in.” . $3. . EH Lug-$1.... 31:.” . 4.1“er s2! 9&1. n ,2! ? 1!! I w 6-. P" r. a... G! ...um.mm.fi..a..fi . . . lo. 7 . mWMWM: ‘ . Lie... Ewan”... 4%...” A ‘ A I .\ 3.50451?! A «an wwfiuugr. 1.: d! .15 1.... . Ixxlkcg. .nflnermq: .. JR. .. 3. a i A... i {fluid} :54. ‘ lit}... 4 . lit .. vL 0x4. :25?! male 200? This is to certify that the dissertation entitled ALLERGIC DISORDERS AND SEX HORMONE DISRUPTERS presented by KEVIN ROYD BROOKS has been accepted towards fulfillment of the requirements for the Doctoral degree in Epidemiology O m L__,, Major Professor’s Signature January , 2007 Date MSU is an Affirmative Action/Equal Opportunity Institution ._.—.-.-._._.__._._.__-.-.__-.-._.___,_._._,-_-._._,_._.-._,_._.-.-.-._.-.-.-._. -- -—._----.—._._-—._._.-—-‘—.-.--nuu LiBRARY Michigan State University PLACE lN 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 6/07 plelRCIDateDue.indd-p.1 EJ/f ALLERGIC DISORDERS AND SEX HORMONE DISRUPTERS By Kevin Royd Brooks A DISSERTATION Submitted to Michigan State University In partial fulfillment of the requirements for the degree of DOCTORAL DEGREE DEPARTMENT OF EPIDEMIOLOGY 2007 ABSTRACT ALLERGIC DISORDERS AND SEX HORMONE DISRUPTERS By Kevin Royd Brooks Allergic disorders are among the most disruptive of childhood. Further, the prevalence and severity of these disorders are increasing. Great strides have been made toward disentangling the etiology of allergies; however a clear understanding is still elusive. Recently, the role of sex hormone disrupters (SHD) in triggering childhood allergies have gained increased attention. Specifically, halogenated organochlorine compounds (HOCs) such as dichlorodiphenyl dichloroethene (DDE) and polychlorinated biphenyls (PCBs) in addition to synthetic hormones such as estrogen and progesterone in the form of contraception have been implicated. We investigated the role of SHD in the etiology of allergic diseases. First we investigated the association between placental p,p’-DDE and cord blood immune markers. p,p'-DDE concentration and cord plasma interleukin (IL)-13, lL-4 and INF-y were determined in 19 neonates from the ongoing Pregnancy Environment and Child Health (PEACH) study. Results showed increased placental p,p’-DDE to be associated with a statistically significant increase in cord plasma interleukin (IL)-13. Furthermore, both cord plasma IL-4/lNF-y and lL-13/lNF-y ratios were significantly positively associated with placental p,p’-DDE concentration. (‘2 F . Second, we estimated associations between maternal oral contraceptive (OCp) use and humoral immune markers in offspring. We hypothesized that maternal OC use increases humoral immune markers of allergy in offspring. Toward this end, data from a cross-sectional investigation including 334 mother child (aged 7—10 years) pairs from Hesse, Germany were used. Results showed that female offspring of mothers who used 00 had significantly lower (p<0.05): lgA (123.43 mg/dL vs. 150.52 mg/dL), and lgE (22.96 kU/L vs. 50.83 kU/L) levels as well as basophilic surface lgE counts (783 vs. 842), compared to those of mothers who did not use 00. For male offspring, statistical significance was only seen in an increased number of basophilic surface lgE (911 vs. 876). Third, using data from a 1986—1987 survey of 11-12 years old Jamaican children, an investigation of the association between maternal OCp use before pregnancy and childhood allergic phenotypes suggest that offspring’s asthma or wheezing may be related to 00p use (aOR: 1.81; 95% CI (1 .25- 2.61)). Fourth, the relationship between childhood DDE and immune markers was quantified using data from the above-mentioned Germany cohort. Higher DDE concentration was significantly related to increased serum lgE levels, lgE count on basophils, and the reduced eosinophilic granula. These findings suggest that SHD may be important in the in etiology of childhood allergy. Of note, SHD may play a role in the prenatal priming of childhood allergy. Exploring the specific immune mechanism behind the possible effect of SHD on allergy may be worthwhile. DEDICATIONS This work is especially dedicated first to my wife Valrie, daughter Nessia, son Kevin and mother Patricia. I also dedicate this monograph to my very promising nephew Jevon 0. Brooks. iv ACKNOWLEDGEMENTS The completion of this work is the result of a team of enthusiastic and devoted individuals. Sincere gratitude to my advisor, Dr. Wilfried Karmaus, for his prudence, and moral support in the process of completing this work. I also thank the other members of my committee, Drs. Joseph Gardiner, David Todem and Venugopal Gangur for taking time from their busy schedule to provide unswerving intellectual support. For their many years of encouragement, example and technical support, I thank Drs. Maureen Samms-Vaughn and Rainford Wilks. It would be remiss of me not to acknowledge my collegues Kathy, Ali and Azfar for their empathy and friendship. This work would not be possible without the contribution of all study participants. I thank you all for your time and patience. TABLE OF CONTENTS LIST OF TABLES ............................................................................................. Vii LIST OF FIGURES ........................................................................................... ix OVERVIEW ......................................................................................................... 1 CHAPTER1 ALLERGIC DISORDERS AND SEX HORMONES DISRUPTERS ................... .5 11Allergy ..........6 1.2Asthma .......................................................................................... 7 1.3Sex hormones disrupters 21 1.4Time-windowofexposure.. 28 1. 5Components of the conceptual model ......29 1. 6Literature review of components comprising the conceptual model ...... 31 References” .36 CHAPTER 2 BACKGROUND AND METHODS FOR THE RESPECTIVE STUDIES ........... .50 2.1 The PEACH study. .. .51 2. 2 Child Health and Environment Cohort Study Germany ..................... 59 2. 3 The Jamaican Perinatal Morbidity, Mortality Survey. .. .61 References. . .. . . 65 CHAPTER3 RESULTS ............................................................................................. 66 Component one ................................................................................ 68 References ....................................................................................... .78 Component two 79 References ...................................................................................... 92 Component three .102 References ................................................................................... .124 Component four .........136 References .................................................................................... 158 CHAPTER4 Future work 174 References 176 vi CO In L T’ -r LIST OF TABLES COMPONENT ONE In utero exposure to SHD affects perinatal biomarkers of allergy Table 1. Descriptive characteristics of the cohort .................................... 76 Table 2. Association (betas ((3) and standard errors (SE)) between p,p'-DDE concentrations and lL—4, lL-13, INF-y, and the ratio of lL-4/ INF-y, and lL-13/ INF-y ................................................... 77 COMPONENT TWO In utero SHD affects postnatal biomarkers of allergy Table 1. Descriptive characteristics of the children’s cohort ............... 95.96 Table 2: Gender specific geometric mean and 5-, 95 values for immunoglobulins by covriates. ................................................. 97 Table 3: Gender specific geometric mean and 5-, 95 values for white blood cell, eosinophilic characteristics, and basophilic surface lgE by covriates. ...................................................... 98,99 Table 4: Maternal oral contraceptive use by immunoglobulins .............. 100 Table 5: Maternal oral contraceptive use by white blood cell, eosinophilic characteristics, and basophilic surface lgE .......... 101 COMPONENT THREE In utero exposure to SHD affects allergic outcomes in infancy Table 1: Comparison characteristics of exposure variables in baseline and linked datasets ................................................................. 129 Table 2: Proportions of simultaneous occurrence of the different atopic manifestations ............................................................... 130 Table 3: Proportion of atopic disorders in relation to their in utero risk factors .............................................................................. 131 Table 4: Proportion of atopic disorders in relation to their perinatal potential risk factors ........................................................ 132, 133 Table 5: Odds ratios and 95% confidence intervals (CI) of antenatal and perinatal risk factors for asthma or wheezing, and frequent nighttime or early morning cough .............................. 134 vii Table 6. LIST or TABLES (CONTINUED) Odds ratios and 95% confidence intervals (CI) of antenatal and perinatal risk factors for eczema, and hay fever or sinus problem or some other allergy .............................................. 135 COMPONENT FOUR Postnatal exposure to SHD affects biomarkers of allergy in infancy Table 1: Descriptive characteristics of the study cohort. ....................... 162 Table 2: Geometric mean and 5-, 95% values for whole blood OC and Pb by covariates ............................................................... 163 Table 3: Spearman correlation coefficients between organochlorine compounds and their geometric means. ................................. 164 Table 4: White blood cell, eosinophilic characteristics, and basophilic surface lgE by DC and Pb (geometric mean) .......................... 165 Table 5: Lymphocyte phenotypes by whole blood DDE, PCBs, HCB, y-HCH and Pb concentration (geometric mean) ...................... 166 Table 6: lmmunoglobulins by whole blood DDE, PCBs, HCB, y—HCH and Pb concentration in children (geometric mean) ................ 167 viii LIST OF FIGURES Figure 1: Sex Hormone Disrupters and Childhood Allergy — Conceptual model .................................................................... 2 Figure 2: Various definitions of asthma ..................................................... 7 Figure 3: Theoretical immune responses .................................................. 8 Figure 4: Map of Michigan showing PEACH Study Site .......................... 52 Figure 5: The Rhine Valley ...................................................................... 59 Figure 6: Jamaica: parishes, regional administrative areas and Distribution of hospitals ............................................................ 62 Figure 7: Diagrammatic representation of the breastfeeding, childhood exposures and immune markers associations ....................... 146 Figure 8: The combined effect of increasing DDE and lead (Pb) on lgE serum levels .................................................................... 150 ix (I) I) (D OVERVIEW Childhood allergies are among the most vexing diseases: purported to be responsible for enormous personal and public health burden. In spite of numerous efforts, the etiology of these diseases continues to evade researchers. The purpose of this monograph is to investigate the role of sex hormone disrupters (SHD) in the etiology of allergic diseases. Specifically, this work explores the prenatal origins of childhood allergies (in utero programming) by probing the possible role of SHD in this process. Next, the association between exposure to SHD and biomarkers of allergic susceptibility in infancy is assessed. The quest for appropriate pieces of the SHD-allergic disorders puzzle started with the development of a conceptual model (Figure 1; more details provided in the “Components of the conceptual model” section). This model depicts areas that speak to the purpose of epidemiology, which is to provide clues to the cause of a health related problem. An inventory of suitable data was then undertaken and the following four components developed. 1. In utero exposure to SHD affects perinatal biomarkers of allergy. 2. In utero SHD affects postnatal biomarkers of allergy. 3. In utero exposure to SHD affects allergic outcomes in infancy. 4. Postnatal exposure to SHD affects biomarkers of allergy in infancy. "Y! Chil: b... .VfV- 5‘ (D .D-PC Vi; . Serum rogesterone markers In utero and childhood immune development Pregnancy (in utero environment) Childhood Cord plasma IL-13, lL-4 & INF-y Placental OC Immune markers * Maternal oral contraceptive use before pregnancy Figure 1 Sex Hormone Disrupters and Childhood Allergy - Conceptual model Data from three cohorts were used to address the above components. These cohorts are: the ongoing Pregnancy Environment and Child Health (PEACH) study from which information on placental dichlorodiphenyl dichloroethene (DDE) concentration and cord plasma interleukin (lL)-13 were derived (component 1). Data on maternal oral contraceptive use and on childhood biomarkers of allergic susceptibility (lmmunoglobulins (lgs), white blood cell, eosinophils, and basophilic surface lgE) were obtained from the Child Health and Environment Cohort Study conducted in 1995 (component 2). This study also provided data on childhood organochlorine exposure and the above mentioned childhood biomarkers of allergic susceptibility (component 4). A geographic sub-cohort from the Jamaican Perinatal Morbidity, Mortality Survey conducted in 1986-1987 provided data on maternal oral contraceptive use before pregnancy and childhood biomarkers of effect such as asthma, wheezing and eczema (component 3). Detailed description for each respective study is presented in Chapter 2. Ethical approval for this work was obtained from the Institutional Review Board of the Michigan State University. This first chapter presents the definition and immunology of asthma, its prevalence and burden, risk factors and the possible effects of various SHD (sex hormone disrupters) on childhood allergy. Next, the importance of time windows of exposures and the main concepts that may explain the increase in the prevalence of asthma are introduced. Then the conceptual model with its four components (see above) followed by their respective backgrounds are presented. Chapter 2 - ‘Methods’ - describes different aspects of the materials and methods not discussed comprehensively in the manuscripts to be found in Chapter 3 - ‘Results’. Additional details on materials and methods can be found in the respective manuscripts in Chapter 3. Furthermore, Chapter 3 showcases four manuscripts; two published, one submitted and one in preparation for publication. These manuscripts provide findings that address the following research questions: 1. Is there evidence for the immunemodulation of cord immune markers of allergy by in utero exposure to sex hormone disrupters? 2. Is maternal oral contraceptive use (a proxy for in utero estrogen/progesterone exposure) associated with immune markers of allergy in the offspring? 3. Is maternal oral contraceptive use a risk factor for allergic diseases in offspring? 4. Does serum organochlorine alters immune markers in children? The fourth and last chapter of this monograph, ‘Discussion and Conclusion’, discusses and present concluding remarks of the results found in chapter 3. Practical public health issues are here presented along with future scientific steps toward a better understanding and early life prevention of childhood asthma. CHAPTER I ALLERGIC DISORDERS AND SEX HORMONE DISRUPTERS else a 1.1 ALLERGY Allergy is a hypersensitivity reaction initiated by immunologic mechanismsz. Allergy can be antibody- or cell-mediated. In most patients, the antibody typically responsible for an allergic reaction belongs to the lgE isotype and these patients may be said to suffer from lgE-mediated allergy. In cell-mediated allergy, as in allergic contact dermatitis, immunologically sensitized lymphocytes play a major role 2. Allergic disease represents a dysregulation of the intact immune system. Box 1 Definitions ALLERGY is a hypersensitivity reaction initiated by immunologic mechanisms 2. The term allergy is also used to denote allergic disorders such as asthma and allergic rhinitis . ASTHMA is a chronic inflammatory disorder of the lungs that is characterized by a reversible obstruction of the airways ATOPY is a personal and/or familial tendency to become sensitized and produce lgE antibodies in response to ordinary exposures t; allergens. As a consequence, asthma, rhinoconjunctivitis, or eczema may occur . WHEEZE is a high pitched sound made when breathing, which probably results from turbulence through narrowed tubes SENSITIZATION is the state where, following exposure to allergens, the immune system is primed to produce IgE antibodies specific to that allergen. Patients usually develop a hypersensitive immune response to environmental antigens, resulting in local tissue damage and Inflammation . Diseases such as allergic rhinitis, dermatitis, and asthma represent a significant personal and public health burden. Approximately 25% of all children experience at least one allergic manifestation 8. Asthma however, has received the most attention seeing that it is the most common chronic disease of childhood and is considered the most debilitating 9. 1.2 Asthma Definition and immunology Definition A universally accepted definition of asthma has been frustrated by a lack of understanding of the mechanisms involved in the disease. The American Thoracic Society defines asthma as "a disease characterized by an increased responsiveness of the airways (bronchial hyperresponsiveness) to various stimuli and manifested by slowing of forced expiration (due to narrowing of the airways) which changes in severity either spontaneously or as a result of therapy". This narrowing of the airways ultimately produces “wheeze”, which is considered to be the necessary clinical criterion for asthma. Of note; not all that wheezes is 10 asthma Asthma Recently, the Nomenclature , , Allergic asthma Nonallergic asthma ReVIew Committee of the . . 2 _ - _ _ . , WOFId Allergy Organization lgE mediated asthma Non lgE mediated allergic asthma Figure 2 various definitions of asthma proposed the use of the term allergic asthma as the basic term for asthma mediated by immunologic mechanisms (Figure 2). The organization further suggests that when there are indications of lgE-mediated mechanisms, the term should be lgE-mediated Ii -I\ .Ih—P ‘0. “'5 ”P- S/Pt: asthma 2. In addition to lgE, several other immunologic factors such as lL-4 and lL-13 are involved in the etiology of asthma. Immunology For the purpose of this work, the immunology of asthma is here discussed within the framework of the type 1 and 2 helper T cells (Th1/Th2) model. This model/paradigm was born out of a series of experiments developed by Tom Wegmann 11 as he perused the placenta in search of clues that migt explain spontaneous abortion. To date, mechanistic explanations for allergy have often invoked the Th1/Th2 paradigm ”'13 ”'15. Theoretically, the immune system ‘idles' in a Th0 or na'i've state until it is required to change response by some external stimuli (Figure 3). These stimuli may be in the form of environmental antigens in which case non-allergic Th1 response is initiated ieading ICO Adopted from Whitacre et al. Science 1999; Non-allergic state . Non- Allergic secretion of lgG W a immune markers / lFN-y, lL-2, etc. antibodies, removal of the allergen, and the Antigen -—> Naive state Allergic immune suppression of lgE ”by," markers: , lL-4, lL-13 etc. Allergic state . 