio. J Theor Biol 118:427-441 James WH 2006 Possible constraints on adaptive variation in sex ratio at birth in humans and other primates. J Theor Biol 238:383-394 James WH 1989 Parental hormone levels and mammalian sex ratios at birth. J Theor Biol 139259-67 Ryan JJ, Amirova Z, Carrier G 2002 Sex ratios of children of Russian pesticide producers exposed to dioxin. Environ Health Perspect 1102A699- 701 Martuzzi M, Di Tanno ND, Bertollini R 2001 Declining trends of male proportion at birth in Europe. Arch Environ Health 56:358-364 Vartiainen T, Kartovaara L, Tuomisto J 1999 Environmental chemicals and changes in sex ratio: analysis over 250 years in finland. Environ Health Perspect 107:813-815 Figa-Talamanca l, Tarquini M, Lauria L 2003 Is it possible to use sex ratio at birth as indicator of the presence of endocrine disrupters in environmental pollution?]. G Ital Med Lav Ergon 25:52-53 Rogan WJ, Gladen BC, Guo YL, Hsu CC 1999 Sex ratio after exposure to dioxin-like chemicals in Taiwan. Lancet 353:206—207 Figa-Talamanca I, Carbone P, Lauria L, Spinelli A, Ulizzi L 2003 Environmental factors and the proportion of males at birth in Italy. Arch Environ Health 58:1 19-124 Jarrell JF, Gocmen A, Akyol D, Brant R 2002 Hexachlorobenzene exposure and the proportion of male births in Turkey 1935-1990. Reprod Toxicol 16:65-70 69 182. 183. 184. 185. 186. 187. 188. 189. 190. 191. van Birgelen AP 1998 Hexachlorobenzene as a possible major contributor to the dioxin activity of human milk. Environ Health Perspect 106:683-688 Potashnik G, Porath A 1995 Dibromochloropropane (DBCP): a 17-year reassessment of testicular function and reproductive performance. J Occup Environ Med 37:1287-1292 Slutsky M, Levin JL, Levy BS 1999 Azoospennia and oligosperrnia among a large cohort of DBCP applicators in 12 countries. Int J Occup Environ Health 5:116-122 Thrupp LA 1991 Sterilization of workers from pesticide exposure: the causes and consequences of DBCP-induced damage in Costa Rica and beyond. Int J Health Serv 212731-757 Wesseling C, Ahlbom A, Antich D, Rodriguez AC, Castro R 1996 Cancer in banana plantation workers in Costa Rica. Int J Epidemiol 25:1125-1131 Meistrich ML, Wilson G, Shuttlesworth GA, Porter KL 2003 Dibromochloropropane inhibits sperrnatogonial development in rats. Reprod Toxicol 17:263—271 Goldsmith JR 1997 Dibromochloropropane: epidemiological findings and current questions. Ann N Y Acad Sci 837:300-306 Sod-Moriah UA, Sror U, Shemi D, Potashnik G, Chayoth R, Shaked I, Kaplanski J 1988 Long term effects of dibromochloropropane (DBCP) on male rats' reproductive system. Andrologia 20:60-66 Bredbacka K, Bredbacka P 1996 Glucose controls sex-related growth rate differences of bovine embryos produced in vitro. J Reprod Fertil 1062169-172 Williams AC, Ford WC 2001 The role of glucose in supporting motility and capacitation in human spermatozoa. J Androl 22:680-695 70 192. 193. 194. 195. 196. 197. 198. 199. 200. 201. Machado AF, Zimmerman EF, Hovland DNJ, Weiss R, Collins MD 2001 Diabetic embryopathy in C57BL/6Jmice. Altered fetal sex ratio and impact of the splotch allele. Diabetes 50:1193-1199 Cameron E2 2004 Facultative adjustment of mammalian sex ratios in support of the Trivers-Willard hypothesis: evidence for a mechanism. Proc Biol Sci 271 :1 549 Agung B, Otoi T, Wongsrikeao P, Taniguchi M, Shimizu R, Watari H, Nagai T 2006 Effect of maturation culture period of oocytes on the sex ratio of in vitro fertilized bovine embryos. J Reprod Dev 522123-127 Cui KH 1997 Size differences between human X and Y spermatozoa and prefertilization diagnosis. Mol Hum Reprod 3:61-67 Broer KH, Winkhaus I, Sombroek H, Kaiser R 1976 Frequency of Y- chromatin bearing spermatozoa in intracervical and intrauterine postcoital tests. Int J Fertil 212181-185 Hotchkiss AK, Vandenbergh JG 2005 The anogenital distance index of mice (Mus musculus domesticus): an analysis. Contemp Top Lab Anim Sci 44246-48 Salazar-Martinez E, Romano-Riquer P, Yanez-Marquez E, Longnecker MP, Hemandez-Avila M 2004 Anogenital distance in human male and female newborns: a descriptive, cross-sectional study. Environ Health 328 Vandenbergh JG, Huggett CL 1995 The anogenital distance index, a predictor of the intrauterine position effects on reproduction in female house mice. Lab Anim Sci 45:567-573 Kerin TK, Vogler GP, Blizard DA, Stout JT, McCleam GE, Vandenbergh DJ 2003 Anogenital distance measured at weaning is correlated with measures of blood chemistry and behaviors in 450-day-old female mice. Physiol Behav 78:697-702 Marois G 1968 Action of progesterone, testosterone and estradiol on the anogenital distance and somatic sexual differentiation in rats. Biol Med (Paris) 57:44-90 71 202. 203. 204. 205. 206. 207. 208. 209. 210. Swan SH, Main KM, Liu F, Stewart SL, Kruse RL, Calafat AM, Mao CS, Redmon JB, Temand CL, Sullivan S, Teague JL 2005 Decrease in anogenital distance among male infants with prenatal phthalate exposure. Environ Health Perspect 113:1056-1061 Kelce WR, Earl Gray L, Jr. 1999 Environmental antiandrogens as endocrine disruptors. In: Naaz A ed. Endocrine dismptors: effects on male and female reproductive systems. Florida: CRC Press; 247-277 Habert R, Picon R 1984 Testosterone, dihydrotestosterone and estradiol- 17 beta levels in maternal and fetal plasma and in fetal testes in the rat. J Steroid Biochem 21:193-198 Clark RL, Antonello JM, Grossman SJ, Wise LD, Anderson C, Bagdon WJ, Prahalada S, MacDonald JS, Robertson RT 1990 External genitalia abnormalities in male rats exposed in utero to finasteride, a 5 alpha- reductase inhibitor. Teratology 42291-100 Evans BA, Griffiths K, Morton MS 1995 Inhibition of 5 alpha-reductase V in genital skin fibroblasts and prostate tissue by dietary lignans and isoflavonoids. J Endocrinol 147:295-302 Swan SH 2006 Prenatal phthalate exposure and anogenital distance in male infants. Environ Health Perspect 1142A88-89 Gallavan RH, Jr., Holson JF, Stump DG, Knapp JF, Reynolds VL 1999 Interpreting the toxicologic significance of alterations in anogenital distance: potential for confounding effects of progeny body weights. Reproductive Toxicology 13:383-390. Levy JR, Faber KA, Ayyash L, Hughes CL, Jr. 1995 The effect of prenatal exposure to the phytoestrogen genistein on sexual differentiation in rats. Proc Soc Exp Biol Med 208260-66 McIntyre BS, Barlow NJ, Foster PMD 2001 Androgen-mediated development in male rat offspring exposed to flutamide in utero: permanence and correlation of early postnatal changes in anogenital distance and nipple retention with malformations in androgen-dependent tissues. Toxicol Sci 62:236-249 72 211. 212. 213. 214. 215. 216. 217. 218. 219. Crescioli C, Maggi M, Vannelli GB, Ferruzzi P, Granchi S, Mancina R, Muratori M, Forti G, Serio M, Luconi M 2003 Expression of functional estrogen receptors in human fetal male external genitalia. J Clin Endocrinol Metab 88:1815-1824 Sohoni P, Sumpter JP 1998 Several environmental oestrogens are also anti-androgens. J Endocrinol 158:327-339 Marsee K, Woodruff TJ, Axelrad DA, Calafat AM, Swan SH 2006 Estimated daily phthalate exposures in a population of mothers of male infants exhibiting reduced anogenital distance. Environ Health Perspect 114:805-809 Borch J, Axelstad M, Vinggaard AM, Dalgaard M 2006 Diisobutyl phthalate has comparable anti-androgenic effects to di-n-butyl phthalate in fetal rat testis. Toxicol Lett 1 63:183-190 Blount BC, Silva MJ, Caudill SP, Needham LL, Pirkle JL, Sampson EJ, Lucier GW, Jackson RJ, Brock JW 2000 Levels of seven urinary phthalate metabolites in a human reference population. Environ Health Perspect 1082 979- 982 ~ Collins TF, Sprando RL, Black TN, Olejnik N, Eppley RM, Alam HZ, Rorie J, Ruggles DI 2006 Effects of zearalenone on in utero development in rats. Food Chem Toxicol 44:1455-1465 ' Kebayashi K, Miyagawa M, Wang RS, Sekiguchi S, Suda M, Honma T 2002 Effects of in utero and lactational exposure to bisphenol A on somatic growth and anogenital distance in F1 rat offspring. Ind Health 402375-381 Fielden MR, Samy SM, Chou KC, Zacharewski TR 2003 Effect of human dietary exposure levels of genistein during gestation and lactation on Iong-terrn reproductive development and sperm quality in mice. Food Chem Toxicol 41:447-454. Bidlingmaier F, Butenandt O, Knorr D 1977 Plasma gonadotropins and estrogens in girls with idiopathic precocious puberty. Pediatr Res 11:91—94 73 220. 221. 222. 223. 224. 225. 226. 227. 228. 229. Tanner JM 1973 Trend towards earlier menarche in London, Olso, Copenhagen, the Netherlands and Hungary. Nature 243295-96 Ruf KB 1973 How does the brain control the process of puberty? Z Neurol 204296-105 Ordog T, Knobil E 1995 Estradiol and the inhibition of hypothalamic gonadotropin-releasing hormone pulse generator activity in the rhesus monkey. Proc Natl Acad Sci U S A 92:5813-5816 O'Byrne KT, Chen MD, Nishihara M, Williams CL, Thalabard JC, Hotchkiss J, Knobil E 1993 Ovarian control of gonadotropin horrnone- releasing hormone pulse generator activity in the rhesus monkey: duration of the associated hypothalamic signal. Neuroendocrinology 572588-592 MacGillivray MH, Morishima A, Conte F, Grumbach M, Smith EP 1998 Pediatric endocrinology update: an overview. The essential roles of estrogens in pubertal growth, epiphyseal fusion and bone turnover: lessons from mutations in the genes for aromatase and the estrogen receptor. Horm Res; 49:2-8 Schoeters G, Den Hond E, Dhooge W, van Larebeke N, Leijs M 2008 Endocrine dismptors and abnormalities of pubertal development. Basic & clinical pharmacology & toxicology 102:168-175 Oken E, Gillman MW 2003 Fetal origins of obesity. Obes Res 112496-506 Hedley AA, Ogden CL, Johnson CL, Carroll MD, Curtin LR, Flegal KM 2004 Prevalence of ovenrveight and obesity among US children, adolescents, and adults, 1999-2002. JAMA 291 :2847-2850 Flegal KM, Carroll MD, Ogden CL, Johnson CL 2002 Prevalence and trends in obesity among US adults, 1999-2000. JAMA 288:1723—1727 Heindel JJ 2003 Endocrine disruptors and the obesity epidemic. Toxicol Sci 76:247-249 74 230. 231 . 232. 233. 234. 235. 236. 237. 238. Schildkraut JM, Demark-Wahnefried W, DeVoto E, Hughes C, Laseter JL, Newman B 1999 Environmental contaminants and body fat distribution. Cancer Epidemiol Biomarkers Prev 8:179-183 Wolff MS, Anderson HA 1999 Environmental contaminants and body fat distribution. Cancer Epidemiol Biomarkers Prev 82951-952 Jones ME, Thorbum AW, Britt KL, Hewitt KN, Wreford NG, Proietto J, Oz OK, Leury BJ, Robertson KM, Yao S, Simpson ER 2000 Aromatase-deficient (ArKO) mice have a phenotype of increased adiposity. Proc Natl Acad Sci U S A 97:12735—12740. Jones ME, A.W. T, Britt KL, Hewitt KN, Misso ML, Wreford NG, Proietto J, Oz OK, Leury B, Robertson KM, Yao S, Simpson ER 2001 Aromatase-deficient (ArKO) mice accumulate excess adipose tissue. J Steroid Biochem Mol Biol 7923-9. Naaz A, Zakroczymski M, Heine P, Taylor J, Saunders P, Lubahn D, Cooke PS 2002 Effect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor alpha (ERalpha): a potential role for estrogen receptor beta (ERbeta). Horm Metab Res 342758-763. Heine PA, Taylor JA, lwamoto GA, Lubahn DB, Cooke PS 2000 Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc Natl Acad Sci USA 97:12729-12734 Naaz A, Zakroczymski M, Heine P, Taylor J, Saunders P, Lubahn D, Cooke PS 2002 Effect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor alpha (ERalpha): a potential role for estrogen receptor beta (ERbeta). Horm Metab Res 34:758-763 Okura T, Koda M, Ando F, Niino N, Ohta S, Shimokata H 2003 Association of polymorphisms in the estrogen receptor alpha gene with body fat distribution. Int J Obes Relat Metab Disord 27:1020-1027 Baillie-Hamilton PF 2002 Chemical toxins: a hypothesis to explain the global obesity epidemic. J Altem Complement Med 8:185-192 75 239. 240. 241. 242. 243. 244. 245. 246. 247. Sweeney T 2002 Is exposure to endocrine disrupting compounds during fetal/post-natal development affecting the reproductive potential of farm animals? Domest Anim Endocrinol 232203-209 Ankley GT, Jensen KM, Makynen EA, Kahl MD, Korte JJ, Homung MW, Henry TR, Denny JS, Leino RL, Wilson VS, Cardon MC, Hartig PC, Gray LE 2003 Effects of the androgenic growth promoter 17-beta- trenbolone on fecundity and reproductive endocrinology of the fathead minnow. Environ Toxicol Chem 22:1350—1360 Le Guevel R, Pakdel F 2001 Assessment of oestrogenic potency of chemicals used as growth promoter by in-vitro methods. Hum Reprod 16:1030-1036 Barbini DA, Vanni F, Pelosi P, Generali T, Amendola G, Stefanelli P, Girolimetti S, Di Muccio A, Mantovani A, Spera G, Silvestroni L 2004 Low levels of organochlorine pesticides in subjects with metabolic disturbances: a survey taken in Rome in 2001-2002. Bull Environ Contam Toxicol 732219-226 Mizejewski GJ, Vonnegut M, Jacobson HI 1983 Estradiol-activated alpha-fetoprotein suppresses the uterotropic response to estrogens. Proc Natl Acad Sci U S A 80:27.33—2737 Szafran H, Smielak-Koromberl W 1998 The role of estrogens in hormonal regulation of lipid metabolism in women. Przel-Lek 55:266-270. Simpson ER, Clyne C, Rubin G, Boon WC, Robertson K, Britt K, Speed C, Jones M 2002 Aromatase—a brief overview. Annu Rev Physiol 64293-127. Cooke PS, Naaz A 2004 Role of estrogens in adipocyte development and function. Exp Biol Med (Maywood) 229:1 127-1 135 Cooke PS, Heine PA, Taylor JA, Lubahn DB 2001 The role of estrogen and estrogen receptor-alpha in male adipose tissue. Mol Cell Endocrinol 178:147-154 76 CHAPTER 3 LITERATURE REVIEW II. 3.1 DEFINITION OF ESTROGENIC ENDOCRINE MODULATORS Wildlife and laboratory animals exposed to exogenous estrogenic chemicals during in utero and postnatal development have adverse effects on reproductive tract development and fertility (1-3). As described in Chapter 2, endogenous estrogen has important roles in the reproductive system, and alteration in its biological function by interfering exogenous estrogenic chemicals via binding to receptors, secretion, synthesis, transport, or elimination of endogenous estrogens necessary for normal function and homeostasis of reproduction, development and/or behavior could lead to functional and morphological adverse effects on reproduction. These endocrine disrupting chemicals, both naturally occurring as well as man-made chemicals, are known by a variety of terms (e.g., environmental estrogens, environmental hormones, endocrine disrupting chemicals, endocrine modulators, endocrine disrupters, and endocrine disruptors) to describe the phenomenon that can mimic endogenous estrogen in the body. The US EPA adopted the definition established by Kavlock et al.(4) that endocrine disrupters interfere with the synthesis, secretion, transport, binding, action, or elimination of endogenous estrogen that is responsible for the maintenance of homeostasis, reproduction, development, and/or behavior (5). In 1997, the European Commission defined an endocrine disrupter as an exogenous substance that causes adverse health effects in an intact organism, or its progeny, secondary to changes in endocrine function; a potential endocrine 77 disruptor is a substance that possesses properties that might be expected to lead to endocrine disruption in an intact organism (6). However, transient effects of some endocrine disrupters may be considered adverse effects such as a temporary reduction in sperm production that may lead to infertility but not sterility. Therefore, Dickerson et al. (7) gave two definitions of endocrine disruptor and endocrine modulator. Based on these definitions, an endocrine disruptor is a compound that causes irreversible or adverse effects on endocrine function. An endocrine modulator is a compound that causes transient and reversible alterations of endocrine function within the physiological (or homeostatic) range, resulting in effects that normally would not be considered adverse. In this paper, the term estrogenic endocrine modulators (EEMs) will be used because 1) this paper will focus on estrogenic chemicals, 2) it is difficult to distinguish transient, non-adverse effects and permanent, adverse effects on the endocrine system, and 3) there is no agreement as to what constitutes low-dose effects of endocrine disrupting chemicals. 3.2 SOURCES AND EXPOSURE OF EEMs Estrogen endocrine modulators are ubiquitous in our environment (i.e., air, water, and soil) because they have been produced naturally or synthetically. Therefore, both humans and animals can be exposed to EEMs through drinking contaminated water, breathing contaminated air, ingesting food, or contacting contaminated soil. According to the US EPA, approximately 87,000 industrial 78 and agricultural chemicals are used in the US alone. Since the mid-1940s, the US plastics industry has grown at the rate of 6 to 12% per year with an annual production of 85 billion pounds in 1996, whereas plastics production in developing countries is expanding at the rate of 40% per year (8). Among these chemicals, only a few chemicals have been tested for endocrine disrupter activity. Currently approximately 60 chemicals have been identified as EEMs (9). Some of them have long environmental half-lives (e.g., 15 years for DDT and PCB) due to their lipophilicity. An increase in the environmental concentrations of chemicals can lead to bioaccumulation in adipose tissue and biomagnification in species at the top of the food chain. Increased concentrations of EEMs in the body of the higher predator can exert toxic effects on the reproductive system. High concentrations of EEMs are often measured in sewage sludge from industrial, agricultural and domestic origin (10-12). The contaminated sewage sludge can be used as fertilizer for farm animals or land application (13, 14). If these EEMs are ingested with food and drinking water by food-producing animals then there is potential for adverse effects in humans via consuming food- produ'cing animals or animal products. Furthermore, low body burden of EEMs, so called “low-dose effects of EEMs” have been debated because the range of endogenous steroid hormones exert their bioactivities is nmol or pmol range (15, 16). Thus, further research and risk assessment for low-dose effects of EEMs are needed to confirm whether low-dose effects of EEMs have potential risk to reproductive health. 79 There have been concerns regarding the deleterious effects of phytoestrogens on reproduction and development in humans and animals (17- 20). Most natural EEMs are generally plant-produced estrogens. Many plants (e.g., legumes, fruits, and vegetables) produce phytoestrogen (e.g., genistein and daidzein). For example, a study demonstrated a detectable concentrations of genistein and daidzein in currants and raisins (2,350 mg and 1,840 mglkg of wet weight of food) among 80 different fruits and nuts (21). Concentrations of phytoestrogens vary in plants but are mainly formed in legumes (genistein and daidzein occur up to 3.8 x 106 mglkg wet weight of soybean) (17, 22, 23). The , other source of natural EEMs is mycotoxins. A mycotoxin, zearalenone, is synthesized by several species of Fusarium (F. graminearum, F. culmorum, F. croolwellense, F. sambucinum and F. equisetr) commonly contaminating poorly stored agricultural products and foodstuffs. Zearalenone is a non-steroidal mycotoxin with estrogen-like activity (24). Synthetically produced EEMs have been used in pharmaceuticals (e.g., diethylstilbestrol, tamoxifen, and 17a-ethynyl estradiol), pesticides (e.g., methoxychlor and atrazine), industrial chemicals (e.g., bisphenol A and phthalate), and occur as environmental pollutants (e.g., polychlorinated biphenyl and benzo[a]pyrene). Details of each EEM mentioned above will be discussed later in this chapter. 80 3.3 MODES OF ACTION OF EEMs 3.3.1 ALTERED SEX STEROID HORMONE SYNTHESIS It has been known that some EEMs affect the reproductive system through altering endogenous estrogen and androgen synthesis via interfering P450 enzymes (Table 2-1 and Figure 2-1). For example, mono-(2-ethylhexyl)— phthalate (MEHP), which is an active metabolite of a plasticizer, di-(2-ethylhexyl)- phthalate (DEHP), may interfere with estrogen synthesis by decreasing transcription of the aromatase enzyme in cultured rat granulosa cells (25, 26). Additionally, Andrade et al. (27) demonstrated that maternal exposure to DEHP during the gestational and lactational periods alters aromatase activity in the hypothalamus with J-shaped dose-response curve, which means that aromatase activity was inhibited by low doses of DEHP (0.135 and 0.405 mglkg bw/d) and stimulated by high doses (15, 45 and 405 mglkg bw/d) in male rats on postnatal day 1. Corresponding to the results of laboratory animal studies, it has been hypothesized that in utero and early postnatal exposure to phthalates may be responsible for testis abnormalities (e.g. cryptorchidism and hypospadias) in human males via stimulation of aromatase activity and suppression of androgen production from fetal Leydig cells (28-30). Unlike aromatase, which is encoded by a single gene in the human and rodent, hydroxysteroid dehydrogenases are encoded by at least 2 to 3 homologous genes. Human 3B-HSD shares 80 to 94% amino acid sequence identity with rat (31) and human 17a-HSD shares 77, 70 and 72% amino acid sequence identity with mouse type 5 17B-HSD, 3a-HSD and 200-HSD 81 respectively (32). Inhibitory effects of phytoestrogens on 36- or 17B-HSD have been reported (33). Some studies demonstrated that the number of hydroxylations of carbon molecules in A ring and B ring (Figure 3-1) of phytoestrogens is positively correlated with the inhibitory effects on the activity of 313- or 17B-HSD type 1 and 5 in human placental microsomes or adrenocortical H295R cells (34, 35). These results suggest that phytoestrogens are potent inhibitors of human placental estrogen biosynthesis. The biological role of Sci-reductase is to catalyze the conversion of testosterone to Soc-dihydrotestosterone (DHT). Both testosterone and DHT can bind to the androgen receptor to promote androgen-dependent gene transcription, which plays important roles for sperrnatogenesis and accessory sex gland function. It is known that DHT is a more potent androgen than testosterone (e.g. 2.4 times more potent at maintaining rat prostate weight) (36, 37). An in vivo study also confirmed that maternal exposure to 3 or 30 ug/kg bw/d of 3,3',4,4’,5- pentachlorobiphenyl (PCB126) and 3,3’4,4’,5,5’-hexachlorobiphenyl (PCB169) increases the relative activity of 3B—HSD, and decreased the relative activity of SOL-reductase at 15 weeks in the testis of male rat offspring (38). In this study, plasma testosterone concentrations in maternally exposed PCB169 male offspring were decreased at 3 weeks (14 vs. 1 ng/dl, respectively) but were increased at 15 weeks (120 vs. 266 ng/dl, respectively). It suggests that fetal exposure to PCB126 or PCB169 has long-term effects on steroidogenic enzymes activities leading to alter action of the concentrations of steroid hormones, which may suppress sperrnatogenesis. Collectively, future studies need to consider 82 effects of EEMs on the reproductive system via through alteration of steroidogenic enzymes. 3.3.2 ALTERED STEROID HORMONE STORAGE AND/OR RELEASE Steroid hormones do not appear to be stored intracellularly within membranous secretory granules (39). They are more likely synthesized and released instantly after gonadotrophin stimulates the gonads. For example, testosterone is rapidly synthesized by the Leydig cells of the testis upon stimulation by LH from the hypothalamus. Catecholamine hormones such as norepinephrine are stored in granular vesicles and thus they can be released quickly on demand (40). Norepinephrine is critical for the preovulatory increase in the pulsatile release of GnRH and the subsequent ovulatory surge of LH. For example, insecticides such as chlordimeforrn and amitraz have been reported. to block norepinephrine binding to the alpha2-adrenergic receptor, thus inhibiting release of GnRH and production of progesterone in human and rodent granulosa cells without affecting estrogen production (41, 42). It indicates that these pesticides may disrupt in the timing of the LH surge, and alter the viability and the quality of the oocytes (43). 3.3.3 ALTERED STEROID HORMONE TRANSPORT AND CLEARANCE Hormones are transported from blood in the free (biologically active form) or bound state. Steroid hormones and EEMs are mostly transported in the blood by albumin and specialized transport proteins (carriers), known as sex-steroid 83 hormone-binding globulin (SHBG) or testosterone-estrogen-binding globulin (T EBG). Therefore, these carrier proteins in blood are important in transporting lipophilic endogenous hormones and EEMs to target organs and making available free steroids to target cells. It suggests that EEM carrier proteins have a role in the detoxification process by retarding the binding of EEMs to ERs, which allows the inactivation and excretion of EEMs to occur without potential adverse effects on reproduction. Albumin is the major serum protein that binds various lipophilic compounds such as steroid hormones, fatty acids, retinoids, and thyroid hormone and prostaglandins as well as many lipophilic EEMs because of its high serum concentration (approximately 45 mg/mL, 0.67 mM in humans) (44). Corresponding to the high concentration of albumin in serum, 60% of E2 is bound to albumin. Among the endogenous estrogens, E3 has 1/3 the affinity of E2 for the ERs but in the presence of albumin, the affinity of E3 is twice that of E2 for the ERs (44-47). Sex-steroid hormone-binding globulin is the major binding protein for sex steroid hormones in plasma, and mediates androgen and estrogen signaling at the cell membrane through CAMP-induced pathways (48). It is known that estrogen increases the plasma concentration of SHBG via increasing synthesis of SHBG in the liver (49). Concentrations of sex-steroid hormone-binding globulin are twice as high in women as they are in men due to relatively high production of endogenous estrogen or synthetic estrogen derived from birth control pills in women. When woman reach at the final menstrual period, 84 approximately 38 to 60% of E2 and 8% of E1 are bound to SHBG (50). Considering E2 binding to both albumin and SHBG, 1 to 2% of total plasma E2 circulates as the free hormone in women (50, 51). In addition to‘albumin and SHBG, estrogens in the amniotic fluid of pregnant women and rodents are bound to alpha-fetoprotein (AFP) (52). Alpha- fetoprotein in human fetal serum was recognized as a fetal component not commonly found in adults (53). Oviductal fluids from rabbits, monkeys, and women secrete a variety of proteins such as albumin and alpha-fetoprotein (54, 55). The production of albumin and AFP found in human oviductal fluid is under hormonal control and concentrations of such protein secretions are positively correlated with the estrogen peak of the menstrual cycle (56). Studies demonstrated that AFP also can bind and transport several ligands, including fatty acids, retinoids, steroids, heavy metals, dyes, phytoestrogens, dioxins, and various organic drugs (57-59). Regulation of the concentration of albumin, SHBG, and AFP in the serum is of practical significance because they either increase or decrease the clearance of endogenous hormones or access of EEMs to target receptors resulting in altered bioavailability to the target organs. Therefore, humans and animals can have a selective advantage in protection against the potential adverse effects of EEMs. 85 3.3.4 ALTERED STEROID HORMONE RECEPTOR RECOGNITION/BINDING Studies demonstrated that hormones elicit responses in their respective target organs through direct interactions with either intracellular receptors or membrane-bound receptors. Specific binding of the endogenous ligand to its receptor is a critical step in hormone function. Intracellular (nuclear) receptors, such as those for sex steroids, adrenal steroids, and thyroid hormones, regulate gene transcription in a ligand-dependent manner through their interaction with specific DNA sequences (response elements). A number of EEMs may alter this process by mimicking the endogenous estrogen and acting as an agonist or by inhibiting binding and acting as an antagonist. The best known examples are methoxychlor, DDT, some PCBs and alkylphenols, which can compete with endogenous estrogen to bind ERs. Interestingly, some EEMs may act through one or more intracellular receptors. For example, o,p’-DDT has estrogenic and androgenic effects became of binding to both ERs and AR, while, p,p’-DDT inhibits androgen binding to the AR in vitro (60, 61). The pesticides dieldrin, endosulfan, and fenarimol are also known to bind both ERs and AR. Additionally, Kelce et al. (62) observed that chlordecone, which is a synthetic chlorinated organic compound and has been used as an insecticide and fungicide, mainly binds to ER but it also binds to AR at higher concentrations. Therefore, it is difficult to categorize a compound as either estrogenic or androgenic endocrine modulator. It has been hypothesized that EEMs induce or reduce the degradation of steroid hormone receptors resulting in altered physiological effects of 86 endogenous steroid hormones. Studies have demonstrated that the half-life of ERs is approximately 1 to 5 hr in uterine tissues and in cultured cells by using dense amino acid or [358]methionine/cysteine incorporation, or radiolabeled tamoxifen aziridine binding (63-65). There have been controversial results regarding the ERa degradation in transcriptional activation. Some studies suggest that ERa degradation may be required to maintain ERa-mediated transcription with turnover of a specific proteasome (e.g. 26$ proteasome) and coactivators (e.g. Fos/Jun) (66), while other studies suggest that ERa degradation has no direct effects on transcription in pituitary cells or in ERa- positive breast cancer cells (67, 68). Collectively, ligands including EEMs may form a stable complex with their receptors, the ligand bound steroid hormone receptor complexes are required to carry on transcription of target genes for extended periods of time. Therefore, - degradation of the ligand bound receptor complex at transcription is important to maintain transcription activation in response to rapid changes in hormone concentration as well as physiological cellular response. 3.3.5 ALTERED STEROID HORMONE POSTRECEPTOR ACTIVATION Once the endogenous ligands or EEMs bind to their receptors, a cascade of events is initiated for the appropriate cellular response, which is necessary for signal transduction across the membrane or the initiation of transcription and protein synthesis via nuclear receptors (49). When hormones bind to their receptors, transduction of a signal across the membrane is mediated by the 87 activation of second messenger systems. These may include (1) alterations in G-protein/cAMP-dependent protein kinase A (e.g., after LH stimulation of the Leydig cell); (2) phosphatidylinositol regulation of protein kinase C and inositol triphosphate (e.g., after GnRH stimulation of gonadotrophs, thyrotropin releasing hormone stimulation of thyrotrophs); (3) tyrosine kinase (e.g., after insulin binding to the membrane receptor); and (4) calcium ion flux (e.g. the initiation of the calcium/calmodulin-dependent cellular response). EEMs thus can disrupt signal transduction of peptide hormones if they interfere with one or more of these processes (69). For example, 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure indirectly down regulates steroid hormone receptor activation (70, 71). Consequently, because of the diverse known pathways of EEMs, any assessment must consider the net result of all influences on hormone receptor function and feedback regulation. 