16,17 . . SyntheSIS - In add'tlon Figure 3 Theoretical immune responses II AL to antigens, hormones may cause a shift in immune response. However, it is believed that sex hormones such as progesterone and high concentration of estrogen may result in an allergic — Th2 type response 1 (Figure 3). There is a general consencious in the literature that a shift to a Th2 polarize state occurs during dramatic changes in immune responses associated with pregnancy. To prevent termination of the fetus (removal of the allergen), the feto- maternal immune system shifts to a Th2 direction 18’19. This Th2 shift begins with the secretion of human chorionic gonadotropin, which in turn stimulates progesterone production. Progesterone then promotes the secretion of Th2 cytokines over the secretion of phagocytic 20 Th1 cytokines. In other words, progesterone is believed to prime the immature fetal immune system towards an allergic direction which may be manifed as allergic diseases (such as asthma) during infancy. Fundamentally, the underlying problem in asthma Box 2 Select markers of immune response appears to be immunologic with children in the early Th1 response: lgG - immunoglobulin G sta es of asthma showin si ns of excessive g g g lFN-y - interferon gamma imflamation in their ainivays. While chemokines (a "=2 - interleukin 2 IgA - immunoglobulin A subset of cytokines that chemically attracts immune ,gM _ immunoglobunn M cells) are important in the recruitment of imflamatory Th2 res onse, lgE — immunoglobulin E lL-4 - interleukin 4 those producing Th2 cytokines — are responsible for lL-13 - interleukin 13 cells to the airway, the lymphocytes — particularly directing and maintaining this inflammatory process 21. Therefore, the cardinal phenotypic features of allergy are associated with the production of the Th2 cytokines lL-4 and IL-13 which are central to the synthesis of IgE; the forerunner of allergic manifestations (Box 2). Of note, recent reports indicate that cord blood mononuclear cells (CBMC) produce lL-13 and that cord blood lL-13 is associated 22,2 with allergy in childhood 3. Further evidence suggests that lL-13 can participate in allergies independent of lL-4 24. As mentioned earlier, another important marker of a Th2 response is lgE, which is central to the etiology of allergic diseases such as allergic asthma. In allergic individuals, specific lgE is bound to high affinity receptors on mast cells and is cross-linked by antigen; mast cell activation/degranulation occurs and sets in motion a cascade of events resulting in the clinical manifestations of allergic disease 25. The interaction of IgE and antigen results in an immediate hypersensitivity reaction may be responsible for the classic asthma symptoms exhibited during acute exacerbation, for example, mucosal edema, mucus production, and smooth muscle constriction. Eventually, these and other reaction cascades can induce the production of cells responsible for airway inflammation that underlies allergic asthma 26. Nonallergic-asthma, conversely, manifests all the clinical feature of allergic asthma without the lgE mediated immunologic aspect. At a population level, it is estimated that 50% of all asthma cases have 10 .5 ad an allergic etiology 8'27. Regarding Non-IgE-mediated asthma (Figure 2) this subtype may be characterized by the following four criteria: 1) clinical phenotype of lgE mediated asthma; 2) absence of other allergic diseases such as allergic dermatitis and rhinoconjunctivitis; 3) negative sensitization (Box 1) to common inhalant and food allergens ; and 4) normal total serum IgE levels. In contrast to a Th2 response, a Th1 response leads to secretion of IgG antibodies, removal of the allergen, and the suppression of lgE synthesis 28-30. Other immune markers are associated with a Th1 type response (Box 2). In general, Th1-responses are highly protective against infections mounted by the majority of microbes. It is widely accepted that changes leading to allergic disorders such as allergic asthma begin in childhood, thus it is important to study the natural history of childhood asthma in an effort to identify risk factors associated with its development. Asthma in children Asthma may develop during the first few months of life, but it is often difficult to make a definite diagnosis until the child is older 4. Approximately 30 % of asthmatic patients are symptomatic by one year of age, 80-90 % have their first symptoms before four to five year of age. The majority of affected children have Il only occasional attacks of slight to moderate severity, which are managed with relative ease. A minority experience severe, intractable asthma, usually perennial rather than seasonal. The relationship of age of onset to prognosis is uncertain. Most severely affected children have an onset of wheezing during the first year of life and a family history of asthma and other allergic diseases (particularly atopic dermatitis) 31 Prevalence and burden of asthma . . 32 Asthma remains an enormous personal and public health concern . Asthma symptoms in children vary in prevalence from 0% to 35% in different populations 33. The prevalence of asthma symptoms is on average 14% among children aged 6-7 years and 18% among those aged 13-14 years 33. Overall, the . . . . . . 4,34 prevalence and severity of asthma and other allergic diseases is increasmg Globally, approximately 300 million people currently have asthma and it is estimated that there may be an additional 100 million more asthmatics by 2025 9. Furthermore, asthma accounts for about one in 250 deaths worldwide. In the United States (US), asthma affects more than 17 million individuals 35 and results in approximately 10 million visits to physicians, 2 million emergency room visits and approximately 7,000 deaths annually 36. The annual cost of caring for 12 asthmatics exceeds six billion dollars per year in the US, and the worldwide market for asthma medication is currently valued at 5.5 billion dollars each year 37. It is noteworthy that the cost of asthma should be divided into direct and indirect cost. Direct costs include the costs of medication, medical bills, and documented episodes of health service utilization such as clinic visits and hospital admissions. Indirect costs include the adverse economic impact of the disease on an individual, family, and society. This includes the “cost" of premature mortality and productivity loss. In the US, the cost of medical treatments for asthma is on average 10% of total family income 38 39. The economic cost of asthma is alarming both in terms of direct medical costs (drugs, hospitalization) and indirect medical costs (time lost from work and premature death). The number of disability-adjusted life years (DALYs) lost due to asthma worldwide is currently estimated about 15 million/year, and asthma accounts for around 1% of all DALYs lost, reflecting the high prevalence and severity of asthma 9. Asthma in general is incapacitating and interferes with school attendance, play activity, and day-to-day functioning. In a 1988 National Health Interview Survey, children with asthma missed 10.1 million days from school (2 times the number of days missed by children without asthma), and had 12.9 million contacts with medical doctors and 200,000 hospitalizations 40. In the same survey, it was determined that almost 30% of children with asthma experienced some limitation of physical activity, compared to only 5% of children without asthma 41. Risk factors for asthma Risk factors for asthma may be classified as host factors that predispose individuals to developing asthma, and environmental factors that influence the susceptibility to the development of asthma in predisposed individuals, precipitate asthma exacerbations, and/or cause symptoms to persist 4. Host factors These include sex and race along with the genetic predisposition to the development of either asthma or allergic sensitization. Sex Sex biases in the susceptibility to, and severity of, autoimmune and allergic . . 7 . diseases are well-recognized . Preadolescent boys have about 1.5 times the . 42-44 . asthma prevalence of girls . However, after adolescence, the sex difference is reversed 45. Some authors suggest that the increased risk for males in childhood is probably related to narrower airways, increased airway tone 46—48, and possibly higher lgE 44 in boys. These factors, it is believed, predispose boys to enhanced airflow limitation in response to a variety of insults. More recently, other studies have suggested that the observed difference may be due to . . 1,7 . . . 49- endocrine influences . In an effort to explain this difference, some authors 51 postulate that the acquired immune system of females differs from that of males, because estrogens stimulate immunologic processes driven by CD4 + TH2 cells and B cells, whereas androgens enhance CD4 + TH1 and CD8 + cell 14 activity. Androgens are also known to promote production of lL-2 by TH1 cells. Interleukin 2 reduces TH2 activity and stimulates CD8 + T cells 52. Further discussion about the possible role of estrogen as an immune modulator is presented in section ‘Estrogen and progesterone.’ Race Most experts agree that race as a social construct represents a continuum rather than a term with clear-cut boundaries, a point that has been illustrated by large- scale genotyping among diverse biogeographic populations 53 Asthma morbidity and mortality is disproportionately high and continues to increase among African Americans 54. As documented by national surveys, there are higher rates of asthma in black than White children, a disparity that has widened since 1980 55. Simon and co-workers also found that among children with asthma, Blacks were more likely than Whites to report asthma-related limitations in physical activity and need for urgent medical services 56 Furthermore, at the molecular level, authors have detected increased expression of the CD80/CDB6 lymphocytes in African American adults following mitogen and antigen stimulation 57. CD80/86 are crucial molecules in T-cell activation 58. This finding was supported by that of Willwerth et al. 58 which showed a more generalized increased T-cell responsiveness that included responsiveness to the mitogen phytohemagglutinin and to the allergens in children of black ethnicity. 15 These reports give credence to the idea that differential neonatal immune responses may underlie the higher incidence of asthma in blacks. Genetic predisposition There is evidence that asthma has genetic components 4. A number of studies have shown an increased prevalence of asthma phenotypes among the offspring of subjects with asthma compared to the offspring of subjects without 59 60. Family studies have convincingly indicated that atopy (as measured by allergen skin tests, total lgE, and/or specific lgE), airway hyper-responsiveness, and asthma as diagnosed by questionnaire are at least partly under genetic 59 61 control . Numerous studies of twins have demonstrated that concordance rates for asthma phenotypes are all substantially higher for monozygotic than for dizygotic twins, suggesting a strong genetic contribution. In population—based studies of twins, the estimated effect of genetic factors is about 35 to 70 %, depending on the population and the design of the study 59’60. Environmental factors Early attention to environmental factors that may influence the susceptibility to the development of asthma in predisposed individuals focused on the effect of allergens, air pollution and tobacco smoke. Recently, the role of environmental SHD (synthetic hormones: estrogen and progesterone as well as toxicants: halogenated organochlorine compounds (HOCs) such as dichlorodiphenyl l6 dichloroethene (DDE) and polychlorinated biphenyls (PCBs)) in the etiology of . . . 62-69 allergy has gained increased attention Exposure to tobacco smoke Tobacco burning, a ubiquitous source of irritants, produces a large and complex mixture of gases, vapors, and particulate matter. More than 4,500 compounds and contaminants have been identified in tobacco smoke, among them respirable particles, polycyclic hydrocarbons, carbon monoxide, carbon dioxide, nitric oxide, . . . . . 4 nitrogen OdeeS, nicotine, and acrolein . There is evidence that both maternal smoking during pregnancy and passive smoke (environmental tobacco smoke (ETS)) exposure after pregnancy affect . . 70—74 . asthma later In offspring . Actually, ETS was reported to increase both prevalence and severity of asthma, as judged by increases in the frequency of attacks, the number of emergency room visits and the risk of intubations 75 Using data from a large cohort study (n~60,000), Jaakkola et al. demonstrated that maternal smoking in pregnancy increases the childhood risk of asthma 76 A large study of 20,000 children (aged 6 to 12 years) from nine countries in Europe and North America, reported that smoking during pregnancy decreases lung function parameters 77. The effects of past and current passive exposure to tobacco smoke were smaller than the effect of smoking during pregnancy. The aforementioned study strongly suggests that maternal smoking during pregnancy 17 subsequently affects the lung function and intuitively asthma in children. However, in a cohort of New Zealand children, Sherill et al., 78 found no significant detrimental effects for absolute forced expiratory volume (FEV), or vital capacity related to ETS (including in utero) exposure. Likewise, Cunningham et al., 79 compared lung function, measured by FEV1, and were unable to find significant reduction. Breast-feeding Ever since Grulee and Sanford in 1936 80 first reported that breastfeeding protects against an infantile allergic response, researchers have been exploring this topic. Some authors reported that breastfeeding appears to protect against 87-92 atopic manifestations 81-86 while others found no such association To- date, despite numerous studies, the search for consensus continues 81’82'93. Breast milk, as a functional food, is thought to contain its own complex immune system 94. The selective colonization of the mammary gland during lactation by a population of T lymphocytes which displays the phenotype and functional characteristics of memory T cells may be one of the mechanisms whereby the suckling infant benefits from its mother's immunological history 95. Cytokines and chemokines derived from milk cells and mammary gland epithelium may 18 contribute to the activation of intestinal T lymphocytes to enhance immunity during the early neonatal period 96. Findings from a population-based case-contral study (cases=2,315 and control =21 ,513) of 6—15 year old Japanese children showed a significantly higher prevalence of asthma among children who had been breastfed (adjusted OR = 1.2; 95% Cl: 1.05, 1.36). Conversely, a systematic review of prospective studies evaluating the association between exclusive breast-feeding during the first 3- months after birth and asthma, reported opposite findings. The aforementioned study found summary odds ratio (OR) for the protective effect of breast-feeding to be 0.70 (95% Cl 0.60, 0.81) thus concluding that exclusive breast feeding during the first months after birth is associated with lower asthma rates during childhood 81. Maternal allergy Children born to parents with allergic diseases present an increased risk of developing similar diseases 75’97. The proportion of children of two allergic parents can be as high as 58 %, when one parent is allergic it is about 29% and a child with neither parents having history of allergy still has about 13 % risk of developing allergic disease sometime during his life 98. However, maternal atopy is a stronger predictor of childhood allergy that paternal. Mother’s atopic history was found to be significantly associated with cord-blood lgE 99. Likewise, maternal history of atopy was associated with an elevated lgE among newborns. l9 For maternal asthma, this association was only evident in infant girls 100. Recently, Kuiper et al. found that maternal asthma was associated with elevated total lgE at all total IgE cut-off levels studied except 0.5 lU/mL 101. This finding was supported by Shah et al. who report a significant association between maternal history of allergic disease and elevated cord serum lgE among 98 newborns Environmental sex hormone disrupters A balanced endocrine system is essential for optimum health. Sex hormones are considered crucial in maintaining this balance. Therefore any disruption/alteration of these hormones could intuitively bring about adverse health effects such as . . 7 . . . allergic diseases or even death. A more extensrve CIISCUSSIOTT on sex hormone disrupters and their possible role in the etiology of diseases is presented on the next page. Summary The prevalence and severity of allergic diseases continue to increase. Sex is an important risk factor with evidence showing that asthma is more prevalent in boys throughout childhood, but in adolescence a gender reversal occurs. African- Americans are disproportionately affected while the idea of a genetic predisposition to allergy continues to intrigue researchers. There is virtually universal agreement that tobacco smoke represents a significant risk factor for allergies. Conversely, the ‘verdict’ on breast-feeding is inconclusive. Mothers 2O with allergies tend to have children with allergies: clinical and/or immunologic (increased lgE). Sex hormone disrupters have been implicated in the etiology of allergic disorders. Recently, a plethora of studies have focused on this area in an effort to explain the rising prevalence of allergies. 1.3 Sex hormone disrupters One difficulty experienced when considering endocrine disruption is the lack of consensus as to an appropriate definition for the term. Various definitions have been suggested including those of the US EPA (Kavlock et al., 1996) and a European workshop (EC, 1997). For this dissertation, the approach taken by the European workshop was adopted and modified to apply specifically to chemical disruption of the endocrine system: “A sex hormone disrupter is an exogenous substance that causes adverse health effects of an intact organism or its progeny, consequent to changes in endocrine function.” Sex hormones are synthesized mainly by endocrine glands such as the gonads (testis and ovary), the adrenals and (during gestation) by the fetoplacental unit, and are then released into the blood circulation. The biological hormonal properties of these substances have earned them a great deal of attention in the past decade as possible etiologic agents of certain diseases. Some of these chemicals display estrogen like activity, hence their importance in endocrine disruption and subsequent immune modification is closely scrutinize. Sources of these xenobiotics includes life style practices such 21 as diet (e.g., phytoestrogens coumestrol and genestein), oral contraceptive use or from the environment (e.g., dichlorodiphenyl dichloroethene (DDE), and polychlorinated biphenyls (PCBs). Sex hormones or similar substances from the above-mentioned sources may be classified as endocrine disrupters (EDs). These endocrine disrupters alter normal hormonal regulation and may be naturally occurring or environmental contaminants‘”. There are two classes of substances that may cause disruption: 1) Synthetic hormones; mainly estrogen and progesterone as in the case of oral contraceptives (Ocp), and 2) toxicants including the halogenated organochlorine compounds (HOCs): DDE & PCBs. It has been broadly suggested that these substances bring about adverse effects in at least two ways: by mimicking the action of a naturally-produced hormone and thereby setting off similar bio- chemical reactions and/or by affecting the synthesis, transport, metabolism and excretion of hormones, thus altering the concentrations of natural hormones‘o‘. However, there is yet to be a clear mechanistic understanding of how these proposed sex hormones disrupt the immune system. Recent attention has focused on the role of the ‘promiscuous’ estrogen receptors (ERs) through which sex hormone disrupters may communicate with cells prior to altering immune response. 22 Estrogen and progesterone There is mounting evidence that the immune system is regulated in part by sex steroid hormones such as estrogen and progesterone. It is known that proper immune function is critical for the delivery of a healthy baby a process that is also believed to be of importance in the etiology of immune diseases. The involvement of estrogen is hinged on the fact that estrogen receptors are found on T lymphocytes: Families of specialized white blood cells that help orchestrate the body's immune responses. Generally, estrogens are seen as enhancers at least of the humoral immunity 49. Furthermore, many of the allergic family of cytokines are under the control of estrogen 105. Estrogen and progesterone appears to work in tandem. In order for progesterone to work on the immune system, progestin receptors must be synthesized on the reticuloendothelial cells (cells which trap and consume foreign agents, except leukocytes circulating in the bloodstream) of the thymus 106. Such receptors are usually the result of estrogen stimulation. Under normal circumstances, increased estrogen leads to an increase in INF-y concentration, presumably as a mechanism of feedback inhibition to prevent unopposed stimulation of TH2 cells 107. Estrogen also stimulates secretion of IL- 4, -5, -6, and -10 by TH2 lymphocytes. These cytokines are potent stimulators of B-cell proliferation, maturation into plasma cells, and synthesis of antibody. 23 III ac Hal .4 O Interleukins 4, 5, 6, and 10 are expressed in greater quantity in an estrogen- . .. 50,52,108,109 dominant hormonal milieu . Progesterone seems to promote the preferential development of Th2 cells and to induce lL-4 production 110-113. Th2 type cytokines are considered vital for the maintenance of pregnancy by controlling the immune and endocrine systems 19’114. Simultaneously, it is this ability of progestin to invoke this Th1/Th2 shift that makes it important in the etiology of allergic diseases. However, the tight interdependence between estrogen and progestin secretions during the ovarian cycle and concomitant sex hormone hypersecretion during pregnancy pose substantial difficulties for the assessment of the individual contribution of physiological changes in progestin levels to immune regulation. Epidemiologically, Michel and colleagues reported that progestin administered during pregnancy increased the levels of cord blood lgE in 136 neonates: when mothers received progesterone, 53% of the newborns had detectable lgE (<0.5 IU/ml) versus only 24% of the newborns in mothers without progesterone . . . 