3.3.6 ALTERED GENE EXPRESSION WITH CHANGING DNA SEQUENCE Epigenetics refers to the molecular phenomena that regulate gene expression without alterations to the deoxyribonucleic acid (DNA) sequence, which contains genomic information of the cell by encoding the sequence of the amino acid residues in proteins. Genes in mammals are transcribed from methylation-free promoters, while keeping noncoding DNA, containing introns suppressed. Therefore, methylation can either increase or decrease the level of transcription, depending on whether the methylation inactivates a positive or 88 negative regulatory element leading to long-term epigenetic silencing of imprinted genes. DNA methylation is specific to cytosines in CpG dinucleotides and achieved by the transfer of a methyl group from S-adenosyl-methionine to the C5 position of cytosine. It is catalyzed by DNA methyltransferases (e.g., DNA cytosine methyltransferases). Approximately, 70 percent of all cytosines in CpG dinucleotides in the human genome are methylated (72). Two DNA methylation patterns happen during the developmental period (73). The first one is the lineage-specific DNA methylation pattern, which occurs during gastrulation, and the second one is the germ-line DNA methylation pattern, which occurs during gonadal development and sex determination. The lineage-specific DNA methylation pattern is based on the genetic material transferred from the egg and sperm, which can result in developmental defects or embryonic lethality by any possible alterations of DNA methylation (73, 74). Unlike the lineage-specific DNA methylation pattern, any potential alteration of the germ-line DNA methylation pattern could be critical for imprinted genes in germ cellsand‘ result in transgenerational effects (75-77). During gonadal development, primordial germ cells in mouse embryos migrate down the genital ridge and a complete DNA demethylation is achieved from embryonic day 10.5 to day 11.5 (78). Thereafter, the germ cells undergo a remethylation involving a sex-specific determination of the germ cells during sex determination in the gonad. There is a hypothesis that EEMs can affect, through an epigenetic process, the germ line by altering DNA methylation status in germ cells during the period 89 of sex determination in the developing gonad (79). Since the estrogen-regulated genes are programmed during development, altered hormone response may be transmitted to subsequent generations. An example of an epigenetic mechanism of DES, such as an increased tumor incidence in the reproductive tract in subsequent generations, has been well documented in human and laboratory animals (76, 80, 81). Newbold et al. (82) observed that 31% of uterine tumor incidence in 18 month-old F1 mice exposed to DES at a dose of 0.002 ug/pup per day on PND 1 through 5, whereas the incidence was 11% in their DES descendants. Corresponding to this increased tumor incidence in subsequent generations, studies demonstrated that altered methylation patterns in uterine genes can be passed to subsequent generations (83) and hypomethylated estrogen-responsive proteins LF and c-fos were permanently up-regulated in the uterus after developmental exposure to DES (84). Additionally, methoxychlor caused transgenerational adverse effects in spennatogenic capacity including cell number and viability, and increased male infertility in rats maternally exposed to 100 or 200 mglkg bw/d from embryonic day 8 to 15 with epigenetic alteration in the DNA hyper— and hypomethylation, which was observed in subsequent generations (F1 to F4) (85). Currently, many studies into transgenerational effects of other EEMs and possible epigenetic mechanisms that govern these effects have been investigated. 90 3.4 PHYTOESTROGEN AND MYCOTOXIN 3.4.1 GENISTEIN (GEN) Genistein (4’,5,7-trihydrozyisoflavone or GEN) is a phytoestrogen, the predominant isoflavonoids in leguminous plants. A study showed that plant concentrations of GEN vary widely depending on genotype of the soybean, location, and temperature. The average concentration of GEN in 31 different genotypes of vegetable soybeans was 34 pg/g (86). GEN is found in fruits and vegetables such as grapefnrit, licorice black, and licorice tea (27, 599, 26 jig/1009 of plant tissues, respectively) (87). Since biochemical structure of GEN is similar to E2, it has been reported that GEN exerts its estrogenic effects via binding to ERs as demonstrated in in vitro and in vivo studies. The basic structural feature of GEN is the flavone nucleus, which is composed of 2 benzene rings, A and B, linked through a heterocyclic pyrane C ring (Figure 3-1). Compared to E2, its estrogenic potency is 104 to 105 times less potent in the luciferase reporter gene assay and the E- screen (88, 89). GEN has 20-fold higher binding affinity for ERB than ERor as determined in an in vitro assay (90). Furthermore, the transactivational potency of GEN at ERa and ERB is approximately 2-fold greater than a similar concentration of E2 (91 ). There have been great concerns of adverse effects on reproductive health of GEN exposure during the developmental period because of its concentrations measured in soy-based infant formula and its similarity of biochemical structure with E2 (92, 93). Some soy-based formulas contains 40 mg/l of soy isoflavones 91 consisting in part of GEN and its glycosidic conjugates (94). The estimated daily intake of total isoflavones in infants fed a soy-based infant formula is 5 to 8 mglkg bw/d, which is approximately 200 times higher compared to that in adults who consumed soy-based products (0.025 to 0.068 mglkg bw/d) (23). Furthermore, the plasma concentration of GEN measured in infants fed soy- based formulas was approximately 684 ng/ml, which was a greater amount than found in infants fed cow’s milk formula (3.2 ng/ml) or human breast milk (2.8 ng/ml) (95). In laboratory animal studies, adverse effects of GEN in the reproductive system have been reported. In utero exposure to 50 mg GEN lkg bw/d results in significant increases in uterine adenocarcinomas in 18-month—old mice, which is roughly middle age (20). Maternal exposure to GEN (100 mglkg of maternal body weight) from GD 7 decreased average live pup weight at birth and offspring body weight (96). Neonatal exposure to GEN (1, 10, or 100 ug/pup/day) for 5 days caused a dose-related multioocyte follicles and induced ERa expression in granulosa cells in mice (97). Additionally, in the same study, an increase in the number of ovulated oocytes was observed in the 1 pg treated mice, whereas a decrease was observed in 10 and 100 pg GEN-treated mice. A dose- and age- related prolonged estrous cyclicity in mice was also observed following neonatal exposure to GEN at doses ranging from 0.5 to 50 mglkg bw/d from PND 1 through PND 5 (98). Fertility and the number of live pups at birth were also decreased. Additionally, maternal exposure to GEN (15 mglkg maternal body weight) increased the estrogen-regulated progesterone receptor in the rat, which 92 has important roles in uterine proliferation, implantation, and maintenance of pregnancy (99). However, a study demonstrated that GEN (0.1, 0.5, 2.5 or 10 mglkg maternal body weight), at concentrations equal to or greater than human dietary exposure does, not interfere with reproductive development, epididymal sperm production, or sperm motion parameters, but in vitro sperm fertilizing ability increased in male offspring (100). Similarly, exposure to GEN (250 and 1000 mglkg diet) from postnatal day 21 to 35 did not alter rat testicular development (101). Collectively, further study is needed to investigate potential adverse effects of in utero and postnatal exposure to GEN. 3.4.2 ZEARALENONE (Zearalenone (ZEA) is a mycoestrogen known as resorcyclic acid lactone (RAL) and F-2 toxin. It is a family of phenolic compounds produced by several species of Fusarium (F. graminearum, F. culmorum, F. croolwellense, F. sambucinum and F. equisetr) that frequently infest many important crops (102). The concentrations of ZEA and its derivatives (e.g. a-zearalenol, B-zearalenol, a- zearalanol (zeranol), B-zearalanol (taleranol)) in cereal grains have been reported by Bottalico (103) at concentrations ranging from 175 to 758 pg/kg food. Additionally, concentrations of ZEA were measured in the range of 4 to 584 ug/kg and 55 to 1400 ug/kg in maize for human consumption and animal feed, respectively, in the UK (104, 105). ZEA was also detected in sewage influents and effluents of an Italian city at levels below 30 ng/I (106). a-Zearalenol is used 93 as an anabolic growth promoter in cattle in the US (trade names Ralgo®). The acceptable daily intake (ADI) of zeranol for humans is 0.05 mglkg bw/d and the limit of zeranol residue in edible meat tissue of cattle implanted with Ralgo® after 70 days is 0.23 mglkg (107, 108). ZEA was detected at a level of 8.7 uglkg in fresh meat and at a level of 6.9 ug/kg in milk samples (109). A Canadian study reported that the range of estimated daily exposure to ZEA from food consumption for young children is 0.05 to 0.10 ug/kg bw/d (110). The chemical structures of ZEA and its metabolites are similar to the structure of E2, and thus they have estrogenic and anabolic activities as documented in in vitro and in vivo tests (111). Consequently, ZEA and its metabolites have been associated with hyperestrogenism and other reproductive disorders by competing with E2 in binding to intracellular ERs present in the uterus, mammary gland, hypothalamus and pituitary gland in various species (rodent, pigs and monkeys) (24, 112-114). ZEA is metabolized rapidly and its half-life in the rat intestine is approximately 5 min (115). Compared to E2, ZEA has an approximately 20-fold lower binding affinity to ERs. a-Zearalenol was as potent as EE2 and DES in the human endometrial lshikawa cell line (116). The relative binding affinity of ZEA and its metabolites for ERs in the pig, rat and chicken have shown that a-zearalenol exhibiting greater affinity than ZEA and while B-zearalenol had the lowest binding affinity in all species tested. Furthermore, the relative binding affinity of a-zearalenol was greater in swine than in the rat and significantly greater than in the chicken because of differences in ZEA metabolites formed (113). 94 It has been well documented that ZEA disrupts early pregnancy in swine. Diekman and Green reviewed the adverse effects of ZEA on swine (117). The age of onset of puberty at 1.8 ppm, ZEA (naturally contaminated milo) fed prepubertal gilts was decreased, while it was increased in gilts fed 10 ppm purified ZEA. This result may indicate that ZEA can act as an estrogenic agonist at a low dose and as an antagonist at a high dose, although it may be due to difference in purity of ZEA and the strain of test animals. In cycling gilts or sows, an extended interestrus interval was observed in all animals, regardless of the purity of ZEA and the treatment period. For the pubertal pig, the no observable adverse effect level (NOAEL) of ZEA is 0.06 mglkg/day (110). In mated gilts or sows, postmating day 7 to 10 is a Critical period of gestation for ZEA to exert its detrimental actions on early embryonic development. Effects of ZEA on reproduction in boars are decreased libido, testes weights, epididymis weight, serum testosterone, spennatogenesis, and sperm motility. On the other hand, the common effects of exposure to ZEA in cattle are abortion, decreased fertility, abnormal estrus cycles (e.g., prolonged, skipped, or irregular heats), swollen vulvas, vaginitis, reduced milk production, and mammary gland enlargement of virgin heifers (118, 119). Weaver et‘al. (120) observed a reduced conception rate in virgin heifers that consumed ZEA at greater concentrations than 25 ppm administrated by gelatin capsule (62% vs. 87%) without changes of serum of progesterone concentrations. Collectively, the potential health risk of ZEA from food will need to be further investigation, specially for young children. 95 3.5 PHARMACEUTICALS 3.5.1 DIETHYLSTILBESTROL (DES) Diethylstilbestrol (DES) is a non-steroidal synthetic estrogen that was first manufactured in London in 1938 (121). In 1947, the US Food and Drug Administration (FDA) approved DES for preventing habitual abortion. Therefore, between the late 19405 and 1971, DES was administered to pregnant women to prevent miscarriage and other pregnancy complications in the US (1.4 to 17.9 9 total dose during pregnancy). As a result, an estimated 4 million pregnant women have been exposed to DES in utero (81, 88). Furthermore, DES has been used to restrain lactation, to manage menopausal symptoms, and to treat breast and prostate cancer (122). In 1953, a study demonstrated that DES did not prevent miscarriages or premature births (123). However, DES continued to be prescribed until 1971 in the US and until the late 19705 in European countries (124). The first adverse reproductive effects of DES, eight cases of vaginal cancer in Boston area between 1966 and 1969, were reported in 1971. A study concluded that prenatal exposure to DES causes vaginal and cervical clear cell adenocarcinoma (CCA) in women younger than 50 (125). When women who were exposed to DES in utero reached puberty, an increase in cases of genital cancer (126) and morphological and functional abnormalities in their ovaries, uterus, and/or oviduct had been reported (127, 128). For example, in utero exposure to DES caused a 40 times higher incidence of CCA of the vagina and cervix in female offspring than woman not exposed to DES (126, 129). 96 Studies suggest that estrogenic compounds can alter Miillerian duct development that occurs between the sixth and eleventh weeks of pregnancy in humans and laboratory animals. Data from epidemiologic studies demonstrate that there is astrong correlation between DES exposures in utero and urogenital and reproductive abnormalities, such as increased cryptorchidism, urogenital abnormalities epidermal cysts, and hypotrophic testes (130, 131). Correspondingly, laboratory animal studies demonstrated that significant increased cryptorchidism (91%) and testicular cancer (8%) occurred in mice that were exposed to 100 pg DES/kg bw/d from gestational day 9 to 16 (132). The exact mechanisms by which DES alter development of the reproductive tract are unknown, but a possible explanation is that DES alters the expression of certain genes (e.g. insulin-like factor 3, homeobox A10 [Hoxa 10], and Wingless-type MMTV integration site family member 7a [Wnt-7a]) that are involved in the development of the reproductive tract during a critical period of development. For example, studies demonstrated that insulin-like factor 3 has an important role in the transabdominal phase of testis descent, and in utero exposure to DES (100 pg/kg maternal body weight) from GD 9 through 17 decreased lnsl3 mRNA expression in the mouse fetal testis (133). Environmental estrogenic compounds at high levels of exposure decrease body weight of laboratory animals but at lower levels of exposure, they may increase body weight (134). Recently, Newbold et al. (135) demonstrated that in utero exposure to an extremely low doses of DES (1 part per billion) significantly increased body weight of mice at 4 months of age. It indicates that there may be 97 an association between in utero exposure to low doses of DES and disruption of the body's natural weight-control mechanisms resulting in interference of the normal developmental and homeostatic controls over adipogenesis and energy balance. 3.5.2 TAMOXIFEN Tamoxifen is a selective estrogen receptor modulator, which was first used to treat early and advanced breast cancer in women via regulating growth of breast cancer cells. It was invented by AstraZeneca Pharmaceutical Company and it has been commercially available since the mid 1970’s. In the 1950’s, its potential use as an oral contraceptive was investigated because of its estrogenic antagonistic activity. It was approved by the US FDA in 1998 for women at risk developing for breast cancer. Tamoxifen has also been used to treat infertility in women with anovulatory disorders (10-40 mg/day in days 3 to 7 of the menstrual cycle). Tamoxifen and its metabolite (4-hydroxytamoxifen, endoxifen) have both estrogenic and antiestrogenic activities (136, 137). Compared to the estrogenic potency of E2 in an recombinant yeast cell assay, tamoxifen is approximately 106 times less potent than E2. Tamoxifen and 4-hydroxytamoxifen have similar estrogenic effects in an uterotropic assay but the estrogenic activity of tamoxifen is increased by hydroxylation in a receptor binding assay (138). Endoxifen has 100-fold greater affinity for the ER and in 30 to 100-fold more potent than tamoxifen in suppressing estrogen-dependent cell proliferation (139). 98 An epidemiologic study reported that endometrial thickness and abnormalities such as endometrial polyps increased with the duration of tamoxifen treatment that was longer than 5 years in postmenOpausal breast cancer patients (140). Suggested mechanisms of action of tamoxifen are to compete with endogenous estrogen in the body for binding to ERs in breast tissue. Therefore, transcription of estrogen-stimulated genes are inhibited. Additionally, the well-known antiestrogen tamoxifen inhibits protein kinase C activity (141), and thus tamoxifen has the growth-inhibitory and cytotoxic effects in cultured mouse fibroblast cells at concentrations as low as 5 pM (69). Effects of tamoxifen on reproduction in laboratory animals have been reported. A dose-dependent decrease in epididymal sperm production and testicular tubular atrophy were observed in adult male rats orally exposed to 400 mg tamoxifeng bw/d for 30 days (142). Additionally, pregnenolone biosynthesis via down regulation of P450$cc gene expression in the testis was decreased beginning at day 60 of treatment (143). Chronic oral exposure to 0.4 mglkg/day reduced (74%) the fertility of the male rats with reduced circulating testosterone and LH (144). However, tamoxifen and endoxifen did not change sperm production. An increase in the number of abnormal eggs at the 2 to 4 cell stage was observed in rats exposed to tamoxifen at 0.4 mglkg bw/d from GD 0 to GD 4 (145). 99 3.5.3 17a-ETHYNYL ESTRADIOL (EE2) Ethynyl estradiol is the major estrogenic component of oral contraceptives and in a carcinostatic agent for prostate and breast carcinomas.” Its synthesis was first reported in 1938 by lnhoffen and Hohlweg (146). Doses of EE2 in oral contraceptives are from 0.4 toZ pg/kg bw/d (147), which doses of 4 to 1400 pg/kg bw/d (0.3 to 100 mg/day) are used for arresting prostate carcinomas in man (148). The lowest teratogenic dose of EE2 in mice, 20 pg/kg bwld, was about 10 to 50 times higher than the does of 'EE2 in an oral contraceptive (147, 149). The maximum tolerated dose (MTD) resulting in reduced body weight gain was 200 pg/kg bwld in male rats treated by gavaging for 28 to 32 days beginning at 7 weeks of age (150). Additionally, the oral lethal doses to 50% of the experimental animals (LDso) in rats and mice are greater than 5,000 and, 2,500 mglkg bw/d, respectively. Concern for the adverse effects of EE2 on reproductive health of humans and animals is increasing, even though there is lack of epidemiological evidence linking oral contraceptive use and malforrnatlons at birth. Each year, approximately 2 million women in the United States (US) and the United Kingdom (UK) are exposed to 0.4 to 2.0 pg EE2/kg bw/d during undetected early pregnancy (147). Slikker et al. (151) demonstrated placental transfer of EE2 by daily intravenous administration to pregnant rhesus monkeys on gestational days 108 through 121, whereas endogenous estrogen E2 in the placenta is metabolized to estrone (E1). In the fetal circulation, 1E1 and EE2 were observed, whereas E1, E2 and EE2 were observed in the maternal circulation. Therefore, 100 fetuses can be exposed to EE2 in utero during critical stages of development of the reproductive and central nervous systems (152-154). Furthermore, there have been human and ecological health concerns about pharmaceuticals and their metabolites present in drinking water supplies because of the inability of sewage treatment facilities to remove them from the waste stream. The major endogenous estrogen metabolite in the urine is 3- methoxy-2-hydroxy-estrone glucuronide. However, sulfated EE2 is almost completely absorbed from the gastrointestinal tract and binds to serum albumin. It appears slowly in the urine, 40% after 5 days, as glucuronides and sulfates, with a minor amount unconjugated (155). Correspondingly, about 90% of a dose of titrated EE2 was recovered in both urine and feces in women and the ratio of eliminated EE2 in urine and feces was 4:6 (156). The half-life'of EE2 in human and primates is variable (approximately 8 to 27 h) due to different conjugation profiles produced by tested subjects and different methods of administration (156-158). Approximately 17 to 29% of sulfate or glucuronide metabolites of ingested EE2 being used as an oral contraceptive is excreted in the urine (159, 160). In March, 2008, an Associated Press investigation reported that pharmaceuticals, including EE2, have been detected in the US drinking water supplies of 24 major metropolitan areas supplying 41 million Americans (161). The concentrations of EE2 in sewage treatment plant effluents were in the range of 0.4 to 15 ng/l and 300 to 1800 ng/l in the UK and US, respectively (162-164). Concentrations of EE2 were measured at 0.48 pg/l in Las Vegas Wash and at 0.52 pg/I in Las Vegas Bay (165). Based on the US Environmental Protection 101 Agency’s (EPA) drinking water exposure estimates, women of childbearing age (15 to 44 years of age), could be exposed to 6.3x10’5 to 2.5x10‘2 pg EE2/kg bwld through water consumption, and children younger than one year of age could be exposed to 2.1x10'5 to 8.3x10’2 pg EE2/kg bwld (166, 167). Therefore, potential adverse effects of exposure of the fetus and neonate to EE2 through consumption of drinking water need to be investigated. Because of different assay methods and different testing cell lines used, it is difficult to determine estrogenic potency of EE2, compared to E2 or DES. Studies demonstrated that EE2 and DES may be similar or more potent than E2 based on in vitro E-screen assays (168). In postmenopausal women who received hormone replacement therapy, E2 and EE2 similarly induced vaginal epithelium (169). Correspondingly, the potencies of DES and EE2 in in vivo assays are similar in terms of increasing uterine luminal epithelium hypertrophy in the immature rat (170). In addition, estrogenic potencies of EE2 in fish have been shown to be 25 times more potent in in vivo assays (e.g., the vitellogenin assay and the uterophic assay) compared to in vitro E-screen results (171). Studies demonstrated that adverse effects of EE2 on the reproductive health of laboratory animals. Yasuda et al. (149) observed ovotestis and intra- abdominal testis with persistent Mt’rllerian and Wolffian ducts in male fetuses exposed in utero to EE2 (0.02, 0.2, or 2.0 mg/kg bw/d) from day 11 through day 17 of gestation, which is prior to gonadal differentiation. Prenatal exposure to EE2 has long-term effects on the testes such as hyperplasia of Leydig cells and atrophy of seminiferous tubules in aged mice (172). Leydig cell atrophy and 102 degeneration of germinal epithelium were observed in male rats treated by gavage with the MT D of EE2 (200 pg/kg bwld) for 28 to 32 days beginning at 7 weeks of age. There was an increase in abnormal spermatozoa and a decrease in epididymal spermatozoa count in the 10 pg/kg treatment group compared to controls but these differences were not statistically significant. The male mammary gland was sensitively affected and feminized by the low dose of EE2 (10 pg/kg) (150). After receiving 10 mg EE2/kg bw/d by daily oral gavage, epididymal sperm production was significantly decreased in 11- to 12-week-old rats, whereas the sperm production was slightly changed in the testis (173). Therefore, high doses of EE2 could decrease sperm production before it affects sperrnatogenesis in the testis. Additionally, sperm motion parameters such as percent motile sperm, velocity, linearity, and amplitude of lateral head displacement (ALH) were significantly decreased (173). 3.6 PESTICIDES 3.6.1 METHOXYCHLOR Methoxychlor (MXC) is a non-steroidal chlorinated hydrocarbon pesticide. It has been used to control insects on crops and livestock. In addition, it is used in gardens and on pets. Its half-life in the water is 20 to 200 days (174). The US EPA reference dose for MXC is 0.005 mg/kg bwld. Estrogenic activity of MXC was discovered by Bulger et al in 1978 (175). Furthermore, many studies have demonstrated its estrogenic effects in different in vivo and in vitro systems (138, 176). Methoxychlor is metabolized by cytochrome P450, and its active estrogenic 103 form is 2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroethane (HPTE) (138, 177). This metabolite has higher affinity for binding to ERs than MXC, and it possesses ER agonist and ER antagonist activities (178, 179). Compared to the estrogen potency of E2, MXC is 1,000 times less potent in the vitellogenin induction assay (180). Since studies demonstrated that MXC suppressed pre-implantation embryo development and increased the rate of embryo abnormalities in female mice, effects of MXC on the female have been extensively studied and well- documented (181, 182). Maternal exposure to MXC (33.0 mglkg bw/d) during the early post-implantation period (days 5—7) significantly decreases sexual arousal, with a concomitant decrease in testosterone concentrations in adult males (183). This suggests that MXC may alter the function of the hypothalamic- pituitary-testicular axis and thus compromise sexual behavior in male offspring. 2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroethane causes a direct inhibition of testosterone biosynthesis by Leydig cells in rats , which may result in decreased male fertility (184). 3.6.2 ATRAZINE Atrazine (2-chIoro-4-ethylamino-6-isopropylamine-1,2,5-triazine) is the most commonly used herbicide in the US with an annual application of about 30,000 tone. It also has been used in more than 80 countries (185, 186). It is produced by Syngenta. Because of concerns about atrazine’s potential ability of disrupt sex hormones, and the presence of residues in drinking water, it has 104 been banned in Germany, France, Italy, Sweden, Norway and Switzeriand. Atrazine is routinely found at concentrations as high as 21 ppb in groundwater, 42 ppb in surface water, 102 ppb in river basins in agricultural areas, and up 40 ppb in rainfall in the US midwestem agricultural areas (185, 187, 188). Drinking water in the US is allowed to have up to 3 ppb atrazine (189). There were controversial effects of atrazine in laboratory animal studies. Atrazine has no observed effect levels (NOEL) for reproductive and developmental toxicity of 5 mglkg bw/d in New Zealand White rabbits, 25 mglkg bwld in Sprague-Dawley rats, and 5 mglkg/day in a 2-generation feeding study in SD rats (190, 191). However, serum and intratesticular concentrations of testosterone were reduced approximately 50% in juvenile rat males orally exposed to atrazine (50 mglkg) either acutely- or chronically (192). Trentacoste et al. also observed that 100 mglkg of atrazine for a similar time period (PND 22 to PND 48) significantly decreased serum and intratesticular concentrations of testosterone in juvenile male rats (193). Furthermore, these in vivo results were confirmed in a in vitro study. Isolated Leydig cells from rats on PND 49 were cultured with 232 pM atrazine which resulted in a 35% decrease in testosterone production (192). This suggests that atrazine may act directly on Leydig cells and decrease testosterone production. Stoker et al. also demonstrated that doses as low as 12.5 mglkg bw/d of atrazine administrated from PND 21 until PND 53 delayed the onset of puberty in the male rat (194). In the female rodent, atrazine (300 mg/kg bw/d) induced mammary tumors and accelerated reproductive aging by inhibiting LH release from the pituitary via inhibition of 105 GnRH signaling in the adult rat hypothalamus (195, 196). However, a low dose of atrazine (25 mglkg bwld) can delay the onset of puberty and alter estrous cyclicity in the female rat (191). Vaginal opening was significantly delayed In peripubertal female rats exposed to 30 or 100 mg atrazine/kg bwld, but uterine weights did not change (197). Further studies are needed to confirm reproductive toxic effects of atrazine to estimate potential human risk. 3.7 INDUSTRIAL CHEMICALS 3.7.1 BISPHENOL A (BPA) Bisphenol A (BPA), 2,2-bis(4—hydroxyphenyl)propane, is made by combining acetone and phenol. It was synthesized by a Russian chemist, A. P. Dianin, in 1914 (198). Bisphenol A has been used to produce polycarbonate plastic and epoxy resins which are high-perfonnance plastics that are lightweight and have high heat resistance. They have a variety of uses including protective coatings, metal cans, PVC pipe, dental material, automobile parts, adhesives, plastic bottles, and electrical equipment. In 2003, more than 6.4 billion lb of BPA was produced globally (199). Heating and washing of polycarbonate products have been shown to result in an increase in the rate of leaching of BPA (200- 202). As a result, BPA contamination is wide spread in the environment through municipal waste water. The range of BPA concentrations in the environment has been reported to be 0.5 to 410 ng/l in rivers and 0.01 to 0.19 mglkg in sediment (106, 203). 106 Due to an increased production of BPA, exposure of humans to BPA through intake from food and drinking water has increased. The half-life of BPA in the human is less than 6 hours and an administrated oral dose of BPA was were completely recovered in the urine as BPA glucuronide (204). Bisphenol A has been found in human urine samples at concentrations of 0.4 to 8.0 pg/l and 0.1 to 10.0 pg/l in blood samples (205, 206). The reference dose of BPA established by the US EPA is 50 pg/kg bwld (207) and the tolerable daily intake (TDI) established by the European Commission’s Scientific Committee on Food (ECSCF) is 10 pg/kg bwld, which Is five times lower than the reference dose established by the US EPA (208). Furthermore, the daily intake of BPA for infants is about 1.6 pg/kg bwld and for children 4 to 6 years old, it is about 1.2 pg/kg bwld (209, 210). The migration limit of BPA in food from BPA containing food containers is 0.6 mglkg in the European Union (EU) (211). The reproductive toxicity of BPA at high doses has been reported in rodents. A continuous breeding toxicity study conducted by the US National Toxicology Program demonstrated that oral exposure to BPA at doses of 875 or 1750 mglkg bw/d decreased the number of live pups per liter and litters per pair in CD—1 mice in the first generation (212). Furthermore, intraperitoneal injection of BPA at 125 mg/kg from gestational day 1 to 15 decreased the number of live fetuses per litter in rat (213). On the contrary, no significant developmental toxicity resulted from oral exposure to BPA at doses of 640 mglkg in rats and at 1000 mglkg bwld in mice administrated from GD 6 though GD 15 (214). 107 Even though a positive uterotrophic effect of BPA was demonstrated in ovariectomized female rats in the 19305 (215, 216), health concerns related to BPA as an EEM was not an issue until its estrogenic activity was accidentally discovered by Krishnan’s group when they observed an increased rate of proliferation of MCF-7 breast cancer cells by BPA that had leached from autoclaved polycarbonate flasks (217). A study demonstrated that the in vitro binding affinity of BPA was approximately 20,000-fold lower than that of DES for both ERor and ERB (90). Additionally, estrogenic activity of BPA is approximately 15,000-fold less than E2, using a yeast-based gene transcription assay (218). There has been a debate whether there are adverse effects of BPA at low doses. A study demonstrated that BPA at 100 pM disrupted maturation of mouse oocytes isolated from the ovary via altering Ca2+ oscillations (219). At maternal doses as low as 2 and 20 ng BPA/kg bw/d from gestation day 11 to 17 decreased of daily sperm production by approximately 20% and fertility in male offspring (220). Stimulated development of the mammary gland in mice exposed in utero to low doses of BPA (25 and 250 pg/kg bwld) was also observed (221 ). However, other researchers reported no low-dose effects of BPA at the similar time of pregnancy and in the same strain of mice (222, 223). In order to confirm potential adverse effects of BPA at low doses, further studies are needed. 4 In addition to reproductive toxicity of induced by BPA, studies demonstrated that there has a strong association between exposure to BPA and increased risk of obesity. Bisphenol A in combination with insulin significantly increased the differentiation of fibroblasts to adipocytes, which means that BPA 108 could increase the risk of obesity (224). A study demonstrated that a 2.5-fold increase in plasma insulin and a 20% decrease in blood glucose levels 30 min after oral administration of BPA in mice (225). Additionally, BPA rapidly decreases serum glucose and increases insulin in mice injected twice daily or orally administrated 10 or 100 pg BPA/kg bw over 4 days (225). BPA treatment in vitro caused increased glucose uptake in mouse 3T3-F442A adipocytes (226). Further cohort studies are needed to investigate the possible mechanism of increased adipocytes by BPA exposure. 3.7.2 PHTHALATES Phthalate esters are the most widely used industrial chemicals. They are used principally as plasticizers to improve flexibility and durability in soft vinyl plastic toys. They are also used in shampoos, soaps, nail polish, and vinyl flooring. More than 60 different phthalates are also found in products such as paints, adhesives, inks, dentures, and cosmetics (227). Among the 60 different phthalates, DEHP in PCV, butyl benzyl phthalate (BBP) in vinyl floor tiles, dibutyl phthalate (DBP) in paints, and diisononyl phthalate (DINP) in children’s sucking toys have been investigated for their developmental and reproductive toxicity because they are able to leach from many phthalate products and thus contaminate food or drinking water (228-235). Environmental concentration of DEHP ranged from 0.33 to 97.8 pg/I in surface water, 1.74 to 182 pg/l in sewage effluents, 27.9 to 154 mglkg in sewage sludge, and 0.21 to 8.44 mglkg in sediment (203). 109 Based on the NHANES 1999-2000, more than 75% of human urine samples had detectable concentrations of four phthalate metabolites; mEP, mono-2-ethylhexyl phthalate (mEHP), mono-n-butyl phthalate (mnBP), and mBzP (236). Four phthalate monoesters were detected in all urine specimens from African-American women in the Washington DC. Concentrations were 31 ng/ml for monobenzyl phthalate (mBzP), 53 ng/mL for monobutyl phthalate (mBP), 211 ng/ml for monoethyl phthalate (mEP), and 7.3 ng/ml for mono- ethylhexyl phthalate (mEHP) (237). The half-life of phthalate monoester and its metabolites is approximately 12 hr in humans (237). The main route of exposure of humans to phthalates is oral. Examples include ingestion of food, or the chewing of toys by small children (238). Studies reported that the estimated daily intake of DBP is approximately 0.48 pg/kg bwld, DEHP is 18 pg/kg bwld, and BBP is 0.11 to 0.29 pg/kg bw/d (239). Other potential exposure routes may include skin contact from constantly handling plastic products. The first documented estrogenicity of a phthalate in vivo was published by Sharpe et al. (240). In this study, gestational and lactational exposure to BBP decreases testis weight and daily sperm production in rats. Even though Sharpe et 3! clearly stated that there was no evidence of a specific estrogenic mechanism, the endocrine disrupting effects of phthalate and its metabolites in laboratory animals have been investigated and demonstrated. These effects include reduced AGD, nipple and areola retention, cleft phallus, hypospadias, and undescended testes via reducing testosterone synthesis by the fetal testis 110 (231, 241, 242). A decreased in testosterone concentrations can be reversed with clearance of DBP and its metabolites from the testis (243). There was a dose-response relationship between urine levels of phthalate metabolites such as mBP and mBzP and lower sperm motility and concentrations in humans (244). Additionally, there was a significant inverse association between human AGD and concentrations of phthalate metabolites (245). Dibutyl phthalate and DEHP treatment decreased of fetal testosterone concentrations by 60 to 85% during the critical period of development resulting in hypospadlas and cryptorchidism (246). In contrast to the apparent adverse effects of phthalate on male reproductive health, females have been thought to be much less sensitive to phthalate-induced disruption. However, there is evidence that at doses (500 or 1000 mg DBP/kg bw/d from weaning to pregnancy) similar to those that affect testis function in male rats increased midpregnancy abortions and decreased the number of delivered live pups (247). The most recent hypothesis on the mechanisms behind the reproductive effects of phthalates is that they may have an inhibitory effect on androgen activity rather than having estrogenic activity. Sohoni and Sumpter demonstrated that BBP has antagonistic activity in a recombinant yeast-based androgen screen (248). However, Zacharewski et al. observed that DBP, BBP, and DHP have weak ER-mediated activity in ER competitive ligand-binding and mammalian- and yeast-based gene expression assays (249). The fetal testis within one hours of in utero exposure to 500 mg DBP/kg bwld from GD 12 to GD19, there was down regulation in the transcription of genes (i.e. SR—B1, StAR, P450scc, and 111 38-HSD) in the tests that are involved in cholesterol transport and the biosynthesis of testosterone (243, 250). Consequently, testosterone concentration in fetuses was decreased by 87% as early as GD 17. Taken together, confirmation of whether current human exposure levels of phthalate have any consequences for human reproduction as well as whether phthalate and its metabolites have estrogenic and/or antiandrogenic effects on reproduction is still needed. 3.8 ENVIRONMENTAL POLLUTANTS 3.8.1 POLYCHLORINATED BIPHENYLS (PCBs) Polychlorinated biphenyls are a family of 209 congeners, which consist of two linked phenyl rings and variable chlorination. PCBs were first introduced and commercially produced in the late 1920s in the US (251). Due to their non- flammability and chemical stability, PCBs were used widely for various industrial and commercial purpose such as electrical, heat transfer system, hydraulic equipment, plastic resins, pigments, dyes, and carbonless copy paper. Approximately 1.5 billion pounds of PCBs were manufactured in the US until they was banned in 1977 (252). However, approximately 70% of the manufactured PCBs are still in use (253, 254). Persistence of PCBs in the environment and in humans has been well documented (255). The half-lives for most PCB congeners in the human body ranged from a few years to 20 years (256). A recent study reported relatively high serum concentration of PCBs in humans residing near a former 112 manufacturing site in eastern Slovakia that ceased PCB production 20 years before (3105 ng/g vs. 871 ng/g of lipid in the reference population) (254). Laboratory animal studies demonstrated that PCBs alter endocrine, immune, and nervous system functions leading to reproductive tract, alterations, decreased embryonic growth, delayed implantation, and increased abortion rates in animals (252, 257, 258). An influence on the sexual differentiation of the brain by maternal PCB-exposure (40 mglkg) resulted in reduced aromatase activity, decreased testis weights, and increased uterotrophic effects in rats (259). Studies indicated that PCB-induced abnormal development of the female reproductive tract in mice resulted from down-regulation of estrogen-mediated Wnt genes (Wingless-type MMTV integration site family), which is a known factor in the reproductive deficits found in mice exposed to DES (252). In vitro studies using the E-SCREEN assay identified weak estrogenic potency of PCB congeners (i.e. PCBs 17, 18, 30, 44, 49, 66, 74, 82, 103, 110, 128, and 179) at environmentally relevant concentrations from a superfund site in Massachusetts (260). Laboratory animal studies related to PCBs as EEMs have been clearly demonstrated and reviewed (261 ). However, even though eidemiological studies have demonstrated that there was strong association between neurodevelopmental delay and prenatal or early postnatal PCB-exposure at environmental concentrations, it is difficult to prove a causative role of PCBs in producing neurodevelopmental deficits in these cohort studies alone (262, 263). 113 Further epidemiological studies are needed to confirm potential adverse effects of PCBs as EEMs on reproductive health in humans. 3.8.2 BENZO[A]PYRENE Benzo[a]pyrene (B[a]P) is a member of the class of lipophilic polycyclic aromatic hydrocarbons (PAHs), which occur naturally in gas. It is also continually released into the environment through various combustion processes, including automobile exhausts, manufacturing of petroleum, aluminum products, and expulsion of fumes from industrial plants. The major route of human exposure to B[a]P is ingestion of meat and dairy foods and the minor route of human exposure is inhalation (264). The estimated average daily intake of B[a]P in the US population is 1.1 to 2.2 pg per day (264). Benzo[a]pyrene is biotransformed to polar metabolites by the cytochrome P450 enzymes, predominantly CYP1A1, 1A2, 2A1, 3A4, and 1B1 (265-269). Main metabolites of B[a]P are 1-,3-,7-, and 9-OH-B[a]P, 4,5-, 7.8-, and 9,10-diOH-B[a]P, and 1,6-, 3,6-, and 6,12-B[a]P-dione (270-272). Most B[a]P studies have been focused on its carcinogenic effects. Laboratory animal studies indicate metabolites of B[a]P, +7,8-oxide (7,8-O), (-)— dihydrodio (DHD), and (+)-diol-epoxide-2 (DE2), which are capable of covalent binding to cellular macromolecules such as DNA, RNA, or protein, induce tumor formation in estrogen-response tissues such as ovary, uterus, and mammary gland (273-275). 114 In addition to the carcinogenic effects of B[a]P, there have been conflicting results related to the estrogenic and/or antiestrogenic effects of B[a]P because B[a]P itself or specific metabolites may be either ER agonists or antagonists. Benzo[a]pyrene decreases ovarian follicle growth and ovulation, and corpora lutea formation may due to its antiestrogenic effects through ER-mediated pathways (276, 277). In vitro studies indicated that B[a]P inhibits estradiol- dependent reporter activity in hER and decreases E2-induced MCF-7 cell proliferation (278, 279). Furthermore, one study demonstrated that B[a]P has estrogenic effects such as uterotrophic effects in the rodent uterus (280) but another study could not demonstrate such effects (281). Because of these inconsistent results, it has been hypothesized that the antiestrogenic effects of B[a]P may be mediated through the AhR. Studies demonstrated that B[a]P possesses high binding affinity for the AhR (282, 283) and the AhR-ligand complex alters the transcription rate of E2-inducible genes (284). Additionally, monohydroxylated metabolites of B[a]P have weak estrogenic activity or antiestrogenic activity via binding to ER. In a receptor competition assay using MCF-cells, only monohydroxylated B[a]P metabolites showed affinity for ERs: 1-, 3-, 7-, and 9-OH-B[a]P caused approximately 20 to 60% displacement of E2 binding to hERor and the IC50 values were 3.3 pM, 90 nM, 10 pM, and 2.0 pM, respectively (IC50 values of E2 were approximately 5.5 nM for ERor and ERB) (281). Overall, 3-OH-B[a]P has the strongest estrogenic affinity for binding ERs. 115 3.9 BIBLIOGRAPHY 1. Iguchi T, Sumi M, Tanabe S 2002 Endocrine disruptor issues in Japan. Congenit Anom (Kyoto) 42:106-1 19 Elias EA, Kincaid RL 1984 Fertility of female mice fed coumestrol and diethylstilbestrol. J Environ Sci Health B 192441 -451 Kincl FA 1966 Influence of steroid treatment during neonatal life on sexual maturation. Proc R Soc Med 592817-818 Kavlock RJ, Daston GP, DeRosa C, Fenner—Crisp P, Gray LE, Kaattari S, Lucier G, Luster M, Mac MJ, Maczka C, Miller R, Moore J, Rolland R, Scott G, Sheehan DM, Sinks T, Tilson HA 1996 Research needs for the risk assessment of health and environmental effects of endocrine disruptors: a report of the US. EPA-sponsored workshop. Environ Health Perspect 104:715—740 EPA US 1997 Special report on Environmental Endocrine Disruption: An Effects Assessment and Analysis. In. Washington DC: Office of Research and Development; EPA/630/R-696/012 European Commission 1997 European Workshop on the Impact of Endocrine Disrupters on Human Health and the Environment. In. DG XII: Environment and Climate Research Programme Dickerson RL, Brouwer A, Earl Gray L, Grothe DR, Peterson RE, Sheehan DM, M Alfred Wiedow CSM 1998 Dose-response relationships. In: KENdall R, Dickerson R, Glesy J, Suk W eds. Principles and processes for evaluating endocrine disruption in wildlife. Danvers, MA: SETAC Press; 69-96 Colbom T 2004 Neurodevelopment and endocrine disruption. Environ Health Perspect 1122944-949 Brevinl TA, Cillo F, Antonini S, Gandolfi F 2005 Effects of endocrine disrupters on the oocytes and embryos of farm animals. Reprod Domest Anim 402291 -299 116 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Abbassy MS, Ibrahim HZ, el-Amayem MM 1999 Occurrence of pesticides and polychlorinated biphenyls in water of the Nile river at the estuaries of Rosetta and Damiatta branches, north of Delta, Egypt. J Environ Sci Health B 342255-267 Nakada N, Nyunoya H, Nakamura M, Hara A, Iguchi T, Takada H 2004 Identification of estrogenic compounds in wastewater effluent. Environ Toxicol Chem 23:2807-2815 Peck M, Gibson RW, Kortenkamp A, Hill EM 2004 Sediments are major sinks of steroidal estrogens in two United Kingdom rivers. Environ Toxicol Chem 232945-952 Wild SR, Jones KC 1992 Organic chemicals entering agricultural soils in sewage sludges: screening for their potential to transfer to crop plants and livestock. Sci Total Environ 1 19285-1 19 Harrison EZ, Oakes SR, Hysell M, Hay A 2006 Organic chemicals in sewage sludges. Sci Total Environ 367:481-497 Melnick R, Lucier G, Wolfe M, Hall R, Stancel G, Prins G, Gallo M, Reuhl K, Ho SM, Brown T, Moore J, Leakey J, Haseman J, Kohn M 2002 Summary of the National Toxicology Program's report of the endocrine disruptors low-dose peer review. Environ Health Perspect 1 102427-431 Almstrup K, Fernandez MF, Petersen JH, Olea N, Skakkebaek NE, Leffers H 2002 Dual effects of phytoestrogens result in u-shaped dose- response curves. Environ Health Perspect 110:743—748 Jefferson WN, Newbold RR 2000 Potential endocrine-modulating effects of various phytoestrogens in the diet. Nutrition 162658-662 North K, Golding J 2000 A maternal vegetarian diet in pregnancy is associated with hypospadias. The ALSPAC Study Team. Avon Longitudinal Study of Pregnancy and Childhood. BJU Int 85:107-113 Setchell KD 2001 Soy isoflavones—benefits and risks from nature's selective estrogen receptor modulators (SERMs). J Am Coll Nutr 2013548- 362$ 117 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. Newbold RR, Banks EP, Bullock B, Jefferson WM 2001 Uterine adenocarcinoma in mice treated neonatally with genistein. Cancer Res 61:4325-4328 Liggins J, Bluck LJ, Runswick S, Atkinson C, Coward WA, Bingham SA 2000 Daidzein and genistein content of fruits and nuts. J Nutr Biochem 1 1 :326-331 Reinli K, Block G 1996 Phytoestrogen content of foods—a compendium of literature values. Nutr Cancer 262123-148 Fukutake M, Takahashi M, lshida K, Kawamura H, Sugimura T, Wakabayashi K 1996 Quantification of genistein and genistin in soybeans and soybean products. Food Chem Toxicol 342457-461 Etienne M, Jemmali M 1982 Effects of zearalenone (F2) on estrous activity and reproduction in gilts. J Anim Sci 5521-10 Davis BJ, Weaver R, Gaines LJ, Heindel JJ 1994 Mono-(2-ethylhexyl) phthalate suppresses estradiol production independent of FSH-CAMP stimulation in rat granulosa cells. Toxicol Appl Pharmacol 128:224-228 Lovekamp TN, Davis BJ 2001 Mono-(2-ethylhexyl) phthalate suppresses aromatase transcript levels and estradiol production in cultured rat granulosa cells. Toxicol Appl Pharmacol 172:217-224 Andrade AJ, Grande SW, Talsness CE, Grote K, Chahoud l 2006 A dose-response study following in utero and lactational exposure to di-(2- ethylhexyl)-phthalate (DEHP): non-monotonic dose-response and low dose effects on rat brain aromatase activity. Toxicology 2272185-192 Sharpe RM 2001 Hormones and testis development and the possible adverse effects of environmental chemicals. Toxicol Lett 1202221-232 Sharpe RM 2006 Pathways of endocrine disruption during male sexual differentiation and masculinization. Best Pract Res Clin Endocrinol Metab 20291-1 10 118 30. 31. 32. 33. 34. 35. 36. 37. 38. Lottrup G, Andersson AM, Leffers H, Mortensen GK, Toppari J, Skakkebaek NE, Main KM 2006 Possible impact of phthalates on infant reproductive health. Int J Androl 292172-180 Labrie F, Simard J, Luu-The .V, Pelletier G, Belanger A, Lachance Y, Zhao HF, Labrie C, Breton N, de Launoit Y 1992 Structure and tissue- specific expression of 3 beta-hydroxysteroid dehydrogenase/S-ene-4-ene isomerase genes in human and rat classical and peripheral steroidogenic tissues. J Steroid Biochem Mol Biol 412421-435 Bellemare V, Faucher F, Breton R, Luu-The V 2005 Characterization of 17alpha-hydroxysteroid dehydrogenase activity (17alpha-HSD) and its involvement in the biosynthesis of epitestosterone. BMC Biochem 6:12 Whitehead SA, Rice S 2006 Endocrine-disrupting chemicals as modulators of sex steroid synthesis. Best Pract Res Clin Endocrinol Metab 20245-61 Ohno S, Shinoda S, Toyoshima S, Nakazawa H, Makino T, Nakajin S 2002 Effects of flavonoid phytochemicals on cortisol production and on activities of steroidogenic enzymes in human adrenocortical H295R cells. J Steroid Biochem Mol Biol 802355-363 Le Bail JC, Champavier Y, Chulia AJ, Habrioux G 2000 Effects of phytoestrogens on aromatase, 3beta and 17beta-hydroxysteroid dehydrogenase activities and human breast cancer cells. Life Sci 6621281- 1291 Wright AS, Thomas LN, Douglas RC, Lazier CB, Rittmaster RS 1996 Relative potency of testosterone and dihydrotestosterone in preventing atrophy and apoptosis in the prostate of the castrated rat. J Clin Invest 98:2558—2563 Grino PB, Griffin JE, Wilson JD 1990 Testosterone at high concentrations interacts with the human androgen receptor similarly to dihydrotestosterone. Endocrinology 126:1 165-1 172 Yamamoto M, Narita A, Kagohata M, Shirai M, Akahori F, Arishima K 2005 Effects of maternal exposure to 3,3',4,4',5-pentachlorobiphenyl (PCB126) or 3,3',4,4',5,5'-hexachlorobiphenyl (PCB169) on testicular 119 39. 40. 41. 42. 43. 45. 46. 47. steroidogenesis and sperrnatogenesis in male offspring rats. J Androl 262205-214 Bretveld RW, Thomas CM, Scheepers PT, Zielhuis GA, Roeleveld N 2006 Pesticide exposure: the hormonal function of the female reproductive system disrupted? Reprod Biol Endocrinol 4230 Stoker TE, Goldman JM, Cooper RL 2001 Delayed ovulation and pregnancy outcome: effect of environmental toxicants on the neuroendocrine control of the ovary. Environ Toxicol Pharmacol 92117-129 Costa LG, Olibet G, Murphy SD 1988 Alpha 2-adrenoceptors as a target for fonnamidine pesticides: in vitro and in vivo studies in mice. Toxicol Appl Phannacol 932319-328 Young FM, Menadue MF, Lavranos TC 2005 Effects of the insecticide amitraz, an alpha2-adrenergic receptor agonist, on human luteinized granulosa cells. Hum Reprod 20:3018-3025 Jongbloet PH 2004 Over-ripeness ovopathy: a challenging hypothesis for sex ratio modulation. Hum Reprod 192769-774 Dunn JF, Nisula BC, Rodbard D 1981 Transport of steroid hormones: binding of 21 endogenous steroids to both testosterone-binding globulin and corticosteroid-binding globulin in human plasma. J Clin Endocrinol Metab 53:58-68 Kragh-Hansen U 1981 Molecular aspects of ligand binding to serum albumin. Pharmacol Rev 33:17-53 Sodergard R, Backstrom T, Shanbhag V, Carstensen H 1982 Calculation of free and bound fractions of testosterone and estradiol-17 beta to human plasma proteins at body temperature. J Steroid Biochem 16:801-810 Baker ME 2002 Albumin, steroid hormones and the origin of vertebrates. J Endocrinol 175:121-127 120 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. Kahn SM, Hryb DJ, Nakhla AM, Romas NA, Rosner W 2002 Sex hormone-binding globulin is synthesized in target cells. J Endocrinol 17521 1 3-1 20 Crisp TM, Clegg ED, Cooper RL, Wood WP, Anderson DG, Baetcke KP, Hoffmann JL, Morrow MS, Rodier DJ, Schaeffer JE, Touart LW, Zeeman MG, Patel YM 1998 Environmental endocrine disruption: an effects assessment andanalysis. Environ Health Perspect Suppl 1:11-56 Jones EJ, DeChemey AH 2003 The female reproductive system. In: Boron WF, Boulpaep EL eds. Medical physiology. Philadelphia: Saunders; 1141-1208 Grow DR 2002 Metabolism of endogenous and exogenous reproductive hormones. Obstet Gynecol Clin North Am 292425-436 Garreau B, Vallette G, Adlercreutz H, Wahala K, Makela T, Benassayag C, Nunez EA 1991 Phytoestrogens: new ligands for rat and human alpha-fetoprotein. Biochim Biophys Acta 1094:339-345 Bergstrand DG, Czar B 1957 Paper electrophoretic study of human fetal serum proteins with demonstration of a new protein fraction. Scand J Clin Lab Invest 9:277-286 Mizejewski GJ 2003 Levels of alpha-fetoprotein during pregnancy and early infancy in normal and disease states. Obstet Gynecol Surv 58:804- 826 Zhu J, Barratt CL, Lippes J, Pacey AA, Lenton EA, Cooke ID 1994 Human oviductal fluid prolongs sperm survival. Fertil Steril 61:360-366 Zhu J, Barratt CL, Lippes J, Pacey AA, Cooke ID 1994 The sequential effects of human cervical mucus, oviductal fluid, and follicular fluid on sperm function. Fertil Steril 6121129-1135 Mizejewski GJ 1995 The phylogeny of alpha-fetoprotein in vertebrates: survey of biochemical and physiological data. Crit Rev Eukaryot Gene Expr 52281-316 121 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. Mizejewski GJ 2004 Biological roles of alpha-fetoprotein during pregnancy and perinatal development. Exp Biol Med (Maywood) 2292439- 463 Hodgert JH, Zacharewski TR, Hammond GL 2000 Interactions between human plasma sex hormone-binding globulin and xenobiotic ligands. J Steroid Biochem Mol Biol 752167-176 Danzo BJ 1997 Environmental xenobiotics may disrupt normal endocrine function by interfering with the binding of physiological ligands to steroid receptors and binding proteins. Environ Health Perspect 105:294-301 Kelce WR, Stone CR, Laws SC, Gray LE, Kemppainen JA, Wilson EM 1995 Persistent DDT metabolite p,p'-DDE is a potent androgen receptor antagonist. Nature 375:581-585 Kelce WR, Stone CR, Laws SC, Gray LE, Kemppainen JA, Jr., Wilson EM 1995 Persistent DDT metabolite p,p'-DDE is a potent androgen receptor antagonist. Nature 3752581 Nardulli AM, Katzenellenbogen BS 1986 Dynamics of estrogen receptor turnover in uterine cells in vitro and in uteri in vivo. Endocrinology 1 19:2038-2046 Pakdel F, Le Goff P, Katzenellenbogen BS 1993 An assessment of the role of domain F and PEST sequences in estrogen receptor half-life and bioactivity. J Steroid Biochem Mol Biol 462663-672 Alarid ET, Bakopoulos N, Solodin N 1999 Proteasome-mediated proteolysis of estrogen receptor: a novel component in autologous down- regulation. Mol Endocrinol 13:1522-1534 Lonard DM, Nawaz Z, Smith CL, O'Malley BW 2000 The 265 proteasome is required for estrogen receptor-alpha and coactivator turnover and for efficient estrogen receptor-alpha transactivation. Mol Cell 52939-948 Alarid ET, Preisler-Mashek MT, Solodin NM 2003 Thyroid hormone is an inhibitor of estrogen-induced degradation of estrogen receptor-alpha 122 68. 69. 70. 71. 72. 73. 74. 75. 76. protein: estrogen-dependent proteolysis is not essential for receptor transactivation function in the pituitary. Endocrinology 144:3469-3476 Fan M, Bigsby RM, Nephew KP 2003 The NEDD8 pathway is required for proteasome-mediated degradation of human estrogen receptor (ER)- alpha and essential for the antiproliferative activity of ICI 182,780 in ERalpha-positive breast cancer cells. Mol Endocrinol 172356-365 O'Brian CA, Liskamp RM, Solomon DH, Weinstein IB 1985 Inhibition of protein kinase C by tamoxifen. Cancer Res 45:2462-2465 Safe SH 1995 Environmental and dietary estrogens and human health: is there a problem? Environ Health Perspect 1032346-351 Safe S, Astroff B, Harris M, Zacharewski T, Dickerson R, Romkes M, Biegel L 1991 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related compounds as antioestrogens: characterization and mechanism of action. Pharmacol Toxicol 692400-409 Strichman-Almashanu LZ, Lee RS, Onyango PO, Perlman E, Flam F, Frieman MB, Feinberg AP 2002 A genome-wide screen for normally methylated human CpG islands that can identify novel imprinted genes. Genome Res 122543-554 Reik W, Dean W, Walter J 2001 Epigenetic reprogramming in mammalian development. Science :0: 293:1089-1093 Jiang YH, Bressler J, Beaudet AL 2004 Epigenetics and human disease. Annu Rev Genomics Hum Genet 52479-510 Allegrucci C, Thurston A, Lucas E, Young L 2005 Epigenetics and the gerrnline. Reproduction 129:137-149 Newbold RR, Padilla-Banks E, Jefferson WN 2006 Adverse effects of the model environmental estrogen diethylstilbestrol are transmitted to subsequent generations. Endocrinology 147:Si 1-17 123 77. 78. 79. 80. 81. 82. 83. 84. 85. Anway MD, Memon MA, Uzumcu M, Skinner MK 2006 Transgenerational effect of the endocrine disruptor vinclozolin on male spennatogenesis. J Androl Epub ahead of print Lee J, Inoue K, Ono R, Ogonuki N, Kohda T, Kaneko-lshino T, Ogura A, lshino F 2002 Erasing genomic imprinting memory in mouse clone embryos produced from day 11.5 primordial germ cells. Development 12921807-1817 Li S, Hursting SD, Davis BJ, McLachIan JA, Barrett JC 2003 Environmental exposure, DNA methylation, and gene regulation: lessons from diethylstilbesterol-lnduced cancers. Ann N Y Acad Sci 9832161-169 Skinner MK, Anway MD 2005 Seminiferous cord formation and germ-cell programming: epigenetic transgenerational actions of endocrine disruptors. Ann N Y Acad Sci 1061218-32 Veurink M, Koster M, Berg LT 2005 The history of DES, lessons to be Ieamed. Phann Worid Sci 272139-143 Newbold RR, Hanson RB, Jefferson WN, Bullock BC, Haseman J, McLachIan JA 1998 Increased tumors but uncompromised fertility in the female descendants of mice exposed developmentally to diethylstilbestrol. Carcinogenesis 19: 1 655-1 663 Li S, Washbum KA, Moore R, Uno T, Teng C, Newbold RR, McLachIan JA, Negishi M 1997 Developmental exposure to diethylstilbestrol elicits demethylation of estrogen-responsive lactoferrin gene in mouse uterus. Cancer Res 57:4356-4359 Li S, Hansman R, Newbold R, Davis B, McLachIan JA, Barrett JC 2003 Neonatal diethylstilbestrol exposure induces persistent elevation of c-fos expression and hypomethylation in its exon-4 in mouse uterus. Mol Carcinog 38:78-84 Anway MD, Cupp AS, Uzumcu M, Skinner MK 2005 Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308:1466-1469 124 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. Mebrahtu T, Mohamed A, Wang CY, Andebrhan T 2004 Analysis of isoflavone contents in vegetable soybeans. Plant Foods Hum Nutr 59:55- 61 Mazur W 1998 Phytoestrogen content in foods. Baillieres Clin Endocrinol Metab 122729-742 Shapiro S, Slone D 1979 The effects of exogenous female hormones on the fetus. Epidemiol Rev 1 :1 10-123 Mussman HC 1975 Drug and chemical residues in domestic animals. Fed Proc 342197-201 Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, van der Saag PT, van der Burg B, Gustafsson JA 1998 Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 1 39:4252-4263 Belcher SM, Zsamovszky A 2001 Estrogenic actions in the brain: estrogen, phytoestrogens, and rapid intracellular signaling mechanisms. J Pharmacol Exp Ther 299:408-414 Murrill WB, Brown NM, Zhang JX, Manzolillo PA, Barnes S, Lamartiniere CA 1996 Prepubertal genistein exposure suppresses mammary cancer and enhances gland differentiation in rats. Carcinogenesis 1 721451-1457 Shen F, Xue X, Weber G 1999 Tamoxifen and genistein synergistically down-regulate signal transduction and proliferation in estrogen receptor- negative human breast carcinoma MDA-MB-435 cells. Anticancer Res 19:1657-1662 Setchell KD, Zimmer-Nechemias L, Cai J, Heubi JE 1998 lsoflavone content of infant formulas and the metabolic fate of these phytoestrogens in early life. Am J Clin Nutr 68:14538-14618 Setchell KD, Zimmer-Nechemias L, Cai J, Heubi JE 1997 Exposure of infants to phyto-oestrogens from soy-based infant formula. Lancet 350223- 27 125 96. 97. 98. 99. 100. 101. 102. 103. 