97 administration Halogenated organochlorine compounds (HOCs) To date studies assessing the role of HOCs in altered immune response have focused mainly on their estrogen-like effects. However, some authors have proposed that the estrogenic activity of environmental toxicants is very low, 24 relative to estradiol, and thus unlikely to make significant contribution to the 115J16 etiology of diseases . This view has been challenged by reports of low dose effects of estrogenic toxicants 117'118 while others have been unable to 119fl20 confirm these effects . Nonetheless, environmental toxicants continue to be regarded as important contributing factors in the pathogenesis of allergic diseases. Dichlorodiphenyldichloroethene (DDE) Dichlorodiphenyltrichloroethane (DDT) was originally prepared in 1873, but it was not until 1939 that Paul Muller of Geigy Pharmaceutical in Switzerland discovered the effectiveness of DDT as an insecticide 121. The use of DDT increased enormously worldwide after World War II, primarily because of its effectiveness against the mosquito and lice. The World Health Organization estimates that during the period of its use approximately 25 million lives were saved predominantly from malaria and typhus. However, many species of insects developed resistance to DDT; it proved to have a high toxicity toward fish; and it was responsible for the near extinction of several bird species because of its interference with the formation of egg shells. For these reasons and because of its environmental persistence, the use of DDT was banned in the United States in 1972. However, DDT is still in use in many countries (including some South . . . . .. 122 American, African countries) as an InsectICIde 25 Humans are exposed to DDT mainly through foods, and infants through the placenta and breast-feeding 123. DDE, a metabolite of DDT, is persistent and is stored in fat tissue. The long half-life of DDE accounts for its ubiquity in the general population 124’125. Though exposure is of relatively low toxicity, DDE does have troubling effects as it is known to have weak estrogenic 126 and considerable anti-androgenic activity resulting in its endocrine disrupting capabilities 127-131. A more detailed description of DDE possible effect on the etiology of allergic diseases is presented in the ‘Literature Review of Components Comprising the Conceptual model’ section. Polychlorinated biphenyls (PCBs) PCBs are mixtures of up to 209 individual chlorinated compounds (known as congeners). There are no known natural sources of PCBs. PCBs are either oily liquids or solids that are colorless to light yellow. Some PCBs can exist as a vapor in air. PCBs have no known smell or taste. Many commercial PCB mixtures are known in the US. by the trade name Aroclor. PCBs have been used as coolants and lubricants in transformers, capacitors, and other electrical equipment because they do not burn easily and are good insulators. The manufacture of PCBs was stopped in the US. in 1977 because of evidence they build up in the environment and can cause harmful health effects. Products made before 1977 that may contain PCBs include old fluorescent 26 lighting fixtures and electrical devices containing PCB capacitors, and old . . . 132 microscope and hydraulic OIIS Residues have been detected in foods and in human adipose tissue, milk, and serum fat 133. Reports of PCBs having estrogenic effects 129 and altering immune responses have been recorded. Reichrtova et al. 65 reported that higher levels of the congener PC8118 and DDE in human placentas are associated with increased cord blood lgE levels. Specific to PCBs and various congeners: A study using pregnant minks documented that serum progesterone concentrations were significantly increased by experimental exposure to PCB-153 134. Similarly, an in vitro study documented increased secretion of progesterone from ovaries after exposure to 135136 PCB-153 . In seals, progesterone metabolism was significantly decreased with increasing liver PCB concentrations”? In polar bear dams, the plasma concentration of total PCBs is associated with an increase in placenta progesterone and accounts for 27% of the variance in the progesterone . 138 concentration Sex hormones and immunophysiology - Conclusion There is an abundance of evidence suggesting that the sex hormone disrupters DDE, PCBs, and synthetic progesterone, and estrogen may bring about alteration of normal endocrine functions resulting in allergic manifestations. 27 However, no clear unifying hypothesis exists to explain how sex hormone disrupters affect the development and function of the immune system. 1 .4 Time-window of exposure In an attempt to sort out the complex interrelationships between sex steroid hormones and allergic diseases, this work will focus on two important time- windows: the in utero and perinatal. The in utero environment sets the stage for prenatal priming of allergic diseases. This concept (prenatal priming) considers fetal life as the critical period of exposure and suggests that in utero exposure to environmental factors such as sex steroid hormones are important for the rise in the prevalence of asthma and 139-141 allergy . The prenatal priming concept ‘Priming or programming’ describes the process whereby a factor at a critical period of development has lasting effects 142. As an example, dramatic changes in maternal immune responses occur during pregnancy. To avoid aborting the fetus, the feto-maternal immune system is shifted in an allergic/Th2 direction 18’19. This Th2 reaction is characterized by the secretion of cytokines which then promote lgE production, resulting finally in allergies later in life. Hormones are believed to be responsible for controlling this shift 105. 28 T) In assessing the in utero impact of sex steroid hormones on allergies, this dissertation investigates associations between: placental organochlorine and cord blood markers of allergy; maternal oral contraceptive use and offspring’s serum allergic markers followed by an investigation of maternal oral contraceptive use and allergic phenotypes. The postnatal priming concept Seeing that hormones may play an important in the prenatal priming of allergies, it is plausible to postulate that postnatal exposure may also have a role in the etiology of allergies. Using serum 00, this work further determined the importance of postnatal exposure to sex steroid hormones in altering allergic markers and henceforth the development of asthma. A theoretical construct outlining the components of both processes (pre- and postnatal), are presented in the forthcoming section. 1.5 Components of the conceptual model The etiology of asthma, for example, is the result of a complex conflation of events. This section presents a model for assessing one of these complex processes: the involvement of sex hormone disrupters. The components of this model span the pre - and postnatal periods while using a combination of qualitative and quantitative measures of exposure and outcome. Specific pieces of evidence provided are: First, evidence that prenatal SHD exposure can disrupt allergic markers measured in utero. For this piece of evidence, placental p, p’ DDE was used as measure of exposure while cord plasma lL-4, lL-13 and 29 INF-y (cytokines typical of the atopic response) as measures outcome. This will show the effect of SHD on allergic markers that cannot be attributed to postnatal influences. Note, both exposure (placental p,p’ DDE) and outcomes (IL-4, lL-13 and INF-ry) are quantitative measures. The hypotheses behind this piece of evidence are: Increased placental p,p’-DDE concentration is associated with an increase in cord plasma IL—4 and lL-13, and that increased placental p,p’-DDE concentration will lead to a reduction in cord plasma lNF-wy. Second, existence of an association between proxy measures of in utero exposure to sex hormone disrupters and postnatal markers of allergic susceptibility. This bit of evidence suggests that the possible in utero immunomodulating effect of sex hormone disrupters may persist into child hood. Third, as an extension of the second condition above, evidence is required linking proxy measures of in utero exposure to sex hormone disrupters (maternal oral contraceptive use) with markers of effect: clinical manifestations of allergy such as asthma-like symptoms and atopic eczema in childhood. This along with evidence from the second condition will provide further support for the idea that sex steroid hormones may be associated with allergic disorders by modulating the immune status, through endocrine disruption. Fourth, serum organochlorines stimulate an allergic response. Results from the serum DDE / markers of allergic susceptibility association add a new level of support to the sex hormone disrupters / immune modulating idea in that both exposure and outcome are measured postnatally. This suggests that the effects of sex hormone disrupters do not only occur during the early stages of immune development (in utero) when 30 *Estrogen I 37:39:“: Progesterone markers @childhood immune WE Pregnancy (in utero environment) I a ChildhoOd log/e * Maternal oral contraceptive use before pregnancy Cord plasma IL-13, lL-4 & INF-q Placental OC Figure 1 Sex Hormone Disrupters and Childhood Allergy - Conceptual model the cells are more susceptible to change but may also produce change in immune development postnatally. A schematic representation of a proposed model that encapsulates the overarching theme of SHD and childhood allergy is presented in Figure 1 and discussed in the following background section. 1.6 Literature review of components comprising the conceptual model. The effect of placental p,p’ DDE on cord plasma lL-4, lL-13 & INF-y. There is evidence that the cytokine lL-13 is both necessary and sufficient to induce all features of allergic asthma 143. In humans, this was supported by consistent associations between tissue lL-13 levels and genetic variants in the lL-13 gene with asthma and related traits 143. Cellular response (including lL-4, lL-13, and lFNq,) to allergens have been reported at week 23 gestation 144 31 ('1 Recent findings suggest that in the etiology of allergic diseases, the Th2/Th1 ratio may be of more importance that individual cytokines 145 DDE is known to cross the placenta 146 potentially interfering with fetal immune development. This interference is thought to be via its ability to reduce the binding of progesterone to its receptor 147 Prenatally, Noakes and co-workers suggest a possible immunomodulating effect of cord blood DDE on phytohaemagglutinin mitogen stimulated cord blood lL-13 and lFN-y 148. However, findings on the effect of placental p,p’ DDE on cord plasma cytokines and their ratios (Th1/Th2) are yet to be reported. In utero exposure to sex hormone disrupters is related to immune markers of allergic susceptibility in childhood. To date, Frye et al. is the only team to report on the association between maternal OCp use and immune markers (specific immunoglobulin E against common inhalant allergens) in offspring. For atopic sensitization, they reported similar effects for maternal OCp use before and after birth. This finding raises 93,105,149 doubts in the previously reported association between maternal hormone level and allergic diseases. In an effort to allay these doubts, this dissertation investigates the association between maternal oral contraceptive use and humoral immune markers (such as serum immunoglobulin (lg)-A, lgE and basophilic surface lgE counts) in offspring. 32 In utero exposure to sex steroid hormones and their relation to childhood markers of allergic effect. Studies assessing the association between in utero exposure to sex hormone disrupters (using maternal oral contraceptive use as proxy) and allergic phenotypes have reported inconsistent findings. In 1987, Peters and Golding, presented findings suggestive of an increased risk of childhood eczema for children of mothers who used OC in the 18 months prior to pregnancy in a cohort of 10,900 5-year-old children 93 Approximately ten years later, using data from an aggregative (ecological) study, stt et al. found a geographic trend between asthma prevalence in children and maternal OCp use. This similarity was most notable during 1970 when a sharp decrease in OCp use was followed by a similar decrease in rate of hospital discharge with a diagnosis of asthma. They concluded that mother’s OCp use fits well into the geographic and temporal background of this increase in asthma 105 prevalence In addition, findings from a geographically defined cohort of 5,188 subjects showed that the prevalence of atopy at age 31 years was lower in children whose mothers reached menarche at a later age, especially after age 15 149. Early age of menarche is associated with higher levels of estrogen in adulthood 150452. 33 Contrary to the above findings, Xu et al. 153 found no differences in the mean concentrations of maternal sex steroid hormones according to the presence of allergic rhinitis or atopic eczema among offspring. In addition, while Frye et al. found maternal OC use before birth to be risk factor for asthma, allergic rhinitis, and atopic eczema; they also found similar effects for 00 use after birth 69. To date, these associations have only been tested using data from predominantly Caucasian cohorts. Assessing this association in a cohort with a different ethnic profile may provide cogency to the controversy. It is in this spirit that we investigated the in utero sex hormone exposure / allergic phenotypes association using data form an African-Caribbean cohort. Immunologic consequence of childhood exposure to organochlorine. Previously, in this monograph, we proposed the assessment of prenatal exposure to DDE and its association with allergic markers in childhood. This, as mentioned, provided evidence in favor of the prenatal priming concept; purporting that DDE is able to alter immune status during its early stages of development. In this section we extend this idea by determining whether DDE is also able to impose a similar effect postnatally. There are reports of DDE being associated with changes in cellular and humoral 154,155 immunity , in particular cytokines related to allergy, such as interleukin-4 156'157. In addition, using data from the Child Health and Environment Cohort 34 Study - Germany, Karmaus et al. showed serum DDE to be strongly related with increases in total lgE 66. All the above-mentioned studies focused on adult populations, with the exception of that of Karmaus et al. However, this study in children only reported on one allergic marker -— total lgE. Here, using the same study of Germany school children, we investigated whether serum DDE concentrations is associated with several immune markers including lgE, IgG, and IgA levels, lgE count on basophils along with eosinophilic granula, and white blood cell count. 35 References 10. 11. 12. Whitacre CC, Reingold SC, O_Looney PA. A gender gap in autoimmunity. Science 1999;283(5406):1277-8. Johansson SG, Bieber T, Dahl R, Friedmann PS, Lanier BQ, Lockey RF, Motala C, Ortega Martell JA, Platts-Mills TA, Ring J, Thien F, Van Cauwenberge P, Williams HC. Revised nomenclature for allergy for global use: Report of the Nomenclature Review Committee of the World Allergy Organization, October 2003. J Allergy Clin lmmunol 2004;113(5):832-836. Galli SJ. Allergy. Curr Biol 2000;10(3):R93-5. National institutes of Health, LaBl. NH. GLOBAL STRATEGY FOR ASTHMA MANAGEMENT AND PREVENTION, 2005. MERCKMedicus. Updated 2006. Available at: http://www.merckmedicus.com Elphick HE, Sherlock P, Foxall G, Simpson EJ, Shiell NA, Primhak RA, Everard ML. Survey of respiratory sounds in infants. Arch Dis Child 2001;84(1):35—39. Shames RS. Gender differences in the development and function of the immune system. Journal of Adolescent Health 2002;30(4):59—70. Kuehr J, Frischer T, Karmaus W, Meinert R, Barth R, Urbanek R. Clinical atopy and associated factors in primary-school pupils. Allergy 1992;47(6):650-5. Humbert M. Asthma, a priority for the allergist. Allergy 2006;61(5):515-7. Murray DM, Lawler PG. All that wheezes is not asthma. Paradoxical vocal cord movement presenting as severe acute asthma requiring ventilatory support. Anaesthesia 1998;53(10):1006-11. Chaouat G, Ledee-Bataille N, Dubanchet S, Zourbas S, Sandra 0, Martal J. TH1/T H2 paradigm in pregnancy: paradigm lost? Cytokines in pregnancy/early abortion: reexamining the TH1/'1' H2 paradigm. Int Arch Allergy lmmunol 2004;134(2):93—119. Maggi E. The TH1/T H2 paradigm in allergy. lmmunotechnology 1998;3(4):233-44. 36 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. Wills-Karp M, Santeliz J, Karp CL. The germless theory of allergic disease: revisiting the hygiene hypothesis. Nat Rev lmmunol 2001;1(1):69-75. Robinson 08, Larche M, Durham SR. Tregs and allergic disease. J Clin Invest 2004;114(10):1389-97. Romagnani S. Immunologic influences on allergy and the TH1/T H2 balance. J Allergy Clin Immunol 2004;113(3):395-400. Beeh KM, Ksoll M, Buhl R. Elevation of total serum immunoglobulin E is associated with asthma in nonallergic individuals. Eur Respir J 2000;16(4):609-14. Baena-Cagnani CE, Teijeiro A. Role of food allergy in asthma in childhood. Curr Opin Allergy Clin lmmunol 2001 ;1(2):145-9. Gaunt G, Ramin K. Immunological tolerance of the human fetus. Am J Perinatol 2001;18(6):299-312. Thellin O, Coumans B, Zorzi W, lgout A, Heinen E. Tolerance to the foeto- placental ’graft': ten ways to support a child for nine months. Curr Opin lmmunol 2000;12(6):731-7. Romagnani S. The role of lymphocytes in allergic disease. journal of allergy and clinical immunology 2000;105(3):399-408. Monaco C, Andreakos E, Kiriakidis S, Feldmann M, Paleolog E. T-cell- mediated signalling in immune, inflammatory and angiogenic processes: the cascade of events leading to inflammatory diseases. Curr Drug Targets lnflamm Allergy 2004;3(1):35-42. Ohshima Y, Yasutomi M, Omata N, Yamada A, Fujisawa K, Kasuga K, Hiraoka M, Mayumi M. Dysregulation of lL-13 production by cord blood CD4+ T cells is associated with the subsequent development of atopic disease in infants. Pediatr Res 2002;51(2):195-200. Williams TJ, Jones CA, Miles EA, Warner JO, Warner JA. Fetal and neonatal IL-13 production during pregnancy and at birth and subsequent development of atopic symptoms. J Allergy Clin lmmunol 2000;105(5):951-9. Wynn TA. lL-13 effector functions. Annu Rev lmmunol 2003;21:425-56. Siraganian RP. Allergy: principles and practice. . 5th ed. ed. St.Louis: C V Mosby, Inc, 1998. 37 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. Oettgen HC, Geha RS. lgE in asthma and atopy: cellular and molecular connections. J Clin Invest 1999;104(7):829-35. Ponsonby AL, Gatenby P, Glasgow N, Mullins R, McDonald T, Hurwitz M. Which clinical subgroups within the spectrum of child asthma are attributable to atopy? Chest 2002;121(1):135-42. Renz H. The central role of T-cells in allergic sensitization and lgE regulation. Exp Dermatol 1995;4(4 Pt 1):173-82. Daser A, Meissner N, Herz U, Renz H. Role and modulation of T-cell cytokines in allergy. Curr Opin lmmunol 1995;7(6):762-70. Erb KJ. Atopic disorders: a default pathway in the absence of infection? lmmunol Today 1999;20(7):317-22. Behrman RE, Kiliegman, R. M., Jenson, H. B. Nelson Textbook of Pediatrics. 17 ed W.B. Saunders Company, 2004. Guilbert T, Krawiec M. Natural history of asthma. Pediatric Clinics of North America 2003;50(3):523-38. Johnson CC, Ownby DR, Zoratti EM, Alford SH, Williams LK, Joseph CLM. Environmental epidemiology of pediatric asthma and allergy. Epidemiologic Reviews 2002;24(2):154-175. Beasley R, Crane J, Lai CK, Pearce N. Prevalence and etiology of asthma. The Journal of Allergy and Clinical Immunology 2000;105(2 Pt 2):S466-72. MMWR Morb Mortal Wkly Rep. Forecasted state-specific estimates of self-reported asthma prevalence-United States. MMWR Morb Mortal Wkly Rep, 1998;47:1022—5. Mannino DM, Homa DM, Akinbami LJ, Moorman JE, Gwynn C, Redd SC. Surveillance for asthma—United States, 1980-1999. MMWR Surveill Summ 2002;51(1):1-13. Palmer LJ, Cookson W. Genomic approaches to understanding asthma. Genome Research 2000; 10(9):1280-1287. Marion RJ, Creer TL, Reynolds RVC. Direct and Indirect Costs Associated with the Management of Childhood Asthma. Annals of Allergy 1985;54(1):31-34. 38 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. Vance VJ, Taylor WF. The financial cost of chronic childhood asthma. Ann Allergy 1971 ;29(9):455-60. Department of Health and Human Services. Action against asthma: A strategic plan for the Department of Health and Human Services. 2000. Taylor WR, Newacheck PW. Impact of childhood asthma on health. Pediatrics 1992;90(5):657-62. Taussig LM, Wright AL, Holberg CJ, Halonen M, Morgan WJ, Martinez FD. Tucson Children's Respiratory Study: 1980 to present. J Allergy Clin Immunol 2003;111(4):661-75; quiz 676. Kuehr J, Frischer T, Karmaus W, Meinert R, Barth R, Herrmann-Kunz E, Forster J, Urbanek R. Early childhood risk factors for sensitization at school age. Journal Of Allergy And Clinical Immunology 1992;90(3 Pt 1):358-63. Sears MR, Burrows B, Flannery EM, Herbison GP, Holdaway MD. Atopy in childhood. 