104. Flynn KM, Ferguson SA, Delclos KB, Newbold RR 2000 Effects of genistein exposure on sexually dimorphic behaviors in rats. Toxicol Sci 55:31 1-319. Jefferson WN, Couse JF, Padilla-Banks E, Korach KS, Newbold RR 2002 Neonatal exposure to genistein induces estrogen receptor (ER)alpha expression and multioocyte follicles in the maturing mouse ovary: evidence for ERbeta-mediated and nonestrogenic actions. Biol Reprod 67:1285—1296 Jefferson WN, Padilla-Banks E, Newbold RR 2006 Studies of the effects of neonatal exposure to genistein on the developing female reproductive system. J AOAC Int 89:1189-1 196 Hughes CL, Liu G, Beall S, Foster WG, Davis V 2004 Effects of genistein or soy milk during late gestation and lactation on adult uterine organization in the rat. Exp Biol Med (Maywood) 2292108-1 17 Fielden MR, Samy SM, Chou KC, Zacharewski TR 2003 Effect of human dietary exposure levels of genistein during gestation and lactation on long-term reproductive development and sperm quality in mice. Food Chem Toxicol 41:447-454. Fritz WA, Cotroneo MS, Wang J, Eltoum IE, Lamartiniere CA 2003 Dietary diethylstilbestrol but not genistein adversely affects rat testicular development. J Nutr 133:2287-2293. Minervini F, Dell'Aquila ME, Maritato F, Minoia P, Visconti A 2001 Toxic effects of the mycotoxin zearalenone and its derivatives on in vitro maturation of bovine oocytes and 17 beta-estradiol levels in mural granulosa cell cultures. Toxicol In Vitro 152489-495 Bottalico A 1998 Fusarium diseses of creals: species complex and related mycotoxin profiles, in Europe. Journal of Plant Pathology 80285-103 Scudamore KA, Nawaz S, Hetmanski MT 1998 Mycotoxins in ingredients of animal feeding stuffs: ll. Determination of mycotoxins in maize and maize products. Food Addit Contam 15:30-55 126 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. Scudamore KA, Patel S 2000 Survey for aflatoxins, ochratoxin A, zearalenone and fumonisins in maize imported into the United Kingdom. Food Addit Contam 172407-416 Lagana A, Bacaloni A, De Leva I, Faberi A, Fago G, Marino A 2004 Analytical methodologies for determining the occurrence of endocrine disrupting chemicals in sewage treatment plants and natural waters. Analytica Chemica Acta 501 :79-88 Doyle E 2000 Human safety of hormone implants used to promote growth in cattle. In: Galbraith H 2002 Hormones in international meat production: biological, sociological and consumer issues. Nutrition Research Reviews 15:293- 314 EI-Hoshy SM 1999 Occurrence of zearalenone in milk, meat and their products with emphasis on influence of heat treatments on its level. Archiv fur Lebensmittelhygiene 502140-143 Kuiper-Goodman T, Scott PM, Watanabe H 1987 Risk assessment of the mycotoxin zearalenone. Regul Toxicol Pharmacol 72253-306 Etienne M, Doumiad JY 1994 Effects of zearalenone or glucosinolates in the diet on reproduction in sows: a review. Livestock Porudction Science 40299-113 Boyd PA, Wittliff JL 1978 Mechanism of Fusarium mycotoxin action in mammary gland. J Toxicol Environ Health 421-8 Fitzpatrick DW, Picken CA, Murphy LC, Buhr MM 1989 Measurement of the relative binding affinity of zearalenone, alpha-zearalenol and beta- zearalenol for uterine and oviduct estrogen receptors in swine, rats and chickens: an indicator of estrogenic potencies. Comp Biochem Physiol C 942691-694 Kiang DT, Kennedy BJ, Pathre SV, Mirocha CJ 1978 Binding characteristics of zearalenone analogs to estrogen receptors. Cancer Res 38:3611-3615 127 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. Ramosa AJ, Hemandezb E, Pla-Delfinac JM, Merino M 1996 Intestinal absorption of zearalenone and in vitro study of non-nutritive sorbent materials. Int J Pharrn 128:129-137 Le Guevel R, Pakdel F 2001 Assessment of oestrogenic potency of chemicals used as growth promoter by in—vitro methods. Hum Reprod 1621030—1036 Diekman MA, Green ML 1992 Mycotoxins and reproduction in domestic livestock. J Anim Sci 70:1615—1627 Kallela K, Ettala E 1984 The oestrogenic Fusarium toxin (zearalenone) in hay as a cause of early abortions in the cow. Nord Vet Med 362305-309 Mirocha CJ, Schauerhamer B, Pathre SV 1974 Isolation, detection, and quantitation of zearalenone in maize and bariey. J Assoc Off Anal Chem 5721104-1 1 10 Weaver GA, Kurtz HJ, Behrens JC, Robison TS, Seguin BE, Bates FY, Mirocha CJ 1986 Effect of zearalenone on the fertility of virgin dairy heifers. Am J Vet Res 47:1395—1397 Dodds EC, Goldberg L, Larson W, Robinson R 1938 Estrogenic activity of certain synthetic compounds. Nature 1412247 Noller KL, Fish CR 1974 Diethylstilbestrol usage: Its interesting past, important present, and questionable future. Med Clin North Am 58:793- 810 Dieckmann WJ, Davis ME, Rynkiewicz LM, Pottinger RE 1953 Does the administration of diethylstilbestrol during pregnancy have therapeutic value? Am J Obstet Gynecol 66:1062-1081 Giusti RM, lwamoto K, Hatch EE 1995 Diethylstilbestrol revisited: a review of the long-term health effects. Ann Intern Med 122:778-788 Herbst AL, Ulfelder H, Poskanzer DC 1971 Adenocarcinoma of the vagina. Association of maternal stilbestrol therapy with tumor appearance in young women. N Engl J Med 284:878-881 128 126. 127. 128. 129. 130. 131. 132. 133. 134. 135. Herbst AL, Ulfelder H, Poskanzer DC 1971 Adenocarcinoma of the vagina. N Engl J Med 284:878-881 DeChemey AH, Cholst I, Naftolin F 1981 Structure and function of the fallopian tubes following exposure to diethylstilbestrol (DES) during gestation. Fertil Steril 1981 Dec;36(6):741-5 362741 -745 Greco TL, Duello TM, Gorski J 1993 Estrogen receptors, estradiol, and diethylstilbestrol in early development: the mouse as a model for the study of estrogen receptors and estrogen sensitivity in embryonic development of male and female reproductive tracts. Endocrine reviews 14259-71 Noller KL 1977 The ”moming-after pill". Fertil Steril 282211 Gill WB, Schumacher GF, Bibbo M, Straus FHn, Schoenberg HW 1979 Association of diethylstilbestrol exposure in utero with cryptorchidism, testicular hypoplasia and semen abnormalities. J Urol 122236-39 Storgaard L, Bonde JP, Olsen J 2006 Male reproductive disorders in humans and prenatal indicators of estrogen exposure. A review of published epidemiological studies. Reprod Toxicol 2124-15 Newbold RR, Bullock BC, McLachIan JA 1987 Testicular tumors in mice exposed in utero to diethylstilbestrol. J Urol 13821446-1450 Emmen JM, McLuskey A, Adham IM, Engel W, Verhoef Post M, Themmen AP, Grootegoed JA, Brinkmann A0 2000 Involvement of insulin-like factor 3 (lnsl3) in diethylstilbestrol-induced cryptorchidism. Endocrinology 141 2846-849 Baillie-Hamilton PF 2002 Chemical toxins: a hypothesis to explain the global obesity epidemic. J Altem Complement Med 8:185—192 Newbold RR, Padilla-Banks E, Snyder RJ, Jefferson WN 2005 Developmental exposure to estrogenic compounds and obesity. Birth Defects Res A Clin Mol Teratol 732478-480 129 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. Jordan VC 1995 Tamoxifen: toxicities and drug resistance during the treatment and prevention of breast cancer. Annu Rev Pharmacol Toxicol 35 Gill-Sharma MK, Gopalkrishnan K, Balasinor N, Parte P, Jayaraman S, Juneja HS 1993 Effects of tamoxifen on the fertility of male rats. J Reprod Fertil 992395-402 Shelby MD, Newbold RR, Tully DB, Chae K, Davis VL 1996 Assessing environmental chemicals for estrogenicity using a combination of in vitro and in vivo assays. Environ Health Perspect10421296-1300 Coezy E, Borgna JL, Rochefort H 1982 Tamoxifen and metabolites in MCF7 cells: correlation between binding to estrogen receptor and inhibition of cell growth. Cancer Res 422317-323 Hann LE, Giess CS, Bach AM, Tao Y, Baum HJ, Barakat RR 1997 Endometrial thickness in tamoxifen-treated patients: correlation with clinical and pathologic findings. AJR Am J Roentgenol 168:657-661 Sabenlval GS, Gill-Sharma MK, Balasinor N, Choudhary J, Padwal V, Juneja HS 2003 Tamoxifen, protein kinase C and rat sperm mitochondria. Cell Biol Int 272761-768 D'Souza UJ 2004 Effect of tamoxifen on spennatogenesis and tubular morphology in rats. Asian J Androl 62223-226 Parte P, Balasinor N, Glll-Sharma MK, Maitra A, Juneja HS 2002 Temporal effect of tamoxifen on cytochrome P450 side chain cleavage gene expression and steroid concentration in adult male rats. J Steroid Biochem Mol Biol 822349-358 Gill-Shanna MK, Balasinor N, Parte P, Aleem M, Juneja HS 2001 Effects of tamoxifen metabolites on fertility of male rat. Contraception 632103-1 09 Balasinor N, Parte P, Gill-Sharma MK, Juneja HS 2001 Effect of tamoxifen on sperm fertilising ability and preimplantation embryo development. Molecular and Cellular Endocrinology 178:199-206. 130 146. 147. 148. 149. 150. 151. 152. 153. ‘1 54. Hyder SM, Chiappetta C, Stancel GM 1999 Synthetic estrogen 17alpha- ethinyl estradiol induces pattern of uterine gene expression similar to endogenous estrogen 17beta-estradiol. J Phannacol Exp Ther 2902740- 747 Thayer KA, Ruhlen RL, Howdeshell KL, Buchanan DL, Cooke PS, Preziosi D, Welshons WV, Haseman J, Vom Saal FS 2001 Altered prostate growth and daily sperm production in male mice exposed prenatally to subclinical doses of 17alpha-ethinyl oestradiol. Hum Reprod 162988-996. Miyamoto Y, Ueda K, Oshida K, Ohmori E 2000 Collaborative work to evaluate toxicity on male reproductive organs by repeated dose studies in rats 2). Testicular toxicity in rats treated orally with ethinylestradiol for 2 weeks. J Toxicol Sci 25:33-42. Yasuda Y, Kihara T, Tanimura T, Nishimura H 1985 Gonadal dysgenesis induced by prenatal exposure to ethinyl estradiol in mice. Teratology 32:219-227. Andrews P, Freyberger A, Hartrnann E, Eiben R, Loof I, Schmidt U, Temerowski M, Folkerts A, Stahl B, Kayser M 2002 Sensitive detection of the endocrine effects of the estrogen analogue ethinylestradiol using a modified enhanced subacute rat study protocol (OECD Test Guideline no. 407). Arch Toxicol 762194-202 Slikker W, Jr., Bailey JR, Newport D, Lipe GW, Hill DE 1982 Placental transfer and metabolism of 17 alpha-ethynylestradiol-17 beta and estradiol-17 beta in the rhesus monkey. J Pharmacol Exp Ther 2232483- 489 Kallen B, Mastroiacovo P, Lancaster PA, Mutchinick O, Kringelbach M, Martinez-Frias ML, Robert E, Castilla EE 1991 Oral contraceptives in the etiology of isolated hypospadias. Contraception 442173-182 Li DK, Daling JR, Mueller BA, Hickok DE, Fantel AG, Weiss NS 1995 Oral contraceptive use after conception in relation to the risk of congenital urinary tract anomalies. Teratology 51:30-36 DeSesso JM 1997 Comparative embryology. Handbook of developmental toxicology. In. New York:: CRC Press; 111-174 131 155. 156. 157. 158. 159. 160. 161. 162. 163. 164. Longcope C, Gorbach S, Goldin B, Woods M, Dwyer J, Warram J 1985 The metabolism of estradiol; oral compared to intravenous administration. J Steroid Biochem 23:1065—1070 Speck U, Wendt H, Schulze PE, Jentsch D 1976 Bio-availability and pharrnacokinetics of cyproterone acetate-14C and ethinyloestradioI-3H after oral administration as a coated tablet (SH B 209 AB). Contraception 142151-163 Goldzieher JW, Mileikowsky G, Newburger J, Dorantes A, Stavchansky SA 1988 Human pharrnacokinetics of ethynyl estradiol 3- sulfate and 17-sulfate. Steroids 51 :63-79 Pasqualini JR, Castellet R, Portols MC, Hill JL, Kincl FA 1977 Plasma concentrations of ethynyl oestradiol and norethindrone after oral administration to women. J Reprod Fertil 492189-193 Maggs JL, Grimmer SF,,Orme ML, Breckenridge AM, Park BK, Gilmore IT 1983 The biliary and urinary metabolites of [3H]17 alpha- ethynylestradiol in women. Xenobiotica 132421-431 Williams MC, Goldzieher JW 1980 Chromatographic patterns of urinary ethynyl estrogen metabolites in various populations. Steroids 362255-282 Donn J, Mendoza M, Pritchard J 2008 AP Probe Finds Drugs in Drinking Water. In: The Associated Press. New York Aheme GW, Briggs R 1989 The relevance of the presence of certain synthetic steroids in the aquatic environment. Pharrn Pharmacol 41:735- 736. Tabak HH, Bloomhuff RN, Bunch RL 1981 Steroid hormones as water pollutants, ll. Studies on the persistence and stability of natural urinary and synthetic ovulation-inhibiting hormones in untreated and treated wastewaters. Dev Ind Microbiol 222497-519 Williams RJ, Johnson AC, Smith JJ, Kanda R 2003 Steroid estrogens profiles along river stretches arising from sewage treatment works discharges. Environ Sci Technol 3721744-1750 132 165. 166. 167. 168. 169. 170. 171. 172. 173. 174. Roofer P, Snyder S, Zegers RE, Rexing DJ, Fronk JL 2000 Endocrine- disrupting chemicals in a so'urce water. J AWWA 92:52-58 US EPA 2000 Estimated per capita water ingestion in the United States. In. Washington DC: US EPA, Office of Water 1301; 40 US EPA 2001 National primary drinking water regulations; arsenic and clarifications to compliance and new source contaminants monitoring; final rule. In. Washington DC: US EPA Gutendorf B, Westendorf J 2001 Comparison of an array of in vitro assays for the assessment of the estrogenic potential of natural and synthetic estrogens, phytoestrogens and xenoestrogens. Toxicology 1 66:79-89. Von Schoultz B 1988 Potency of different oestrogen preparations. In: Studd JWW, Whitehead Ml eds. The menopause. Oxford: Blackwell Scientific Publications; 130-137 Branham WS, Zehr DR, Sheehan DM 1993 Differential sensitivity of rat uterine growth and epithelium hypertrophy to estrogens and antiestrogens. Proc Soc Exp Biol Med 203:297-303 Van den Belt K, Berckmans P, Vangenechten C, Verheyen R, Witters H 2004 Comparative study on the in vitro/in vivo estrogenic potencies of 17beta-estradiol, estrone, 17alpha-ethynylestradiol and nonylphenol. Aquat Toxicol 662183-195 Yasuda Y, Ohara I, Konishi H, Tanimura T 1988 Long-term effects on male reproductive organs of prenatal exposure to ethinyl estradiol. Am J Obstet Gynecol 1 59:1246-1250 Kaneto M, Kanamori S, Hishikawa A, Kishi K 1999 Epididymal sperm motion as a parameter of male reproductive toxicity: sperm motion, fertility, and histopathology in ethinylestradiol-treated rats. Reprod Toxicol 13(4): 2279-289. Lafuente A, Marquez N, Pousada Y, Pazo D, Esquifino Al 2000 Possible estrogenic and/or antiandrogenic effects of methoxychlor on prolactin release in male rats. Arch Toxicol 742270-275 133 175. 176. 177. 178. 179. 180. 181. 182. 183. Bulger WH, Muccitelli RM, Kupfer D 1978 Interactions of chlorinated hydrocarbon pesticides with the BS estrogen-binding protein in rat testes. Steroids 322165-177 Suzuki M, Lee HC, Chiba S, Yonezawa T, Nishihara M 2004 Effects of methoxychlor exposure during perinatal period on reproductive function after maturation in rats. J Reprod Dev 502455-461 Laws SC, Carey SA, Ferrell JM, Bodman GJ, Cooper RL 2000 Estrogenic activity of octylphenol, nonylphenol, bisphenol A and methoxychlor in rats. Toxicol Sci 5421 54-167 Gaido KW, Leonard LS, Maness SC, Hall JM, McDonnell DP, Saville B, Safe S 1999 Differential interaction of the methoxychlor metabolite 2,2- bis-(p-hydroxyphenyI)-1,1,1-trichloroethane with estrogen receptors alpha and beta. Endocrinology 140:5746-5753 Gaido KW, Maness SC, McDonnell DP, Dehal SS, Kupfer D, Safe S 2000 Interaction of methoxychlor and related compounds with estrogen receptor alpha and beta, and androgen receptor: structure-activity studies. Mol Phannacol 582852-858 Smeets JM, van Holsteijn I, Giesy JP, Seinen W, van den Berg M 1999 Estrogenic potencies of several environmental pollutants, as determined by vitellogenin induction in a carp hepatocyte assay. Toxicol Sci 50:206- 21 3 Cummings AM 1997 Methoxychlor as a model for environmental estrogens. Crit Rev Toxicol 272367-379 Tiemann U 2008 In vivo and in vitro effects of the organochlorine pesticides DDT, TCPM, methoxychlor, and lindane on the female reproductive tract of mammals: a review. Reproductive toxicology (Elmsford, NY 252316-326 Amstislavsky SY, Amstislavskaya TG, Eroschenko VP 1999 Methoxychlor given in the periimplantation period blocks sexual arousal in male mice. Reprod Toxicol 132405-411 134 184. 185. 186. 187. 188. 189. 190. 191. 192. Akingbemi BT, Ge RS, Klinefelter GR, Gunsalus GL, Hardy MP 2000 A metabolite of methoxychlor, 2,2-bis(p-hydroxyphenyI)-1,1, 1- trichloroethane, reduces testosterone biosynthesis in rat leydig cells through suppression of steady-state messenger ribonucleic acid levels of the cholesterol side-chain cleavage enzyme. Biol Reprod 622571-578 Dalton R 2002 Frogs put in the gender blender by America's favourite herbicide. Nature 4162665-666 Steeger T 2003 White paper on potential developmental effects of atrazine on amphibians. In. Washington, DC: Office of Prevention, Pesticides and Toxic Substances, Office of Pesticide Programs, Environmental Fate and Effects Division; 1-95 Gammon DW, Aldous CN, Carr WCJ, Sanbom JR, Pfeifer KF 2005 A risk assessment of atrazine use in California: human health and ecological aspects. Pest Manag Sci 612331-355 Solomon KR, Baker DB, Richards RP, Dixon KR, Klaine WJ, LaPoint TW, Kendall RJ, Weisskopf CP, Giddings JM, Giesy JP, Hall LWJ, Williams WM 1996 Ecological risk assessment of atrazine in North American surface waters. Environ Toxicol Chem 15:31-76 Du Preez LH, Jansenvan Rensburg PJ, Jooste AM, Carr JA, Giesy JP, Gross TS, Kendall RJ, Smith EE, Van Der Kraak G, Solomon KR 2005 Seasonal exposures to triazine and other pesticides in surface waters in the western Highveld com-production region in South Africa. Environ Pollut 135:131-141 Hauswirth JW, Wetzel LT 1998 Toxicity characteristics of the 2- chlorotriazines atrazine and simazine. In: Ballantine LG, McFarland JE, Hackett DS eds. Triazine herbicides: Risk assessmen. Washington, DC: Oxford University Press; 370-383 Laws SC, Ferrell JM, Stoker TE, Schmid J, Cooper RL 2000 The effects of atrazine on female wistar rats: an evaluation of the protocol for assessing pubertal development and thyroid function. Toxicol Sci ;() 582366-376 Friedmann AS 2002 Atrazine inhibition of testosterone production in rat males following peripubertal exposure. Reprod Toxicol 162275-279 135 193. 194. 195. 196. 197. 198. 199. 200. 201. 202. Trentacoste SV, Friedmann AS, Youker RT, Breckenridge CB, Zirkin BR 2001 Atrazine effects on testosterone levels and androgen-dependent reproductive organs in peripubertal male rats. J Androl 222142-148 Stoker TE, Laws SC, Guidici DL, Cooper RL 2000 The effect of atrazine on puberty in male wistar rats: an evaluation in the protocol for the assessment of pubertal development and thyroid function. Toxicol Sci 58:50-59 Cooper RL, Stoker TE, Tyrey L, Goldman JM, McElroy WK 2000 Atrazine disrupts the hypothalamic control of pituitary-ovarian function. Toxicol Sci 532297-307 Eldridge JC, Wetzel LT, Stevens JT, Simpkins JW 1999 The mammary tumor response in triazine-treated female rats: a threshold-mediated interaction with strain and species-specific reproductive senescence. Steroids 642672-678 Ashby J, Tinwell H, Stevens J, Pastoor T, Breckenridge CB 2002 The effects of atrazine on the sexual maturation of female rats. Regul Toxicol Pharmacol 352468-473 Dianin AP 1914 Molecular structure in relation to oestrogenic activity. Compounds without a phenanthrene nucleus. Russ Phys Chem Soc 3621310 Burridge E 2003 Bisphenol A: product profile. Eur Chem News 14-20217 Kang JH, Kito K, Kondo F 2003 Factors influencing the migration of bisphenol A from cans. J Food Prot 6621444-1447 Olea N, Pulgar R, Perez P, Olea-Serrano F, Rivas A, NovilIo-Fertrell A, Pedraza V, Soto AM, Sonnenschein C 1996 Estrogenicity of resin-based composites and sealants used in dentistry. Environ Health Perspect 1042298-305 Kang JH, Kondo F 2002 Determination of bisphenol A in canned pet foods. Res Vet Sci 732177-182 136 203. 204. 205. 206. 207. 208. 209. 210. 211. 212. Fromme H, Kuchler T, Otto T, Pilz K, Muller J, Wenzel A 2002 Occurrence of phthalates and bisphenol A and F in the environment. Water Res 3621429-1438 Volkel W, Colnot T, Csanady GA, Filser JG, Dekant W 2002 Metabolism and kinetics of bisphenol a in humans at low doses following oral administration. Chem Res Toxicol 15:1281-1287 Calafat AM, Kuklenyik Z, Reidy JA, S.P. C, Ekong J, Needham LL 2005 Urinary concentrations of bisphenol A and 4-nonylphenol in a human reference population. Environ Health Perspect 113:391-395 Ikezuki Y, Tsutsumi O, Takai Y, Kamei Y, Taketani Y 2002 Determination of bisphenol A concentrations in human biological fluids reveals significant early prenatal exposure. Hum Reprod 17:2839—2841 vom Saal FS, Hughes C 2005 An extensive new literature concerning low-dose effects of bisphenol A shows the need for a new risk assessment. Environ Health Perspect1132926-933 ECSCF 2002 Opinion of the Scientific Committee on Food on Bisphenol A. SCF/CS/PM/3938 Final. In: BrusselszEuropean Commission Scientific Committee on Food Degen GH, Bolt HM 2000 Endocrine disruptors: update on xenoestrogens. Int Arch Occup Environ Health 732433-441 Bolt HM, Janning P, Michna H, Degen GH 2001 Comparative assessment of endocrine modulators with oestrogenic activity: I. Definition of a hygiene-based margin of safety (HBMOS) for xeno-oestrogens against the background of European developments. Arch Toxicol 74:649- 662 Kang JH, Kondo F, Katayama Y 2006 Human exposure to bisphenol A. Toxicology Epub ahead of print Morrissey RE, Lamb JCt, Morris RW, Chapin RE, Gulati DK, Heindel JJ 1989 Results and evaluations of 48 continuous breeding reproduction studies conducted in mice. Fundam Appl Toxicol 132747-777 137 213. 214. 215. 216. 217. 218. 219. 220. 221. Hardin 80, Bond GP, Sikov MR, Andrew FD, Beliles RP, Niemeier RW 1981 Testing of selected workplace chemicals for teratogenic potential. Scand J Work Environ Health 7:66-75 Morrissey RE, George JD, Price CJ, Tyl RW, Marr MC, Kimmel CA 1987 The developmental toxicity of bisphenol A in rats and mice. Fundam Appl Toxicol 82571-582 Dodds EC, Lawson W 1936 Synthetic estrogenic agents without the phenanthrene nucleus. Nature 1372996 Dodds EC, Lawson W 1938 Molecular stmcture in relation to oestrogenic activity. Compounds without a phenanthrene nucleus. Proceedings of the Royal Society London B 1252222-232 Krishnan AV, Stathis P, Pennuth SF, Tokes L, Feldman D 1993 Bisphenol-A2 an estrogenic substance is released from polycarbonate flasks during autoclaving. Endocrinology 132:2279-2286 Gaido KW, Leonard LS, Lovell S, Gould JC, Babai D, Portier CJ, McDonnell DP 1997 Evaluation of chemicals with endocrine modulating activity in a yeast-based steroid hormone receptor gene transcription assay. Toxicol Appl Pharmacol 1432205—212 Mohri T, Yoshida S 2005 Estrogen and bisphenol A disrupt spontaneous [Ca(2+)](i) oscillations in mouse oocytes. Biochem Biophys Res Commun 326:166-173 vom Saal FS, Cooke PS, Buchanan DL, Palanza P, Thayer KA, Nagel SC, Parrnigiani S, Welshons WV 1998 A physiologically based approach to the study of bisphenol A and other estrogenic chemicals on the size of reproductive organs, daily sperm production, and behavior. Toxicol Ind Health 142239-260 Markey CM, Luque EH, Munoz De Toro M, Sonnenschein C, Soto AM 2001 In utero exposure to bisphenol A alters the development and tissue organization of the mouse mammary gland. Biol Reprod 65:1215-1223 138 222. 223. 224. 225. 226. 227. 228. 229. 230. 231. Ashby J, Tinwell H, Haseman J 1999 Lack of effects for low dose levels of bisphenol A and diethylstilbestrol on the prostate gland of CF1 mice exposed in utero. Regul Toxicol Phannacol 3021 56-166 Cagen SZ, Waechter JMJ, Dimond SS, Breslin WJ, Butala JH, Jekat FW, Joiner RL, Shiotsuka RN, Veenstra GE, Harris LR 1999 Normal reproductive organ development in CF-1 mice following prenatal exposure to bisphenol A. Toxicol Sci 50236-44 Masuno H, Kidani T, Sekiya K, Sakayama K, Shiosaka T, Yamamoto H, Honda K 2002 Bisphenol A in combination with insulin can accelerate the conversion of 3T3-L1 fibroblasts to adipocytes. J Lipid Res 432676-684 Alonso-Magdalena P, Morimoto S, Ripoll C, Fuentes E, Nadal A 2006 The estrogenic effect of bisphenol A disrupts pancreatic beta-cell function in vivo and induces insulin resistance. Environ Health Perspect 1142106- 1 12 Sakurai K, Kawazuma M, Adachi T, Harigaya T, Saito Y, Hashimoto N, Mori C 2004 Bisphenol A affects glucose transport in mouse 3T3-F442A adipocytes. Br J Pharmacol 141:209-214 Thomas JA, Thomas MJ 1984 Biological effects of di-(2-ethylhexyl) phthalate and other phthalic acid esters. Crit Rev Toxicol 132283-317 Stroheker T, Cabaton N, Nourdin G, Regnier JF, Lhuguenot JC, Chagnon MC 2005 Evaluation of anti-androgenic activity of dl-(2- ethylhexyl)phthalate. Toxicology 20821 15-121 Mahood IK, Hallmark N, McKinnell C, Walker M, Fisher JS, Sharpe RM 2005 Abnormal Leydig cell aggregation in the fetal testis of rats exposed to di (n-butyl) phthalate and its possible role in testicular dysgenesis. Endocrinology 146261 3-623 Fennell TR, Krol WL, Sumner SC, Snyder RW 2004 Phannacokinetics of dibutylphthalate in pregnant rats. Toxicol Sci 822407-418 Hotchkiss AK, Parks-Saldutti LG, Ostby JS, Lambright C, Furr J, Vandenbergh JG, Gray LEJ 2004 A mixture of the "antiandrogens" 139 232. 233. 234. 235. 236. 237. 238. 239. 240. Iinuron and butyl benzyl phthalate alters sexual differentiation of the male rat in a cumulative fashion. Biol Reprod 71:1852-1861 Salazar V, Castillo C, Ariznavarreta C, Campon R, Tresguerres JA 2004 Effect of oral intake of dibutyl phthalate on reproductive parameters of Long Evans rats and pre-pubertal development of their offspring. Toxicology 2052131-137 Masutomi N, Shibutani M, Takagi H, Uneyama C, Takahashi N, Hirose M 2003 Impact of dietary exposure to methoxychlor, genistein, or diisononyl phthalate during the perinatal period on the development of the rat endocrine/reproductive systems in later life. Toxicology 1922149—170 Sohoni P, Sumpter JP 1998 Several environmental oestrogens are also anti-androgens. J Endocrinol 1582327-339 Jobling S, Reynolds T, White R, Parker MG, Sumpter JP 1995 A variety of environmentally persistent chemicals, including some phthalate plasticizers, are weakly estrogenic. Environ Health Perspect 1032582-587 Silva MJ, Barr DB, Reidy JA, Malek NA, Hodge CC, Caudill SP, Brock JW, Needham LL, Calafat AM 2004 Urinary levels of seven phthalate metabolites in the US. population from the National Health and Nutrition Examination Survey (NHANES) 1999-2000. Environ Health Perspect 1 122331 -338 Hoppin JA, Brock JW, Davis BJ, Baird DD 2002 Reproducibility of urinary phthalate metabolites in first morning urine samples. Environ Health Perspect 110:515-518 Schettler T 2006 Human exposure to phthalates via consumer products. Int J Androl 292134-139 Meek M, Chan PJ 1994 Bis(2-ethylhexyl)phthalate: evaluation of risks to health from environmental exposure in Canada. J Environ Sci Health Part C2179-194 Sharpe RM, Fisher JS, Millar MM, Jobling S, Sumpter JP 1995 Gestational and lactational exposure of rats to xenoestrogens results in 140 241 . 242. 243. 244. 245. 246. 247. 248. 249. reduced testicular size and sperm production. Environ Health Perspect 10321136-1143 Ema M, Miyawaki E, Kawashima K 2000 Critical period for adverse effects on development of reproductive system in male offspring of rats given di-n-butyl phthalate during late pregnancy. Toxicol Lett 1112271-278 Moore RW, Rudy TA, Lin TM, Ko K, Peterson RE 2001 Abnormalities of sexual development in male rats with in utero and lactational exposure to the antiandrogenic plasticizer Di(2-ethylhexyl) phthalate. Environ Health Perspect 1092229-237 Thompson CJ, Ross SM, Gaido KW 2004 Di(n-butyl) phthalate impairs cholesterol transport and steroidogenesis in the fetal rat testis through a rapid and reversible mechanism. Endocrinology 14521227—1237 Duty SM, Silva MJ, Barr DB, Brock JW, Ryan L, Chen Z, Herrick RF, Christiani DC, Hauser R 2003 Phthalate exposure and human semen parameters. Epidemiology 142269-277 Marsee K, Woodruff TJ, Axelrad DA, Calafat AM, Swan SH 2006 Estimated daily phthalate exposures in a population of mothers of male infants exhibiting reduced anogenital distance. Environ Health Perspect 114:805-809 Fisher JS 2004 Environmental anti-androgens and male reproductive health: focus on phthalates and testicular dysgenesis syndrome. Reproduction 127:305-315 Gray LEJ, Laskey J, Ostby J 2006 Chronic di-n-butyl phthalate exposure in rats reduces fertility and alters ovarian function during pregnancy in female Long Evans hooded rats. Toxicol Sci 932189-195 Sohoni P, Sumpter JP 1998 Several environmental oestrogens are also anti-androgens. J Endocrinol 1582327-339 Zacharewski TR, Meek MD, Clemons JH, Wu ZF, Fielden MR, Matthews JB 1998 Examination of the in vitro and in vivo estrogenic activities of eight commercial phthalate esters. Toxicol Sci 462282-293 141 250. 251. 252. 253. 254. 255. 256. 257. 258. 259. Thompson CJ, Ross SM, Hensley J, Liu K, Heinze SC, Young SS, Gaido KW 2005 Differential steroidogenic gene expression in the fetal adrenal gland versus the testis and rapid and dynamic response of the fetal testis to di(n-butyl) phthalate. Biol Reprod 732908-917 Battershill JM 1994 Review of the safety assessment of polychlorinated biphenyls (PCBs) with particular reference to reproductive toxicity. Hum Exp Toxicol 132581 -597 Ma R, Sassoon DA 2006 PCBs exert an estrogenic effect through repression of the Wnt7a signaling pathway in the female reproductive tract. Environ Health Perspect 114:898-904 Davidson NE 1998 Environmental estrogens and breast cancer risk. Curr Opin Oncol 102475-478 Petrik J, Drobna B, Pavuk M, Jursa S, Wimmerova S, Chovancova J 2006 Serum PCBs and organochlorine pesticides in Slovakia: age, gender, and residence as determinants of organochlorine concentrations. Chemosphere 652410-418 Kimbrough RD, Krouskas CA 2003 Human exposure to polychlorinated biphenyls and health effects: a critical synopsis. Toxicol Rev 222217-233 Ogura 12004 Half-life of each dioxin and PCB congener in the human body. Organohalogen compounds 66:3376-3384 Colbom T, Smolen MJ 1996 Epidemiological analysis of persistent organochlorine contaminants in cetaceans. Rev Environ Contam Toxicol 146291 -1 72 Kholkute SD, Rodriguez J, Dukelow WR 1994 Reproductive toxicity of Aroclor-1254: effects on oocyte, spermatozoa, in vitro fertilization, and embryo development in the mouse. Reprod Toxicol 82487-493 Hany J, Lilienthal H, Sarasin A, Roth-Harer A, Fastabend A, Dunemann L, Lichtensteiger W, Winneke G 1999 Developmental exposure of rats to a reconstituted PCB mixture or aroclor1254: effects on organ weights, aromatase activity, sex hormone levels, and sweet preference behavior. Toxicol Appl Pharmacol 1 582231 -243 142 260. 261. 262. 263. 264. 265. 266. 267. 268. 269. DeCastro BR, Korrick SA, Spengler JD, Soto AM 2006 Estrogenic activity of polychlorinated biphenyls present in human tissue and the environment. Environ Sci Technol 4022819-2825 Winneke G, Walkowiak J, Lilienthal H 2002 PCB-induced neurodevelopmental toxicity in human infants and its potential mediation by endocrine dysfunction. Toxicology 181—182:161-165 Jacobson JL, Jacobson SW 1996 Intellectual impairment in children exposed to polychlorinated biphenyls in utero. N Engl J Med 335:783—789 Newman J, Aucompaugh AG, Schell LM, Denham M, DeCaprio AP, Gallo MV, Ravenscroft J, Kao CC, Hanover MR, David D, Jacobs AM, Tarbell AM, Worswick P, Environment. ATFot 2006 PCBs and cognitive functioning of Mohawk adolescents. Neurotoxicol Teratol 282439-445 Hattemer-Frey HA, Travis CC 1991 Benzo-a-pyrene: environmental partitioning and human exposure. Toxicol Ind Health 72141-157 Hall M, Forrester LM, Parker DK, Grover PL, Wolf CR 1989 Relative contribution of various forms of cytochrome P450 to the metabolism of benzo[a]pyrene by human liver microsomes. Carcinogenesis 1021815- 1 821 Shimada T, Gillam EM, Oda Y, Tsumura F, Sutter TR, Guengerich FP, Inoue K 1999 Metabolism of benzo[a]pyrene to trans-7,8-dihydroxy-7, 8- dihydrobenzo[a]pyrene by recombinant human cytochrome P450 1B1 and purified liver epoxide hydrolase. Chem Res Toxicol 122623-629 Shou M, Korzekwa KR, Crespi CL, Gonzalez FJ, Gelboin HV 1994 The role of 12 cDNA-expressed human, rodent, and rabbit cytochromes P450 in the metabolism of benzo[a]pyrene and benzo[a]pyrene trans-7,8- dihydrodiol. Mol Carcinog 102159-168 Kim JH, Stansbury KH, Walker NJ, Trush MA, Strickland PT, Sutter TR 1998 Metabolism of benzo[a]pyrene and benzo[a]pyrene-7,8-diol by human cytochrome P450 1B1. Carcinogenesis 1921847-1853 Keshava C, Divi RL, Whipkey DL, Frye BL, McCanlies E, Kuo M, Poirier MC, Weston A 2005 Induction of CYP1A1 and CYP1 B1 and 143 270. 