1. Gender and allergen related risks for development of hay fever and asthma [see comments]. Clinical and Experimental Allergy 1993;23(11):941-8. Camargo CA, Jr., Schatz M. The relationship of gender to asthma prevalence, healthcare utilization, and medications in a large managed care organization. Acad Emerg Med 2003;10(5):508. Gissler M, Jarvelin MR, Louhiala P, E. H. Boys have more health problems in childhood than girls: follow-up of the 1987 Finnish birth cohort. Acta Paediatr 1999;88:310-4. LeSouef PN. Expression of predisposing factors in early life. Asthma: physiology, immunopharmacology and treatment. . London: Academic Press 1993. Smith JM, Harding LK, G. C. The changing prevalence of asthma in school children. . Clin Allergy 1971;1z57-61. Cutolo M, Sulli A, Capellino S, Villaggio B, Montagna P, Seriolo B, Straub RH. Sex steroid hormones influence on the immune system: basic and clinical aspects in autoimmunity. Lupus 2004;13(9):635-638. Beagley KW, Gockel CM. Regulation of innate and adaptive immunity by the female sex steroid hormones oestradiol and progesterone. FEMS Immunol Med Microbiol 2003;38(1):13—22. 39 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. Sell E, Arici A. Sex steroids and the immune system. Immunology and Allergy Clinics of North America 2002;22(3):407-433. Parham P. The Immune System. NY: Elsevier Science Inc, 2000. Barnes KC. Genetic epidemiology of health disparities in allergy and clinical immunology. Journal of Allergy and Clinical Immunology 2006; 1 1 7(2):243-254. Joseph CLM, Williams LK, Ownby DR, Saltzgaber J, Johnson CC. Applying epidemiologic concepts of primary, secondary, and tertiary prevention to the elimination of racial disparities in asthma. Journal of Allergy and Clinical Immunology 2006;117(2):233-240. Centers for Disease Control and Prevention. Measuring childhood asthma prevalence before and after the 1997 redesign of the National Health Interview Survey - United States (Reprinted from MMWR, vol 49, pg 908- 911, 2000). Jama-Journal of the American Medical Association 2000;284(18):2312-2313. Simon PA, Zeng ZW, Wold CM, Haddock W, Fielding JE. Prevalence of childhood asthma and associated morbidity in Los Angeles County: Impacts of race/ethnicity and income. Journal of Asthma 2003;40(5):535- 543. Hutchings A, Purcell WM, Benfield MR. Peripheral blood antigen- presenting cells from African-Americans exhibit increased CD80 and CD86 expression. Clinical and Experimental Immunology 1999;118(2):247-252. Willwerth BM, Schaub B, Tantisira KG, Gold DR, Palmer LJ, Litonjua AA, Perkins DL, Schroeter C, Gibbons F K, Gillman MW, Weiss ST, Finn PW. Prenatal, perinatal, and heritable influences on cord blood immune responses. Annals of Allergy Asthma & Immunology 2006;96(3):445-453. Holloway JW, Beghe B, Holgate ST. The genetic basis of atopic asthma. Clin Exp Allergy 1999;29(8):1023-32. Wiesch DG, Meyers DA, Bleecker ER. Genetics of asthma. J Allergy Clin lmmunol 1999; 1 04(5):895-901. Holgate ST. Genetic and environmental interaction in allergy and asthma. J Allergy Clin Immunol 1999;104(6):1139-46. Weisglas_Kuperus N, Sas TC, Koopman_Esseboom C, van_der_Zwan CW, De_Ridder MA, Beishuizen A, Hooijkaas H, Sauer PJ. Immunologic 40 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. effects of background prenatal and postnatal exposure to dioxins and polychlorinated biphenyls in Dutch infants. Pediatric Research 1995;38(3):404-10. Dewailly E, Ayotte P, Bruneau S, Gingras S, Belles_lsles M, Roy R. Susceptibility to infections and immune status in Inuit infants exposed to organochlorines. Environmental Health Perspectives 2000;108(3):205—1 1. Belles_lsles M, Ayotte P, Dewailly E, Weber JP, Roy R. Cord blood lymphocyte functions in newborns from a remote maritime population exposed to organochlorines and methylmercury. 2002;65(2):165-82. Reichrtova E, Ciznar P, Prachar V, Palkovicova L, Veningerova M. Cord serum immunoglobulin E related to the environmental contamination of human placentas with organochlorine compounds. Environ Health Perspect 1999;107(11):895-9. Karmaus W, Kuehr J, Kruse H. Infections and atopic disorders in childhood and organochlorine exposure. Arch Environ Health 2001;56(6):485-92. Karmaus W, Brooks KR, Nebe T, Witten J, Obi-Osius N, H. K. Immune function biomarkers in children exposed to lead and organochlorine compounds: a cross-sectional study. Environ Health. 2005 Apr 14;4(1):5. . Environ Health. 2005;I4(1):5. Brooks K, Samms-Vaughan M, Karmaus W. Are oral contraceptive use and pregnancy complications risk factors for atopic disorders among offspring? Pediatric Allergy and Immunology 2004;15(6):487-496. Frye C, Mueller JE, Niederrneier K, stt M, Heinrich J. Maternal oral contraceptive use and atopic diseases in the offspring. Allergy 2003;58(3):229-32. Gold DR, Burge HA, Carey V, Milton DK, PIatts-Mills T, ST. W. Predictors of repeated wheeze in the first year of life: the relative roles of cockroach, birth weight, acute lower respiratory illness, and maternal smoking. . Am J Respir Crit Care Med 1999;160:227-36. Nafstad P, Kongerud J, Botten G, Hagen JA J, JJ. a. The role of passive smoking in the development of bronchial obstruction during the first 2 years of life. . Epidemiology 1997;82293—7. Jaakkola JJ, MS. J. Effects of environmental tobacco smoke on the respiratory health of children. . Scand J Work Environ Health 2002;28(Supp| 2):71-83. 41 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. London SJ, James Gauderrnan W, Avol E, Rappaport EB, JM. P. Family history and the risk of early-onset persistent, early-onset transient, and late-onset asthma. . Epidemiology 2001;12:577-83. Gilliland FD, Li YF, JM. P. Effects of maternal smoking during pregnancy and environmental tobacco smoke on asthma and wheezing in children. Am J Respir Crit Care Med 2001;163:429-36. Arruda LK, Sole D, Baena-Cagnani CE, Naspitz CK. Risk factors for asthma and atopy. Curr Opin Allergy Clin Immunol 2005;5(2):153-9. Jaakkola JJ, Gissler M. Maternal smoking in pregnancy, fetal development, and childhood asthma. . Am J Public Health 2004;94:136- 40. Moshammer H, Hoek G, Luttmann—Gibson H, al. e. Parental Smoking and Lung Function in Children: an International Study. . Am J Respir Crit Care Med 2006. Sherrill DL, Martinez FD, Lebowitz MD, Holdaway MD, Flannery EM, Herbison GP, Stanton WR, Silva PA, Sears MR. Longitudinal effects of passive smoking on pulmonary function in New Zealand children. Am Rev Respir Dis 1992; 1 45(5):1 1 36-41. Cunningham J, Dockery D, Speizer FE. Maternal Smoking during Pregnancy as a Predictor of Lung Function in Children American Journal of Epidemiology 1994;139(12):1139-1152 Takemura Y, Sakurai Y, Honjo S, Kusakari A, Hara T, Gibo M, Tokimatsu A, Kugai N. Relation between breastfeeding and the prevalence of asthma : the Tokorozawa Childhood Asthma and Pollinosis Study. American Journal of Epidemiology 2001;154(2):115-9. Gdalevich M, Mimouni D, David M, Mimouni M. Breast-feeding and the onset of atopic dermatitis in childhood: A systematic review and meta- analysis of prospective studies. Journal of the American Academy of Dermatology 2001;45(4):520-7. Gdalevich M, Mimouni D, Mimouni M. Breast-feeding and the risk of bronchial asthma in childhood: a systematic review with meta-analysis of prospective studies. Journal of Pediatrics 2001;139(2):261-6. Saarinen UM, Kajosaari M. Breastfeeding as prophylaxis against atopic disease: prospective follow-up study until 17 years old. Lancet 1995;346(8982):1065-9. 42 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. Peat JK. Prevention of asthma. Eur Respir J 1996;9(7):1545—55. Oddy WH, Holt PG, Sly PD, Read AW, Landau Ll, Stanley FJ, Kendall GE, Burton PR. Association between breast feeding and asthma in 6 year old children: findings of a prospective birth cohort study. ij 1999;319(7213):815-9. Wafula EM, Limbe MS, Onyango FE, Nduati R. Effects of passive smoking and breastfeeding on childhood bronchial asthma. East Afr Med J 1999;76(11):606-9. Taylor B, Wadsworth J, Golding J, Butler N. Breast feeding, eczema, asthma, and hayfever. Journal Of Epidemiology And Community Health 1983;37(2):95-9. Midwinter RE, Morris AF, Colley JR. Infant feeding and atopy. Arch Dis Child 1987;62(9):965-7. Pratt WR. Allergic diseases in pregnancy and breast feeding. Ann Allergy 1981 ;47(5 Pt 1):355-60. Pratt HF. Breastfeeding and eczema. Early Hum Dev 1984;9(3):283-90. stt M, Dold S, Reitmeier P, Wulff A, Nicolai T, von Mutius E. [Does breast feeding prevent asthma and allergies? Results of the Munich asthma and allergy study]. Monatsschr Kinderheilkd 1992;140(10):769—74. Rusconi F, Galassi C, Corbo GM, Forastiere F, Biggeri A, Ciccone G, Renzoni E. Risk factors for early, persistent, and late-onset wheezing in young children. SIDRIA Collaborative Group. Am J Respir Crit Care Med 1999;160(5 Pt 1):1617-22. Peters TJ, Golding J. The epidemiology of childhood eczema: lI. Statistical analyses to identify independent earIy predictors. Paediatric and Perinatal Epidemiology 1987; 1 (1 )280-94. Hanson LA, Korotkova M, Haversen L, Mattsby-Baltzer l, Hahn-Zoric M, Silfverdal SA, Strandvik B, Telemo E. Breast-feeding, a complex support system for the offspring. Pediatr Int 2002;44(4):347-52. Bertotto A, Gerli R, Fabietti G, Crupi S, Arcangeli C, Scalise F, Vaccaro R. Human breast milk T lymphocytes display the phenotype and functional characteristics of memory T cells. Eur J lmmunol 1990;20(8):1877-80. 43 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. Takahata Y, Takada H, Nomura A, Nakayama H, Ohshima K, Hara T. Detection of interferon-gamma-inducible chemokines in human milk. Acta Paediatr 2003;92(6):659-65. Michel FB, Bousquet J, Greillier P, Robinet_Levy M, Coulomb Y. Comparison of cord blood immunoglobulin E concentrations and maternal allergy for the prediction of atopic diseases in infancy. The Journal of Allergy and Clinical Immunology 1980;65(6):422-30. Shah 8, MM. B. Parental history of allergy, maternal serum lgE & cord serum lgE. Indian J Med Sci. 2006 60(1):13-8. Bergmann RL, Schulz J, GUnther S, Dudenhausen JW, Bergmann KE, Bauer CP, Dorsch W, Schmidt E, Luck W, Lau S, al e. Determinants of cord-blood lgE concentrations in 6401 German neonates [see comments]. Allergy 1995;50(1):65-71. Johnson CC, Ownby DR, Peterson EL. Parental history of atopic disease and concentration of cord blood lgE. Clin Exp Allergy 1996;26(6):624-9. Kuiper S, Muris JWM, Dompeling E, van Schayck CP, Schonberger HJAM, Wesseling G, Knottnerus JA. Association between first-degree familial predisposition of asthma and atopy (total lgE) in newborns. Clinical & Experimental Allergy 2006;36(5):594-601. Steimer T. Reproductive health. Ares-Serono Symposia Series - Frontiers in Endocrinology. Vol. 2. Rome: Ares Serono Symposia Publications, 1993. Waring RH, Harris RM. Endocrine disrupters: a human risk? Mol Cell Endocrinol 2005;244(1-2):2-9. Choi SM, Yoo SD, Lee BM. Toxicological characteristics of endocrine- disrupting chemicals: Developmental toxicity, carcinogenicity, and mutagenicity. Journal of Toxicology and Environmental Health-Part B- Critical Reviews 2004;7(1):1-32. stt M, Dold S. Is asthma an endocrine disease? Pediatric Allergy and Immunology 1997;8(4):200-4. Druckmann R. Review: female sex steroid hormones, autoimmune diseases and immune response. Gynecol Endocrinol 2001;15 Suppl 6:69- 76. Ackerman LS. Sex steroid hormones and the genesis of autoimmunity. Arch Dermatol 2006;142(3):371-6. 44 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. Verthelyi D. Sex steroid hormones as immunomodulators in health and disease. INTERNATIONAL IMMUNOPHARMACOLOGY 2001;1(6):983- 993. Wilder RL, Elenkov IJ. Hormonal regulation of tumor necrosis factor-alpha, interleukin-12 and interleukin-10 production by activated macrophages. A disease-modifying mechanism in rheumatoid arthritis and systemic lupus erythematosus? Ann N Y Acad Sci 1999;876:14-31. Szekeres-Bartho J, Wegmann TG. A progesterone-dependent immunomodulatory protein alters the Th1/Th2 balance. J Reprod lmmunol 1996;31(1-2):81-95. Piccinni MP, Giudizi MG, Biagiotti R, Beloni L, Giannarini L, Sampognaro S, Parronchi P, Manetti R, Annunziato F, Livi C. Progesterone favors the development of human T helper cells producing Th2-type cytokines and promotes both lL-4 production and membrane CD30 expression in established Th1 cell clones. Journal of Immunology (Baltimore, Md. : 1950) 1995;155(1):128-33. Szekeres-Bartho J, Par G, Szereday L, Smart CY, Achatz I. Progesterone and non-specific immunologic mechanisms in pregnancy. American Journal Of Reproductive Immunology 1997;38(3):176-82. Hamano N, Terada N, Maesako K, Hohki G, Ito T, Yamashita T, Konno A. Effect of female hormones on the production of lL-4 and lL-13 from peripheral blood mononuclear cells. Acta Otolaryngol Suppl 1998;537:27- 31. Piccinni MP, Maggi E, Romagnani S. Role of hormone-controlled T-cell cytokines in the maintenance of pregnancy. Biochem Soc Trans 2000;28(2):212-5. Safe SH, Zacharewski T. Organochlorine exposure and risk for breast cancer. Progress In Clinical And Biological Research 1997;396:133-45. Safe SH. Xenoestrogens and breast cancer. N Engl J Med 1997;337(18):1303-4. vom Saal FS, Timms BG, Montano MM, Palanza P, Thayer KA, Nagel SC, Dhar MD, Ganjam VK, Parmigiani S, Welshons VW. Prostate enlargement in mice due to fetal exposure to low doses of estradiol or diethylstilbestrol and opposite effects at high doses. Proc Natl Acad Sci U S A 1997;94(5):2056-61. 45 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. 130. Welshons WV, Nagel SC, Thayer KA, Judy BM, Vom Saal FS. Low-dose bioactivity of xenoestrogens in animals: fetal exposure to low doses of methoxychlor and other xenoestrogens increases adult prostate size in mice. Toxicol Ind Health 1999;15(1-2):12-25. Ashby J, Tinwell H, Haseman J. Lack of effects for low dose levels of bisphenol A and diethylstilbestrol on the prostate gland of CF1 mice exposed in utero. Regul Toxicol Pharmacol 1999;30(2 Pt 1):156-66. Odum J, Ashby J. Neonatal exposure of male rats to nonylphenol has no effect on the reproductive tract. Toxicol Sci 2000;56(2):400-4. Africa). ACfEAaGC. Approaches to Effective Malaria Control that Avoid DDT in Kenya: Use of Bacillus thuringiensis israelensis (BTi). 2006. ATSDR. Toxicological Profile for DDT/DDD/DDE. Update. (Draft for Public Comment). In: Registry AfTSaD, ed. Atlanta, GA, 2000;433 pp. Sunyer J, Torrent M, Munoz-Ortiz L, Ribas-Fito N, Carrizo D, Grimalt J, Anto JM, Cullinan P. Prenatal dichlorodiphenyldichloroethylene (DDE) and asthma in children. Environ Health Perspect 2005;113(12):1787-90. Woodruff T, Wolff MS, Davis DL, Hayward D. Organochlorine exposure estimation in the study of cancer etiology. Environ Res 1994;65(1):132-44. Longnecker MP, Rogan WJ, Lucier G. The human health effects of DDT (dichlorodiphenyltrichloroethane) and PCBS (polychlorinated biphenyls) and an overview of organochlorines in public health. Ann Rev Public Health 1997;18:211-44. Chedrese PJ, Feyles F. The diverse mechanism of action of dichlorodiphenyldichloroethylene (DDE) and methoxychlor in ovarian cells in vitro. Reprod Toxicol 2001;15(6):693-8. Kelce WR, Wilson EM, Lambright CR, Gray LE, Jr., Roberts KP, Stone CR, Laws SC, Gray LE, Kemppainen JA.. J Mol Med 1997;75(3):198-207. Kelce WR, Lambright CR, Gray LE, Jr., Roberts KP, Stone CR, Laws SC, Gray LE, Kemppainen JA, Wilson EM. Toxicol Appl Pharmacol 1997;142(1):192-200. Hansen LG. Stepping backward to improve assessment of PCB congener toxicities. Environmental Health Perspectives 1998;106 Suppl 1:171-89. Ulrich EM, Caperell-Grant A, Jung SH, Hites RA, Bigsby RM. Environmentally relevant xenoestrogen tissue concentrations correlated to 46 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. biological responses in mice. Environ Health Perspect 2000;108(10):973- 7. Burow ME, Tang Y, Collins-Burow BM, Krajewski S, Reed JC, McLachlan JA, Beckman BS. Effects of environmental estrogens on tumor necrosis factor alpha- mediated apoptosis in MCF-7 cells. Carcinogenesis 1999;20(11):2057-61. ATSDR. ToxFAQsTM for Polychlorinated Biphenyls (PCBs) Safe 8. Polychlorinated biphenyls (PCBs), dibenzo-p-dioxins (PCDDs), dibenzofurans (PCDFs), and related compounds: environmental and mechanistic considerations which support the development of toxic equivalency factors (TEFs). Crit Rev Toxicol 1990;21 (1 ):51-88. Patnode KA, Curtis LR. 2,2',4,4’,5,5'- and 3,3',4,4',5,5'-hexachlorobiphenyl alteration of uterine progesterone and estrogen receptors coincides with embryotoxicity in mink (Mustela vision). Toxicol Appl Pharmacol 1994;127(1):9-18. Wojtowicz AK, Gregoraszczuk EL, Lyche JL, Ropstad E. Time dependent and cell-specific action of polychlorinated biphenyls (PCB 153 and PCB 126) on steroid secretion by porcine theca and granulosa cells in mono- and co-culture. J Physiol Pharmacol 2000;51 (3)1555-68. Wojtowicz A, Ropstad E, Gregoraszczuk E. Estrous cycle dependent changes in steroid secretion by pig ovarian cells in vitro to polychlorinated biphenyl (PCB 153). Endocr Regul 2001;35(4):223-8. Troisi GM, Mason CF. PCB-associated alteration of hepatic steroid metabolism in harbor seals (Phoca vitulina). J Toxicol Environ Health A 2000;61(8):649-55. Haave M, Ropstad E, Derocher AE, Lie E, Dahl E, Wiig O, Skaare JU, Jenssen BM. Polychlorinated biphenyls and reproductive hormones in female polar bears at Svalbard. Environ Health Perspect 2003;111(4):431- 6. Holt PG, Macaubas C. Development of long-term tolerance versus sensitisation to environmental allergens during the perinatal period. Curr Opin lmmunol 1997;9(6):782-7. Strachan DP. ls allergic disease programmed in early life? Clinical And Experimental Allergy 1994;24(7):603—5. Bjorksten B, Kjellman NI. Perinatal environmental factors influencing the development of allergy. Clin Exp Allergy 1990;20 Suppl 3:3-8. 47 —_.._ --.“--7 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. Barker DJ. In utero programming of chronic disease. Clin Sci (Lond) 1998;95(2):115—28. Wills-Karp M, Chiaramonte M. Interleukin-13 in asthma. Curr Opin Pulm Med 2003;9(1):21-7. Evans PC, Lambert N, Maloney S, Furst DE, Moore JM, Nelson JL. Long- term fetal microchimerism in peripheral blood mononuclear cell subsets in ‘ healthy women and women with scleroderma. Blood 1999;93(6):2033-7. Gabrielsson S, Soderlund A, Nilsson C, Lilja G, Nordlund M, Troye- Blomberg M. Influence of atopic heredity on |L-4-, IL-12- and IFN-gamma- producing cells in in vitro activated cord blood mononuclear cells. Clinical and Experimental Immunology 2001;126(3):390-396. t;.‘___ 1' Cohn B. DDT and DDE: Second Generation Time to Pregnancy Effects. Lundholm CE. The effects of DDE, PCB and chlordane on the binding of progesterone to its cytoplasmic receptor in the eggshell gland mucosa of birds and the endometrium of mammalian uterus. Comp Biochem Physiol C 1988;89(2):361-8. Noakes PS, Taylor P, Wilkinson S, Prescott SL. The relationship between persistent organic pollutants in maternal and neonatal tissues and immune responses to allergens: A novel exploratory study. Chemosphere 2006;63(8):1304-1311. Xu B, Jarvelin MR, Hartikainen AL, Pekkanen J. Maternal age at menarche and atopy among offspring at the age of 31 years. Thorax 2000;55(8):691-3. Vihko R, Apter D. Endogenous steroids in the pathophysiology of breast cancer. Critical Reviews in Oncology/Hematology 1989;9(1):1-16. Apter D, Reinila M, Vihko R. Some endocrine characteristics of early menarche, a risk factor for breast cancer, are preserved into adulthood. International Journal of Cancer. Journal International Du Cancer 1989;44(5):783-7. Moore JW, Key TJ, Wang DY, Bulbrook RD, Hayward JL, Takatani 0. Blood concentrations of estradiol and sex hormone - binding globulin in relation to age at menarche in premenopausal British and Japanese women. Breast Cancer Research and Treatment 1991 ;18 Suppl 12847-50. 48 153. 154. 155. 156. 157. 158. 159. Xu B, Pekkanen J, Husman T, Keski-Nisula L, Koskela P. Maternal sex steroid hormones in early pregnancy and asthma among offspring: a case- control study. The Journal of Allergy and Clinical Immunology 2003;112(6):1101-4. Cooper GS, Martin SA, Longnecker MP, Sandler DP, Gerrnolec DR. Associations between plasma DDE levels and immunologic measures in African-American farmers in North Carolina. Environ Health Perspect 2004;112(10):1080-4. Vine MF, Stein L, Weigle K, Schroeder J, Degnan D, Tse CK, Backer L. Plasma 1,1-dichloro-2,2-bis(p-ch|orophenyl)ethylene (DDE) levels and immune response. Am J Epidemiol 2001;153(1):53-63. Bilrha H, Roy R, Moreau B, Belles-Isles M, Dewailly E, Ayotte P. In vitro activation of cord blood mononuclear cells and cytokine production in a remote coastal population exposed to organochlorines and methyl mercury. Environ Health Perspect 2003;111(16):1952-7. Daniel V, Huber W, Bauer K, Suesal C, Conradt C, Opelz G. Associations of dichlorodiphenyltrichloroethane (DDT) 4.4 and dichlorodiphenyldichloroethylene (DDE) 4.4 blood levels with plasma IL-4. Arch Environ Health 2002;57(6):541-7. Kavlock RJ, Daston GP, Derosa C, et al., Research needs for the risk assessment of health and environmental effects of endocrine disruptors Environ Health Perspect 1996;104 (suppl 4), 715-740. EC (1997) European Workshop on the Impact of Endocrine Disrupters on Human Health and Wildlife: Report of the Proceedings (EUR 17549), Weybridge, UK, European Commission 49 kw...” CHAPTER 2 BACKGROUND AND METHODS FOR THE RESPECTIVE STUDIES 50 The four research questions posed in this monograph utilizes data from three cohorts: namely the Pregnancy Environment and Child Health (PEACH) study, the Jamaican Perinatal Morbidity, Mortality Survey and the Child Health and Environment Cohort Study-Germany. This chapter describes the background and methods used the respective studies. Methodologic details not covered in this hf-‘IW'I . chapter are presented in the four respective manuscripts found in the ‘Results’ secfion. 2.1 The PEACH study - Background PEACH is a multi-center longitudinal study of mother-infant pairs, which had its inception in Berrien County Michigan (Figure 4) at the Benton Harbor and St. Joseph study sites. Situated along the South East shore of Lake Michigan in Berrien County, Benton Harbor and St. Joseph are two adjoining but very different cities. According to City-Data.com 1, in 2004, Benton Harbor had over 10, 000 residence (92.4% are African-American) with a median household income 0f approximately $17,500. 51 E Michigan State University _ Berrien County - PEACH Study Site Other Counties (Not Involved) Figure 4. Map of Michigan showing PEACH Study Site 52 In 1894, the Baushke family, prominent citizens in Benton Harbor, built America’s first car, with 4-wheels and seating for 5 people. This was the inception of Benton Harbor’s industries which now includes the headquarters for the world renowned Whirlpool Corporation formally known as the Upton Machine Co. As in any other towns or cities, the introduction of industries is good news for the economy; l however their possible impact on public health is of grave concern. St. Joseph on the other hand in 2004 had an estimated population of 9,000 of aim” .1. which 89.6% are Caucasian. Median household income for the year 2000 was $37,032 1. Its ideal location at the mouth of the St. Joseph River (linking it to Chicago) earned the city the title ‘The Riviera of the Midwest’. This location encourages much commerce and tourism. Fertile lands boast orchards full of apples, peaches and cherries along with vineyards of grapes, fields of corn, rows of strawberries and blueberries. Little wonder St. Joe (as it is affectionately called) is the largest non-citrus fruit-growing region in the United States. There are also several industries (In 1911 the Uptons started their company, now Whirlpool - later moved to Benton Harbor) dotting the landscape, hence as is the case of its ‘sister’ city Benton Harbor, a possible risks to public health looms. The cities of Benton Harbor and St. Joseph are ideal sites for studying the human health effects of exposure to environmental sex steroid hormones for the following reasons. First, both cities are adjacent to each other; second, they are located along the South East shore of Lake Michigan, a known ‘reservoir’ for environmental toxicants and; third, the discrete ethnic and socio-economic differences between cities. It is in this setting that the PEACH study had its ‘genesis’ in May of 2003. The target population was women in their first trimester of pregnancy living in Benton Harbor or St. Joseph. The study population was women from target population who met the inclusion criteria of being primiparous (expecting first live birth), having no previous stillbirth, no multiple births, no diabetes, thyroid or adrenal disorders, and age 18-34 years. The PEACH study - Methods Questionnaires At the time of enrollment, a brief face-to-face interview was conducted to collect data on the mother’s lifestyle and mediation usage. A more detailed telephone interview was then conducted two weeks after to collect data on participants’ general and reproductive health. Approximately two weeks after delivery, another telephone interview was conducted that focuses on the mother’s pregnancy experience such as duration of pregnancy and delivery, and complications during pregnancy. Information on the mother’s general health along with that of her newborn was also ascertained. Cord blood and placenta tissue sampling Following delivery of the placenta, the external surface of the cord was washed to avoid contamination with maternal cells, and then approximately 1 OmL cord blood was collected by cordocentesis in heparinized tubes. The tube with blood 54 is then gently shaken to allow proper mixing of heparin and cord blood. The mixture was then refrigerated and prepared for mononuclear cell separation within 24 hours of collection. Within an hour of delivery, approximately one quarter of placental tissue (1009) was cut and stored in a glass container for the determination of DDE and other organochlorine compounds. The sample was then stored at —20°C until analyzed. Laboratory analyses of placental samples All organochlorine analyses were done at the Analytical Chemistry Section (ACS), Michigan Department of Community Health. Compounds were extracted, fractionated and quantified according to procedures described by Najam et al. 2, with modifications. Laboratory analyses of cord plasma samples Fresh cord blood was diluted with equal volume of sterile saline, carefully layered onto Histopaque-1077 (Sigma Chemical Company, St. Louis, MO) and centrifuged 30 minutes (at 600 G) at room temperature. The cells harvested from the interface were washed in serum free saline, then re-suspended into 2-3 ml of RPMI 10 (Gibco BRL, Life Technologies Inc., New York, USA) and the number of viable cells determined using trypan blue exclusion. CBMC staining will also be performed with Turk’s stain to exclude nucleated erythrocytes from cell counts and determine the degree of contamination. PHA-P (Sigma Chemical Company), 55 as a polyclonal activator at 5 ug/mL in culture, and dust mite allergen (Greer Labs), at 100 and 500 ug/mL, were used in cell cultures. Cells were cultured in triplicates using 96-well flat bottom plates in a final volume of 200 ill/well at a final concentration of 1 x 106 leukocytes/mL. Cytokine assays: Cytokine protein levels were measured using cytokine- specific, sandwich enzyme-linked immunoassays (ELISAs). IL-4 and lL-13: Recombinant human lL-4 and lL-13 standards of specific activity equivalent to that of WHO standard 88/656 (21) were included in each ELISA. The sensitivity of this assay was 0.9 to 1.9 pg/ml and the linear range of detection between 0.9 to 250 pg/mL. IFN-y. Similar procedure to that used for lL-13 was used to quantify IF N-y. However, the lower limit of detection for the assay was typically 0.3 mm and the linear range for quantitation was between 0.3 to 25 UlmL. This assay was calibrated using human lFN-y reference reagent G923—901—530 (specific activity 7 x 105 U/mg, 1 NIH unit = 115 pg, provided by Dr. C. Laughlin, NIAID, Bethesda, Md.). The Institutional Review Board of the Michigan State University approved the study in addition; all participants gave written, informed consent. 56 firm—- *1! Statistical analysis Adjusted betas (l3) and standard errors (SE) for the associations between exposure and outcomes were determined using linear regression models. Statistical tests are 2-sided and declared significant if p value < 0.05. The statistical software package SAS (version 9.1; SAS Institute Inc, Cary, NC) 3 was used for all analyses . Linear regression In its simplest form a linear regression analysis involves finding the best straight line relationship that explains how the variation in an outcome (or dependent) variable, Y, depends on the variation in a predictor (or independent) variable, X, and is expressed by the following equation: Y = b0 + b1X where b1 is called the slope of the equation and b0 the intercept. From a sample {(Yi, Xi) : | = 1, ...,n}, b1, be are estimated by 3 XXX. — (2X.- KEY.) 1 2 2X12 — (2X: )2 be is called the intercept and estimated by the following formula; A ZY—QZX b0: n This method is called the process of Least Squares that calls for minimization n n with respect to (b0, b,) of the error sum of squares; Zeiz : 20”. — b0 — b139,)2 i=1 i=1 57 ' Linn -__. which leads to the expression forbAO. Ii, . Since most outcomes are dependent on more than one factor (variable) it is necessary to explain the outcome in terms of the combined effect of these multiple independent factors or variables. This is done by multiple regression which is an extension of simple linear regression in which the dependent variable is predicted by a linear combination of the explanatory variables. For example, if an outcome Y depends on two independent variables X1 and X2, the regression equation will be Y = be + b1X1 + bzxz where the Do is the intercept, and b1 and b2 are slopes of X1 and X2 respectively. The amount of variation in the dependent variable that is accounted for by variation in the predictor variables is measured by the value of the coefficient of determination, or R2 (R squared). The R2 from multiple regression models is called Adjusted R2 when penalized for the number of variables in the model. This Adjusted R2 is the total percent variation in the dependent variable that is explained by the predictor variables together. 58 2.2 Child Health and Environment Cohort Study - Germany. The Child Health and Environment Cohort Study was conducted in 1994, targeting children residing in 18 townships in Hesse, Germany. Hesse is one of Germany's sixteen federal states and has an area of 21,1 10 km2 and just over six million inhabitants. Most inhabitants live in the southernmost part of Hesse between the rivers Main and Rhine. The latter is one of the longest and most important rivers in Europe and borders Hesse on the southwest without running through the state. The valley formed by the Rhine (Rhine Valley) in Hesse is where two of the regions targeted in this study are situated 4. Figure 5 shows the location of Worms Rhine Valley-control Toxlc Waste lnclnerator-reglon Odenwald-control O Darmstadt Figure 5: The Rhine Valley study sites and the toxic waste incinerator in the Rhine Valley. Two regions are situated in the Rhine Valley with low mountains on both sides. 59 For details regarding the Questionnaires and Laboratory procedures used in this study, please see the manuscript entitled ‘Immune function biomarkers in children exposed to lead and organochlorine compounds: a cross-sectional study in the ‘Results’ section. Statistical analysis The statistical software package SAS (version 9.1; SAS Institute Inc, Cary, NC) was used for all analyses 3. The linear regression method is described in pages 39 and 40. Statistical tests are 2-sided and decleared important if p value < 0.05. 60 2.3 The Jamaican Perinatal Morbidity, Mortality Survey - Background The Jamaican Perinatal Morbidity, Mortality Survey targeted all women who had a live birth, or stillbirth of 500 g or more in Jamaica during the period September 1, 1986 to October 31, 1986. Jamaica is an island nation 240 kilometres (150 miles) in length and 85 kilometres (50 miles) in width, which is slightly smaller than Connecticut-USA. The island is situated in the Caribbean Sea, approximately 804 kilometres (500 miles) south south east of the southern tip of Florida (Figure 5). Jamaica, with its mostly mountains interior and narrow discontinuous coastal plains, enjoys a mainly tropical climate with a temperate interior (average minimum temperature 220°C (71.6°F) and average maximum of 303°C (86.5°F)). The population (N=2,758,124) is mainly African-Caribbean (90.9%) of West African descent with a labor force (approximately 1.2 million) made up of workers in services (64.1%; mainly tourism), agriculture (19.3%) and industry (16.6%; mainly bauxite/alumina). The Jamaican Perinatal Morbidity, Mortality Survey was motivated by results of the Child Mortality Study of 1972 that clearly demonstrated the extent of under- reporting of early infant deaths. Shortly after the island wide survey (Figure 6) was designed using the First British Perinatal Mortality Survey 5 as a blue print. 61 .quoE. “mun ”322.4 5 ecu :33:qu _. ”octofino «mu—z. "Summ 530m .ucqun—ue ”has. “mum Ec< umuo swam 5.52 doused—On: “Layne—.052"? ”finned—m ”mu—... ”530w .Eiflofinh HoEu... «mum ro>oca1um 65.955335qu Soc; .3533: .0 5:45:35 use use; 3:23.58? .2369. 623:3 ”saunas .0 2:9“. I 00:8 O_oow (mm Zdwmmamdo 08 Any 40323.. 88 2.538 952 23238: 100m 0 . u ~.¢§ e 0 In, 258 8238 .4... 3535 a o 33:43. p .o oOh — 62 The overall aim of the study was to provide information to help improve maternal and childcare services and reduce perinatal morbidity and mortality in Jamaica. The Jamaican Perinatal Morbidity, Mortality Survey - Methods At enrollment, a face-to-face interview was conducted with each prticipant and their babies examined, usually within the first 48 hours after delivery. A standardized questionnaire was used to get at data during the antenatal, labor and delivery, and perinatal periods. Data was also abstracted from the newborn nursery charts and mother’s medical record to supplement interview data where necessary. Overall, 10,310 (94%) of the births in the two-month period were identified and included in the study (main cohort study) 5. The first follow-up occurred six weeks after delivery when another face-to-face interview was conducted aimed at information about breastfeeding practices and the infant’s health status. The study included in this monograph utilizes data from a geographic sub-sample (n= 1,720) of eleven to twelve year old children, representing those born in Kingston and St. Andrew. In a cross-sectional survey of these children, data on health outcomes were collected from the guardian or parent; who was often time the mother. The Research Ethics Committees of the University of the West Indies Jamaica and Michigan State University approved the assembly of the cohort and analysis of data for the included manuscript, respectively. 63 Statistical analysis The statistical software package SAS (version 9.1; SAS Institute Inc, Cary, NC) was used for all analyses 3. The method of statistical analysis used was logistic regression with 2-sided statistical tests declaring associations significant if p value < 0.05. Logistic regression Logistic regression is an adaptation to dichotomous outcomes of the classical linear regression model for continuous outcomes. It is primarily used to describe the relationship to the outcome of a primary exposure after adjusting for the effects of other (independent) variables that may influence both the outcome and exposure. Formally, in formulating a logistic model let Y (0 or 1) denoting the dichotomous outcome of interest with Y =1 labeling the presence of the condition under study. Then the logistic model for P[Y = l | X 1 , X 2 X k] is written simply as P(X) where X is a shorthand for X1 through Xk, The model formula is given by __ 1 PL)_ -(0t+Zl3iXi) I+e in which or and B, are unknown parameters to be estimated based on the observed exposure variables X1 through Xk and the outcome variable Y. The coefficients [31 through [3,. can be interpreted as adjusted log-odds ratios. For example, if X1 is the primary exposure, coded X1 = 1 for “exposed” and X1 =0 for “not exposed”, then assuming the variables X2 through Xk do not include X1, the adjusted odds ratio is given by exp(i31). 64 References 1. 2. City-Data.com. www.City-Data.com. Najam AR, Kover MP, Williams CC, Burse VW, Needham LL. Analysis of a Mixture of Polychlorinated Biphenyls and Chlorinated Pesticides in Human Serum by Column Fractionation and Dual-Column Gas Chromatography with Electron Capture Detection. Journal of the AOAC International 1999;82:177-185. SAS Institute. Statistical Analysis System, Version 9.1. In: Institute S, ed. Gary, NC, 2002-2003. Wikipedia. Hesse. 2006. Ashley D, McCaw-Binns A, Golding J, Keeling J, Escoffery C, Coard K, Foster-Williams K. Perinatal mortality survey in Jamaica: aims and methodology. Paediatr Perinat Epidemiol 1994;8 Suppl 1:6—16. 65 '* h;m w .' ,1 A CHAPTER 3 RESULTS 66 In this chapter, four manuscripts are presented. Their titles and order are: . Placental p, p'-dichlorodiphenyldichloroethylene and cord blood immune markers (forthcoming). Maternal oral contraceptive use and immune markers in the offspring (In progress). Are oral contraceptive use and pregnancy complications risk factors for atopic disorders among offspring? (Published) Immune function biomarkers in children exposed to lead and organochlorine compounds: a cross-sectional study (Published). These manuscripts correspond to the four research questions: 1. Is there evidence for the immunemodulation of cord immune markers of allergy by in utero sex hormone exposure? . Is maternal oral contraceptive use (a proxy for in utero estrogen/progesterone exposure) associated with immune markers of allergy in the offspring? . Is maternal oral contraceptive use (a proxy for in utero estrogen/progesterone exposure) a risk factor for markers of allergic susceptibility in offspring? 4. Does serum organochlorine alters immune markers in children? 67 COMPONENT ONE In utero exposure to SHD affects perinatal biomarkers of allergy 68 Placental p, p'-dichlorodiphenyldichloroethylene and cord blood immune markers Kevin Brooks1, Hanem Hasanz, Sridhar Samineniz, Venu Gangur2 and Wilfried Karmaus1 1 Department of Epidemiology and Biostatistics, Norman J. Arnold School of Public Health, University of South Carolina 800 Sumter Street, Columbia, SC., 29208-0001 2 Food Allergy & Immunology Laboratory, Food Science 8. Human Nutrition; National food Safety & Toxicology Center, Michigan State University, East Lansing, MI 48824, USA Abstract Placental p, p'-dichlorodiphenyldichloroethylene (p,p’-DDE) concentration and cord blood atopic markers were determined in 19 neonates. Increased placental p,p’-DDE was associated with a statistically Significant increase in cord plasma interleukin (lL)-13. Furthermore, both cord plasma IL-4/lNF-y and lL-13/lNF-y ratios were Significantly positively associated with placental p,p’-DDE concentration. Key words: DDE, placenta, cytokines, cord, allergy 69 The possible immune modulating role of ‘hormone-like substances’ is under investigation 1'2. Previously, in a cohort of German school children, we demonstrated significant associations between serum DDE and biomarkers of allergic susceptibility; including serum immunoglobulin E 2. In addition, for a group of 26 mother-infant pairs, Noakes et al. reported on the ability of maternal DDE and other ‘hormone-like’ substances to alter the fetal immune response 1. More recently, Sunyer and co-workers showed that in utero DDE exposure is related to increased incidence of asthma in children 3. It is therefore very timely to present the yet-to-be-reported relation between placental p,p’-DDE and cord plasma levels of prototypic type-1 and type-2 cytokines. Our objective was to test the hypothesis whether increased placental p,p’-DDE concentration is associated with an increase in cord plasma IL-4 and lL-13. In addition, since type-1 and type-2 cytokines often antagonize each others activity, we hypothesize that increased placental p,p’-DDE concentration is associated with a reduction in cord plasma INF-y. The Th2/Th1 cytokine ratio has been used as a measure of relative atopic reactivity 4. We therefore further hypothesize that increased placental p,p’-DDE concentration is associated with an increase in the ratio of cord plasma lL-4/lNF-y as well as the ratio of lL-13/lNF-y. 70 Methods and results Primiparous women in their first trimester were enrolled in the ongoing Environment and Child Health (PEACH) study. Ethical approval was granted by the Michigan State University Committee on Research Involving Human Subjects. Telephone interview data on passive smoking (ETS), mother’s age, and allergic status (defined as mother ever had one or any combination of: asthma, hay fever, itchy rash, or wheezing) were collected. Cord blood samples were drawn from the umbilical vein and transported to the lab within 24 hours. Cytokine protein levels were measured using ultrasensitive Elisa based assays optimized using paired monoclonal antibodies (PharMingen). Typical assay sensitivities; IL- 4: 0.05pg/ml; |L-13:1.5 pg/ml; and INF-y. 0.5pg/ml). Placental p,p’-DDE was extracted into diethyl ether-hexane (1 :1 v/v), passed over a Florisil column and quantified by high-resolution gas chromatography with electron capture detection. p,p’-DDE values below the detection limit (DL = 0.50 pg/g) were replaced with half the DL and all concentrations were lipid adjusted. We used linear regression models (PROC GLM) with cytokines as dependent and p,p’-DDE as independent variables; while controlling for mother's age, atopic status, and ETS. In order to obtain a multivariate normal distribution we log- transformed cytokine levels. 71 Complete data (interview, cytokine and DDE concentration) are available for 19 mother-infant pairs. Mothers were on average 29 years of age while 2 of 19 were exposed to passive cigarette smoke in their homes during pregnancy (Table 1). Median value for placental p,p’-DDE was 58.3 pg/g. For lL-4 and lL-13 their median values were 4.2 pg/mL and 61.4 pg/mL respectively (Table 1). Adjusting for maternal age, allergy and exposure to tobacco smoke, we found that placental p,p’-DDE statistically significantly increase the level of cord plasma Phil-HM fl lL-13 (B = 0.007 pg/g; p=0.03, Table 2). Placental p,p’-DDE was also found to increase lL-4 levels while reducing INF-y; however these associations were not statistically significant. Nevertheless, both cord plasma lL—4/INF-y and lL-13/INF- y ratios were Significantly increased with increasing placental p,p’-DDE concentration (8 = 0.03 pg/g; p=0.007 and i3 = 0.17 pg/g; p=0.02 respectively; Table 2) Discussion Using placental and cord blood samples of 19 mother-infant pairs we demonstrated, a statistically significant association between placental p,p’-DDE and cord plasma lL-13. The results support our hypotheses that placental DDE is related to increased cord plasma lL-13 and may be associated with increased IL- 4 and reduced INF-y. Considering the aforementioned results for lL-4, |L-13 and INF-y, we expected placental DDE to be associated with the ratio of lL-4/lNF-y along with that of lL-13/lNF-y. The associations (increased DDE and increased Th2fTh1 ratios) were found to be statistically Significant. 