271. 272. 273. 274. 275. 276. 277. 278. 279. formation of carcinogen-DNA adducts in normal human mammary epithelial cells treated with benzo[a]pyrene. Cancer Lett 221 :21 3-224 Estabrook RW, Saeki Y, Chacos N, Capdevila J, Prough RA 1980 Polycyclic hydrocarbon metabolism: a plethora of phenomena. Adv Enzyme Regul 1923-17 Gelboin HV 1980 Benzo[alphalpyrene metabolism, activation and carcinogenesis: role and regulation of mixed-function oxidases and related enzymes. Physiol Rev 6021 107-1166 Pelkonen O, Nebert DW 1982 Metabolism of polycyclic aromatic hydrocarbons: etiologic role in carcinogenesis. Phannacol Rev 342189-222 Huggins CB, Pataki J, Harvey RG 1967 Geometry of carcinogenic polycyclic aromatic hydrocarbons. Proc Natl Acad Sci U S A 58:2253-2260 Yang NC, Castro AJ, Lewis M, Wong TW 1961 Polynuclear aromatic hydrocarbons, steroids and carcinogenesis. Science 1342386-387 Grover PL, MacNicoll AD, Sims P, Easty GC, Neville AM 1980 Polycyclic hydrocarbon activation and metabolism in epithelial cell aggregates prepared from human mammary tissue. Int J Cancer 262467- 475 Mattison DR, Singh H, Takizawa K, Thomford PJ 1989 Ovarian toxicity of benzo(a)pyrene and metabolites in mice. Reprod Toxicol 32115-125 Swartz WJ, Mattison DR 1985 Benzo(a)pyrene inhibits ovulation in 057BL/6N mice. Anat Rec 212:268-276 Tran DQ, lde CF, McLachIan JA, Arnold SF 1996 The anti-estrogenic activity of selected polynuclear aromatic hydrocarbons in yeast expressing human estrogen receptor. Biochem Biophys Res Commun 229:101-108 Arcaro KF, O'Keefe PW, Yang Y, Clayton W, Gierthy JF 1999 Antiestrogenicity of environmental polycyclic aromatic hydrocarbons in human breast cancer cells. Toxicology 13321 15-127 144 280. 281. 282. 283. 284. Cook JH, Dodds EC 1933 Sex hormones and cancer-producing compounds. Nature 131:205-206 Fertuck KC, Matthews JB, Zacharewski TR 2001 Hydroxylated benzo[a]pyrene metabolites are responsible for in vitro estrogen receptor- mediated gene expression induced by benzo[a]pyrene, but do not elicit uterotrophic effects in vivo. Toxicol Sci 592231-240 Bigelow SW, Nebert DW 1982 The Ah regulatory gene product. Survey of nineteen polycyclic aromatic compounds' and fifteen benzo[a]pyrene metabolites' capacity to bind to the cytosolic receptor. Toxicol Lett 102109- 1 18 Okey AB, Dube AW, Vella LM 1984 Binding of benzo(a)pyrene and dibenz(a,h)anthracene to the Ah receptor in mouse and rat hepatic cytosols. Cancer Res 4421426-1432 Duan R, Porter W, Samudio I, Vyhlidal C, Kladde M, Safe S 1999 Transcriptional activation of c-fos protooncogene by 17beta-estradiol: mechanism of aryl hydrocarbon receptor-mediated inhibition. Mol Endocrinol 1321511-1521 145 CHAPTER4 Effects of In Utero and Lactational Exposure of Female Mice to Diethylstilbestrol, 17a-Ethynyl Estradiol, and Genistein on Ovulation and In Vitro Fertilizing Ability of Eggs. 4.1 ABSTRACT In this study, the effects of in utero and lactational exposure of mice to diethylstilbestrol (DES), 17a-ethynyl estradiol (EE2), and genistein (GEN) on ovulation and in vitro fertilizing ability of eggs collected from female offspring (F- 1) were examined. Pregnant C57BL/6 mice (F-0), bred with DBA/2 males, were gavaged with DES (0, 0.1, 1.0, or 10 pg/kg of body weight per day (bw/d)), EE2 (0, 0.1, 1.0, or 10 pg/kg bw/d), or GEN (0, 0.1, 0.5, 2.5, or 10 mglkg bw/d) from gestational day (GD) 12 to postnatal day (PND) 20. The female offspring were examined at two different stages of reproductive development: PND 21 to 42 (peripuberty) and PND 56 to 105 (postpuberty). There was no significant change in the number of eggs ovulated by peripubertal female offspring exposed to DES, EE2, or GEN. However, the number of eggs ovulated by female offspring on PND 56 to 105 significantly increased (46%) in the 0.1 pg DES/kg bw/d treatment group, while the number of eggs ovulated decreased (31 %) in the 10 pg EE2/kg bw/d group, compared to the control group. Fertilizing ability of eggs at peripuberty was significantly decreased in the 10 pg EE2/kg bw/d treatment group (P < 0.01). Maternal exposure to GEN did not affect ovulation rate nor the fertilizing ability of eggs ovulated by F-1 mice. Our results suggest that in utero and lactational exposure to DES and EE2 altered ovulation and fertilization, whereas GEN had no observable effects on the number of ovulated 146 eggs and fertilizing ability of eggs from female offspring. 4.2 INTRODUCTION Estrogen plays a key role in regulating ovarian development and function during growth of the fetus. The primary function of the ovaries is to produce eggs, which originate from a permanent cell population established before birth. The ovaries of the fetus are constantly exposed to hormonal stimuli during the critical periods of fetal and perinatal development to acquire competence for ovulation and for fertilizing ability of eggs. Therefore, in utero and perinatal exposure to exogenous chemicals, which mimic endogenous hormones, may disrupt fertility in adulthood. Fetal and perinatal exposure to exogenous chemicals known as estrogenic endocrine modulators (EEMs) has been .linked to a variety of adverse effects on reproductive health in both humans and wildlife (1-4). Estrogenic endocrine modulators are naturally occurring or synthetic chemicals that may cause transient alteration of endocrine function within the physiological or homeostatic range. In this study, we examined the effects of three: EEMs: diethylstilbestrol (DES), 17a-ethynyl estradiol (EE2), and genistein (4’,5,7- trihydrozylsoflavone or GEN) on female mice after in utero and lactational exposure. Diethylstilbestrol is a synthetic estrogen, which was administered to pregnant women to prevent miscarriages in the late 19403 and 19503. Reduced fertility and greater reproductive tract abnormalities and genital cancer have been 147 documented as consequences of fetal exposure to DES In humans (5-7). Total doses of DES prescribed to women ranged from 1.4 to 17.9 9 during pregnancy (8, 9). DES was banned in 1971 because of carcinogenic effects in offspring attributed to in utero exposure. Because of the adverse reproductive effects, many epidemiological and toxicological studies have been conducted assessing DES to examine and compare toxicities in various tissues (10-14). Interest in the effects of the synthetic estrogen EE2 on the reproductive health of humans and animals is increasing. The chemical is used as the major component of oral contraceptives (15, 16). Each year, there are approximately 2 million women in the United States (US) and the United Kingdom (UK) exposed to 0.4 to 2.0 pg EE2/kg bw/d during undetected early pregnancy (15), thus resulting in exposure of the fetus to this synthetic estrogen. In animal studies, matemally administrated EE2 can cross the placenta and be transferred to the fetus resulting in decreased fetal body weight, increased number of fetal deaths, hyperplasia of follicular cells and degeneration of primordial follicles in a dose- dependent manner (17-20). Furthermore, there have been human and ecological health concerns about pharmaceuticals and their metabolites present in drinking water supplies because of the inability of sewage treatment facilities to remove them from the waste stream. In March, 2008, an Associated Press investigation reported that pharmaceuticals, including estrogenic compounds such as EE2, have been detected in the US drinking water supplies of 24 major metropolitan areas supplying 41 million Americans (21). Approximately 17 to 29% of sulfate or 148 glucuronide metabolites of ingested EE2 being used as an oral contraceptive are excreted in the urine (22, 23). The concentrations of EE2 in sewage treatment plant effluents were in the range of 0.4 to 15 ng/l and 300 to 1800 ng/l in the UK and US, respectively (24-26). Concentrations of EE2 were measured at 0.48 pg/l in Las Vegas Wash and at 0.52 pg/l in Las Vegas Bay (27). Based on the US Environmental Protection Agency’s (EPA) drinking water exposure estimates, women of childbearing age (15 to 44 years of age), could be exposed to 6.3x10'6 to 2.5x10'2 pg EE2/kg bw/d through water consumption, and children younger than one year of age could be exposed to 2.1x10'5 to 8.3x10’2 pg EE2/kg bwld (28, 29). Therefore, potential adverse effects of exposure of the fetus and neonate to EE2 through consumption of drinking water need to be investigated. There are also concern and controversy regarding the potential adverse effects of fetal and neonatal exposure to GEN. Genistein is a phytoestrogen isoflavonoid found in the seeds of leguminous plants. Some soy-based infant formulas contain as much as 40 mg/I of soy lsoflavones of which approximately 65% is GEN and its glycosidic conjugates (30). The estimated consumption of total isoflavones by infants fed a soy-based formula is 4.5 to 8.0 mglkg bwld. Mouse studies have shown that in utero exposure to GEN decreased body weight at birth (31) and increased the incidence of uterine adenocarcinomas at adulthood in mice (32). Furthermore, neonatal exposure to GEN increased multioocyte follicles (33), and prolonged the estrous cycle in mice (34) in a dose- dependent fashion. Female mice exposed to GEN as neonates have reduced fertility and litter sizes as adults. Based on animal studies, further study is 149 needed to examine potential adverse effects of GEN exposure at human dietary concentrations. Ovarian response to EEMs may differ at different stages of biological maturation (e.g. prepubertal, pubertal, and postpubertal) (35). In the present study, we tested the hypothesis that in utero and lactational exposure to DES, EE2 and GEN causes long-term alterations in ovarian function and affects ovulation and fertility of female offspring (F-1). We examined the hormonal response of ovaries to superovulation at two different ages in female offspring (PND 21 to 42 [peripuberty; PEP] and PND 56 to 105 [postpuberty; POP). Egg fertilizing ability was assessed using an in vitro fertilization (IVF) assay to avoid confounding treatment effects on sexual behavior. Doses of DES, EE2 and GEN were similar to clinical and recommended dietary doses used by humans. 4.3 MATERIALS AND METHODS Animals EIeven-week-old virgin C57BL/6 female (F-O) and seven- to eight-week- old DBA/2 male (F-0) were obtained from Charles River Laboratories (Portage, MI, USA) for each of three trials assessing the effects of DES, EE2, and GEN independent of one another (each test compound has its own control group for comparison). Previous studies from our laboratory have shown that B602 F-1, offspring from the C57BL/6 x DBA/2 cross mating, superovulate a large number of eggs and have a consistently high rate of fertilization (36-38). All mice were housed in polycarbonate cages with cellulose fiber chips (Aspen Chip Laboratory 150 Bedding, Northeastern Products, Warrensberg, NY, USA) as bedding. The animal room was maintained at 23 °C with 30-40% humidity. The lights were on a 12-hour cycle. All animals had unlimited access to deionized water provided in glass bottles with rubber stoppers and AlN-76A rodent feed (Research Diets, New Brunswick, NJ, USA). AlN-76A rodent feed is a casein-based, open-forrnula purified diet that contains non-detectable concentrations of the estrogenic isoflavones genistein, diadzein and glycitein (39). Offspring were provided with standard 2019 rodent feed (Harlen Teklad 22/5, Madison, WI, USA).. Maternal treatment When animals arrived, CS7BL/6 female and DBA/2 male mice (F-O) were separated by sex, housed two per cage and acclimatized for three to seven days. At the beginning of the study, two females were housed with a male for mating. Following evidence of pregnancy, dams were separated from the males, housed individually, and randomly assigned to one of the three estrogenic endocrine modulators treatment groups. Ten to twelve pregnant females per each treatment were used. Diethylstilbestrol, 17a-ethynylestradiol, and corn oil were obtained from Sigma Chemical Co. (St. Louis, MO, USA). Genistein (>98% pure) was obtained from lndofine Chemical Company (Somerville, NJ, USA). Pregnant C57BL/6 F-0 mice were treated by daily gavage with diethylstilbestrol (0, 0.1, 1.0, or 10 pg/kg bw), 17a-ethynylestradiol (0, 0.1, 1.0, 10 pg/kg bw), or genistein (0, 151 0.1, 0.5, 2.5, or 10 mglkg bw) in 0.1 ml corn oil from GD 12 to postnatal day (PND) 20 (Figure 4-1). Dams were not treated on the day of parturition (PND O) to avoid cannibalism of pups by the dams. Body weights of dams were measured daily between 09:00 AM and 12:00 AM to administer treatment. Offspring were separated by sex on PND 21 (weaning) and fed AlN-76A until necropsy. All F-O mice were euthanized on PND 21. Paiing female (c5731.) and male pert) Birth I302) We in Mating Gestation Lactation F.0 llllllllllllllllll llllllllllllllllll Offspring Maternal treatment period GesMionaldaylZ PostnataldayZO . Dams were treated by gavage with DES (0, 0.1, 1.0, 10 pglrg maternal bwlday, EE2 (D, 0.1, 1.0, 10119119 maternal bwlday), GEN (0, 0.1, 0.5, 2.5, 10 mglkg mat emal bwlday) from gestational day 12 to postnatal day We try 20 Fflllllllllllll F1 Offspring - _ Age Dayo Day? Day 14 Day21 Day211039 Day56t0105 Litter size Anogenital distance Body weight Body weight (AGD) Organ weight Organ weight Body weights Ovulalion Ovulation Egg fertilizing ability Egg fertilizing abil'ly Figure 4-1. Schedule of maternal treatments and parameters measured in their female offspring (F-1). 152 Offspring (F- 1) development and necropsy B602 F-1 body weight and anogenital distance (AGD; the length of the perineum from the base of the genital tubercle to the center of the anus when the skin was naturally extended without stretching) were measured on PND 7, PND 14, and PND 21. A dissecting microscope (Nikon, Melville, NY, USA) with an ocular micrometer was used for measurement of AGD. The anogenital area was extended slightly without stretching and measured to the nearest 0.1 mm. At the times of the in vitro fertilization assay at PEP or POP, female F-1 body weights were recorded. Superovulatr'on To examine the responsiveness of female offspring exposed to DES, EE2 or GEN in utero and during lactation to ovulation induction stimuli, female offspring at PEP or POP were injected intraperitoneally (i.p.) with 10 international units (IU) of pregnant mare’s serum gonadotropin (Sigma Chemical Co., St. Louis, MO, USA) in 0.1 ml saline followed 48 h later with 10 IU human Chorionic gonadotropin (Sigma Chemical Co., St. Louis, MO, USA) in 0.1 ml saline. The superovulated eggs from each female were collected from the ampulla 14 to 16 h later and incubated in a 1-ml organ culture dish (60 mm diameter x 15 mm high) (Becton Dickinson Falcon, Franklin Lakes, NJ, USA) containing 0.9 ml Brinster’s BMOC-3 medium supplemented with penicillin and streptomycin (Gibco/BRL, Grand Island, NY, USA). 153 In vitro fertilization assay Sperm used for the in vitro fertilization assay were collected by puncturing the caudal epididymides of reproductively mature non-treated male mice with a 25-gauge needle and placed in a 1-ml organ culture dish containing 1.0 ml BMOC—3. The sperm suspension was incubated at 37 °C under a humidified 5% 002 air environment for 60 min before insemination. Sperm was diluted in BMOC-3 medium with the concentration being measured using a CellSoft computer (CASA; CRYO Resources Inc, New York, NY, USA). Diluted sperm (0.1 ml) was added to the 0.9 ml medium containing the eggs in an organ culture dish to achieve a final sperm concentration of 3 x 104 sperm per 1-ml dish. After insemination, eggs were incubated for 24 h, and 50 pl of 35 pM bisBenzimide (Sigma Chemical Co., St. Luis, MO, USA) was added to the dish. After 20 min incubation with the stain, the eggs were examined using a Nikon Optophot fluorescent microscope (Melville, NY, USA) equipped with a 100-watt mercury bulb, a 365/10-nm excitation filter, a 400-nm dichromic mirror, and 400-nm barrier filter. Eggs were counted and scored as fertilized if they were at the two- cell stage or at the one-cell stage containing spindle, two pronuclei, and a second polar body. Additionally, eggs were evaluated for fragmentation and other signs of degeneration. Statistical analysis SAS version 8.2 (SAS Inc, Cary, NC, USA) was used for all data analyses. All data were tested for normality by using the Shapiro-Wilk test and residual plot. 154 Litter was considered to be the experimental unit. Comparisons of offspring body weight, AGD, number of eggs ovulated, percent of egg fertilizing ability and percent of egg degeneration between control and treatment groups were analyzed with ANOVA using the MIXED procedure of SAS, where treatment was a fixed effect and dam was a random effect within treatment (a nested design). Dunnett’s test was used for comparisons between control and treatment groups for each compound. No within dose comparisons were made, nor were EEMs treatments compared to one another. When parameters were analyzed, litter size on PND 0, body weight, and age at necropsy were included as a covariate, if significant correlations were confirmed (40). In vitro fertilization data were analyzed in two age groups (PEP and POP) and were adjusted by age of animal. Eggs were not included in data analysis, if it was not possible to determine the stage of development. Statements of significance are based on a P value less than 0.05. 4.4 RESULTS Body weights of F-1 female mice Body weights on PND 7 and 21 were analyzed with and without adjustment for litter size on PND 0, because, as expected, body weight was negatively correlated with litter size (P < 0.05) (Table 4-1). With litter size adjustment, body weight in the 10 pg DES treatment group was significantly decreased on PND 21. Conversely, the 1.0 pg DES treatment increased body weight at POP (P < 0.05). Maternal EE2 treatment significantly decreased F-1 female body weight in the 1.0 and 10 pg treatment groups at PND 7 and this 155 effect was persistent only in the 10 pg treatment groups until PND 21. After litter size adjustment, there was no significant difference in body weights observed, compared to the control group. Unlike the DES and EE2 treatments, there were no significant body weight changes in the GEN treatment groups with or without litter size adjustment. Anogenital distance DES, EE2, or GEN treatment did not affect AGD regardless of AGD adjustment by body weight (Table 4-2). Ovulation The number of ovulated eggs and in vitro fertilizing ability was determined at two different ages (PEP and POP). There was no significant change in the number of ovulated eggs from DES-, GEN-, and EE2-exposed female offspring at PEP (Table 4-3). The number of ovulated eggs was greater than controls in the 0.1 pg DESI kg bwld treatment group (P < 0.05). There was no significant ' change in the number of ovulated eggs in the EE2 or GEN maternal treatment groups. The number of ovulated eggs was less than controls in the 10 pg EE2/kg bwld treatment group at POP but the P value was 0.09. Fertilizing ability of eggs 156 The percent of fertilized eggs in the DES treatment groups was not different from controls at PEP and POP (Table 4-4). Fertilizing ability of eggs was less than controls in the 10 pg EE2 female offspring PEP (P < 0.05). GEN treatment did not affect fertilizing ability of eggs. The relative percentage of fertilizing ability of eggs is presented in Table 4-4a (when the control = 100%). Percentage of degenerated eggs DES, EE2, or GEN did not affect degeneration of eggs regardless of age (Table 4-5). 157 35.9.05: 5o 5E: «an an E25253 5555 55.5 n 55.5 5555 55.5 n 55.5 5555 5.55 H 55.5 5555 55.5 n 55.5 5555 55.5 n 55.5 a 525 5555 55.5 n 55.5 5555 55.5 n 55.5. 5555 55.5 n 5:. 5555 55.5 n «5.5 5555 55.5 n 55.5 5 525 5 oze :5 3.5 .25. 5. 5525555! 5555 55.5 n 3.5 5555 :55 n 55.5 5555 5.5 n 5.5 5555 55.5 H 5.5 5555 55.5 n 55.5 a 525 5:555 55 n «I. 5555 55.5 a 55.5 5555 55.5 n 53V 5555 55.5 n 55.5 5555 R5 5 55.5 5 525 55255555 555555 5. 25.5; 5555. 55 5.5 5.5 5.5 5 $335 55.qu5 535.com he soon 352.55. sz 55: 55.5 n «5.55 5:55 555 a 55.55 3555 55.5 n E55 3555 55.5 n «5.55 55559555 55: 55.5 e 55.55 3:55 555 e 55.55 35: 55.5 n 55.3 A :5: 55.5 5 R55 5.5555555 5555 35.5 n F: 5:55 555 n 5.5 5:555 555 n 55.5 3555 3.5 n 3.5 35$ .55 n 3.5 35$ 95 n {.5 >32er 5o 5E: am one an EoEuwaF< 525$ 55.5 ... 55.5 a 525 35$ 35 n 55.5 5 525 5 52.. 55 on.» .35. 5 52555.3 .5555 55.5 n 55.5 5:555 55.5 n 55.5 35$ 55.5 n 55.5 35$ 55 n 55.5 a 525 .5355 55.5 n 55.5 .3555 25 n 55.5 35$ 55.5 5 55.5 35$ 55.5 n 53 5 525 EoEamafia 5.85:5 5. 25.5; 5.8 55 5.5 ..5 5 $5555 9:9: .2525: $550.82. .0 one: 3535:. «mm 35: 55.5 e 55.55 .555 55.5 e 55. a 355 55.5 H 55.55 55: 55.5 n 3.55 55559555 55: 55.5 n 5.55 :5: 55.5 n 55. 3 £555 :55 n 55.: 5:55 555 n 55.: 5.855555 255$ 55.5 a 55.5 $58 :20 H mod 3555 55.5 n 55.5 3555 55.5 n 55.5 55$ 55.5 e 3.5 55$ 35 n 55.5 >323: ..o 5E: as one 3 E25325 358 55.5 n 55.5 a 525 3555 S5 5 55.5 5 525 5 oze :5 en.» .3... 5. 52555.3 .5598. 55595 55 55 55555855555 zmo 555 .mm .35 .5? 52555 5555 55555555 5.552 5 £555; 585 .3 5.555 55$ :5 n 5.5 5:555 55.5 a 55.5 55$ :5 n 55.5 3555 55.5 n 55.5 a 525 5555 55.5 n 55.5 3555 55.5 n 55.5. 55$ :5 H 5.5 3555 3.5 n 55.5 5 025 E25253 505:5 5. 2.5.5; 558 55 5.. 5.5 5 mmo .5235 9.53 .2585553525 2.5 855 .5535: 158 .55 v 1: ”mod v n? ”5595 65:8 :55 5:25.56 25:85:55 . $55 .3558: ”02.5 .3555: ,6 :onEac \ ......mEEm 50 558:5 50 mm H 5:55.: 95:55 5555. mm 52:55.95 Em 505.55 __< 55: 55.5 H 55.55 55: 55.. H 55.55 :5: 55.5 H 55.55 35 55.5 H 55.5. 55: 55.5 5 E55 55: 55.5 a 2.55 5:: t5 5 3.53 5555985 555 5: a 55.3 55$ 5: H 55.5. 555 5: H 55.3 55555555 5.58 5-5 5.55: 159 .28 .9958 ”02a .Emz: Ho 59:5: H mEEEm .0 55:55 .6 mm H game 993 .32 nwucmmma 2m mm:_m> __< 53 «to H H3 889 2.0 H one 82.2 2.0 H 9} 853 :d H Be 859 02. H H? 3 02¢ 8:3 Hod H o: 332 Hod H a: 3:3 Hod H :3 8:3 Hod H :2 858 Hod H 9: H 02¢ :55 £90; 2.3 3 “5.53.2 33$ 36 H 9} 888 «3 H 36 53v :6 H m: 853 «to H H3 683 N3 H one a 32.”. 8.53 mod H HHH 858 mod H x: 833 23 H 84 $9: 8.0 H x: 658 Hod H m3 H 02¢ :55 “5.52.?“ 522.5 ow< 3 3 m... to c 335 9:95 ESmEam Ho anon 353a: zmo 8:99 2d H m3 533 Ed H 3m 35$ are H de 8:2: H; H Xe to. 02¢ 8:5 «to H 3N 6:3 2.0 H H? 35$ :6 H 2: 53$ :6 H 84 H oza :55 £925 .62. 3 .5563? 6:3 8.0 H «me $83 Ed H and 358 mad H 8n 3:03 5.0 H 31‘ E oza 6:3 «:0 H vow 3:va o; H H3 858 :.o H Hg 8:93 Ed H 23 H 02¢ :55 “cassava 285:5 ao< 2 3 to o .23.. 9.53 352.8 355ng .o 8% .233: «mm 8:333 H N? 3:8 2.0 H N? 33$ 2.0 H Re 359 mod H N; 5 02¢ 832 8.0 H :3 3:08 mod H H? am: «no H 23 2:89 vod H 23 H ozn. .55. £225 :68. 3 “552.3 8S3 HE H HIV GEE m; H m: 33$ H; H H3 558 Ed H m: 3 02¢ 659 mod H 8H 3:3 mod H 8H 8:9; 8d H SH 389 mod H H3 H oza i=5 “5.58%.“ .855: oo< 3 3 to o .23 9.53 ....zoeznaszu Ho 38 352a: mmo 8:38. 5:25 9 H3 .2253.“ so: 2.5 Ba .Nmm .mmo 5E 38: 2:3 Ho gumbo 23m. Ho 80$ 8.5% 2%,: .3 2an 160 .86 v or 539m .928 E0: EmHmtfi >_Emoc_cm_m . A925 Ho .0983: : 2955 Ho .8825 Ho mm H .888 993 $8. “825me 9m .82? __< 88: 8.0 H 3.2 EN: 8; H cm. 5 88: 88 H 8. 5 88: 8.8 H 2.3 8:: 8.”. H 3.8 82368 88: 8.8 H 8.8 85 SH H H88. 85 8.8 H 3.9 83 H; H 8.3. 8:8 88 H 8.8 883:8 on? 082:3 Ho .3532 2 m a m a H a o 23 :55 9:95 533:3 Ho awou 35322 833 mm.» H 8.: 8S8 22 H 8.8 38$ 88 H mHHN 88: «NH H 88m 85% 88 H 8.3, :5: 88 H 8H,? 8:88 8.8 H Hv.H~ b.8368 8:0: 5.8 H 8.8 83:98 mama Boa—2:3 Ho .3532 3 3 2. 8:3 9:83 682.8 3553: Ho 3% .2522 9 «mm Emu SH H 8.2 8:3 8.8 H 3.8 . 8:3 38 H 8.8 88: 8+ H 3.3 85: «3 H 5.9. 85% a? H 9.3 88.: 88 H 8.8 833988 8:3 one H NH. 2. 823:8 uuuo $2326 “—0 53:52 3 3 3 8:2. 9.31. .23352268 .0 83 .2332 o mwo .cozflofl H3395 Nwflmc .9833me Eat sz ucm .wa .wmo 5:5 umfimb mEmo Ho macamto 29:3 E9. $3 8535 Ho .3832 .mé mfim... 1 6 1 .80 v Q. ”0:05 6.2.00 E0... 0.0.00.0 2.0005005 .. 8.00.. .0 505:... . 208.00 00 50:55 .0 mm H 2.00.: 90:8 #80. 08:80.0 0.0 829 __< 6.9. 8.8 H 88 EN: 8.... H 8.8 .92. 8... H 8.8 8.2. 88 H 2.5 SE 8.8 H 8.8 383.83 .8: 8..” H 5.8 it 88 H 3.8 $8. 8... H 8.8 33. 8... H 8.8 68. 3+ H 8.8 5.838.. 0......“ 8.3.5“. .x. 2 3 ...... H... .. .035 9:95 520.000 .0 0000 .0530: zme $8: 8.8 H 8.8 823. 8.8 H 83 32%. 8.8 H 8.8 8:8. «8 H 8.8 383.83 .88: 8.. H 8.8 5.3. 8.. H 8.8 :8: 8.. H 8.8 . ES. 8... H 8.8 38383 0......“ 83:5“. ... 3 3 H... a .23: 9:8. 3.02.8 35.53: .0 80: 353...: «mm :8: 9: H 8.8 $8. 8.2 H 8.8 :8. 8... H 8...... 8.9. E H 8.8 2.83.83 68. 8.8 H 8.2. .8 H. Bx H 8.8 E8. .38 H 8.8 8.5. 8.8 H 8.8 38383 8.8 83.03“. .x. S 3 H... o .23: 9:8. 383.838.: .0 one: 3522... mm: .388. 83.... 9 ..8 83.588 59. 2me 8m .88 .80 5.; 8.8.. 28... ..o 8.380 298. 52.. 88 u.0 ......8 88.8. 9..: s ..1 2an 162 P1 .86 v Q. EH65 63:8 E0: Embtfi 228535 . .me: ho .mnEsc \ 295cm ho .3835 Ho mm H 259: Ems—um “mam. 3:533 05 829 __< Ana: «.0 H N8 EN: no H 0.3 as: to H 0:: 68: Yo H v.02 Er: 3.2 E2338 :5: 00 H 3.2 $5 no H $2 $8 to H 08. S3 to H «.8 35V 3.2 3.83%; 3......“ 9.3.3:“. .x. 3 ....N m... 2. o .23.. 9:95 503.com .0 32. 3:3qu zww 3mm 3 H 3”: 858 No H Q8 22.0.3 90 H 32 8:3 32 5.3338 .83: E H mam 5:8 No H H. 2: EB: E H «.8 EB: 92: €835“. 3:3.“ 9.3.33". .x. 3 3 3 o .23.. 9:9: 33.32. 3:38.52 3 Son .253: «mm :5 3 0o H ”.8 3516 H 52 43$ 3 H 32 8a: 92: émaamon. 65: mm H 6% En: «N H «.8 £53 to H 0.8? 8:3 92: 3.33:8 3......“ 9.3.3.9. .\. 3 3 E. o .23.. 9:9: .3893286 .0 82. .333: mmo .5388. 298.5 N? awn .mco_§wmm 82. zmo Em NH. .80 55, Emma memo Ho actuate 22.2 52. m8» 8 5% 35:5 E: s 3 $3528 9%? HI. 2an 163 Awe... .o .383. \ «.mEEm .o .3835 .0 mm H 9.8:. 933 “mam. 8.535 2m 93.? __< 8a: SN H 8..” Eu: 3.3 H 3d 8.333 H 8.0 a.” a? H 8.» .3333 H cm. 3 3.8398,. .33. 8a H 9.... 3.5 mom H Be .3. an H 3: 53 So H B... as. 3.m H 23 2.83.5. mono 33.2895 ox. 3 3 m... 3 o 5:5 9:95 583:3 .0 300 3532.. zmo .23. mod H 8.0 .35. 6a H .3 saw. m5 H 3... 6:8. 8N H 9... 2.83.8. 8.3. 3.3 H 3..” .333 8... H mm... fa: 8.0 H on... 2:3. No.3 H 8.3 2.835.. ammo wanna—890 .x. 3 3 3... a u m .23.. 9.3... 352.8 3:332 .0 38 3532... m in: mud H :3 33. 3.... H 38 $3. 8... H 3.3 8.9. an H 8.3 3.2398. .23. 3.3 H no... .5: 8.. H and E8. 3... H SN 65. X! H no... 3.83:8 ago ocuflacouco .x. 3 o 3 3 o a mug .23 9.3... .2.§..=u..§o.u .0 88 352“: .8598. case... 3 E. .mcoasmmmrefi zmo 2m .mm .80 5.; 8.8.. 25.. 39.5%.. 