72 DDE is known to cross the placenta. However, in their Western Australian samples, Noakes et al. were unable to detect placental p,p’-DDE and used maternal levels instead. This was not the case in our North American samples. The fact that we determined p,p’-DDE in placental samples may be explained by different exposure in the southern and northern hemisphere, resulting from a long-range atmospheric transport of organochlorines enhanced by the meridional flow 5. Whereas both |L-4 and IL-13 play critical roles in the initiation as well as maintenance of allergic inflammation and asthma 6, INF-y dominated responses are associated with a ‘non-allergic’ response 7. Notably, lL-4 and lL-13 Share many but not all proallergenlc activities. Recent studies suggest that lL-13 is a key player in asthma independent of lL-4 6. It is suggested that decreased lFN-y in cord blood mononuclear cells is the hallmark of newborns from high-risk population 8. We are uncertain whether the relationships observed in this study between cytokines and placental p,p’-DDE are direct or indirect. Maternal and . . . . 9 fetal immune responses during pregnancy are under endocrine Influence . Thus, it is possible that an explanation for our finding may be that DDE introduce an endocrine disrupting effect 10, which in turn might alter the fetal T-cell cytokine profile. 73 We are not aware of studies implicating direct effect of DDE on Th2fih1 cell development or cytokine responses. Our findings suggest that the assessment of DDE exposure on Th2/Th1 ratios may be of more importance than the investigation of individual cytokines. Noakes et al. reported a significant inverse correlation (r =0.406, p = 0.049) between maternal adipose tissue levels of OC p,p’-DDE and maternal INF-y reaction to mitogens 1. The authors also found increased phytohaemagglutinin mitogen stimulated |L-13 and decreased IF N-y with increasing cord blood p,p’- DDE levels. Though these findings, from their small sample (n=26), were not statistically significant; they were in the same direction as those we presented using a similar size cohort (n=19). Since these small studies reported comparable findings, the effects are likely to gain statistical significance in a study with a larger sample size and improved statistical power. Our findings suggest that DDE may play a role in the prenatal priming of allergic diseases via promoting lL-13 dominance. This priming — to a greater extent - may also be through the overall dominance of the ratio of Type-2 cytokines (both lL-4 and lL-13) over Type-1 (lFN-y) cytokine. Larger epidemiologic studies are needed to explicate possible mechanistic explanations for our findings. 74 Acknowledgement This work was supported by United States Environment Protection Agency STAR grant number R830825—01-0. 75 Table 1. Descriptive characteristics of the cohort Stuchohort (n=19) Maternal Everhad: Asthma Yes 1/19 Hay fever Yes 3/19 Itchy rash Yes 2/19 Itchy eyes or stuffy nose Yes 7/19 Wheezing Yes 3/19 Allergy Yes 10/19 Exposure to passive smoke in Yes 2/19 home Ethnicity Caucasian 15/19 Age (mean, range, years) 29.3 (21-42) Placental p,p’-DDE (median, 5-95% value, pg/g) 58.3 (40.2 — 388.4) Cord plasma (median, 5-95% value, pg/mL): lL-4 4.2 (0.7 — 29.8) lL-13 61.4 (9.2 - 963) INF-y 119.8 (0.35 — 1398.1) lL-4/ INF-y ratio 0.05 (0.003 - 15.41) lL-13/ INF-y ratio 0.72 (0.04 — 614.14) 76 683 a co ummmn mod v Q .. 8:5: E 96:5 8283 2 mSmoaxm new >925 .mmm 9559: Lo.— noumaua. m 68.8 .too 88.3 .83 68.923. 88.9 Loco 88.983 a 8513. 88.9 Sod 83.9 Rod 68980.0- $8.8 Sod $8.8 Boo 820 33 moaned Emu a $9 a me a mg a mg n oee $2. a 3. 25. i2. 21.: 225.9 I2. SEES 2-... SEE: E. 875 $2. 83. as... .52. :1. So one 9: oco .rmz. .35: T... ucm mcosmbcmocoo moaned cmmzcmn 2mg Bate Emucflm vcm av mmumnv cozfloommd. .N 833. 77 References 10. Noakes PS, Taylor P, Wilkinson 8, Prescott SL. The relationship between persistent organic pollutants in maternal and neonatal tissues and immune responses to allergens: A novel exploratory study. Chemosphere 2006: 63: 1304-1311. Karmaus W, Brooks KR, Nebe T, Witten J, Obi-Osius N, Kruse H. Immune function biomarkers in children exposed to lead and organochlorine compounds: a cross-sectional study. Environ Health 2005: 4: 5. Sunyer J, Torrent M, Garcia-Esteban R, et al. Early exposure to DDE, breastfeeding and asthma at age six (in print). Clin Exp Allergy 2006. Tanaka T, Kouda K, Kotani M, et al. Vegetarian diet ameliorates symptoms of atopic dermatitis through reduction of the number of peripheral eosinophils and of PGE2 synthesis by monocytes. J Physiol Anthropol Appl Human Sci 2001: 20: 353-361. Tanabe S, Tatsukawa R, Kawano M, Hidaka H. Global distribution and atmospheric transport of chlorinated hydrocarbons: HCH (BHC) isomers and DDT compounds in the Western Pacific, Eastern Indian and Antarctic Oceans. Journal of Oceanography 1982: 38: 137-148. Wills-Karp M. Interleukin-13 in asthma pathogenesis. lmmunol Rev 2004: 202: 175—190. Romagnani S. Immunologic influences on allergy and the TH1/1' H2 balance. J Allergy Clin lmmunol 2004:1132 395-400. Kondo N, Kobayashi Y, Shinoda S, et al. Reduced interferon gamma production by antigen-stimulated cord blood mononuclear cells is a risk factor of allergic disorders—6-year follow- up study. Clin Exp Allergy 1998: 28: 1340-1344. Piccinni MP, Maggi E, Romagnani S. Role of hormone-controlled T-cell cytokines in the maintenance of pregnancy. Biochem Soc Trans 2000: 28: 212-215. Sonnenschein C, Soto AM. An updated review of environmental estrogen and androgen mimics and antagonists. J Steroid Biochem Mol Biol 1998: 65: 143-150. 78 COMPONENT TWO In utero SHD affects postnatal biomarkers of allergy 79 rfiha‘.‘--- -' 7‘ Maternal oral contraceptive use and immune markers in the offspring. BACKGROUND: Maternal oral contraceptive (OC) use is Shown to be associated with clinical manifestations of allergic diseases in offspring. The aim of this study was to assess the, yet to be reported, association between OC use and humoral immune markers in offspring. We hypothesized that maternal 00 use increases humoral immune markers of allergy in offspring. METHODS: A cross-sectional investigation included 334 mother child (aged 7— 10 years) pairs from Hesse, Germany. In 1995, detailed self-administered questionnaires were used to collect information on maternal OC use and atopy, passive smoking, breastfeeding, offspring’s gender and number of infections in the previous 12 months. lmmunoglobulins (lgs) along with white blood cell, eosinophils, and basophilic surface lgE were also quantified. The data was analyzed using linear regression, while controlling for confounders. Since allergic reactions are different for boys and girts, we stratified by sex. RESULTS: In children ages 7 to 10, female offspring of mothers who used OC had Significantly lower (p<0.05): IgA (123.43 mg/dL vs. 150.52 mgldL), and lgE (22.96 kU/L vs. 50.83 kU/L) levels as well as basophilic surface lgE counts (783 vs. 842), compared to those of mothers who did not use 00. For male offspring, statistical Significance was only seen in an increased number of basophilic surface lgE (911 vs. 876). 80 CONCLUSIONS: This study suggests that maternal 00 use may result in sex- related differences of the immunomodulating effects in offspring: a finding that could play a role in the etiology of allergic diseases. 81 Allergic/atopic diseases are among the most vexing disorders of childhood. The . . . . . 1,2 IncreaSIng prevalence and severity of asthma and other allergic diseases render them of enormous personal and public health concern 3. Early efforts to elucidate the etiology of these conditions focused on factors such as genetic predisposition, air pollution and tobacco smoke. The role of synthetic hormones (estrogen and progesterone) as risk factors for allergic diseases has gained increased traction 46. Specifically, the importance of prenatal (in utero) exposure to these hormones has been presented 1’7. Xu and co-workers assessed the effect of maternal age at menarche on the occurrence of atopy among offspring. In a follow-up study of 5188 subjects, they collected data during pregnancy and a follow up examination was completed at 31 years of age. Atopy was determined by skin prick tests while maternal age at menarche was obtained from perinatal data. The authors reported that the prevalence of atopy was lower in children whose mothers reached menarche at a later age 7: a finding that may be explained by the fact that early age of menarche is associated with higher levels of estrogen in adult women 8'10. Further support for the possible prenatal endocrine disruption-allergic diseases idea comes from the work of Frye et al. They reported that maternal oral contraceptive (00) use before birth was associated with a higher risk of atopic 82 diseases in the offspring compared with children of mothers who had never taken 00 4. Moreover, in a recent study, we demonstrated significant associations for asthma or wheezing, and cough between 11-12 year old offspring of 00 users 5 . . . . . and non-users . These studies Show aSSOCIatIons between OC use and clinical manifestations of allergic diseases in offspring; however, a relationship between OC use and humoral immune markers in offspring is yet to be presented. Using data from a cross-sectional investigation conducted in Hesse, Germany, we assess the relationship between maternal OC use and immune markers in offspring. We hypothesized that maternal 00 use Significantly increases humoral allergic markers in offspring. Methods Subjects and materials In 1995, parents of 1091 second-grade school children in 18 townships of Hesse, Germany were invited to participate in the study. Children were allowed to participate only if passive smoking in the private household did not exceeded 10 cigarettes per day during the previous 12 months. The study was approved by the Data Protection Agency of the State of Hamburg, Germany, the Ministry of Cultural Affairs of Hesse, Germany, and the participating local school committees. 83 Questionnaires Four self-administered parental questionnaires were used in the survey: one regarding the living condition and nutrition of the family, one for each parent, and one regarding information on the child. Maternal oral contraceptive use was determined in an interview on reproductive history. The question posed was “In the 12 months before conception of the index child, did you use oral contraceptives at any time (yes\no)?” Duration of breastfeeding was recorded in weeks of total and in weeks of exclusive nursing. Environmental tobacco smoke (ETS) was graded as smoking in the child’s home in the previous 12 months (no cigarettes, 1-20 cigarettes, 11-20 cigarettes, 21-30 cigarettes, more than 30 cigarettes per day). We recorded age, gender, and the number of infections, defined as cold, coughing, and sore throat with or without fever in the last 12 months (none, less than 5 infections, 5-10 infections, more than 10 infections). Immunologic assays For phlebotomy, we used the 'Vacutainer System' (Becton, Dickinson 8. Company, San Jose, California). Approximately 25 mL were drawn and separated into different aliquots. lmmunoglobulin (lg) E in serum was quantified at the Medical, Alimentary and Veterinary Institute for Research Middle Hesse, Division of Human Medicine, Dillenburg, Germany, using a florescence- immunoassay (CAP, Pharmacia, Uppsala, Sweden). We also measured IgA, G, and M by laser immunonephelometry (Dade Behring, Liederbach, Germany). The results for IgA, G and M were provided in mg/dL and for lgE in kU/L serum. 84 For leukocyte subsets, we collected 8 mL of blood in tubes containing EDTA and mixed them to prevent clotting. This aliquot was transported to the Central Laboratory of the University Clinic of Mannheim and analyzed on the same day. We used 200 pL of blood for the automated differential (laser-based hematology analyzer CD3500, Abbott Diagnostics, Santa Clara, California), and 100 p.L for each of the nine three-color test tubes analyzed by flow cytometry (FACScan, Becton, Dickinson, 8. Company, San José, California, equipped with a 488 nm air-cooled argon ion laser). Eosinophils were determined according to their specific depolarisation characteristics and their eosinophilic granula content by the intensity of light scatter by flow cytometry. lgE on basophils were identified by their high density on the basophil surface using immunofluorescence with a Phycoerythrin labeled anti-lgE antibody. Statistical methods Since data for leukocytes (VVBC) and their subsets (eosinophils), immunoglobulins were not normally distributed, the geometric mean, 5-, 95- percentiles are provided. In order to obtain a multivariate normal distribution, we log-transformed the number of cells and the immunoglobulins before testing associations with possible predictors. All statistical analyses were performed using SAS software versions 9.1 11. We used multiple linear regression models (PROC GLM) stratified by sex with 85 immune markers of allergic diseases as dependent variables. The potential confounders; maternal atopy (positive if mother reported a history of asthma, Eczema, or hay fever), age of offspring, ETS, number of infections during the last 12 months, and breastfeeding were controlled for in our models. Age of the child was divided into three groups: 7, 8 and 9-10 years of age. Our main exposure of interest, OC use in the 12 months before conception of index child, was coded ‘yeS/no’. Information on passive smoking (ETS) in the child’s home in the previous 12 months was divided into three categories (no cigarettes, 1-20 cigarettes (a pack), 20 cigarettes per day and more). For the number of infections we considered three categories (none, 1-5, more than 5). Duration of breastfeeding was considered as either: 0 weeks, 1-5 weeks or more than 5 weeks. From the results of the regression analyses, we calculated adjusted geometric means of immune markers for the categories within each covariate. T-tests were used to compare the statistical effect of maternal OC use with no maternal 00 use (reference). Results Of 1091 subjects invited, 671 (61.5%) agreed to participate. Of these, only ‘natural' mother-child pairs (n=663) were selected for this paper. Phlebotomy was performed on 350 of the 663 children; 340 of whom had immunoglobulins quantified. Complete information (questionnaires and immune markers) was available for 334 children. 86 A comparison of potential confounders’ characteristics between the sub-cohort of ‘natural’ mother-child pairs and that with complete information is presented in Table 1. Approximately 40 % of mothers who gave birth to boys used OC before pregnancy compared to about a third of those who gave birth to girls. Of the maternal atopy ‘family’ of variables, hay fever was the most prevalent at 15%. Ninety Six percent of the children were between 7 and 8 years of age and approximately 74 % experienced 1 to 5 infections during the last 12 months (Table 1). Due to the eligibility criterion of passive smoking of less than 10 cigarettes in the child’s home for being included in blood sampling, the prevalence of passive smoking was also lower in the group with phlebotomy than in the total group (Table 1). Nevertheless, the fact that parents were separated or divorced and Shared cohabitation for their child, resulted in a re-assessment of the passive smoking status after phlebotomy. Eligibility was determined on the information provided by one parent (mother or father) for their household. In the case of separate dwellings, we re-assessed the exposure by taking the average number of cigarettes smoked in both homes. As a consequence, 24 (7.5%) children who were exposed to more than 10 cigarettes per day at home had a phlebotomy and were included in the analyses. Tables 2 reports that for girls whose mothers used OC before pregnancy, their level of lgE was half that of girls whose mothers did not used OC before pregnancy. The level of IgA was also decreased in girls of 00 users compared to girls of none-OC users. This decreased level however was less dramatic than that reported for lgE. lgG, 19M and lgE showed an increase with increasing age 87 in boys while only lgM showed this pattern in girls. lgE showed an increase with age in girls but not in a dose dependent fashion. Regarding lgE count on basophils; a higher number was evident for boys of mother who use OC compared to boys of mother who were none-OC users. The opposite was true for girls: higher in girls of none-OC-users compared to OC ' users. In addition, lgE count on basophils was found to increase with increasing age and, conversely, decrease with increasing number of infections (Table 3). Results from adjusted regression models showed significantly decreased IgA (p=0.04; 123.43 mg/dL vs. 150.52 mg/dL) and lgE (p=0.04; 22.96 kU/L vs. 50.83 kU/L ) in girls ofimothers who used 00 before pregnancy (Table 5). In boys of OC users, lgE count on basophils was significantly increased (p=0.04; 911 vs. 876); concomitantly, they were significantly decreased in girls (p<0.01; 783 vs. 842; Table 5). Discussion The data for children 7-10 years of age suggest, for the first time, sex-related differences in the association between maternal OC use before pregnancy and serum immune markers in offspring. Decreased serum IgA, and lgE levels as well as basophilic surface lgE counts were observed in female offspring of OC users compared to their female counterpart. In addition, increased basophilic surface lgE counts was present for boys if their mother used 00 before pregnancy. 88 The cross-sectional design of the study is a limitation as it brings into focus the possibility of recall bias. However, mothers did not know the result from their child’s serum immunologic analyses therefore the possibility of recall bias in this instance was minimized. Furthermore there is no major public interest about DC . . 4 use and atopic diseases . Contrary to our a priori hypothesis, the data did not reveal an overall significant association between maternal OC use and humoral immune markers in offspring. Similarly, Xu and co-workers failed to find significant associations between maternal sex hormone concentrations (in serum) during early pregnancy and onset of allergic disease in early childhood 12. Furthermore, a study by Frye et al. found no significant difference for atopic manifestations between offspring of mothers who used 00 before and those of mothers who used 00 after pregnancy 4. Nonetheless, our significant sex-related findings are intriguing and consistent with the established hypothesis that there are sex-related differences . . 13-16 . . . in immune response ,Suggesting the Involvement of sex sterOId hormones 1647 OCp have two major components: progesterone, which is known to stimulate the development of TH2 (allergic type) cells 18, and estrogen that is suppose to increase the production of TH1 (non-allergic type) cells 19’20. To date, there is no clear explanation of how sex steroid hormones affect the development and 89 function of the immune system. Nonetheless, it is know that some cells have estrogen and progesterone receptors; creating binding sites for these hormones . . . 14,15 and ultimately altering Immune responses . Regarding IgA, Sell and Arici suggest that estrogen may increase or decrease IgA levels in an organ dependent manner 15. We found that maternal OC use may decrease serum IgA levels in girls. IgA was also decreased in boys of 00 users though not statistically significant. For lgE, our finding of decreased levels in girls of mothers who used 00 suggests that hormones may be influencing lgE levels. Since lgE iS a strong determinant of asthma 34, this finding supports the existing hypothesis that hormonal factors may be involved in the pathogenesis of asthma. Concerning basophil-bound lgE, studies have Shown increased counts in atopic children 29'30 and deduced that they can serve as an indicator of allergic sensitization 31. Our finding of decrease counts in girls of mothers who used 0C is in concordance with our findings for lgE. Similar explanation may apply. On the other hand, boys of OC users had an increased number of basophil-bound lgE. An increase was also seen for lgE in boys of OC users, however this was not statistically significant. These findings may be due to the estrogen/androgen ratio in boys, which is known to modulate the activities of all cells involve in an 90 . 