23.... EB. mum» 8.99.qu .o 28.9. .3 mam» a 1 4.5 DISCUSSION The present study demonstrated that in utero and lactational exposure to the high dose (10 ug/kg bwld) of DES decreased body weight on PND 21, while the low-dose (1.0 of ug/kg bwld) of DES increased postpubertal body weight in female offspring (Table 4—1). High dose (10 pg/kg bw/d) of EE2 decreased body weights in female offspring until weaning. These results suggest that in utero and lactational exposure to EEMs may alter body weights of infants and wildlife at an early stage of development and during the postpubertal period. Maternal exposure to estrogen reduces fetal body weight due to reduced placental blood flow during late pregnancy (41, 42). Yasuda et al. (18) demonstrated that daily oral gavage of mice with EE2 (0.02, 0.2, and 2.0 mglkg bw) from GD 11 to GD 17 decreased average fetal body weight. Even though our doses used in the present study were 2- to ZOO-fold less than in the study by Yasuda et al. (18), we also observed decreased body weight at the high doses of DES and EE2 until weaning. On the other hand, based on dramatically increased obesity after the 1980’s and an important role of estrogen in the natural weight-control mechanisms, a hypothesis has been formulated that EEMs at low concentration alter the body's natural weight-control mechanisms resulting in interference of the normal developmental and homeostatic controls over adipogenesis and energy balance (43-45). In this study, the increase in body weight of female offspring maternally exposed to 1.0 of pg DES/kg bw/d at POP is in agreement with the results reported by Newbold et al. (43). In their study, in utero exposure to DES (1 part per billion or ppb) and GEN (50 parts per million 165 or ppm) in drinking water significantly increased body weight at 4 months of age (PND 112). Furthermore, ERa knockout mice have increased body fat content with a 10-fold increase in E2 (46, 47). A proposed mechanism is that low doses of EEMs increase signaling of ERB or block signaling of ERa and contribute to obesity by altering the body's natural weight-control mechanisms. Taken together, further research is needed to investigate whether in utero and lactational exposures to low doses of EEMs are a potential cause of obesity. Anogenital distance is considered a sensitive indicator of demasculinization of male genitalia. However, it may not be a sensitive biomarker to examine estrogenicity of EEMs in female offspring. We observed that in utero and lactational exposure to DES, EE2, and GEN treatment did not significantly affect AGD of F-1 females regardless of AGD adjustment for body weight (Table 4-2). In contrast to our results, AGD was significantly increased in female rats exposed in utero to 0.2 pg DES/kg bw on GD 11 through GD 17 by daily subcutaneous injection (48). Clark et al. (49) confirmed that the growth rate of the AGD was controlled by dihydroxytestosterone rather than by testosterone. Consequently, EEMs that interfere with the synthesis of dihydroxytestosterone or decrease its bioactivity can decrease the growth rate of AGD. Evan et al. (50) reported that GEN inhibits the activity of 5a-reductase, which converts testosterone to dihydroxytestosterone in genital skin fibroblasts. A GEN-induced decrease of AGD in female offspring on PND 7 was observed in female rats exposed in utero to 5,000 pg GEN/kg bw on GD 16 through GD 20 by daily subcutaneous injection (51). The difference in AGD results observed in 166 this study compared to results from other studies may be due to species differences, differences in doses, and the effectiveness of different routes of administration to pregnant dams (gavage vs. subcutaneous injection). Results from the present study suggest that in utero and lactational exposure to synthetic estrogens and phytoestrogens may have different effects on ovarian responses of postpubertal female offspring to pregnant mare’s serum gonadotropin injection followed by human chorionic gonadotropin injection, which induces ovulation. We observed that in utero and lactational exposure to DES (0.1 pg DES/kg bwld) altered the number of ovulated eggs in female offspring at POP, while EE2 and GEN has no effects on ovulation in female offspn'ng at the same stage of development. The different ovarian responses to superovulation at later stages of life may be due to differences in estrogen potency between DES and the phytoestrogen. Compared to the potency of endogenous estrogen, the relative potencies of DES were similar or greater compared to 17B-estradiol (E2) (if E2 is assigned an arbitrary relative potency value of 1, then the relative potency of DES and EE2 is 1.25 to 2.5) (52). The relative potency of GEN was 10'4 to 10'5 less than that of E2. Furthermore, our studies indicated that exposure to low doses of DES and EE2 in utero and during lactation enhanced ovulatory capacity when compared with control animals, whereas animals exposed to higher doses ovulated fewer eggs. Oocytes are a permanent cell population generated before birth. Therefore, the total number of oocytes present in the mature ovary originates from a definite number of primordial germ cells, which migrate into the 167 developing fetal ovary (53). Studies demonstrated that in utero or neonatal exposure to an EEM induces multioocyte follicles in adult female offspring (34, 54, 55). Iguchi et al. (56) observed significantly decreased ovulation and increased incidences of multioocyte follicles (up to nine oocytes per follicle) in 7-week-old female mice subcutaneously injected daily with 1.0 pg DES/kg bw from PND 1 to PND 5, compared to age-matched control mice. The incidence of multioocyte follicles was significantly increased when neonatal DES treatment was begun on PND 0 to PND 3, but was decreased when started after PND 1O (54). These results indicate that in utero and lactational exposure to EEMs may decrease the total population of germ cells through direct action on the fetal ovary via induction of multioocyte follicles and/or indirectly through negative feedback on the fetal hypothalamic-pituitary axis (57, 58). Further study is necessary to more fully elucidate the mechanism of induction of multioocyte follicles. In addition to the exposure concentration, the timing of exposure is an important factor in terms of the increased incidence of multioocyte follicles in adult females through alteration of the genetic program of the fetal and neonatal ovary. However, similar to results in the present study, Wordinger et al. (59) observed increased ovulation in 6- to 8-week-old female mouse offspring exposed in utero to DES (10 pg/kg maternal bw) on GD 15 or GD 16 by daily subcutaneous injection. The underlying mechanisms for the biphasic effects of DES and EE2 reported here need to be investigated further. In contrast to our results for the GEN-exposed animals, Jefferson et al. (33) reported that neonatal exposure to GEN (1.0, 10, or 100 pg/pup/d, 168 equivalent to 667, 6,667 or 66,667 jig/kg bwld) by subcutaneous injection for 5 days increased the number of ovulated oocytes in the 1.0 pg GEN treated mice, while the number of ovulated eggs decreased in the 10 and 100 pg GEN-treated mice on PND 19. They observed a dose-related increase in multioocyte follicles (up to four oocytes per follicle). They also demonstrated that ERB plays a role in the occurrence of multioocytes in that ERB knockout mice did not have multioocytes in the ovaries. In addition, epidemiological studies indicated that the consumption of isoflavone-rich soy foods extends the length of the follicular phase and decreases serum follicle-stimulating hormone and luteinizing hormone concentrations (60, 61). This suggests that GEN may alter oocyte maturation via a delay in follicular development. Taken together, this study demonstrated that in utero and lactational exposure to GEN at relevant human exposure concentrations did not have adverse effects on ovulation. Our in vitro fertilizing results from in utero and lactational exposure to DES are in agreement with those from epidemiological studies, which indicated that DES-exposed women are less likely to become pregnant than unexposed controls (62). In the present study, there was a decreasing trend in in vitro fertilizing ability of eggs collected from female offspring exposed to high doses of DES. In addition, the high dose of EE2 significantly decreased in vitro fertilizing ability during the peripubertal period. Studies demonstrated that multioocyte follicles from female mice injected daily with 1.0 pg DES for 5 days from birth, had lower embryo cell division (8-cell stage embryos: 77% in the control animals vs. 47% in the DES-exposed mice, respectively) (56, 63). These results 169 indicate that neonatal exposure to DES may significantly increase multioocytic follicles leading to decreased embryo cell division and thus fertilization. Conversely, Wordinger et al. (59) observed that there was no significant difference in the in vitro development of 2-cell embryos to the blastocyst stage between control and female offspring of females subcutaneously injected with 10 pg DES /kg bw on GD 15 or GD16. On the other hand, there were no significant effects of in utero and lactational exposure to GEN on fertilizing ability of eggs. Further study needs to be done to explain the effects of maternal DES and EE2 exposure on fertilizing ability of eggs of female offspring. The present study demonstrated that in utero and lactational exposure to DES, EE2, and GEN did not affect the number of degenerated eggs. Our results are different from those of other researchers who have shown that the numbers of degenerated eggs increased in female offspring exposed to EEMs during the in utero and/or neonatal period (19, 63). These researchers suggest that EEMs delay cell cycles by degrading centrosomal proteins and perturbing the spindle microtubule organization and chromosome segregation (64, 65). Acknowledgements This work was supported by a grant from US EPA (R827-402-01-0) to KC, PMS, and T2. 170 4.6 BIBLIOGRAPHY 1. 10. 11. Norgil Damgaard I, Main KM, Toppari J, Skakkebaek NE 2002 Impact of exposure to endocrine disrupters in utero and in childhood on adult reproduction. Best Pract Res Clin Endocrinol Metab 16:289-309 Hutchinson TH 2002 Reproductive and developmental effects of endocrine disrupters in invertebrates: in vitro and in vivo approaches. Toxicol Lett 131:75—81 Brevini TA, Cillo F, Antonini S, Gandolfi F 2005 Effects of endocrine disrupters on the oocytes and embryos of farm animals. Reprod Domest Anim 40:291-299 Kaiser J 2005 Developmental biology. Endocrine dianpters trigger fertility problems in multiple generations. Science 308:1391-1392 Herbst AL, Ulfelder H, Poskanzer DC 1971 Adenocarcinoma of the vagina. N Engl J Med 284:878-881 DeChemey AH, Cholst l, Naftolin F 1981 Structure and function of the fallopian tubes following exposure to diethylstilbestrol (DES) during gestation. Fertil Steril 1981 Dec;36(6):741-5 36:741-745 Greco TL, Duello TM, Gorski J 1993 Estrogen receptors, estradiol, and diethylstilbestrol in early development: the mouse as a model for the study of estrogen receptors and estrogen sensitivity in embryonic development of male and female reproductive tracts. Endocrine reviews 14:59-71 Smith OW, Smith GVS 1949 Usa of diethylstilbestrol to prevent fetal loss from complications of late pregnancy. N Engl J Med 241 :562-568 Smith CW 1948 Diethylstilbestrol in the prevention and treatment of complications of pregnancy. Am J Obstet Gynecol 56:821-834 McLachIan JA 1977 Prenatal exposure to diethylstilbestrol in mice: toxicological studies. J Toxicol Environ Health 2:527—537 McLachIan JA, Newbold RR, Shah HC, Hogan MD, Dixon RL 1982 171 12. 13. 14. 15. 16. 17. 18. 19._ 20. Reduced fertility in female mice exposed transplacentally to diethylstilbestrol (DES). Fertil Steril 382364-371 Newbold RR, McLachIan JA 1982 Vaginal adenosis and adenocarcinoma in mice exposed prenatally or neonatally to diethylstilbestrol. Cancer Res 42:2003-2011 Haney AF, Newbold RR, McLachIan JA 1984 Prenatal diethylstilbestrol exposure in the mouse: effects on ovarian histology and steroidogenesis in vitro. Biol Reprod 30:471-478 Yasuda Y, Ohara I, Konishi H, Tanimura T 1988 Long-term effects on male reproductive organs of prenatal exposure to ethinyl estradiol. Am J Obstet Gynecol 1 59: 1 246-1 250 Thayer KA, Ruhlen RL, Howdeshell KL, Buchanan DL, Cooke PS, Preziosi D, Welshons WV, Haseman J, Vom Saal FS 2001 Altered prostate growth and daily sperm production in male mice exposed prenatally to subclinical doses of 17alpha-ethinyl oestradiol. Hum Reprod 16:988—996 Hyder SM, Chiappetta C, Stancel GM 1999 Synthetic estrogen 17alpha- ethinyl estradiol induces pattern of uterine gene expression similar to endogenous estrogen 17beta-estradiol. J Pharmacol Exp Ther 290:740- 747 Slikker W, Jr., Bailey JR, Newport D, Lipe GW, Hill DE 1982 Placental transfer and metabolism of 17 alpha-ethynylestradioI-17 beta and estradiol-17 beta in the rhesus monkey. J Pharmacol Exp Ther 2232483- 489 Yasuda Y, Kihara T, Nishimura H 1981 Effect of ethinyl estradiol on development of mouse fetuses. Teratology 23:233—239 Yasuda Y, Kihara T, Tanimura T, Nishimura H 1985 Gonadal dysgenesis induced by prenatal exposure to ethinyl estradiol in mice. Teratology 322219-227 Yasuda Y, Konish H, Tanimura T 1987 Ovarian follicular cell hyperplasia in fetal mice treated transplacentally with ethinyl estradiol. Teratology 172 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 36:35-43 Donn J, Mendoza M, Pritchard J 2008 AP Probe Finds Drugs in Drinking Water. In: The Associated Press. New York Maggs JL, Grimmer SF, Onne ML, Breckenridge AM, Park BK, Gilmore IT 1983 The biliary and urinary metabolites of [3H]17 alpha- ethynylestradiol in women. Xenobiotica 13:421-431 Williams MC, Goldzieher JW 1980 Chromatographic patterns of urinary ethynyl estrogen metabolites in various populations. Steroids 36:255-282 Aheme GW, Briggs R 1989 The relevance of the presence of certain synthetic steroids in the aquatic environment. Pharrn Pharmacol 41:735- 736 Tabak HH, Bloomhuff RN, Bunch RL 1981 Steroid hormones as water pollutants, II. Studies on the persistence and stability of natural urinary and synthetic ovulation-inhibiting hormones in untreated and treated wastewaters. Dev Ind Microbiol 22:497-519 Williams RJ, Johnson AC, Smith JJ, Kanda R 2003 Steroid estrogens profiles along river stretches arising from sewage treatment works discharges. Environ Sci Technol 37:1744-1750 Roefer P, Snyder S, Zegers RE, Rexing DJ, Fronk JL 2000 Endocrine- disrupting chemicals in a source water. J AWWA 92:52-58 US EPA 2000 Estimated per capita water ingestion in the United States. In. ‘ Washington DC: US EPA, Office of Water 1301; 40 US EPA 2001 National primary drinking water regulations; arsenic and clarifications to compliance and new source contaminants monitoring; final rule. In. Washington DC: US EPA Setchell KD, Zimmer-Nechemias L, Cai J, Heubi JE 1998 Isofiavone content of infant formulas and the metabolic fate of these phytoestrogens in eariy life. Am J Clin Nutr 68:14538-14618 173 31. 32. 33. 34. 35. 36. 37. 38. 39. Flynn KM, Ferguson SA, Delclos KB, Newbold RR 2000 Effects of genistein exposure on sexually dimorphic behaviors in rats. Toxicol Sci 55:311-319 Newbold RR, Banks EP, Bullock B, Jefferson WM 2001 Uterine adenocarcinoma in mice treated neonatally with genistein. Cancer Res 61:4325-4328 Jefferson WN, Couse JF, Padilla-Banks E, Korach KS, Newbold RR 2002 Neonatal exposure to genistein induces estrogen receptor (ER)alpha expression and multioocyte follicles in the maturing mouse ovary: evidence for ERbeta—mediated and nonestrogenic actions. Biol Reprod 67:1285-1296 Jefferson WN, Padilla-Banks E, Newbold RR 2006 Studies of the effects of neonatal exposure to genistein on the developing female reproductive system. J AOAC Int 89:1189-1196 Fitzgerald C, Elstein M, Spona J 1999 Effect of age on the response of the hypothalamo-pituitary—ovarian axis to a combined oral contraceptive. Fertil Steril 71 :1079-1084 Chou K, Cook RM 1994 Paraoxon inhibits fertilization of mouse gameters in vitro. Bull Environ Contm Toxicol 53:863-868 Fielden MR, Halgren RG, Fong CJ, Staub C, Johnson L, Chou K, Zacharewski TR 2002 Gestational and lactational exposure of male mice to diethylstilbestrol causes long-term effects on the testis, sperm fertilizing ability in vitro, and testicular gene expression. Endocrinology 143:3044- 3059 Fielden MR, Samy SM, Chou KC, Zacharewski TR 2003 Effect of human dietary exposure levels of genistein during gestation and lactation on long-term reproductive development and sperm quality in mice. Food Chem Toxicol 41 :447-454 Odum J, Tinwell H, Jones K, Van Miller JP, Joiner RL, Tobin G, Kawasaki H, Deghenghi R, Ashby J 2001 Effect of rodent diets on the sexual development of the rat. Toxicol Sci 61 :115-127 174 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. Gallavan RH, Jr., Holson JF, Stump DG, Knapp JF, Reynolds VL 1999 Interpreting the toxicologic significance of alterations in anogenital distance: potential for confounding effects of progeny body weights. Reproductive Toxicology 13:383-390 Beydoun SN, Abdul-Karim RW, Haviland ME 1974 The regulatory effect of estrogens on fetal growth. III. Placental deoxyribonucleic acid, ribonucleic acid, and proteins. Am J Obstet Gynecol 120:918-921 Shepherd RW, Stanczyk FZ, Bethea CL, Novy MJ 1992 Fetal and maternal endocrine responses to reduced uteroplacental blood flow. J Clin Endocrinol Metab 75:301-307 Newbold RR, Padilla-Banks E, Snyder RJ, Jefferson WM 2005 Developmental exposure to estrogenic compounds and obesity. Birth Defects Res A Clin Mol Teratol 73:478-480 Grun F, Blumberg B 2006 Environmental obesogens: organotins and endocrine disruption via nuclear receptor signaling. Endocrinology 147:850-55 Baillie-Hamilton PF 2002 Chemical toxins: a hypothesis to explain the global obesity epidemic. J Altem Complement Med 8:185-192 Naaz A, Zakroczymski M, Heine P, Taylor J, Saunders P, Lubahn D, Cooke PS 2002 Effect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor alpha (ERalpha): a potential role for estrogen receptor beta (ERbeta). Horm Metab Res 34:758-763 Heine PA, Taylor JA, lwamoto GA, Lubahn DB, Cooke PS 2000 Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc Natl Acad Sci USA 97:12729-12734 Honma S, Suzuki A, Buchanan DL, Katsu Y, Watanabe H, Iguchi T 2002 Low dose effect of in utero exposure to bisphenol A and diethylstilbestrol on female mouse reproduction. Reprod Toxicol 16:117- 122 Clark RL, Antonello JM, Grossman SJ, Wise LD, Anderson C, Bagdon WJ, Prahalada S, MacDonald JS, Robertson RT 1990 External genitalia 175 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. abnormalities in male rats exposed in utero to finasteride, a 5 alpha- reductase inhibitor. Teratology 42:91 -1 00 Evans BA, Griffiths K, Morton MS 1995 Inhibition of 5 alpha-reductase in genital skin fibroblasts and prostate tissue by dietary lignans and isofiavonoids. J Endocrinol 147:295-302 Levy JR, Faber KA, Ayyash L, Hughes CL, Jr. 1995 The effect of prenatal exposure to the phytoestrogen genistein on sexual differentiation in rats. Proc Soc Exp Biol Med 208:60-66 Gutendorf B, Westendorf J 2001 Comparison of an array of in vitro assays for the assessment of the estrogenic potential of natural and synthetic estrogens, phytoestrogens and xenoestrogens. Toxicology 166279-89 Byskov AG 1994 Embryology of mammalian gonads and ducts. In: Knobil E, Neill JD eds. The physiology of reproduction. New York: Raven Press; 487-540 Iguchi T, Takasugi N, Bern HA, Mills KT 1986 Frequent occurrence of polyovular follicles in ovaries of mice exposed neonatally to diethylstilbestrol. Teratology 34:29-35 Guillette LJJ, Moore BC 2006 Environmental contaminants, fertility, and multioocytic follicles: a lesson from wildlife? Semin Reprod Med 24:134- 141 Iguchi T, Fukazawa Y, Uesugi Y, Takasugi N 1990 Polyovular follicles in mouse ovaries exposed neonatally to diethylstilbestrol in vivo and in vitro. Biol Reprod 43:478-484 Gharib SD, Wierrnan ME, Shupnik MA, Chin W 1990 Molecular biology of the pituitary gonadotropins. Endocr Rev 11 :177-199 Shupnik MA 1996 Gonadotropin gene modulation by steroids and gonadotropin-releasing hormone. Biol Reprod 542279-286 Wordinger RJ, Brown D, Atkins E, Jackson FL 1989 Superovulation 176 60. 61. 62. 63. 64. 65. and early embryo development in the adult mouse after prenatal exposure to diethylstilboestrol. J Reprod Fertil 85:383-388 Cassidy A, Bingham S, Setchell KB 1994 Biological effects of a diet of soy protein rich in isofiavones on the menstrual cycle of premenopausal women. Am J Clin Nutr 60:333-340 Cassidy A, Bingham S, Setchell K 1995 Biological effects of isoflavones in young women: importance of the chemical composition of soyabean products. Br J Nutr 74:587-601 Bibbo M, Gill WB, Azlzi F, Blough R, Fang VS, Rosenfield RL, Schumacher GF, Sleeper K, Sonek MG, Wied GL 1977 Follow-up study of male and female offspring of DES-exposed mothers. Obstet Gynecol 49:1-8 Iguchi T, Kamiya K, Uesugi Y, Sayama K, Takasugi N 1991 In vitro fertilization of oocytes from polyovular follicles in mouse ovaries exposed neonatally to diethylstilbestrol. In Vrvo 5:359-363 Can A, Semiz O 2000 Diethylstilbestrol (DES)—induced cell cycle delay and meiotic spindle disruption in mouse oocytes during in-vitro maturation. Mol Hum Reprod 6:154-162 Can A, Semiz O, Cinar O 2005 Bisphenol-A induces cell cycle delay and alters centrosome and spindle microtubular organization in oocytes during meiosis. Mol Hum Reprod 112389-396 177 CHAPTER 5 Effects of in utero and Lactational Exposure of Male Mice to 17a-Ethynyl Estradiol on Sperm Production, Quality and In Vitro Fertilizing Ability. 5.1 ABSTRACT The objective of this study was to examine the effect of in utero and lactational exposure of male mice to 17a-ethynyl estradiol (EE2), the estrogenic component of oral contraceptives, on sperm production, quality and fertilizing ability. Pregnant CS7BU6 mice (F-O), bred with DBA/2 males, were gavaged with O, 0.1, 1.0, or 10 pg EE2/kg body weight per day (bwld) from gestational day (GD) 12 to postnatal day (PND) 20. The male offspring (F-1) were examined for sperm production and quality in young adult (PND 105 to 117) and middle-aged (PND 315 to 337) mice by using computer-aided sperm analysis and an in vitro fertilization (IVF) assay. Body weights were decreased in the 1.0 and 10 pg/kg bwld groups on PND 7. Testis weights were decreased in the 10 pg/kg bwld group on PND 21. Kidney weights were decreased in the 10 pg/kg bwld group in middle-aged males. There was no significant treatment effect on anogenital distance (AGD), regardless of dose and age. Sperm production in the 10 pg/kg bwld group was significantly decreased in middle-aged males, as compared to the control group. Even though sperm production was decreased in the 10 pg/kg bwld group, there was no difference in fertilizing ability of sperm. The results demonstrated that in utero and lactational exposure to 10 pg EE2/kg bw/d could decrease sperm production without changing sperm motion patterns and sperm fertilizing ability. 178 5.2 INTRODUCTION Estrogens have been considered the predominant female sex steroid hormones, but many studies have demonstrated that estrogens are also produced in a variety of tissues in mammalian males such as adipose tissue, brain, liver, adrenal glands, mammary glands, and testes (1-5). The serum concentrations of estrogens are about 20 to 40 pg/ml in male humans (6), 35 to 45 pg/ml in male rats (7), and 2 to 180 pg/ml in males of other species (8, 9). Ten to 20% of the circulating estrogens in males is produced by the testes; the remaining 85% comes from peripheral aromatization of androgen precursors in different tissues (10, 11). Similar to human testes, testes of the adult rat synthesize approximately 10 to 25% of circulating estrogens (11, 12). Estrogens in the male are derived by irreversible conversion of testosterone or androstenedione into 176-estradiol (E2) by aromatase coded in the CYP19 gene (13). To exert their biological effects, estrogens bind to the intracellular estrogen receptors (ERs) or putative plasma membrane ER (14). Studies using ER knockout mouse models demonstrated that estrogens are essential for the development and normal functioning of testes (9), as well as for the processes of sperrnatogenesis (15) and steroidogenesis (16) in males. Concerns have been raised over the potential adverse effects of estrogenic endocrine modulators (EEMs) on the development of the male reproductive system. Humans and animals can be exposed to many estrogenic and anti-estrogenic chemicals, including polychlorinated biphenyls (PCBs), dichloro-diphenyl-trichloroethane (DDT), diethylstilbestrol (DES) and 179 phytoestrogens through environmental media, food and pharmaceutical products. Our previous DES and genistein (GEN) studies demonstrated that in utero and lactational exposure to DES (10 pglkg bwld) decreased sperm production, the number of Sertoli cells, and the in-vitro fertilizing ability of sperm without apparent abnormalities in testes and sperm motion parameters (17). On the other hand, in utero and lactational exposure to GEN at doses of 0.1, 0.5, 2.5 or 10 mglkg bw/d increased in vitro fertilizing ability of sperm from male offspring without changing sperm production (18). In the present study, we examined the effects of in utero and lactational exposure of mice to 17a-ethynylestradiol (EE2) on male reproductive development. Additionally, we compared results of the present study with results from our previous DES and GEN studies. EE2, a synthetic estrogen, is widely used as a component of oral contraceptives and as a carcinostatic agent for prostate and breast carcinomas. The doses of EE2 used for oral contraceptives are approximately 0.4 to 2.0 pglkg bw/d (19) and for arresting prostate carcinomas, doses range from 4.0 to 1400 pglkg bw/d (20). It has been estimated that approximately two million women per year in the United States (US) and the United Kingdom (UK) are exposed to EE2 during undetected early pregnancy (19, 21). Slikker et al. (22) demonstrated placental transfer of EE2 by daily intravenous administration to pregnant rhesus monkeys on gestational days 108 through 121, whereas endogenous estrogen E2 in the placenta is metabolized to estrone (E1). In the fetal circulation, E1 and EE2 were observed, whereas E1, E2 and EE2 were observed in the maternal circulation. These .180 results indicated that EE2 can be transferred to the fetus and affect development of the fetal reproductive and central nervous systems (23—25). In addition, there have been human and ecological health concerns over pharmaceuticals and their metabolites present in drinking water supplies because of human wastes. Significant proportions (approximately 17 to 29%) of ingested sulfate or glucuronide metabolites of EE2 excreted in the urine (26, 27) are present in effluents from wastewater treatment facilities. Concentrations of EE2 in sewage treatment plant effluent has been measured at concentrations ranging from less than 0.4 ng/l to 15 ng/l and from 300 to 1800 ng/l in the UK and US, respectively (28-30). In 1997, concentrations of EE2 were measured at 0.48 pg/I in the Las Vegas Wash and at 0.52 pg/l in Las Vegas Bay (31). In 2006, it was reported in a study that 4.7 ng EE2/I, 1.2 ng EM and 0.83 ng E2/l were detected in seawater from the Acushnet River Estuary (Massachusetts, USA) (32, 33). Furthermore, in March, 2008, an Associated Press report indicated that pharmaceuticals including estrogenic compounds such as EE2 have been detected in the US drinking water supplies of 24 major metropolitan areas supplying 41 million Americans (34). Based on the US Environmental Protection Agency’s (EPA) drinking water exposure estimates, women of childbearing age (15 to 44 years of age), could be exposed to 6.3x10'6 to 2.5x10‘2 pg EE2/kg bw/d through water consumption, and children younger than one year old could be exposed to 2.1x10’5 to 8.3x10‘2 pg EE2/kg bw/d (35, 36). To examine effects of in utero and lactational exposure to EE2 on the reproductive development of 181 male mouse offspring, we selected doses of 0.1, 1.0 and 10 pglkg bw/d based on potential environmental exposures. 5.3 MATERIALS AND METHODS Animals Eleven-week-old virgin C57BL/6 female and 7- to 8-week-old DBA/2 proven breeder male mice were obtained from Charles River Laboratories (Portage, MI, USA) and housed in polycarbonate cages with cellulose fiber chips (Aspen Chip Laboratory Bedding, Northeastern Products, Warrensberg, NY, USA) as bedding. BGD2 F-1 mice, offspring from the CS7BL/6 x DBA/2 cross mating, were used for the IVF assay since a large number of oocytes obtained from superovulated B6D2 F-1 mice had consistent and reproducible fertilization in control animals (37). The animal room was maintained at 30 to 40% humidity and 23 °C. Room lighting was on a 12:12 lightzdark cycle. All animals had free access to deionized water provided in glass bottles with mbber stoppers and AlN-76A rodent feed (Research Diets, New Brunswick, NJ, USA). AlN-76A rodent feed is a casein-based, open-fomiula purified diet that contains non- detectable concentrations of the estrogenic isoflavones genistein, diadzein and glycitein (38, 39). B6D2 F-1 female mice that were used as oocyte donors were provided with standard rodent feed (Harlen Teklad 22/5, Madison, WI, USA). 182 Treatments Upon arrival, C57BL/6 females and DBA/2 males were separated by sex, housed two per cage and acclimatized for three to seven days. At the beginning of the study, two females were housed with a male for mating. Following evidence of pregnancy, dams were separated from the males, housed individually, and randomly assigned to one of four treatment groups (0, 0.1, 1.0, or 10 pglkg bwld). Corn oil and 17a-ethynylestradiol (298% pure) were obtained from Sigma Chemical Co. (St. Louis, MO, USA). Pregnant dams were treated by daily gavage of 17a-ethynylestradiol (0, 0.1, 1.0, 10 pglkg bw) in 0.1 ml corn oil from gestational day (GD) 12 to postnatal day (PND) 20 (Figure 5-1). The dose of test chemical was adjusted each day to the dam’s body weight, which was measured before dosing between 0900 and 1200 h. Offspring were separated by sex on PND 21 and fed AlN-76A until necropsy. F-0 mice were euthanized on PND 21. Measurements: a. Anogenital distance The anogenital distance (AGD) is the distance between the anus and the base of the external genitals. AGD is considered a sensitive indicator of demasculinization of male genitalia. Therefore, F-1 AGD was measured with body weight on PND 7, PND 14, and PND 21. Measurements were made with a dissecting microscope equipped with an ocular micrometer (Nikon, Melville, NY, USA). Measurements were recorded to the nearest 0.1 mm. To minimize 183 operator variation and increase precision, two individuals measured AGD independently. AGD were averaged when the two independent readings were different. Pa'ring female (CSIBL) and male (DBA) Birth (3502) Wearl'ng Mating Gestation Lactation IIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIIII Offspring Maternal treatment period Gestational day12 Postnatal day 20 oDams were heated by gavage with EE2 (o, 0.1, 1.0, 10 pgfrg maternal bwlday) from gestational day 12 to postnatal dav 20 B'rth We 'ng Fol-IIIIIIIIII orrspn'ng F-1 — Age Dayll oayr Day14 Day 21 Day 105t0117 Day315to337 Litter size Anogenital distance Bodyweight Body weight (AGD) Organ weight Organ weight Body weights Sperm concentration Sperm concentration Sperm motion parameters Sperm motion parameters Sperm fertilizing ability Sperm fertilizing ability Figure 5-1. Schedule of maternal treatments and parameters measured in their male offspring (F-1). 184 b. Testis weight, sperm production and motion analysis Testis weight of male offspring (F-1) was measured on PND21 and when animals were sacrificed for the in vitro fertilization assay at young adulthood or middle age. Male offspring were sacrificed by cervical dislocation. Sperm were collected by piercing the pair of cauda epididymides with a 25 gauge needle in an organ culture dish (Becton Dickinson, Franklin Lakes, NJ, USA) that contained 1 ml of BMOC-3 medium (37°C) (Gibco/BRL, Grand Island, NY, USA), which is a capacitation supporting medium. Sperm suspensions were incubated for 30 min at 37°C in humidified air (95% Oz, 5% C02) before sperm concentration and motion parameters were analyzed. Sperm suspensions (20 pl) were placed on a warmed 20 pm counting chamber and analyzed using a Series 4000, computer- assisted digital image analysis system (CellSoft, Series 4000, CRYO Resources Ltd., New York, NY, USA). For motility, number of motile sperm, velocity, linearity, amplitude of lateral head displacement, and beat/cross frequency, a minimum of 100 sperm cells was analyzed for each animal. Sperm concentration is expressed as the number of spermatozoa/ml suspension medium. “Motile count” refers to the number of sperm that moved faster than 20 pm/s/ml. Velocity is defined as the average distance (pm) traveled by motile sperm in 1 s. Linearity represents the ratio of the length of a straight line to the actual distance traveled (x10) by each sperm cell. Amplitude of lateral head displacement is a measurement of the lateral movement of the sperm head from the curve mean of its track. The beat/cross frequency (Hz) is the number of beats (or crosses) of 185 the sperm across its curve mean/s. All measurements for each sperm collection were performed in duplicate and averaged. c. Sperm fertilizing ability Non-treated females (3-week-old B602 F-1) serving as oocyte donors were injected intraperitoneally with 10 IU of pregnant mare’s serum (PMSG) (Sigma Chemical Co., St. Louis, MO, USA) followed by 10 IU of human chorionic gonadotropin (Sigma Chemical Co., St. Louis, MO, USA) 48 h later. Fourteen to 16 h later, the superovulated oocytes from each female were collected from the proximal oviducts and incubated in a 1 ml organ culture dish (60 mm diameter x 15 mm high) (Becton Dickinson Falcon, Franklin Lakes, NJ, USA) containing 0.9 ml Brinster’s BMOC-3 medium supplemented with penicillin and streptomycin (Gibco/BRL, Grand Island, NY, USA). Sperm diluted with Brinster’s BMOC—3 medium (0.1 ml) were added to the 0.9 ml medium organ culture dish containing the oocytes to achieve a final sperm concentration of 3 x 104 sperm/ml/dish. In order to measure sperm fertilizing ability, the optimal concentration of sperm was selected in the IVF assays to increase the assay sensitivity for detecting positive and negative changes in sperm fertilizing ability. After insemination, oocytes were incubated for 24 h, and then 50 pl of 35 pM bisBenzimide (Sigma Chemical Co., St. Louis, MO, USA) was added to the dish. The oocytes were incubated with stain for an additional 20 min before being examined for fertilization, using a Nikon Optophot fluorescent microscope (Melville, NY, USA) equipped with a 100- watt mercury bulb, a 365/10-nm excitation filter, a 400-nm dichromic mirror, and 186 400-nm barrier filter. Oocytes were counted and scored as fertilized if the oocytes were at the two-cell stage or at the one-cell stage containing spindle, two pronuclei, and a second polar body. Fragmentation and other signs of degeneration of oocytes were also recorded. Statistical analysis All data analyses were performed using SAS (version 8.2, SAS Inc, Cary, NC, USA). Data were tested for normality by using the Shapiro-Wilk test and a residual plot. When data were not normally distributed such as velocity and amplitude of later head displacement of sperm, log transformation was applied to the data with subsequent retesting. Data were analyzed using litter as the experimental unit to estimate litter effect as experimental error. The MIXED procedure of SAS was used for analyzing a nested design, which treatment was a fixed effect and dam was a random effect within treatment. To examine correlation between testis weights and kidney weights, Pearson’s correlation test was used. When analyzing offspring body weight, organ weight, and AGD, litter size at PND 0 and age at necropsy were included as covariates (40). For analyzing IVF data, data set were separately analyzed by two age groups (young adult or middle-aged males) and adjusted by age of each animal. The MIXED procedure was also used for IVF data. Dunnett’s test was used for comparisons between control and treatment groups. The level of significance was set at P < 0.05. 