32 . . . . Immune response . The pattern of androgenic and estrogenic stimulation during early development (in utero) 33 may also be of importance. Further studies are needed to examine these hypotheses. In conclusion, our study supports the notion that there are sex-related differences in the susceptibility to allergic diseases. Furthermore, the study adds evidence to the idea that maternal OC (oral contraceptive) use may have immunomodulatory effects in offspring. There are yet unanswered questions. Studies are needed to elucidate the mechanisms by which sex steroid hormones alter immune function. In addition, clarity is needed on how the levels of sex steroids correlate with disease severity. An idyllic setting to begin to answer these questions is that of a follow-up study where women are enrolled early in pregnancy and hormone levels determined from samples taken at different time-points (at least 3) during pregnancy. Cord immune marker of allergy should then be quantified and the association between hormone levels and these markers assessed. In addition to testing for an overall effect, analyses should be stratified by sex to get at possible sex-related differences. 9] References 10. 11. Beasley R, Crane J, Lai CK, Pearce N: Prevalence and etiology of asthma. The Journal of Allergy and Clinical Immunology 2000;105:8466- 472. National institutes of Health, LaBl. NH: GLOBAL STRATEGY FOR ASTHMA MANAGEMENT AND PREVENTION. In, 2005. Guilbert T, Krawiec M: Natural history of asthma. Pediatric Clinics of North America 2003;50:523-538. Frye C, Mueller JE, Niedermeier K, stt M, Heinrich J: Maternal oral contraceptive use and atopic diseases in the offspring. Allergy 2003;58:229-232. Brooks K, Samms-Vaughan M, Karmaus W: Are oral contraceptive use and pregnancy complications risk factors for atopic disorders among offspring? Pediatric Allergy and Immunology 2004;15:487-496. stt M, Dold S: Is asthma an endocrine disease? Pediatric Allergy and Immunology 1997;8z200—204. Xu B, Jarvelin MR, Hartikainen AL, Pekkanen J: Maternal age at menarche and atopy among offspring at the age of 31 years. Thorax 2000;55:691-693. Vihko R, Apter D: Endogenous steroids in the pathophysiology of breast cancer. Critical Reviews in Oncology/Hematology 1989;921-16. Apter D, Reinila M, Vihko R: Some endocrine characteristics of early menarche, a risk factor for breast cancer, are preserved into adulthood. International Journal of Cancer. Journal International Du Cancer 1989;44:783-787. Moore JW, Key TJ, Wang DY, Bulbrook RD, Hayward JL, Takatani 0: Blood concentrations of estradiol and sex hormone -binding globulin in relation to age at menarche in premenopausal British and Japanese women. Breast Cancer Research and Treatment 1991;18 Suppl 1:S47-50. SAS Institute: Statistical Analysis System, Version 8. In Institute S (ed):Gary, NC, 2002-2003. 92 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Xu B, Pekkanen J, Husman T, Keski-Nisula L, Koskela P: Maternal sex steroid hormones in early pregnancy and asthma among offspring: a case- control study. The Journal of Allergy and Clinical Immunology 2003;112:1101-1104. Inman RD: Immunologic sex differences and the female predominance in systemic lupus erythematosus. Arthritis Rheum 1978;21:849-852. Bouman A, Heineman MJ, Faas MM: Sex steroid hormones and the immune response in humans. Hum Reprod Update 2005;11:411-423. Seli E, Arici A: Sex steroids and the immune system. Immunology and Allergy Clinics of North America 2002;22:407-433. Shames RS: Gender differences in the development and function of the immune system. Journal of Adolescent Health 2002;30:59-70. Whitacre CC, Reingold SC, O_Looney PA: A gender gap in autoimmunity. Science 1999;283:1277-1278. Piccinni MP, Giudizi MG, Biagiotti R, Beloni L, Giannarini L, Sampognaro S, Parronchi P, Manetti R, Annunziato F, Livi C: Progesterone favors the development of human T helper cells producing Th2-type cytokines and promotes both lL-4 production and membrane CD30 expression in established Th1 cell clones. J Immunol 1995;155:128-133. Fox HS, Bond BL, Parslow TG: Estrogen regulates the IFN-gamma promoter. J lmmunol 1991;146:4362-4367. Whitacre CC: Sex differences in autoimmune disease. Nature Immunology 2001;2:777-780. Klein KO, Baron J, Colli MJ, McDonnell DP, Cutler GB, Jr.: Estrogen levels in childhood determined by an ultrasensitive recombinant cell bioassay. J Clin Invest 1994;94:2475-2480. ' Garnett SP, Hogler W, Blades B, Baur LA, Peat J, Lee J, Cowell CT: Relation between hormones and body composition, including bone, in prepubertal children. American Journal of Clinical Nutrition 2004;80:966- 972. Sears MR, Burrows B, Flannery EM, Herbison GP, Holdaway MD: Atopy in childhood. |. Gender and allergen related risks for development of hay fever and asthma [see comments]. Clinical and Experimental Allergy 1993;23:941-948. 93 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Maziak W, Behrens T, Brasky TM, Duhme H, Rzehak P, Weiland SK, Keil U: Are asthma and allergies in children and adolescents increasing? Results from ISAAC phase I and phase III surveys in Munster, Germany. Allergy 2003;58:572-579. Obendorf M, Patchev VK: Interactions of sex steroids with mechanisms of inflammation. Curr Drug Targets Inflamm Allergy 2004;3z425—433. Akdis M, Blaser K, Akdis CA: T regulatory cells in allergy: novel concepts in the pathogenesis, prevention, and treatment of allergic diseases. J Allergy Clin lmmunol 2005;116:961-968; quiz 969. Stock P, Akbari O, DeKruyff RH, Umetsu DT: Respiratory tolerance is inhibited by the administration of corticosteroids. Journal of Immunology 2005;175:7380-7387. Jansson L, Holmdahl R: Estrogen-mediated immunosuppression in autoimmune diseases. Inflamm Res 1998;47:290-301. Stallman PJ, Aalberse RC: Quantitation of basophil-bound lgE in atopic and nonatopic subjects. Int Arch Allergy Appl lmmunol 1977;54:114-120. Wada T, Toma T, Shimura 8, Kudo M, Kasahara Y, Koizumi S, Ra C, Seki H, Yachie A: Age-dependent increase of lgE-binding and Fcepsiloan expression on circulating basophils in children. Pediatr Res 1999;46:603- 607. Lander F, Meyer HW, Norn S: Serum lgE specific to indoor moulds, measured by basophil histamine release, is associated with building- related symptoms in damp buildings. Inflamm Res 2001;50:227-231. Cutolo M, Seriolo B, Villaggio B, Pizzorni C, Craviotto C, Sulli A: Androgens and estrogens modulate the immune and inflammatory responses in rheumatoid arthritis. Ann N Y Acad Sci 2002;966:131-142. Martin JT: Sexual dimorphism in immune function: the role of prenatal exposure to androgens and estrogens. Eur J Pharmacol 2000;405:251- 261. Jaakkola MS, Ieromnimon A, Jaakkola JJ.K: Are atopy and specific lgE to mites and molds important for adult asthma? J Allergy Clin lmmunol 2006; 117:642-648 94 0.0 E 28> 2-0 00¢ o.m~ 900> w 0.0.. 0.8 98> > 00:06-02 08 mg 00> >82 0. E 0.0 00> 05080 .0>0.. 02 0.2 9% >0: 3.. 0.0 we, 0552 .0505). 0N we 00.00:). 0.8 in 00> 20 ed m.v 9.00.5. 0.? 0.00 00> >00 5000090 0.0.00 00: 02000090000 .90 80.0.05. :0 «.8 >00 x0m as as €89. somuzv 900 .90me 002:8. 0:0 02000090000 .90 .0599: 5.2, 030505 9.00 2.0000508 .930: .0 030.0 t0...00 0.00.0..00 05 .0 8.6.0.0905 0200:0000 ... 0.00... 95 0.0 0.0 9.00.5. 0.00 0.00 0 :05 0.0.2 0.0V 0.0.. 0-? ...: 0.0. 0 00.003. 0500000090 .900 00 :00930 0.0V 00? 0A 0.: 0.: 0 -F 0.0 0.0 0:02 05:2: N. .00. 05 0:000 0:00002. .0 09:32 me 0.. 0500.2 0.0 for 00 :05 0.0.2 500 ......0 om-.. N00 0. .0 0:02 $00 .00 0000006. 05:08 N? 000. 05 0:030 0600 0.2.00 05 :_ 0:0.0E0 0>.000n. o\o o\o .vmmuc. .000nz. 900 0:00.008 0::EE. 0:0 0>00009E00 00.00 0.....000508 .90 .0599: 5.? 020.0000 .930: n.0 0:000 :0000 0:90:00 000 n.0 000000009000 0200:0000 0000:3000. ., 0.00 .. 96 ...000 I 0.0. ..00 ...000 I ..0. 0.00 0.007000. 0.00 .0.0.0 I ..0. 0.00 .0000 I 0.0. 0.0 .0000 I 0... ..00 .000. I 0.0. 0.00 .0000 I 0... 0.00 .0.0.0 I 0.0. 0.00 0000.00. ...0 0000-00. ..00 ...0000... 0.0 .000 I 00. 00. .000 I 00. 0.... .000 I 00. ...0. .000 I .0. ..00. .000 I 00. 0.00. .0000I00. 0.00. .000 I 00. 0.00. .0.0 I 00. 0.0. ..00 I 00. ..00. .00000. 0.00. .00000. 0.00. .00000. 0. .0. . .00 I 00. ..00. .000 I 00. 0.00. .000 I 00. 0.00. .000 I 0.. 0.00. .000 I 00. 0.00. .000 I 00. 0.00. .00. I .0. 00. .000 I 00. 0.00. .00 I 00. 000. 000-00. 0.00. .000. I 000. 0.0.. .0.0. 0 :9. 0.0.2 .000. I 000. 0.00.. .00. 0-. .000. I 000. 0.00.. .00. 0 00.003. 0500000090 .90. .0 :0..90n. .000. I 000. 0.0... .00. 0A .000. I 000. 0.00.. .000. 0 -. .0000. I .00. ..000. .00. 0.52 05:0... 0. .00. 0... 0:..00 0:0..00.:. .0 00:52 .000. I 000. 0.000. .0. 00 :0... 0.0.2 .000. I 00.. 0.00.. .00. 00-. .000. I 00.. 0. . . .. .000. 0002 900 .00 0000.006. 00.:0E N. .00. 0... 0:..00 0E0: 0.0..00 0... :. 0:...0E0 020000 800.000. 0.00.. .0.. 98> 0.0 .00... M9 30.00.. .29 .0000. 0.00. 800.000. .. .0.. .00.. 38> 0 E000. 0.00. 600.00.. 0.00.. .00.. 28> 0 0000.0-00< 30.0.... <0_ 30.0.... 00_ 0.8800 .00.0..>00 >0 0:..000.00:0EE. .0. 000.05 00 .-0 0:0 :00... 0.000.000 00.0000 00:00 ”N 0.00... 97 .000 I .00. 0.00 .000 I 00. 00. .0000 I 00. 00. .000 I 000. 000 £00 I 000. 0.000 .000 I 000. 0. .00 .0. .00 I .00. 0.000 ..00 I 000. 0.000 ..00 I 000. 000 .0000. I 00.0. 0.0.00 .0000. I 0000. 0.0.00 .0000. I 00.0. 0.0000 .0. 00 :0... 0.0.2 .00.. 00-. .000.0002 900 .00 0000.000. 00.00... N. .00_ .000 I 000. .00 . .00 I 000. 0.000 .000 I 000. 0.000 .000 I 0.000. 0.000 .000 I 0.000. 0.000 .000 I 000. 0.000 .000 I 00 0.000 . ..00 I 00 .0000 .000 I 000. 0.000 m m .000 I 000. 0.000 .000 I 000. 0.000 .000 I 000. 0.0 .0 .000 I 000. 000 .000 I 000. 0.000 .000 I 000. 0.000 ..00 I 000. 0.000 .000 I 000. 0.000 .000 I 000. .00 .000 I 000. 0.000 .000 I 000. ..000 .000 I 000. 0.000 .000 I 000. 0.000 .000. I 00. 0.0.0 .000 I .0. 0.0. .00 I 00. 000. .000 I 00. 0.00. .000 I 00. 0.00. .000 I .0. ...0. .000 I 00. ..00. .000 I 00. 0. .00 .000 I 00. 0.000 .000 I .0. 0.00. .000 I 00. 0.000 00. 00000 00.00 00:00 00. 0. 00200.0 020000 .0000. I 0000. 0.. .00 .0000. I 0000. 0.00.0 .0000. I 0000. ..0000 .0000. I 0000. 0.0000 .000.. I 00.0. 0.0000 .000.. I 0000. 0.0000 .0000. I 0000 0.0000 . .00.0. I 0000. 0.0.00 .0000. I 0000. 0.0000 .0.. 98> 0.0 .00.. 98> 0 .00.. 98> 0 000 02.0000..000 _0.0 050.02 .000.. I 0000. 0.0000 .0000. I 0000. 0.0000 0:000000 00 .0000 m0_ 0300.0 0___0000_00m 3.0.x .008 :00 __..000_000 3.0.x 0:00 0020 0.22. 0000.0-00< ..00. oz ..0. 00> >00.0 .00.0.02 .00. oz .00. 00> _._0 .00.. 02 >00 ..0. 00> .00.. _._0 .00..>00 x0w 0.000>00 .00.00>00 >0 m9 000000 05000000 000 02.000.00.000 050005000 .__00 0020 0.0.3 .0. 000_0> 00 .-m 000 0000. 00.0.0000 000000 .00000 .0 0.00-.- 98 .000 I 000. 0.000 .000 I 000. 0.000 .000 I 00. 0. .0. 400.0. I 0000. 0.0000 .0.0. 0 :0... 0.05. .000 I 0.0. 0.000 .000 I 000. ..000 .000 I 00. 0.00. .0000. I 0000. ..0000 .00. 0-. .000 I ..000. 000 ..00 I 000. 0.000 .000. I 00. 0.00. .0000. I 0000. 0.0000 .00. 0 .3003. 0500000005 _0.0. .0 0000.00 .000 I 000. 0.000 .000 I 000. 0.000 .000 I 00. 0. .00 .0000. I 0000. 0000 .00. 0A .000 I0.000. 0.000 .000 I 000. 0.000 .000 I 00. ..00. .000.. I 0000. 0.0000 .000. 0 -. .000 I 000. 0.000 .000 I 000. 000 .0.000 I 0.00. 0.00. .000.. I 00.0. 0.0000 .00. 0002 00000 00.000. N. .00_ 00. 05000.5 .0 000.02 0:000000 01.0.0... .008 30,0... 00 .0000 m0_ 0_000.0 0___0000_00m =00 __0000_00m 0:00 0020 0:03 0.00050 .00.00>00 >0 m9 000000 05000000 000 02.000.00.000 050005000 .__00 0020 0.0.3 .0. 000_0> 00 .-m 000 000E 00.00.000 050000 .00000 ".0000_.000. 0 0.00... 00020.0. 00. 00 000 0>_.0000..000 _0.0 _00.0.00. 00 0. 00.000.00.00.-. 0 00 00000 00.90 .. 0500000005 _0.0. .0 0000.00 000 00.00.: .0.. .00_ 00. 00000 00080.5 .0 .00.000 00.00:. N. .00_ 00. 00000 00.00 00:00 00. 0_ 00200.0 020000 .000 00:00 $00.0 _00.0.0E .0. 00.00.00. 0 .m 00 mm. mm. .0. S. .00. 3.. 0z 0 00.00.04. .00 m... .0. mm. .mm. 00. .mm. mm .. 00> 0m 0m 0m. 0.. 0m. 0m. 00.. 000. 02 000.0 v. 00 mm. 0.. 0.. 00. 00.. 000. 00> 000 0>_.0000..000 _0.0 00.0.02 2 005 0>0m 005 0>0m 005 0>0m 005 0>0m 00.50.00 .00... 09 .00.0.... 29 .00.0.... <9 30.00.. 00. 00__000_00000.0._ >0 000 0>_.0000..000 _0.0 00.0.0.2 "v 0.00... 100 0800.0. 00. 00 000 0>_.0000..000 _0.0 _00.0.0E 00 0. 00.000.00.00.-. 0 00 00000 00.0V0 . 020000000... _0.0. .0 0000.00 000 00.00.: N. .00_ 00. 00000 00260.0. .0 .0060: 00.00:. N. .00_ 00. 00000 00.00 00:00 00. 0_ 02200.0 020000 .000 00:00 .>00.0 _00.0.00. .0. 00.00.00. 0 N00 000 000 000 0N. 00N 0000 0000 02 a 00.00_0< .00. .. .0 ..00 000 .N. .mN .0000 N. .0 00> 000 000 N00 .00 .0. NON 0N8 0.00 02 000.0 ..00 000 .00 000 N0. 00N 0000 00N0 00> 000 0>00000..000 _0.0 00.0.00. 2 005 0>0m 005 0>0m 005 0.80 005 0>0m 2000000 0000.0 .100... .008 3.0... 00.00.00 00 .0000 09 2__0000_00m_ =00 __0000_0om 0:00 0020 0._0>> 00_I 000000 2:000000 000 02.000.00.000 050000000 .__00 0020 0002. >0 000 0>00000..000 _0.0 _00.0.0_>_ ”0 200... 101 COMPONENT THREE In utero exposure to SHD affects allergic outcomes in infancy 102 Are oral contraceptive use and pregnancy complications risk factors for atopic disorders among offspring? Kevin Brooks 1, Maureen Samms-Vaughan 2, Wilfried Karmaus 1 1 Department of Epidemiology, School of Human Medicine, Michigan State University, East Lansing, United States; Department of Obstetrics, Gynecology and Child Health, University of the West Indies, Mona campus, Jamaica. 103 Abstract In utero programming of atopic manifestations has been suggested. We investigated the association between oral contraceptive (OC) use before, and complications during pregnancy (GDP) and asthma, along with other atopic manifestations. The study is based on neonates from Kingston and St. Andrew, a geographic sub-cohort from the Jamaican Perinatal Morbidity, Mortality Survey conducted in 1986-1987. Information on 00 use and CDP was extracted from maternal interviews and medical records. In a follow-up in 1997-1998, via interviews with mothers, trained nurses collected information on asthma/wheezing, coughing, eczema, and hay fever. Data, specific to this paper, from birth and 11 - 12 years of age was available for a total of 1040 of the 1720 members of the geographic sub-cohort. Using logistic regression, controlling for confounders, we estimated adjusted odds ratio (aOR) and corresponding 95 % confidence intervals (CI). For asthma or wheezing, and coughing, aOR for 00 use were 1.81 (95% Cl 1.25- 2.61), and 2.72 (95% CI 1.41-5.24), respectively. CDP was only shown to be a significant risk factor for hay fever. Additionally, a higher number of older siblings were protective for hay fever. The results suggest that asthma in childhood may be programmed in utero. Key words: asthma; eczema; hay fever; atopy; oral contraceptives; pregnancy complications 104 The prevalence of allergies is increasing in many parts of the world, and asthma has become the most common chronic disease of childhood. The etiology of asthma and allergic disease remains poorly understood, despite considerable research 1. Recent work has expanded to include the study of novel factors such as in utero and perinatal exposures that may program initial susceptibility 2. Some authors have reported that maternal estrogen levels and maternal health complications during pregnancy are associated with asthma and other atopic manifestations later in life 3'6. The most appropriate design to test an association between perinatal conditions and asthma in offspring is a longitudinal study commencing with mothers in the antenatal period and following their offspring through childhood. The Jamaican Perinatal Mortality and Morbidity Survey (JPMMS) 7, and the subsequent follow— up studies offered a unique opportunity for testing these hypotheses. The JPMMS, originally designed to ascertain causes of maternal and neonatal mortality and morbidity, obtained detailed information about maternal lifestyle before and during pregnancy, the labor and delivery process. The subsequent follow-up studies of the children were designed to identify factors promoting and preventing optimum childhood outcomes, including child health. 105 Women who participated in the JPMMS in 1986 were contacted at six weeks postpartum and again eleven to twelve years later. We investigated whether in utero and perinatal factors contributed to the development of asthma and other atopic manifestations in children 11 to 12 years of age. Specifically, we tested the association between two risk factors: (1) Maternal use of oral contraceptives (OC) before pregnancy (2) Maternal complications during pregnancy (GDP) and the prevalence of four atopic manifestations: (a) Asthma/wheezing, (b) frequent nighttime/early morning cough, (0) eczema, and (d) hay fever/sinus problems/other allergy Methods Study population The Jamaican Perinatal Morbidity and Mortality Survey included all pregnant women who had a live or stillbirth during the two-month period from September 1St to October 315‘, 1986. Women were interviewed and their babies examined, usually within the first 48 hours after delivery. Data were gathered during this phase using the main questionnaire (MQ). A total of 10392 babies or 94 % of the births in the two-month period were identified and included in the study (main cohort study) 8. The first follow-up study of the children occurred at six weeks of age when 8,800 (84.7 %) mother-child pair from the main cohort were 106 interviewed. Information on breastfeeding and the infant’s health was obtained using the first follow-up questionnaire (FF UQ). These 8800 mother-child pairs were used as the baseline cohort (entire island) for the analyses. Follow-up A geographic sub-sample of 1,720 eleven to twelve-year-old children, representing those residing in Kingston or St. Andrew, the two most urban of Jamaica’s fourteen parishes, was selected for a second follow-up study. In a cross-sectional survey of these children, data on health outcomes were collected from participating parents or guardians (n=1,163), mostly the mothers, and recorded using a second follow-up questionnaire (SFUQ). We linked the three datasets using a unique identification number assigned at birth and dates of birth for both mother and child. Questionnaires and examinations During the JPMMS, trained nurse midwives administered face-to-face interviews with mothers and conducted a brief examination of the babies shortly after birth. Babies were also weighed and measured. Hospital delivery notes and maternal health records were consulted to verify and supplement the data obtained by interview. The MO provided data on past obstetric history and complications during pregnancy (CDP), labor and delivery. Data on the current status of the baby (alive/healthy, alive/ill, dead or don’t know) and what the baby was currently being fed (breast, breast and formula or formula only) was extracted from the FFUQ. Information on the presence of asthma or 107 wheezing, coughs, eczema and hay fever/ sinus problem/ other allergy in the children at 11 to 12 years of age was collected from the general health section of the SFUQ. Trained nurse interviewers administered the questionnaire to either the parents or guardians. Definition of variables Maternal age at birth was calculated from the mother’s date of birth and child’s date of delivery. Based on questions from the MD, a yes, no, or don’t know response was used to define the following: - Did you smoke tobacco regularly at any time during this pregnancy? - Have you ever used contraception? If yes, which methods have you used? (Contraceptive pill was among a list of contraception.) - Were you admitted to hospital or rural maternity center during this pregnancy but before going into labor? (Used to determine maternal hospital admissions.) - Are any of the following services available within 1-2 miles walking distance of where you live?’ (Health center was among a list of services. - Used to determine access to health care.) Mother’s education level was grouped into three categories for the purpose of this study (less than secondary, secondary, and more than secondary educafion) Maternal CDP was defined as a positive response to any of the following quesfions: 1) Did you have vaginal bleeding in the first 28 weeks of pregnancy? 2) Did you have vaginal bleeding after 28 weeks (7 months)? 108 3) 4) 5) 5) 7) After starting antenatal care, were you referred for treatment during pregnancy for any reason? Were you referred during pregnancy for any reason? Was the mother diagnosed as having hypertension, pre-eclampsia or eclampsia during this pregnancy? Did the mother have any of the following during this pregnancy; epilepsy, eclamptic fits or heart disease and lastly, Did the mother have any other complications, disorder or serious illness during this pregnancy? Responses to the first five questions above were obtained by interview, while the remainder was obtained from health records. Maternal infection during pregnancy was defined as a positive answer to any of the following question based on information from medical records: 1) 2) Did the mother have a vaginal discharge or infection during this pregnancy? Did the mother have urinary tract infection, tuberculosis, rubella (German measles), gonorrhea, syphilis, a positive Venereal Disease Research Laboratory test or genital sores or blisters during pregnancy? The following variables were used to define maternal health complications during labor or delivery (positive if indicated in the medical records): 1) 2) 3) Did the mother have eclamptic fits? Did she hemorrhage? Was labor obstructed? 