187 5.4 RESULTS Developmental effects on F-1 male mice Maternal exposure to 1.0 and 10 pg EE2/kg bwld resulted in changes in male F-1 body weight while changes in organ weights were restricted to the 10 pg EE2/kg bwld group. Body weights in both the 1.0 and 10 pg EE2/kg bwld groups were significantly decreased at PND 7 (P < 0.05) (Table 5-1). There was a trend of decreased body weight in young adult males. Kidney weights were decreased in the 10 pglkg bwld group in middle-aged males. Testis weights of males in the 10 pg EE2/kg bw/d group were significantly less compared to controls on PND 21. The correlations between body weights and kidney weights were P = 0.19 on PND 21 and P < 0.01 at middle age, respectively. The correlations between body weights and testis weights were P < 0.01 both on PND 21 and at middle age, respectively. The correlation between testis weights and kidney weights were P = 0.08 on PND 21 and P = 0.03 at middle age, respectively. When organ weights were adjusted for body weight, however, there were no statistically significant EE2 treatment effects in organ weights. With a single exception, in utero and lactational exposure to EE2 did not result in gross abnormalities in the reproductive tract of male offspring. Furthermore, there were no significant differences in AGD (Table 5-2). 188 Sperm production and quality In utero and lactational exposure to EE2 at doses of 10 pglkg bwld had a significant effect on epididymal sperm concentration of sperm in middle-aged mice (Table 5-3). There was no significant change in sperm motion parameters. Sperm fertilizing ability Although there was a statistically significant decrease in epididymal sperm concentrations (Table 5-3), in utero and lactational exposure to EE2 did not affect in vitro fertilizing ability of sperm from F-1 male offspring (Table 5-4). The relative percentage of fertilizing ability of sperm was presented in Table 5-4a (when the control = 100%). 189 Em: 88 H .8... 8:8: 88 H 8... 88v 88 H 8... 88: 88 H 8... 88-8.85 8.9 88 H 8... Ea 8.8 H 8.8 88. 8.8 H .88 88: 8.8 H R... 9.88 88> 888 88 H 8... c... .v .88 H 8... 888 88 H 8.8 8.8: R. H 8.8 .8 828 o 888 .o .8. 2.8.83 .2... 8.8.2 E8: ... .. H 8.8. 8.8.: 88 H 858 888 B. .. H 858 88: 88. H 8. .8 8888.822 88. 8.8 H 8.8. Se .88. H 8.8. 688 8.8. H 888. 88: .88. H 8.8. 89.88 88> .85 88. H 8.8 A. ... .v 8... H 88.3. 88. 8.8 H .8. .8 8.8: 8:. H .38 .N 828 3.5 2925 no.3» .58: 8.8 H 8. .8 8.8.: 8.8 H 88.88 68. 8.8 H 8.88 88: 8.8 H 888 888-2822 58. 8.8 H 8.88 Se 88. H 88.88 88. 8.8 H 8.8.. 68: 88. H 8.8.. 89.88 888> 53 8.8 H 8.8. is 88. H 8.8 E: 88.8 H 88.. 58 8.8 H 888... .N 828 as. .8833 8:28. £8: .88 H ....8 8.2.: 88 H 8.3 88. 88 H ....E as: 8.8 H 8.... 888.2822 88. 88 H 88.8 E8 88 H 888.. 8.8 8... H 8.8 88: 88 H 8.8.. 88.88 88> 88. #8. H 8.8 A. ... .V 8.8. H 8.8 .88. 88 H 8.8 8.8: 8.8. H 8.8 .N 828 3.5 23.83 8:83... ca: 8.8 H 8.. 8:8: ...8 H 8.. 88. ..8 H 8.8 88: 8.8 H ...8 8888.825. 88. 888 H 8... E8 88 H 8.. 6.8V 88 H 8.. 68: 88 H 8.. 89.88 888> 88. 88 H 88 :2. .v 88 H 88 8.3 88 H 88 8.8: 8.8 H 8.8 .N 828 a. £8.83 .2... E8: 8.. H .88 8:8: 8.. H 8.8 6.8 .88 H 8.8 $8: 8.. H 8.8.. 8 888.2822 588 8.. H 8.8 SE 8.. H 88 A85 8.. H 8.8 68: 8.. H :8 8 88.38 888> 8.8. 88 H 8.. 8.83 88 H ...8 888 88 H 8.8 8:89 88 H 8.8 .8 828 8.83 88 H 88 3:3 .88 H 88 A888 88 H 88 8.83 88 H 88 ... 828 .689 8.8 H 88 8:83 ..8 H 88 888 88 H 8.... 8.83 :8 H 8... . 828 .3 £8.83 88 8. 8.. .8 8 6.38 9:83 3.82.8 .8388: .8 Son .8538: .coonflicmaocfi N. >88 .93sz% E9. «mm 53> no.8... 8888 .o matawto o_mE .o 8.599. compo ucm mango; room ..A». 038. 190 .modva . ”8:05 68:00 E9. €932: 3:85:05 . .8898: .0 8E: 8.. E995 ..on ..o. 3.83.8.8. o ._o.ooE omx=2 :_ .3998: .o 9:: .m mom :0. 3.3.6.4 n 885 888.2 s 8 828 88 .88. 8 8.8.8 8; £98; .88 m .92.: .0 :3E:: . m_mE_:m .o .8835 .0 mm H mcmoE 28:88 .82 En mo:_m> __< E3 88 H 88 48...... 888 H 88 .8388 H 88 .88.. 88 H .88 88.285 .88. 888 H 88 E8. 88 H 88 .88. 88 H «.8 .88.. 88 H ..8 9.88 88> .88. 88 H .88 .. ... 5 888 H 88 .88. 88 H .88 8.8.. 88 H 88 .N 828 8 8.38 .o .8. £88; 8.8. 8.8.3 :8: 88 H 8. 6...... 88 H .8. 6.8. 88 H 8.. 48.8.. 88 H 8.. 88-2822 .88. 88 H E. E8. .88 H 8.. .88. 88 H E. .88.. 88 H 8.. 9.88 88> EN. 88 H 8.. E... 88 H 8.. ...: 88.8 H 8.. .88. 88 H 8.. .8 828 8 p88 .8 .8. £8.83 8:8. 8.8.3. €2.88 H 8.8 48...... .88 H «.8 .88. 88.8 H 8.8 88: 88 H ..8 888822 .88. .88 H 8.8 .8. .88 H :8 6.8. .88 H v.8 .88.. .88 H 8.8 8.38 88> .88. 88 H R8 .. ... .. 8.8 H 88 .88. 8.8 H 88 8.8: 88 H 88 .8 828 o 8.38 .8 .8. £8.83 8:55 8.8.3 8.88 .-8 28b 191 .58988888 09. .o 8:... .8 £982, >83 .0 .09 3:0 8:. 80. 8288.68. 8 .88... 88.8.2 5 8.8888 8 88 E88; 888 88 888.8 88; :9. 8 .29.: .0 .3E:: . 8_mE_:m .o 85:83:. .0 mm H 8:92.. 98:88 .888. 2.8 8829 =< 8:88. ...8 H 88.8 8.8... 8.8 H 88.8 8.8. 8.8 H 888 8.88. 8.8 H 8.8 .8 82.. 8:88. 8.8 H 8.8 8.83. 8.8 H 88.. A888. 8.8 H 38.. 8:88. 3.8 H 38.8 3. 82: 8:88. 8.8H38. A.:3. ...8H88. A888. 8.8H88. 8:3. ...8H88. 882: V 8A8..8.EE. 898 8.8:. 8.88.28 8:88. 88.8 H .88 8:8... 88.8 H 88.8 A888. 88.8 H 8.8 8.8... 88.8 H 83.8 .8 82: 8:88. 88 H 38.8 8.83. 88.8 H 838 A888. 88.8 H .88 8:88. 88.8 H 88.8 3. :2: 8:88. 8.8 H 83.8 A. :3. 8.8 H 88 A888. 8.8 H 88.8 8:3. 8.8 H .88 8 oz: 88:... oo< 8.8.2 8:88. 88.8 H .88 8.8.. 88.8 H 88.8 888. 88.8 H 8.8 88:83. 88.8 H 88.8 .8 :2: 8:88. 88 H .88 8.83. 88.8 H 88.8 A888. .88 H 88.8 8:88. 88.8 H 88.8 3. :2: 8:88. 8.8 H 88.8 A. :3. 8.8 H 38 A888. 8.8 H 88.8 8:3. 8.8 H 38.8 8 :2: Ass. :98 8.8: 2. o._. F... 0 A28... 9:8... .2882. 38858.88. .8 888 .8528: .5388. :wfiefi 8. >80 .m:o_.m.8mm E9. «mm 5.3 80.80.. 8E8: .o m::amto 28:: .o 08:38:. _m._:ma:< 8.8 8.3.8... 192 .00.0vn_ . ”0:90 .0008 E0: $9006 20:85:05 . .mw H 0009: 90:00 .000. 20 mm:_m> =< «v.0 H 0.20.. 3.0 H 9.0? 00.0 H 3.8. 0Y0 H V0.0? 0000-202: 0Y0 H 00.0.. 00.0 H 00.0w 0v.0 H 3.0? 00.0 H 0.0.? 02:00 93> 3.: 0.000360% 000.8 “mam 5.0 H 0.0 3.0 H 00.»~ 00.0 H 006 «0.0 H 00.0 300-2025. 2.0 H 3.0 9.0 H :3. 00.0 H 004. 9.0 H 00.0 9.300 93> 3.1355320»... cam: .30. .0 0033.003 30 H 00.0 3.0 H 0:. 00 H 5v 0'0 H and 800-2025. 0P0 H 006 9.0 H 006 0V0 H 004q 2.0 H 00.0‘ $300 93> 5:35.. 000 H 00.00? 0.0.0 H 00.0: 00.0 H v0.02. ~00 H 00.03 800.2025. 0N.N H 00.0: 0:. H 00.5? 00.~ H 00.0NF VON H N009 02:00 0::o> £53 $8.; .3; H V0.00 00.? H 00.00 00.? H v0.0.0 00.? H v0.0a 0000.062: N0; H 05.8 3H H v0.00 00.? H 00.0w F0... H 00.00 0:000 0::o> ...-:00 :0 003223000 .5on #0? Acta 800 400: 803622 3;: 6%: So: $0: 258 98> A905. 00 50:5: \ 0.9020 00 .00.:an 0... 9.0 0 3 .225 9.33.2 00.33 $550.3... .0 «not 3530! 000800. 50:25 me >00 .mcoafimmd E0: 000 55, 09000 0800 ..ometmwto 90E 00 5030 0:0 0000:0808 .5QO 3,5223 .00 2an 193 00000 ..0002 05 2 000020 00: 20 “05 0000 “—0 E0200 0 .mm H 0000:. 000000 0000. 0.0 000_0> __< 00.0 H 00.0 00.0 H VV.3 0: H V0.0 V0.0 H V0.0 00000.02: 00.0 H 0: 00.0 H 30.0 00.0 H 0: 00.0 H V0.0 0:000 0008» 0000 0300800.: .3063... .x. V0.0.H 03.0 00.0 H 00.0 0V0 H 0: 00.0 H 03.0 00000.02: 00.0 H 00.3 00.0 H 00.0 V0.0 H 00.0 0V.0 H 00.0 0:000 0:0o> 00000 08009000.: ..00000 ox. 0V3 H 00.0 0V3 H 00... 00.3 H 30.0 00.3 H 0..... 00000.02: 00.3 H 3.0 00.0 H 00.0 00.0 H 30.3 V0.0 H 00... 0:000 0008» 0000 00000500.“. .x. 00.0 H 00.0V 00.0 H 00.0V V0.0 H 00. V 00.0 H 00.0V 00000.02: 00.0 H 00.00 00.x. H 00.00 00.0 H 00.30 0V0 H 00.V0 0:000 0c0o> 003.00”. =00.0:O .x. 30.0 H 00.00 03.0 H 00.00 V0.V H 00.00 0V0 H V0.00 00000.02: 03.0 H 00.00 V0.0 H 00.00 00V H 30.00 V0.0 H 00.00 0:000 0c0o> 35% 9.3.0.5". .x. Em: SS: @100 000: 0000-000: 50: So: GE :0: 0 0:80 93> 0.00: 00 .00E0: \ 0.0.500 .0 0000.02: 03 0.3 3.0 0 :22. 9:03 .2258 .2053: .o 3% .0520: .8038. 00:25 «2% .mcozsmmefi 000 .0; 02000 0500 03:30.8 0.05 EB. 50% 0:02.28 .0 $5320.00. S: 5 V0 003 194 .mm H 0:00E 0.0000 0000. 0.0 0029 ..< 0.0 H 0.00 0.0 H 0.00 0.V H 0V0 0.00.. 00000.02: 00 H v.0: 0.3 H 0.3 0.0 H V2 0.02 93%. 28> 03.0.8 __3.0=0 ... 0.0 H 0.3 0.0 H 0.5 0.0 H32 0.02 800.002.). 3 H002 to H 0.02 3 H 0.5 0.02 0:30 88> . 0:30 050.03". V. $2. .20.: 8.0. .00: 800-0002 .20. E2. .05 .0: 0 0:80 28> .000.. “.0 .0200: . 0.0.0.00 .0 000.002: 2 3 2. o .23.. 9:0... .2058 3550.5: .0 080 .0530: 8.000 50:25 2 :00 00005000 80.... 0mm 5.? 000000 00.00 00 0000000 0.08 E0... 0:000 .0E>0_0_00 00 3:50 00.0.0.0. 00.3 E “.0 0000.00.00 02.0.00 .0V.0 0.00.. 195 5.5 DISCUSSION The present study demonstrated that in utero and lactational exposure to EE2 at high doses (1.0 and 10 pglkg bw/d) decreased early postnatal body weights in male offspring. Commercially available oral contraceptives provide daily doses ranging from 0.4 to 2.0 pg EE2/kg bw (19). Therefore, early developmental delays in an infant may result from the mother taking oral contraceptives during an undetected early pregnancy. We expected that in utero and lactational exposure to EE2 have a similar effect on body weights of male offspring as we observed in our previous DES study because studies indicated that the effective doses of DES and EE2 in cell proliferation and ER binding assays were similar or greater compared to E2 (41-45). However, the decrease in early postnatal body weights reported here is not in agreement with our previous results from the DES study in that there were no significant treatment effects on body weights of male offspring (17). The reasons for these conflicting results may be that EE2 has a different estrogenic mode of action in the in vivo assay system compared to DES. Therefore, it is difficult to predict the in vivo estrogenic effects of EEMs. Epidemiological and laboratory animal studies demonstrated that in utero exposure to EEMs at low doses increases body weights in adulthood via interference of the normal developmental and homeostatic controls over adipogenesis and energy balance (46-49). For example, Newbold et al. (50) demonstrated that in utero exposure to DES (1 part per billion or ppb) and GEN (50 parts per million or ppm) in drinking water significantly increased mouse body 196 weight at 4 months of age. Furthermore, ERa knockout mice have increased body fat content with a 10-fold increase in E2 (51, 52). A proposed mechanism is that low doses of EEMs increase signaling of ERB or block signaling of ER): and contribute to obesity by altering the body's natural weight-control mechanisms. However, in the present study, we observed a trend of decreased body weights of male offspring in both young adult and middle-aged males at 1 and 10 pg EE2/kg bwld. Taken together, further research is needed to investigate whether in utero and lactational exposure to low doses of EEMs is a potential cause of obesity. In addition to decreased early postnatal body weights by in utero and lactational exposure to EE2, decreases in kidney weights were observed in the 10 pg EE2/kg bw/d male offspring at middle age as well as decreases in testis weights were observed at PND 21. However, when organ weights were adjusted for body weight, there were no statistically significant EE2 treatment effects in organ weights. These results suggest that low doses of in utero and lactational exposure to EE2 may not affect organ weight, which is in agreement with the results of a 28-day repeatedoral toxicity study in the rat (53). The development of reproductive organs is a complex process that occurs during early gestation and is related to hormonal mechanisms that control the differentiation of bipotential gonads into the testes or ovaries. In the present study, we observed one incidence of herrnaphroditic organs in a male in the 0.1 pg EE2/kg bw/d treatment group on PND 105 (1 out of 113 mice maternally exposed to EE2). This animal’s testis weight was approximately 47% less than 197 the mean testes weight of the control group. The cauda epididymis contained no sperm cells. Similar to our observation, Yasuda et al. (54) demonstrated ovotestis and intra-abdominal testis with persistent Miillerian and Wolffian ducts in matemally—exposed male mouse fetuses whose dams were exposed to EE2 (20, 200, or 2000 pglkg bwld) from day 11 through day 17 of gestation, which was prior to gonadal differentiation. Additionally, studies demonstrated that high doses of EE2 (200 pglkg bw/d) administered for 2 or 7 days to pregnant mice caused 17 to 20% cryptorchidism in fetal and postnatal mice (55, 56). In our previous study (17), there was only one incidence of unilateral cryptorchidism on PND 315 in a total of 116 male mice offspring maternally exposed to 10 pg DES/kg bw/d. Since spontaneous herrnaphroditism in the common laboratory rodent is rare (57-59), our case may be related to the effects of exposure to low doses of EE2 from GD 12 through lactation. However, further investigation is needed to determine whether this incidence of herrnaphroditic organs occurred by chance. In general, AGD is considered to be a sensitive indicator of demasculinization of male genitalia. The AGD in males is approximately twice as long as the AGD in females. This is because dihydroxytestosterone produced by the gonads increases the length of the perineum separating the anus from the testicles via stimulation of cell proliferation (60-62). The growth rate of AGD in male rats is increased around gestational day 15, when testosterone synthesis begins (63). In the present study, administration of EE2 to pregnant dams was initiated on GD 12, when the gonads begin to produce dihydroxytestosteone. 198 However, AGD in male offspring was not changed. Our finding corresponds to the results from our previous studies with DES and GEN (17, 18). Furthermore, few studies demonstrated that EE2 exposure affects AGD in males. (64, 65). Collectively, in utero and lactational exposure to EE2 at potential environmental exposure concentration dose not result in demasculinization of male genitalia. There have been concerns about decreases in sperm concentration due to in utero exposure to EEMs. In this study, we observed an approximate 20% decrease in sperm production that was associated with a 10% decrease in testes weight at middle age in the 10 pglkg bwld group. Similarly, in our previous studies, a 20% decrease in sperm production was associated with an 11% decrease in testicular weight due to a DES-induced decrease in the number of Sertoli cells (25%, 17%, and 32% relative to controls on PND 21, 105, and 315, respectively) (17). In contrast, a 31% increase in sperm production associated with an approximate 10% increase in testicular weight was observed in the 10 mg GEN/kg bw/d treatment group (18). However, clinically relevant doses of EE2 (0.002, 0.02, 0.20, and 2.0 pglkg bw/d) from GD 0 to GD 17, which is equivalent to gestation week 16 in humans, decreased sperm production without changing testis weights in 50-day-old mice (19). Collectively, the data indicate that in utero exposure to EE2 has long-term effects on sperm production and testicular weight, which may be useful criteria to evaluate male reproductive toxicity. In utero exposure to EE2 resulting in decreased testis weight and a concomitant decrease in sperm production may be at least in part a result of 199 inhibition of testicular steroidogenesis and local loss of ERa-mediated estrogen action, which are important in testicular development. Leydig cells and Sertoli cells are the main synthesis sites of steroid hormones. During PND 10 to 26, Leydig cells and Sertoli cells in the immature rat are dividing and undergoing functional maturation (66). Synthesis of steroid hormones in the testis is dependent on two key groups of steroidogenic enzymes; cytochrome P450 enzymes (e.g. cholesterol side chain cleavage enzyme and aromatase) and hydroxysteroid dehydrogenases (HSD) (e.g. 3B-HSD and 17B-HSD) (67). An in vitro study demonstrated that E52 inhibits activities of 3B-HSD and 17a—HSD in human testicular tissue (68). At 2- or 200-fold higher doses of EE2 compared to the highest dose (10 pg EE2/kg bw/d) used in the present study, atrophy of Sertoli cells in fetal mouse testes on GD 18 as well as atrophy of seminiferous tubules, Leydig cell hyperplasia, and a 67% decrease in testosterone concentrations in aged (20- to 22-month-old) male mice resulted from in utero exposure to 20, 300, or 2,000 pg EE2/kg from GD 11 to GD 17 (54, 69, 70). At the maximum tolerated dose (200 pglkg bw/d) of EE2, Leydig cell atrophy and degeneration of germinal epithelium were observed in rats (71). After receiving 10,000 pg EE2/kg bw/d, which is 1,000-fold higher dose in the highest dose used in the present study, by oral gavage for 3 and 5 days, epididymal sperm production was significantly decreased in 11- and 12-week—old rats (72). Furthermore, estrogen receptor or knockout (aERKO) mice had normal prenatal development of the reproductive tract, but at approximately 20 weeks of age, there was a significant decrease in testis weights (73). Compared with age- 200 # J matched, wild-type male mice, there was an approximately 2-fold increase in LH concentrations, whereas FSH concentrations remained unchanged in aERKO mice (74). Increased LH secretion resulted in Leydig cell hyperplasia and a 2- fold increase in testosterone concentrations. These data suggested that in utero and lactational exposUre to EE2 decrease weights of the testes and reproductive tract via local loss of ERa-mediated estrogen action. Additionally, the effects of in utero and lactational exposure to EE2 would be occurring at the level of the hypothalamus through the negative feedback loop to decrease GnRH pulse frequency and at the level of the pituitary to decrease responsiveness to GnRH (9, 75, 76). Therefore, synthesis of LH and FSH, which were stimulated by GnRH, would result in decreased concentrations of testosterone leading to altered testicular develOpment, which in turn would alter sperrnatogenesis in the adult testis. On the other hand, there is a possibility of a direct action of estrogen on germ cells during various stages of germ cell development. Li et al. (77) demonstrated that increased differentiating sperrnatogonia type A cells were stimulated by a 1 pM dose of E2 but not by higher doses. The present study demonstrated that in utero and lactational exposure to EE2 at relevant doses for the human did not affect fertilizing ability of sperm without changing sperm motion parameters. However, this finding is contrary to results from our previous DES and GEN studies. A dose of 0.1 pg DES/kg bw/d resulted in increases in sperm fertilizing ability and beat cross frequency on PND 315 (17). On the other hand, GEN (10 mglkg bw) significantly increased in vitro fertilizing ability of sperm without changes in any sperm motion parameters (18). 201 WIT At a 1,000-fold higher dose of EE2, Kaneto et al. (70) reported decreased percentages of motile sperm, velocity and ALH displacement in rats orally exposed to 10,000 pg EE2/kg bw/d for 4 weeks as well as reduced male fertilizing ability in vivo (72). In conclusion, this study demonstrates that in utero and lactational exposure to relevant doses of EE2 for the human decrease postnatal body weight and has long-term effects on sperm production and without changing fertilizing ability of sperm and sperm motion parameters in male mice. Acknowledgements This work was supported by a grant from US EPA (R827-402-01-0) to KC, PMS, and T2. 202 5.6 REFERENCES 1. Schlinger BA, Arnold AP 1991 Brain is the major site of estrogen synthesis in a male songbird. Proc Natl Acad Sci 88:4191-4194 Roselli CE, Resko JA, Stormshak F 2003 Estrogen synthesis in fetal sheep brain: effect of maternal treatment with an aromatase inhibitor. Biol Reprod 682370-374 Patchev AV, Gotz F, Rohde W 2004 Differential role of estrogen receptor isofonns in sex-specific brain organization. The FASEB Journal Express Article Brodie AM 1979 Recent advances in studies on estrogen biosynthesis. J Endocrinol Invest 2:445-460 Kula K, Walczak-Jedrzejowska R, SIowikowska-Hilczer J, Wranicz JK, Kula P, Oszukowska E, Marchlewska K 2005 Important functions of estrogens in men—breakthrough in contemporary medicine. Przegl Lek 62:908-915 Nagata C, Takatsuka N, Shimizu H, Hayashi H, Akamatsu T, Murase K 2001 Effect of soymilk consumption on serum estrogen and androgen concentrations in Japanese men. Cancer Epidemiol Biomarkers Prev 102179-184 Brewster ME, Anderson WR, Pop E 1997 Effect of sustained estradiol release in the intact male rat: correlation of estradiol serum levels with actions on body weight, serum testosterone, and peripheral androgen- dependent tissues. Physiol Behav 61:225-229 Weinstein RL, Kelch RP, Jenner MR, Kaplan SL, Grumbach MM 1974 Secretion of unconjugated androgens and estrogens by the normal and abnormal human testis before and after human chorionic gonadotropin. J Clin Invest 53:1-6 Akingbemi BT 2005 Estrogen regulation of testicular function. Reprod Biol Endocrinol 3:51 203 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Gennari L, Nuti R, Bilezikian JP 2004 Aromatase activity and bone homeostasis in men. J Clin Endocrinol Metab 89:5898—5907 Levine AC, Kirschenbaum A, Gabrilove JL 1997 The role of sex steroids in the pathogenesis and maintenance of benign prostatic hyperplasia. Mt Sinai J Med 64:20-25 Dorrington JH, Fritz IB, Armstrong DT 1978 Control of testicular estrogen synthesis. Biol Reprod 18:55-64 Robertson KM, O'Donnell L, Jones ME, Meachem SJ, Boon WC, Fisher CR, Graves KH, McLachIan RI, Simpson ER 1999 lmpairrnent of sperrnatogenesis in mice lacking a functional aromatase (cyp 19) gene. Proc Natl Acad Sci U S A 96:7986—7991 Benten WP, Lieberherr M, Giese G, Wunderlich F 1998 Estradiol binding to cell surface raises cytosolic free calcium in T cells. FEBS Lett 422:349—353 O'Donnell L, Robertson KM, Jones ME, Simpson ER 2001 Estrogen and sperrnatogenesis. Endocr Rev 222289-318 Simpson E, Rubin G, Clyne C, Robertson K, O'Donnell L, Jones M, Davis S 2000 The role of local estrogen biosynthesis in males and females. Trends Endocrinol Metab 1 1 :184—188 Fielden MR, Halgren RG, Fong CJ, Staub C, Johnson L, Chou K, Zacharewski TR 2002 Gestational and lactational exposure of male mice to diethylstilbestrol causes long-term effects on the testis, sperm fertilizing ability in vitro, and testicular gene expression. Endocrinology 143:3044- 3059 Fielden MR, Samy SM, Chou KC, Zacharewski TR 2003 Effect of human dietary exposure levels of genistein during gestation and lactation on long-term reproductive development and sperm quality in mice. Food Chem Toxicol 41 :447-454 Thayer KA, Ruhlen RL, Howdeshell KL, Buchanan DL, Cooke PS, Preziosi D, Welshons WV, Haseman J, Vom Saal FS 2001 Altered prostate growth and daily sperm production in male mice exposed 204 20. 21. 22. 23. 24. 25. 26. 27. 28. prenatally to subclinical doses of 17alpha-ethinyl oestradiol. Hum Reprod 16:988-996 Miyamoto Y, Ueda K, Oshida K, Ohmori E 2000 Collaborative work to evaluate toxicity on male reproductive organs by repeated dose studies in rats 2). Testicular toxicity in rats treated orally with ethinylestradiol for 2 weeks. J Toxicol Sci 25:33—42 Smithells RW 1981 Oral contraceptives and birth defects. Dev Med Child Neurol 23:369-372 Slikker W, Jr., Bailey JR, Newport D, Lipe GW, Hill DE 1982 Placental transfer and metabolism of 17 alpha-ethynylestradiol-17 beta and estradiol-17 beta in the rhesus monkey. J Pharmacol Exp Ther 223:483- 489 Kallen B, Mastroiacovo P, Lancaster PA, Mutchinick O, Kringelbach M, Martinez-Frias ML, Robert E, Castilla EE 1991 Oral contraceptives in the etiology of isolated hypospadias. Contraception 44:173-182 Li DK, Daling JR, Mueller BA, Hickok DE, Fantel AG, Weiss NS 1995 Oral contraceptive use after conception in relation to the risk of congenital urinary tract anomalies. Teratology 51 :30-36 DeSesso JM 1997 Comparative embryology. Handbook of developmental toxicology. In. New York:: CRC Press; 111-174 Maggs JL, Grimmer SF, Onne ML, Breckenridge AM, Park BK, Gilmore IT 1983 The biliary and urinary metabolites of [3H]17 alpha- ethynylestradiol in women. Xenobiotica 13:421-431 Williams MC, Goldzieher JW 1980 Chromatographic patterns of urinary ethynyl estrogen metabolites in various populations. Steroids 36:255-282 Aheme GW, Briggs R 1989 The relevance of the presence of certain synthetic steroids in the aquatic environment. Pharrn Pharmacol 41:735- 736 205 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. Tabak HH, Bloomhuff RN, Bunch RL 1981 Steroid hormones as water pollutants, ll. Studies on the persistence and stability of natural urinary and synthetic ovulation-inhibiting hormones in untreated and treated wastewaters. Dev Ind Microbiol 222497-519 Williams RJ, Johnson AC, Smith JJ, Kanda R 2003 Steroid estrogens profiles along river stretches arising from sewage treatment works discharges. Environ Sci Technol 37:1744-1750 Reefer P, Snyder S, Zegers RE, Rexing DJ, Fronk JL 2000 Endocrine- disrupting chemicals in a source water. J AWWA 92252-58 Zuo Y, Zhang K, Deng Y 2006 Occurrence and photochemical degradation of 17alpha-ethinylestradiol in Acushnet River Estuary. Chemosphere 63:1 583-1 590 Zuo Y ZK, Deng Y. 2006 Occurrence and photochemical degradation of 17alpha-ethinylestradiol in Acushnet River Estuary. Chemosphere ;(): 63:1583-1590 Donn J, Mendoza M, Pritchard J 2008 AP Probe Finds Drugs in Drinking Water. In: The Associated Press. New York US EPA 2000 Estimated per capita water ingestion in the United States. In. Washington D.C.: US EPA, Office of Water 1301; 40 US EPA 2001 National primary drinking water regulations; arsenic and clarifications to compliance and new source contaminants monitoring; final rule. In. Washington DC: US EPA Chou K, Cook RM 1994 Paraoxon inhibits fertilization of mouse gameters in vitro. Bull Environ Contm Toxicol 53:863-868 Reeves PG 1997 Components of the AlN-93 diets as improvements in the AlN-76A diet. J Nutr 127:8388-8418 Thigpen JE, Setchell KD, Ahlmark KB, Locklear J, Spahr T, Caviness GF, Goelz MF, Haseman JK, Newbold RR, Forsythe DB 1999 206 40. 41. 42. 43. 45. 46. 47. 48. Phytoestrogen content of purified, open- and closed-formula laboratory animal diets. Lab Anim Sci 49:530-536 Gallavan RH, Jr., Holson JF, Stump DG, Knapp JF, Reynolds VL 1999 Interpreting the toxicologic significance of alterations in anogenital distance: potential for confounding effects of progeny body weights. Reproductive Toxicology 13:383-390 Gutendorf B, Westendorf J 2001 Comparison of an array of in vitro assays for the assessment of the estrogenic potential of natural and synthetic estrogens, phytoestrogens and xenoestrogens. Toxicology 166279-89 A Branham WS, Zehr DR, Sheehan DM 1993 Differential sensitivity of rat uterine growth and epithelium hypertrophy to estrogens and antiestrogens. Proc Soc Exp Biol Med 203:297-303 Van den Belt K, Berckmans P, Vangenechten C, Verheyen R, Witters H 2004 Comparative study on the in vitro/in vivo estrogenic potencies of 17beta-estradiol, estrone, 17alpha-ethynylestradiol and nonylphenol. Aquat Toxicol 66:183-195 * Shapiro S, Slone D 1979 The effects of exogenous female hormones on the fetus. Epidemiol Rev 1 :1 10-123 Mussman HC 1975 Drug and chemical residues in domestic animals. Fed Proc 34:197-201 Schildkraut JM, Demark-Wahnefried W, DeVoto E, Hughes C, Laseter JL, Newman B 1999 Environmental contaminants and body fat distribution. Cancer Epidemiol Biomarkers Prev 8:179-183 Wolff MS, Anderson HA 1999 Environmental contaminants and body fat distribution. Cancer Epidemiol Biomarkers Prev 8:951-952 Baillie-Hamilton PF 2002 Chemical toxins: a hypothesis to explain the global obesity epidemic. J Altem Complement Med 8:185-192 207 49. 50. 51. 52. 53. 54. 55. 56. 57. Rubin BS, Murray MK, Damassa DA, King JC, Soto AM 2001 Perinatal exposure to low doses of bisphenol A affects body weight, patterns of estrous cyclicity, and plasma LH levels. Environmental health perspectives 109:675-680 Newbold RR, Padilla-Banks E, Snyder RJ, Jefferson WN 2005 Developmental exposure to estrogenic compounds and obesity. Birth Defects Res A Clin Mol Teratol 73:478-480 Naaz A, Zakroczymski M, Heine P, Taylor J, Saunders P, Lubahn D, Cooke PS 2002 Effect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor alpha (ERalpha): a potential role for estrogen receptor beta (ERbeta). Horm Metab Res 34:758-763 Heine PA, Taylor JA, lwamoto GA, Lubahn DB, Cooke PS 2000 Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc Natl Acad Sci USA 97:12729-12734 Yamasaki K, Sawaki M, Noda S, lmatanaka N, Takatsuki M 2002 Subacute oral toxicity study of ethynylestradiol and bisphenol A, based on the draft protocol for the "Enhanced OECD Test Guideline no. 407". Arch Toxicol 76:65-74 Yasuda Y, Kihara T, Tanimura T, Nishimura H 1985 Gonadal dysgenesis induced by prenatal exposure to ethinyl estradiol in mice. Teratology 322219-227 Yasuda Y, Konishi H, Matuso T, Tanimura T 1986 Accelerated differentiation in seminiferous tubules of fetal mice prenatally exposed to ethinyl estradiol. Anat Embryol (Berl) 174:289-29 Walker AH, Bernstein L, Warren DW, Warner NE, Zheng X, Henderson BE 1990 The effect of in utero ethinyl oestradiol exposure on the risk of cryptorchid testis and testicular teratoma in mice. Br J Cancer 62:599-602 Eicher EM, Beamer WG, Washbum LL, Whitten WK 1980 A cytogenetic investigation of inherited true hennaphroditism in BALB/th mice. Cytogenet Cell Genet 28: 1 04-1 1 5 208 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. Imaizuml K, Imado N, Minato Y, Wada I, Takeshita M, Okaniwa A 1987 Case report of hennaphroditism in a rat. Nippon Juigaku Zasshi 4921171- 1 173 Kai K, Satoh N, Watanabe A, Shiraiwa K, Sasano H, Furuhama K 2003 Case report of rat true herrnaphroditism: colocalization of oocytes and granulosa and sertoli cells in the germinal cord. Toxicol Pathol 312290-294 Hotchkiss AK, Vandenbergh JG 2005 The anogenital distance index of mice (Mus musculus domesticus): an analysis. Contemp Top Lab Anim Sci 44246-48 Salazar-Martinez E, Romano-Riquer P, Yanez-Marquez E, Longnecker MP, Hemandez—Avila M 2004 Anogenital distance in human male and female newborns: a descriptive, cross-sectional study. Environ Health 328 Clark RL, Antonello JM, Grossman SJ, Wise LD, Anderson C, Bagdon WJ, Prahalada S, MacDonald JS, Robertson RT 1990 External genitalia abnormalities in male rats exposed in utero to finasteride, a 5 alpha- reductase inhibitor. Teratology 42:91-100 Habert R, Picon R 1984 Testosterone, dihydrotestosterone and estradiol- 17 beta levels in maternal and fetal plasma and in fetal testes in the rat. J Steroid Biochem 212193-198 Ryan BC, Vandenbergh JG 2006 Developmental exposure to environmental estrogens alters anxiety and spatial memory in female mice. Horm Behav 50:85-93 Sawaki M, Noda S, Muroi T, Mitoma H, Takakura S, Sakamoto S, Yamasaki K 2003 In utero through lactational exposure to ethinyl estradiol induces cleft phallus and delayed ovarian dysfunction in the offspring. Toxicol Sci 75: 402-411 de Kretser DM, Kerr JB 1988 The cytology of the testis. In: Knobil E NJ ed. The Physiology of Reproduction. New York: Raven Press Ltd; 837-932 Sanderson JT 2006 The steroid hormone biosynthesis pathway as a target for endocrine-disrupting chemicals. Toxicol Sci 94:3-21 209 68. 69. 70. 71. 72. 73. 74. 75. 76. Daehlin L, Hammar M, Damber JE, Berg AA, Petersson F 1986 Effects of oestradiol-17 beta and ethinyl oestradiol on human testicular steroidogenesis in vitro. Scand J Urol Nephrol 202177-181 Yasuda Y, Kihara 'I’, Nishimura H 1981 Effect of ethinyl estradiol on development of mouse fetuses. Teratology 23:233-239 Yasuda Y, Ohara I, Konishi H, Tanimura T 1988 Long-term effects on male reproductive organs of prenatal exposure to ethinyl estradiol. Am J Obstet Gynecol 1 59:1246-1250 Andrews P, Freyberger A, Hartmann E, Eiben R, Loof I, Schmidt U, Temerowski M, Folkerts A, Stahl B, Kayser M 2002 Sensitive detection of the endocrine effects of the estrogen analogue ethinylestradiol using a modified enhanced subacute rat study protocol (OECD Test Guideline no. 407). Arch Toxicol 76:194-202 Kaneto M, Kanamori S, Hishikawa A, Kishi K 1999 Epididymal sperm motion as a parameter of male reproductive toxicity: sperm motion, fertility, and histopathology in ethinylestradiol-treated rats. Reprod Toxicol 13(4): :279-289 Eddy EM, Washbum TF, Bunch DO, Goulding EH, Gladen BC, Lubahn DB, Korach KS 1996 Targeted disruption of the estrogen receptor gene in male mice causes alteration of sperrnatogenesis and infertility. Endocrinology 13724796-4805 Lindzey J, Wetsel WC, Couse JF, Stoker T, Cooper R, Korach KS 1998 Effects of castration and chronic steroid treatments on hypothalamic gonadotropin-releasing hormone content and pituitary gonadotropins in male wild-type and estrogen receptor-alpha knockout mice. Endocrinology 1 3924092-4101 Franchimont P, Chari S, Demoulin A1975 Hypothalamus-pituitary-testis interaction. J Reprod Fertil 442335-350 Hayes FJ, Seminara SB, Decruz S, Boepple PA, Crowley WFJ 2000 Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. J Clin Endocrinol Metab 85:3027-3035 210 77. Li H, Papadopoulos V, Vidic B, Dym M, Culty M 1997 Regulation of rat testis gonocyte proliferation by platelet-derived growth factor and estradiol: identification of signaling mechanisms involved. Endocrinology 13821289- 1298 211 CHAPTER 6 Effects of Gestational and Lactational Exposure of Female Mlce to 17a- Ethynyl Estradiol on Reproduction. 6.1 ABSTRACT Previously, in our diethylstilbestrol (DES) study with mice (1), fecundity, body weight, litter size, litter weight, and the percentage of male offspring were significantly decreased in the 10 pg DES/kg bw/d treatment group. The aim of the present study was to examine the effects of gestational and lactational exposure to 17a-ethynyl estradiol (EE2) on reproduction of dams. Ethynyl estradiol is a widely used estrogenic component of oral contraceptives (2). Pregnant CS7BL/6 mice, bred with DBA/2 males, were gavaged with EE2 (0, 0.1, 1.0, or 10 pglkg body weight per day (bw/d)) from gestational day (GD) 12 to postnatal day (PND) 20. Average pup weight in the 1 and 10 pg EE2/kg bw/d treatment groups on PND 1 was significantly decreased (P < 0.01) without a change in litter weight on PND 1. Four-day and 21-day pup survival rates were significantly decreased in the 10 pg EE2/kg treatment group (P < 0.01). The percentage of male offspring was not changed by gestational and lactational exposure to EE2. Our results suggest that gestational and lactational exposure to EE2 treatment decreased perinatal body weight and survival rate of offspring through weaning. 212 6.2 INTRODUCTION There have been human and ecological health concerns about a pharmaceutical estrogenic compound and its metabolites present in drinking water supplies due to the inability of sewage treatment facilities to remove them from the waste stream. Ethynyl estradiol is a widely used estrogenic component of oral contraceptives (2). Each year, there are approximately 2 million women in the United States (US) and the United Kingdom (UK) exposed to 0.4 to 2.0 pg EE2/kg bw/d during undetected early pregnancy (2). EE2 has been known as an environmental estrogenic modulator (EEM) because it and its metabolite are excreted in the urine and are found in effluents from wastewater treatment facilities (3-5). Approximately 17 to 29% of sulfate or glucuronide metabolites of EE2 ingested as an oral contraceptive are excreted in the urine (3, 4). In March, 2008, the Associated Press reported that pharmaceuticals, including estrogenic compounds such as EE2, had been detected in the drinking water supplies of 24 major US metropolitan areas supplying 41 million Americans (6). The concentration ranges of EE2 in sewage treatment plant effluents were in the range of 0.4 to 15 ng/l and 300 to 1800 ng/l in the UK and US, respectively (7-9). Concentrations of EE2 were measured at 0.48 pgll in Las Vegas Wash and at 0.52 pg/l in Las Vegas Bay (10). In addition, the concentrations of EE2 in surface water and rivers are about 5 to 50 pg/ml (11). Based on the US. Environmental Protection Agency’s (USEPA) drinking water exposure estimates, women of childbearing age (15 to 44 years of age) could be exposed to 6.3 x 10'6 to 2.5 x 10'2 pg EE2/kg bw/d through water consumption, and children younger 213 than one year of age could be exposed to 2.1 x 10'5 to 8.3 x 10'2 pg EE2/kg bwld (12, 13). Therefore, potential adverse reproductive effects of exposure to EE2 through consumption of drinking water in women and wildlife need to be investigated. Environmental estrogenic modulators have been linked to a variety of adverse effects on reproductive health in both humans and wildlife. Approximately 8 to 12% of all pregnancy losses in humans are the result of endocrine factors (14). Since exposure to estrogen during pregnancy plays a key role in regulating female reproductive process, exposure to EEMs during a critical period of fetal and neonatal development could permanently disrupt fertility of the offspring. For example, Yasuda et al. (15) demonstrated that daily oral gavage of mice with EE2 (0.02, 0.2, and 2.0 mglkg bw) from GD 11 to GD 17 decreased average fetal body weight and increased the number of fetal deaths in a dose-related manner. 6.3 MATERIALS AND METHODS Animals EIeven-week-old virgin C57BL/6 females and seven- to eight-week-old DBA/2 males were obtained from Charles River Laboratories (Portage, MI, USA). All mice were housed in polycarbonate cages with cellulose fiber chips (Aspen Chip Laboratory Bedding, Northeastern Products, Warrensberg, NY, USA) as bedding. The animal room was maintained at a humidity of 30 to 40% and a temperature of 23 °C. Lighting in the room was on 12-h light/dark cycle. All 214 animals were given free access to deionized water in glass bottles with rubber stoppers and AlN-76A rodent feed (Research Diets, New Brunswick, NJ, USA) ad libitum. This diet is a casein and sucrose-based open-forrnula purified diet with non-detectable concentrations of the estrogenic isoflavones genistein and diadzein (16). Treatment All mice were housed two per cage on arrival and acclimatized for at least three days before mating. Each pair of females was housed with a male within a week of arrival. Females were separated from the male following evidence of pregnancy and housed individually and randomly assigned to one of treatment groups. Corn oil and Wet-ethynylestradiol were obtained from Sigma Chemical Co. (St. Louis, MO, USA). Pregnant females were treated by daily gavage with Wan-ethynylestradiol (0, 0.1, 1.0, 10 pglkg bw/d) in 0.1 ml corn oil from GD 12 to PND 20 (Figure 6-1). On the day of parturition (PND 0), animals were not gavaged to avoid cannibalism of pups by the dams. The dose of test chemical was adjusted each day to the dam’s body weight, which was measured between 09:00 and 12:00 before dosing. Reproductive performances Body weights of females were recorded prior to mating and on PND 1, 7, 14, and 21. Fecundity [(number of pregnant females giving birth to live 215 Pairing female (057BL) and male (DBA) BlflhliBGDZI Wea Ing Mating Gestation Lactation F-O IIIIIllllllllllIIIIIIIIIIIIIIIIIIIII Offspring F.1 Treatment period Gestaflonal day 12 Postnatal day 20 - Pregnant females were treated by gavage with EE2 (0, 0.1, 1.0, 10 pglkg maternal bwlday) from gestational day 12 to postnatal day 20 m We hg F-OIIIIIIIIIIIIIIIIIIIIII Ofispflng F-1 Age Day 0 Day? Day 14 Day 21 Fecundity Body weights Body weights Organ weights Litter size Litter size Litter weight Survival rate of pups Survival rate of pups Sex ralio Figure 6-1. Schedule of treatments and parameters measured in females. young/number of pregnant females) x 100, %] and body weight gain (difference between pre-mating body weight and final body weight on PND 21) were calculated. The ratio of the number of pups born live and stillborn pups was recorded on PND 0. Litter weight and average pup weight were recorded on PND 1. Litter size was recorded daily for the pup survival analysis. Sex of the pups was determined on PND 21 and the sex ratio (% of male pups) was 216 calculated. Dams were euthanized and their organ weights were measured on PND 21. Statistical analysis SAS version 8.2 (SAS Inc, Cary, NC, USA) was used for all data analyses. The litter was considered as the experimental unit. All data were tested for normality by the Shapiro-Wilk test and residual plot. The MIXED procedure of SAS was used for analysis, where treatment was a fixed effect and dam was a random effect. Organ weights were adjusted by body weights at time of necropsy. Litter size on PND 0 and sex ratio on PND 21 were included as a covariate due to significant correlations with average pup weights. Dunnett’s test was used for comparisons between control and treatment groups. Statements of significance are based on a P value less than 0.05. 6.4 RESULTS Fecundity In EE2-exposed females, there was a decreasing trend in fecundity in the 0.1 and 10 pglkg bwld treatment groups (4% and 16%, respectively) when compared to the control group (Table 6-1). In our previous DES study, fecundity was significantly decreased in the 10 pglkg bwld treatment group (34%) (Table 6- 2)(1). 217 Body weights and organ weights Body weights in the our previous DES study were significantly decreased in the 0.1 and 10 pg DES/kg bw/d treatment groups on PND 7 and 20 (Table 6- 2)(1), whereas body weights and weight gain were not affected by EE2 exposure from gestation through lactation (Table 6-1). Correspondingly, there were no differences in liver and thymus weights of dams on PND 21 regardless of body weight-adjustment. Reproductive performance The number of live and stillborn pups was not significantly changed by EE2 treatment when compared to the control group. However, there was a trend toward an increase in the number of stillborn pups in EE2-exposed groups (Table 6-1). Average pup weight in the 1.0 and 10 pg EE2/kg bwld treatment groups on PND 1 was significantly decreased (P < 0.01) without a change in litter weight on PND 1. Pup body weight in the 1.0 and 10 pg EE2/kg bwld treatment groups was depressed compared to controls. Body weight was negatively correlated with litter size (P < 0.05). Sex ratio was positively correlated with average pup weight (P < 0.05). Four-day and 21-day pup survival rates were significantly decreased in the 10 pg EE2/kg treatment group (P < 0.01). In our previous study, the high dose of DES decreased average pup weight on PND 1 and litter size on PND 0 and 20 (Table 6-2)(1). The percentage of male offspring was not changed in the EE2 treatment, in contrast to the lower percentage of males in the 10 pg DES/kg treatment of our previous DES study. Overall, gestational and 218 lactational exposure to GEN did not affect any reproductive parameters (Table 6- 3)(17). 219 .ww H CNOE 2.69.: 0.53"“ ummg ..O r505 95 mm:_w> =< 0.0 H 0.~0 0.0 H 0.00 0.~ H 0.0.. 2.0 H 0.~0 8.0... 008.00 :00 H 0.00 0.2 H 02 DH H 0.00 0.0 H 0.00 0.90300 00.. .600 :02 H 0.0 .0 H 0.20 0... H N~0 0.0 H 3.0 .0323 0:0 .021. :30 H 0~2 :30 H ~02 00.0 H 03 3.0 H 00.. 0.0 .2 0200 £0.03 ...... 00223 ...00.0 H 20.2 .000 H 00.. 00.0 H 03 00.0 H 20.. 0.0 .0 :20. £0.03 00.. 000.24. 100.0 H 00.0 2.00.0 H 00.0 00.0 H D; 00.0 H 00.0 .0 .r 020. £0.03 0:0 000.0>< 00.0 H 00.0 2.0 H .~. 3 00.0 H 30 ~00 H 00.2 .0 .H 020. £0.03 .20... 0.0 H ~.v 0.0 H 2.0 0.0 H ~.0 0.0 H ~.0 :~ 020. an.» .30... 0.0 H 0.0 0.0 H 0.0 0.0 H 0.0 0.0 H 0.0 . A0 020. 0N? .03... 0.0 H 0.0 0.0 H 0.0 0.0 H 0.0 .020 H 0.0 .0 0200 00:0 €00.50 ho ..0nE:z 0.0 H 0.0 0.0 H _..0 0.0 H 0.0 0.0 H 0.0 A0 0200 00:0 0>__ ho .0nE:z 00.0~ H 00.0 00.0~ H 00.02 00.00 H 00.00 E- H v~..0 0.0.5 000...... 00.00.00. 00.0 H 2.. 00.0 H 00.. 00.0 H 0: 00.0 H ~02 0.00 .9: 0900.2 0000 H 00.00 00.~0 H 2.02 030 H 00.00 .0. 20 H ~02 .000 005.3» 2005 2.0H 8.2 30:02 2.0H 20.. arms: E 020 2.0.0; 0005 00.0 H ~10 00.0 H -.~ ~00 H 0: 00.0 H 00.0 0500 20.02. 04.0 H S0~ 00.0 H 00.0~ 00.0 H 20.5 00.0 H 0v.0~ 282.. «0.0 H 00.0~ 00.0 H 0~.0~ 30 H 00- 0.0 H H0- 3020 00.0 H 0v0~ 00.0 H 00.0~ 00.0 H ..0.0~ 00.0 H 0~.0~ 520 00.0 H 00.0~ 20.0 H ~0.0~ ~40 H 0~.0~ 20.0 H 0~.0~ 520 00.0 H 00.0N 00.0 H 00.0.. 00.0 H 00.00 00.0 H 00.00 0:_00E-0.0 20.25 .600 02 0.00 0.00 ~00 03.0008". 2 3 3 2 0.2... 200.30 30:52 00 00 9 t 00.... “00:00.0 .3 .3052 2 0.2 2.0 0 $0200.00. 3.02.8 $03.32 0000000. 50:25 N_. 30 000000000 Ea... 66000.0 30050.8: 5.2. 000000 00.080. 0o 02.00.0200 0200202000 .70 0.00.. 220 ...0.0 v Q: 60.0 v n... ”0:90 .0008 0.0.. 2.0.050 2.0005005 .. 0.00 0.0508 020 .o\o .000 x .0 020 :o 03.0 00:0 .0 .0>E:E..~ 020 :o 02.0 00:0 .0 008:2. 0 00 .000 x .0 020 02.0 00:0 .0 .3050? 020 :0 02:“. 0:0 .0 .3802. H~ 02. 00 000. x00 .0. 8.00.2 M .0 020 00 000 .00.. .0. 0900.2 .000.... me=2 0. 0000.00: .3 0...... .0 £0.03 >08 .0. 00.09.90.” ...N 020 :0 2.0.03 .62. .00... .000 £0.03 >000 0:..0E0.0 0002000 0000.005 .00 .000 x .00.0E0. 0.0.00.0 .0 0053.059. 02. o. 5.5 00.20 00.0.00. E00020 .o .0952. M 0.08 2-0 2000 221 .3. o v 0.: .00. o v 0... 0:90 .0008 E9. 00.0.00 2.0005005 . .00 0.0580 .020 ..x. .03 x 8 020 00 02.0 00:0 .0 .0>.0:0.FN 020 00 02.0 00:0 .0 .3052. 0 ..x. 03 x 8 020 02.0 00:0 .0 .0250? 020 00 02.0 0:0 .0 .000.:2.0 .FN 020 00 £0.03 >000 .00.. 000 0.0.0.5 >000 0020.000 0003.00 0000.00.00 0 .00 02 x 00.000. 00000.0 .0 .00E:0.00:0> 02. 0. 00.0 00.20 00.0.00. E00000 .0 .000.:2.0 .mm H 000.0 0000.0 0.0:00 .000. .0 000.0 0.0 00:.0> _.< .00 H F. 3 m... H 0.00 F... H 0.; F. v H 0. 0v 00.0.0 0.00.00 0.0 H 0.8 0... H 0.3 0.0 H 0.5 3‘ H :0 02.0 .0223 0:0 2.030 0... H 0.8 0... H 0.3 ...0 H 3.0 N... H .00 03.0 .0203 0:0 0.30.0 00.0 H 00F 00.0 H 0.1 00.0 H :1 00.0 H 00F .0 F nwzn: £0.03 0:0 000.0>< :Fnd H 00.0 00.0 H 0N.FF 0..... H mmNF 00.0 H 0m.NF .0. F n20. 2.0.0.... .00... :00 H 0.0 0.0 H 0.0 .0 H 0.0 0.0 H 0.0 :0 020. an.» .00... :0... H 0.0 0.0 H 0.0 .0 H 0.0 0.0 H 0.0 ... 020. on.» .25 00... H 00.0 00.0 H 2.0 E... H 8.0 00.0 H 5.2 00.00 000.5 :00... H R0... 0v... H 00.00 .000 H :00 00.0 H 3.00 00020 0..... H 00.00 00.0 H 00.00 .00 H :00 :3 H 8.00 3020 .3... H 00.8 00.0 H 00.00 .00... H 3.00 .0... H 5.00 .020 .00... H 00.00 00.0 H 0.00 «to H .000 00.0 H 0000 .020 ~00 H 00.9 00.0 H «0.9 00.0 H 00.9 3.0 H 00.3 05.0.0-0... 0.0.2: 230 .000 .00 0.0. 0.00 02.00300 2 N. o. E 0.2... 2:030 .o .3052 0F 0F 9 0F 00.0. 30000.0 .0 000.0:2 0F 0.. F.0 0 02.0.00 008.... N. .00 .mcoumwj0 0008.00.30... .... 0.00.9.0... .2330003020 .s 000000 00.0000. .0 0000000000 0200:0900”. 0-0 0.00... 222 .52.. 33:. 3 cmuzmcm new b99093 MimamficomN >595... .5 53, 83983.8 s 32.85 mm; Ema Sun .9958 Hoza 3 .03 x 8 02¢ co m>__m maaa ho 525553 02a :0 m>__m 83a .0 .wnsszv o ..x. do? x 8 02m m>__m 83a ho 3583:? 021 :o w>=m can “_o 5953 n ..x. .02 x AmmfiEE Emcmma ho 89:35:33 02. 2 5.5 9.35 $563 €9.35 *0 35:55 m .mw H 23.: 23am “mam. 8 :99: 9m 82.? __< 5.0 H 33 5.0 H 8.2. 8.“ H 3% is H 8.3 «I. H 8.8 no.2. .523. mm...” H 8.3 new H 8. 5 :5 H 5.3 «on H 83 «3 H 8.8 use .32..» ea :2...“ S.» H 8.8 5.» H 2. a 8.” H 8% :8 H 8.5 t.» H 8.8 ass .33...» 9:. 283. So H 84 8.0 H m: 8.0 H 93 85 H o: 8.0 H v? a .. oz... 292., ea $202 56 H 3d 3.0 H m; Be H 8.: «3 H 8.2 NS H 8. : a .52“: £925 5:: No H 3 3 H 3 3 H 3 3 H 3 9o H 3 E“ nzn: on» an... E H to 3 H 3 2. H No 3 H 3 ed H 2 3 oz... can as... md H 3 9o H I no H 3 ed H o.» no H E 3 oza. 3.» 3:: QE 2: m3 8. new «£2.83. 3 5 a S S 22:. 233.. 33:2 2 3 3 2 NH 8.... 2.29.... .o .352 S 3 ...... to a $529.55. £2£50 .5398. 3:95 N? >8 .mcozflwmm E9; EQmEmm 5? ommmzwm mmfiEmz ho 8cm§otwa w>=o=uoawm .m-o 29m... 223 6.5 DISCUSSION The present study was the third study of a series of experiments to examine effects of gestational and lactational exposure to EEMs (DES, GEN, and EE2, respectively) on reproduction in mice. Average pup weights on PND 1 were decreased in the high doses (1 .O and 10 pglkg bwld) of EE2, as were pup survival rates on PND 4 and PND 21 in the high dose group compared to the control group. These results suggest that maternal exposure to EE2 during the gestational and lactational periods may alter body weights and survival rates of offspring at an early stage of life. Gestational exposure to EEMs at low-doses may alter fecundity. In the present study, we observed an approximate 16% decrease in fecundity in female mice exposed to 10 pg EE2/kg bwld. This result is consistent with our previous DES study in that the high dose (10 pglkg bw/d) of DES resulted in a 34% decrease in fecundity, whereas the phytoestrogen GEN did not affect fecundity (1,, 17, 18). The decrease in fecundity induced by EEM exposure during gestation may be due to an altered uterine environment. It has been demonstrated that estrogen administration (5 mglanimal by intramuscular injection on gestational day 9 and 10), before the conceptus secretes estrogen on gestational day 12, alters the pattern of insulin-like growth factor (IGF) gene expression through the nuclear factor kappa B (NF-KB) system. This desynchronizes the uterine environment for conceptus implantation resulting in embryonic loss in pigs (19). Yasuda et al. (15) demonstrated that daily oral gavage of mice with EE2 (0.02, 0.2, and 2.0 mg/kg bw) from GD 11 to GD 17 increased the number of fetal 224 deaths in a dose-related manner. These results suggest that gestational exposure to EEMs may lead to embryonic loss due to alteration of the uterine environment. A possible reason for different effects between synthetic estrogens (DES and EE2) and phytoestrogens (GEN) on fecundity could be that estrogenic transactivation of GEN is 104 to 105 times less potent in the luciferase reporter gene assay and in the cell proliferation assay in estrogen response cell lines (20, 21). Litter size on PND O in the 1.0 pg EE2/kg bwld treatment group was increased, while it was decreased in the 10 pg DES/kg bw/d treatment group (P < 0.01) (18). The increase in litter size in the 1.0 pg EE2/kg bwld treatment group may be a result of increased fetal retention. An increase in litter size after gestational exposure to EE2 was also reported by Edgren et al. (22) in rats treated with 1.25 pglkg maternal bwld. Litter weight on PND 1 was significantly decreased in the 10 pg DES/kg treatment group (P < 0.05) without changing average pup weight on PND 1 (1 , 18). Gestational and lactational exposure to EE2 decreased average pup weights on PND 1, while exposure to DES and GEN treatment did not. Decreased perinatal body weights corresponded to decreased fetal body weights in primates maternally exposed to estrogen due to reduced placental blood flow during late pregnancy (23, 24). Yasuda et al. (15) demonstrated that daily oral gavage of mice with EE2 (0.02, 0.2, and 2.0 mglkg bw) from GD 11 to GD 17 caused a dose-dependent decreased in average fetal body weight. 225 The 10 pg EE2/kg bw/d treatment decreased survival at 4 and 21 days of age, while DES and GEN treatment did not affect pup’s survival (1, 17, 18). In the present study, body weight-adjusted maternal liver weights were significantly decreased in the high EE2 treatment group (Table 6-1). Pup survival rate was positively correlated with the dam’s liver weight and litter size PND 0 (P < 0.05) and negatively correlated with thymus weight (P < 0.01). These results are consistent with other studies that indicate that estrogen induces cytochrome P450 in the liver, thus increasing liver weight (25, 26). Therefore, the hepatic metabolism of estrogenic compounds by the dam could enhance the pup’s survival rate. Contrary to the protective effect of the dam’s hepatic metabolism of estrogenic compounds, estrogenic compounds suppress the dam’s immune system and increase thymus weight (27). Interestingly, the percentage of males was not changed in the EE2 and GEN treatment groups, but it was slightly greater compared to the control group in the 1.0 pg DES/kg bwld treatment group (P < 0.05) (18). There was a large and significant decrease (32%; P < 0.05) in the percentage of males in the 10 pg DES/kg treatment group. This result is consistent with a study that indicated that the sex ratio at birth-was decreased at doses 0.02 and 0.2 pg DES/kg bw. Administered from GD 11 to 17 in CF-1 mice (P < 0.01) (28). Furthermore, it is known that EEMs have estrogenic and antiestrogenic properties, thus altering the sex ratio via changing parental biological factors. For example, high serum concentrations of 2,3,7,8-tetrachlorochlibenzo-p-dioxin (TCDD) in fathers exposed during the Seveso accident in 1976 were significantly associated with a 226 decline in sex ratio up to eight years after the incident in two areas of highest contamination (Meda and Seveso) (P = 0.008) (29). The effect of TCDD on the sex ratio was detected at serum concentrations lower than 20 ng/kg bw in the fathers who were exposed to TCDD (30). However, since pregnant females in our studies and in the study of Palanza et al. (28) were randomly assigned to treatment groups and administered compounds after conception, this precludes a direct effect on the fertilization of eggs by XX- or XY-bearing sperm. However, published studies addressing possible association or effects of EEMs on mammalian sex ratios have still produced contradictory results because of unknown mechanisms. Further studies are needed to confirm the effects of EEMs on sex-selection and associated biological mechanisms. In conclusion, the present study indicates that gestational and lactational exposure of female mice to 10 pg EE2/kg bwld leads to a decrease in offspring body weight and survival. Considering the environmental exposure concentrations of EE2 through both drinking water and the diet, further studies are needed to investigate potential adverse effects of EE2 on reproduction in the human and wildlife. Acknowledgements This work was supported by a grant from US EPA (R827-402-01-0) to KC, PMS, and T2. 227 6.6 REFERENCES 1. Fielden MR, Halgren RG, Fong CJ, Staub C, Johnson L, Chou K, Zacharewski TR 2002 Gestational and lactational exposure of male mice to diethylstilbestrol causes long-term effects on the testis, sperm fertilizing ability in vitro, and testicular gene expression. Endocrinology 143:3044- 3059 Thayer KA, Ruhlen RL, Howdeshell KL, Buchanan DL, Cooke PS, Preziosi D, Welshons WV, Haseman J, Vom Saal FS 2001 Altered prostate growth and daily sperm production in male mice exposed prenatally to subclinical doses of 17alpha-ethinyl oestradiol. Hum Reprod 162988-996 Maggs JL, Grimmer SF, Orme ML, Breckenridge AM, Park BK, Gilmore IT 1983 The biliary and urinary metabolites of [3H]17 alpha- ethynylestradiol in women. Xenobiotica 13:421-431 Williams MC, Goldzieher JW 1980 Chromatographic patterns of urinary ethynyl estrogen metabolites in various populations. Steroids 36:255-282 Ranney RE 1977 Comparative metabolism of 17alpha-ethynyl steroids used in oral contraceptives. J Toxicol Environ Health 3:139-166 Donn J, Mendoza M, Pritchard J 2008 AP Probe Finds Drugs in Drinking Water. In: The Associated Press. New York Aheme GW, Briggs R 1989 The relevance of the presence of certain synthetic steroids in the aquatic environment. Pharm Pharmacol 41:735- 736 Tabak HH, Bloomhuff RN, Bunch RL 1981 Steroid hormones as water pollutants, ll. Studies on the persistence and stability of natural urinary and synthetic ovulation-inhibiting hormones in untreated and treated wastewaters. Dev lnd Microbiol 22:497-519 Williams RJ, Johnson AC, Smith JJ, Kanda R 2003 Steroid estrogens profiles along river stretches arising from sewage treatment works discharges. Environ Sci Technol 37:1744-1750 228 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Roofer P, Snyder S, Zegers RE, Rexing DJ, Fronk JL 2000 Endocrine- disrupting chemicals in a source water. J AWWA 92:52-58 Fent K, Weston AA, Caminada D 2006 Ecotoxicology of human pharmaceuticals. Aquat Toxicol 76:122-159 US EPA 2000 Estimated per capita water ingestion in the United States. In. Washington DC: US EPA, Office of Water 1301; 40 US EPA 2001 National primary drinking water regulations; arsenic and clarifications to compliance and new source contaminants monitoring; final rule. In. Washington DC: US EPA Arredondo F, Noble LS 2006 Endocrinology of recurrent pregnancy loss. Semin Reprod Med 24:33-39 Yasuda Y, Kihara T, Nishimura H 1981 Effect of ethinyl estradiol on development of mouse fetuses. Teratology 23:233—239 Odum J, Tinwell H, Jones K, Van Miller JP, Joiner RL, Tobin G, Kawasaki H, Deghenghi R, Ashby J 2001 Effect of rodent diets on the sexual development of the rat. Toxicol Sci 61 :1 15-127 Fielden MR, Samy SM, Chou KC, Zacharewski TR 2003 Effect of human dietary exposure levels of genistein during gestation and lactation on long-term reproductive development and sperm quality in mice. Food Chem Toxicol 41 :447-454 Fielden MR 2002 Reprodictive and genomic effects of gestational and lactational exposure to estrogenic endocrine disruptors in male mice. In: Department of biochemistry and molecular biology. East Lansing: Michigan state university; 3-57, 99-138 Geisert RD, Ross JW, Ashworth MD, White FJ, Johnson GA, DeSiIva U 2006 Maternal recognition of pregnancy signal or endocrine disruptor“. the two faces of oestrogen during establishment of pregnancy in the pig. Reprod Suppl 62:131-145 229 20. 21. 22. 23. 24. 25. 26. 27. 28. Shapiro S, Slone D 1979 The effects of exogenous female hormones on the fetus. Epidemiol Rev 1:110-123 Mussman HC 1975 Drug and chemical residues in domestic animals. Fed Proc 34:197-201 Edgren RA, Clancy DP 1968 The effects of norgestrel, ethinyl estradiol, and their combination (ovral) on the young of female rats treated during pregnancy. Int J Fertil 13:209-214 Beydoun SN, Abdul-Karim RW, Haviland ME 1974 The regulatory effect of estrogens on fetal growth. Ill. Placental deoxyribonucleic acid, ribonucleic acid, and proteins. Am J Obstet Gynecol 120:918-921 Shepherd RW, Stanczyk FZ, Bethea CL, Novy MJ 1992 Fetal and maternal endocrine responses to reduced uteroplacental blood flow. J Clin Endocrinol Metab 75:301-307 Sundstrom SA, Sinclair JF, Smith EL, Sinclair PR 1988 Effect of 17 alpha-ethynylestradiol on the induction of cytochrome P-450 by 3- methylcholanthrene in cultured chick embryo hepatocytes. Biochem Phannacol 37:1003—1008 Jager W, Correia MA, Bomheim LM, Mahnke A, Hanstein WG, Xue L, Benet L2 1999 Ethynylestradiol-mediated induction of hepatic CYP3A9 in female rats: implication for cyclosporine metabolism. Drug Metab Dispos 27:1505-1511 Salem ML, Matsuzaki G, Kishihara K, Madkour GA, Nomoto K 2000 Beta-estradiol suppresses T cell-mediated delayed-type hypersensitivity through suppression of antigen-presenting cell function and Th1 induction. Int Arch Allergy Immunol 121:161-169 Palanza P, Parmigiani S, vom Saal FS 2001 Effects of prenatal exposure to low doses of diethylstilbestrol, o,p'DDT, and methoxychlor on postnatal growth and neurobehavioral development in male and female mice. Horm Behav 40:252-265 230 29. 30. Figa-Talamanca I, Tarquini M, Lauria L 2003 Is it possible to use sex ratio at birth as indicator of the presence of endocrine disrupters in environmental pollution?]. G Ital Med Lav Ergon 25:52-53 Mocarelli P, Gerthoux PM, Ferrari E, Patterson DGJ, Kieszak SM, Brambilla P, Vincoli N, Signorini S, Tramacere P, Carreri V, Sampson EJ, Turner WE, Needham LL 2000 Paternal concentrations of dioxin and sex ratio of offspring. Lancet 355:1858—1863 231 CHAPTER 7 7.1 CONCLUSIONS The aims of the present study was to examine and to compare the effects of maternally administrated three different EEMs, DES, EE2, and GEN, at human exposure related concentration during in utero and lactation on sperm production, sperm quality, and sperm fertilizing ability in male offspring (F-1) and on superovulation and egg fertilizing ability in female offspring (F-1). This study demonstrated that in utero and lactational exposure to EEMs at human exposure related concentrations may have persistent or latent effects in sperm production in male offspring. The high doses (10 pglkg) of DES and EE2 decrease sperm production in middle-aged male offspring. This result suggests . that in utero and lactational exposure to synthetic and potent EEMs may decrease the total population of germ cells through affecting directly on the fetal testis and/or indirectly on the fetal hypothalamic-pituitary axis. On the other hand, in utero and lactational exposure to the EEMs could persistently alter ovarian function. The low dose (0.1 pglkg) of DES increases the number of ovulated eggs in postpubertal female offspring and the high dose (10 pglkg) of EE2 decreases fertilizing ability of eggs in peripubertal female offspring. However, in utero and lactational exposure to a phytoestrogen, GEN at human exposure related concentration may have no health risk to reproduction. Additionally, this study demonstrated that gestational and lactational exposure to EEMs at human exposure related concentrations decreases body weights of offspring from early postnatal period through lactation. Taken together, 232 our results indicate that potential effects on reproduction of EEMs may not be predictive as results of different adsorption, metabolism, disposition, excretion, potency, duration of exposure and dose of each EEMs. Collectively, it is important to consider and to examine potential effects of an EEM both in vivo as well as in vitro studies. In vivo assessment of EEMs involves multigenerational studies, could provide the entire facts essential for health hazard assessment required because degree of adverse effects would be different with age as well as effects of EEMs may be persistent and/or latent. 233