109 4) Did the uterus rupture? 5) Was mother transferred during labor? 6) Did the cord prolapse? 7) Was there meconium in the liquor? 8) Were there any other complications? The number of preceding live births determined birth order of the index child. The child’s birth weight was also recorded during this period (MO) and grouped into three categories for the purpose of our analyses (<2.5kg, 2.5-4.0 kg and >40 kg). The variable child's illness or health problems in first week of life was defined as a positive response to any of the following questions from the FF U0: 1) Was the baby referred to a medical officer? 2) Was the baby admitted to hospital or special care baby unit? 3) Were any abnormal symptoms noted in the baby? Outcomes of interest at 11 to 12 years of age were ascertained from the SF U0. The original question to the parents was, ‘Has your child ever had...’ This question was followed by a list of conditions; including asthma or wheezing, frequent nighttime or early morning cough, eczema, and hay fever sinus problem or other allergy. Statistical analysis We tabulated the prevalence of the four atopic manifestations for the different ante- and perinatal exposures. Differences in proportions between the different sub-samples were assessed. Adjusted odds ratios (aOR) and their 95 % 110 confidence intervals (95% Cl) were estimated by logistic regression analysis. Statistically, we controlled for maternal infection, hospital admissions and smoking during pregnancy, as well as maternal complications during labor/delivery, induced labor, anesthetic/analgesic during labor, gender, birth weight, breastfeeding, child’s illness in the first week of life, access to health care and mother’s level of education. These potential confounders were included based on previous publications and after satisfactorily, checking for collinearity between all predictors (risk factors and all confounders) 9. We estimated aOR for all four outcome variables independently. Further steps were taken to determine whether having both, asthma or wheezing and frequent nighttime or early morning cough, was associated with 00 use and complications during pregnancy. All calculations were carried out using SAS for Microsoft Windows (release 8.2) 10. Results From the three data sets, we successfully linked 1040 (60.4 %) of the 1720 available mother-child pairs from the Kingston and St. Andrew sub-cohort. Compared characteristics between exposure variables from the baseline cohort (entire island) and the linked data set (urban population) are presented in Table 1. Approximately 50 % of mothers used OC, and approximately, 50 % had CDP. In the baseline sample, maternal education was lower than in our sub-sample (Table 1: less than secondary level of education - baseline: 35.6%, subsample: 111 25.4%). The proportion of children born with low birth weight (<2.5 kg) was higher in the more urban setting of Kingston and St. Andrew (12.2 %), where the second follow-up cohort originated. A significant rural-urban difference was also indicated by the difference in breastfeeding categories. When comparing the follow-up group for whom health information was available, (n=1163 of 1720) with the linked sample (n=1040) there was no substantial difference (asthma or wheezing: 17.0 % vs. 16.8 %, frequent nighttime or early morning cough: 4.8 % vs. 5.3 %, eczema: 6.5 % vs. 6.3 %, hay fever or sinus problem: 19.6 % vs. 20.1 %), Table 2 presents the extent to which atopic manifestations occur with each other. The proportion of atopic disorders in relation to in utero risk factors is presented in Table 3. Children of mothers, who used, compared to those who did not use OC before pregnancy, had a higher prevalence of asthma (20 °/o vs. 13.4 %), frequent nighttime or early morning cough (7.4 % vs. 3.6 %) and eczema (7.4 % vs. 5.5 %), Asthma and wheezing, as well as hay fever or sinus problems were more common in children whose mothers had complications during pregnancy (CDP) than in those who did not. Of the perinatal risk factors, induced labor was related to a higher prevalence of eczema (11.4 % vs. 6 %, Table 4). Children of mothers who had complications during labor or delivery had a greater proportion of frequent nighttime or early 112 morning cough (8.1 °/o vs. 4.8 %), In children who had illness or health problems in their first week of life, eczema (10.5 % vs. 5.3 %) and hay fever sinus problem or some other allergy (27.4 °/o vs. 18.3 %) were more frequently reported at 11- 12 years of age. Information on breastfeeding was available for only 763 children (Table 4). For this reason, breastfeeding was not considered when testing the hypotheses with a full model. A possible confounding effect of breastfeeding was investigated in additional analyses. Adjusted odds ratios for asthma or wheeze, and frequent nighttime or early morning cough are reported in Table 5 while those for eczema and hay fever, sinus problems or other allergy are reported in Table 6. Maternal 00 use before pregnancy was significantly associated with both asthma (aOR = 1.81) and frequent nighttime or early morning cough (aOR = 2.72) in the offSpring. The association between maternal OC use before pregnancy and asthma was stronger for girls (aOR = 2.02, 95% Cl 1.21 -3.37) than for boys (aOR = 1.60, 95% CI 0.92- 2.76). Regarding eczema, hay fever, and sinus problem or other allergy at age 11-12 years of age, the confidence limits of the aOR for maternal OC use included the null-value (Table 6). Additionally, it is well established that ‘All that wheezes is not asthma’ 11 therefore we conducted further analysis with a combined variable, defined as the 113 conditions asthma or wheezing and frequent nighttime or early morning cough occurring concurrently. Of children whose mothers used CC, 5.1 % had asthma or wheezing, and coughing compared to 1.9 % of children of mothers who did not. This association was then assessed controlling for confounders in the full model. When compared to the two separate effects, the association of the combined outcome was stronger (aOR=3.32, 95% CI 1.33-8.32). Complications during pregnancy gained statistical importance only for hay fever or sinus problems (Table 6). When type of feeding was introduced into the models, analyses were then based on 763 children. For CDP, this resulted in an increased odds ratio for hay fever (aOR=1.72, 95% CI 1.13-2.63) (data not shown). However, the significant odds ratio for oral contraceptive use did not change. A higher number of older siblings (birth order) showed a significant protective effect for hay fever (Table 6). For asthma or wheezing and for coughing, similar association was found though not statistically significant (Table 5). Discussion The findings support the hypothesis that children whose mothers used OC (oral contraceptive) before pregnancy, are at a higher risk for developing asthma or wheezing, and frequent nighttime or early morning cough at 11 to 12 year of age. The data also supported the hypothesis that a maternal complication during pregnancy is a risk factor for atopic manifestations in offspring. 114 The association between OC use and asthma or wheezing and coughing is in concordance with a suggestion of stt et al. who, based on aggregative data on OC sales and hospital discharge data, were the first to hypothesize that maternal use of OC is a risk factor for asthma in the offspring 3. To the best of our knowledge, the present study is the first to prospectively assess the association between, maternal OC use and asthma or wheeze in offspring, using individual data. The most frequently used 00 in Jamaica during the study period was the Pearl ®, in which the active components were ethinyloestradiol (30 pg) and Ievonorgestrel (150 pg), an estrogen and progesterone respectively. It has been suggested that women who use OC have higher estrogens levels after discontinuing their use 12, however, the evidence is not consistent 13. The OC-atopy association may be explained by endocrine effects on T-helper 2 (Th2) cells. Th2 cells produce interleukin (IL)-4, lL-5, and lL-13, which promote the production of IgE and eosinophil infiltration in the ainivays and ultimately orchestrate the development of allergy and asthma 14-16. stt and co-workers suggest that estrogen might enhance the activity of Th2 type cytokines 3, predisposing the fetus to atopic disorders later in life. Furthermore, in vitro studies suggest that progesterone promotes the preferential development of Th2- type cells and to function as a potent inducer of the production of the Th2 type 115 cytokines 1749. Michel et al. reported that lgE levels in cord blood were significantly increased among neonates whose mothers had taken progesterone . 20 during pregnancy We suggest two possible scenarios for the endocrine mechanism, which may explain our OC-atopy association. Either maternal exposure to estrogen via OC use may trigger an increase Th2 activity continuing into pregnancy (prolonged Th2 effect), consequently predisposing the fetus to a higher risk of developing atopy later in life, a Th2-mediated disorder 15’21’22. Alternately, women who used OC continuously, discontinuing before becoming pregnant, may have created an environment of higher estrogen or progesterone levels (prolonged endocrine effect), resulting in an increased Th2 activity impacting the fetus transplacentally. Evidence for the prolonged Th2 effect is supported by findings that women exposed in utero to the estrogen diethylstilbestrol, were found to have alterations in their T-cell-mediated immunity 23. In addition, children of mothers who used OC in the 18 months prior to being pregnant were found to be more likely to have eczema at age five 24. This finding was later corroborated by Xu et al. 25. Evidence in support of an prolonged endocrine effect: Xu et al. reported that atopy in adults was more common among those whose mothers experienced an early age of menarche 4. A likely explanation for the atopy — age at menarche 116 association is that early age of menarche is associated with higher levels of . 1 - - estrogen In adult women 3’ 26 28. Hence, the fetus of women wrth an early age at menarche may experience higher exposure to estrogens. This could then lead to the programming of the immune system of the fetus affecting atopic status later in life 29. In a cross-sectional study, Frye et al. 30 provided data to support the prevailing OC - atopy association. They however downplayed the significance of this finding on the basis that similar results were found in the offspring of women who used OC exclusively after they delivered. It is noteworthy to accentuate the fact that Frye and co-workers used a cross-sectional design wherein OC use was assessed retrospectively. This design will no doubt be plagued with biases. These results may be an artifact of ‘reverse causation’ introduced by behavior modification of the parents. An example of this is women in the ‘exclusive OC use after birth’ group may have chosen to resume the use of OC after the index child developed atopic eczema. The reason is that a child with eczema will require more attention at nights. Therefore, a mother with a demanding child may decide to delay having another by using OC. There is a paucity of studies on the relationship between maternal complications during pregnancy and the risk of having asthma or any atopic disorders. Nafstad et al. in a population-based, four-year, cohort study involving 2531 children. Asthma was assessed at age four 5. The authors reported that uterus-related 117 complication (antepartum hemorrhage, preterm contractions, insufficient placenta, and restricted growth of the uterus) increased the risk of having asthma (OR = 3.0, 95 % CI 1.8-5.4) and allergic rhinitis (OR = 2.9, 95 % CI 1.6-5.2). Annesi-Maesano et al. also found maternal complications during pregnancy to be a risk factor of asthma (OR = 2.01, 95 % CI 1.52-2.67) in the offspring using a large British birth cohort (4065 natural children of 2583 mothers) 6. More 31,32 recently, two studies have supported these findings , one by using data on Norwegian live births (1967-1993, n=1,548,429) 31. Our study found maternal complications during pregnancy (CDP) to be a significant risk factor for hay fever and sinus problems (aOR = 1.5, 95 % CI 1.05-2.16, p=0.03). Further analyses for items included in the predictor complication during pregnancy (CDP) indicated that only the variable ‘have there been any other complications, disorder or serious illness during this pregnancy?’ conferred a significantly increased risk for asthma in the child (p< 0.001), but none of the specific items. This finding is in agreement with a recent report by Nafstad et al. who also found that pregnancy complications in general might present risk factors of stress for the developing fetus 31. It might be that CDP (complications during pregnancy) is reflective of factors such as a threatened pregnancy, found to be associated with frequent wheezing 33, which will alter the Th1 /Th2 balance 34'35. This hypothesis warrants further investigations with adequate perinatal and breastfeeding information. 118 Our study adds to the body of evidence, which suggest that the number of siblings, in particular older siblings, is important in the etiology of atopic disorders 28. We found number of older siblings to be protective for hay fever or sinus problems (Table 6). Number of older siblings and oral contraceptive use were statistically significantly correlated (rspe,,.rm,,.n = 0.3, p=0.0001, n=1020). The lack of association between low birth weight and asthma is in contrast to some previous studies 636-38 and in agreement with others 3942. If is worth mentioning that the finding from our Afro-Caribbean cohort is similar to that of an African-American cohort 40. The strengths of the current investigation are in its prospective design, being derived from a large population based cohort, with all the exposure information collected during pregnancy, at birth and shortly after. This design presents a clear time-order, as outcomes were not assessed until approximately 11 years after. The impact of recall bias is also reduced. Additionally, controlling for a broad range of confounders minimize the possibility of these findings occurring purely by chance. A limitation of this study is the follow-up proportion of 60.4 %. This is largely explained by parental participation in the second follow-up study. Though 119 parental consent was obtained for the 1720 children evaluated using the child’s completed reports, only 1163 parents provided interview and questionnaire data. The significant differences found between the characteristics of some of the variables from the baseline cohort when compared with those of the linked dataset were expected. This, we infer, is due to the rural-urban difference between these groups. The difference between reported occurrence of CDP in the baseline cohort (38.4%) and our sub-sample from Kingston/St. Andrew (49.2%) is a possible source of selection bias. The baseline cohort has both rural and urban populations, whereas the sub-sample if predominantly urban. Different proportions of maternal complications in these samples can be explained by two factors, namely maternal education level and access to health care. For instance, of the women in our sub-sample who reported having complications during pregnancy, 3.6% were characterize as having low level education (less than secondary) and less access to health care compared to 8.9% of those with similar characteristics from the baseline cohort. Recently, McCaw—Binns et al reported that access to health care in Jamaica differs from rural to urban areas 43. Since these two factors (maternal education level and access to health care) seem to affect the reported proportion of maternal complications, and as the prevalence of asthma, eczema, and sinus problems/allergies increase with level of education, they were included as confounders in our models to minimize the impact of a selection effect (Tables 5 and 6). 120 Another limitation is the absence of information on how long before pregnancy OC was used and if it was the last method of birth control used. This limitation introduces a non-differential misclassification, which tends to underestimate the association between OC and asthma or wheezing, and coughing. To evaluate, whether the OC hypothesis holds true, we investigated data of children from pregnancies that occurred despite birth control (n=90), 42 with and 35 without using oral contraceptives. Asthma was more frequently reported at 11-12 years of age in children whose mothers used 00, 23.6 %, compare to 17.1 % in children whose mothers did not use OC when they conceived. We did not collect information on parental atopy and thus could not adjust for it in our model. While there is data showing parental history of atopy to be a risk factor for atopic disorders in the offspring, data supporting an association between parental history of atopy and oral contraceptive use is lacking and the plausibility of such an association is questionable. Hence, it is highly unlikely that parental history of atopy would have confounded the association between oral contraceptive and atopy, since confounding requires both, association with 44 exposure and outcome The present study was not originally designed to address our hypotheses. However, an advantage of the cohort study design is that data collected can be used to provide answers to newly emerging research questions. We therefore 121 lJfi-_ took advantage of the utility and efficiency of this study to test our respective hypotheses. Additionally, the unique nature of this cohort makes this the first study to test perinatal risk factors for asthma in an Afro-Caribbean population. Regarding coughing, we asked for ‘frequent night time or early morning coughing’, but did not include the adjective ‘dry'. Nevertheless, the validity of “early morning coughing’ as a marker for asthma has been found to be sufficient 45'46. On the other hand, it has been shown that bronchial hyperreactivity was not related to dry cough or to nocturnal cough 47. According to Chang, coughing and bronchoconstriction may be caused by different mechanisms. Hence, different asthma-like symptoms may indicate different features 48 The definition or measurement of asthma or wheezing may also be of concern as wheezing is a heterogeneous disorder 49 and not all that wheezes is asthma 11. The impact this misclassification may have, was assessed by combining the variables asthma or wheezing and frequent nighttime or early morning cough to create a compound outcome variable. The combined condition was less prevalent and represented a more specific state of bronchial reactivity. The association between the combined variable and maternal OC use before pregnancy was assessed and results showed a stronger association (aOR = 3.15, 95 % Cl 1.29- 7.71). Additionally, we investigated whether the association with asthma was more likely due to an atopic response. For this purpose, we 122 combined two variables: having both, asthma and allergies. The association between maternal OC and the new combined variable was stronger (aOR 2.26, 95% CI 1.18-4.35) compared to the association with only asthma (aOR 1.81, 95% CI 1.25-2.61). This strengthening suggests that the association is likely to have been the result of an atopic mechanism. In summary, this data contributes to existing literature suggesting that maternal oral contraceptive use may be important in the development of asthma in offspring. In particular, the data helps to address the ongoing question as to whether it is ‘hormones or hygiene’ 50 that is responsible for atopy. There are only a few existing studies that allow for the investigation of maternal OC use and the child’s risk of atopic manifestation. The hypothesis that oral contraceptive use may contribute to an increase of asthma in children as put forward by stt 3 is very thought provoking. We suggest this hypothesis be tested in future studies with more precise exposure assessment. Acknowledgements Funding for the Jamaican Perinatal Morbidity, Mortality Survey was provided by the International Development Research Center of Canada while the Jamaica Cohort study was supported by the Policy Development Unit of the Planning Institute of Jamaica. 123 References 10. 11. 12. ASHER Ml, KEIL U, ANDERSON HR, et al. International Study of Asthma and Allergies in Childhood (ISAAC): rationale and methods. The European Respiratory Journal : Official Journal of the European Society For Clinical Respiratory Physiology 1995;82483-91. BEASLEY R, CRANE J, LAI CK, PEARCE N. Prevalence and etiology of asthma. The Journal of Allergy and Clinical Immunology 2000;105:8466- 72. WJST M, DOLD S. ls asthma an endocrine disease? Pediatric Allergy and Immunology 1997;82200-4. XU B, JARVELIN MR, HARTIKAINEN AL, PEKKANEN J. Maternal age at menarche and atopy among offspring at the age of 31 years. Thorax 2000;55:691-3. NAFSTAD P, MAGNUS P, JAAKKOLA JJ. Risk of childhood asthma and allergic rhinitis in relation to pregnancy complications. J Allergy Clin lmmunol. 2000;106:867-73. ANNESl-MAESANO l, MOREAU D, STRACHAN D. In utero and perinatal complications preceding asthma. Allergy 2001;56:491-7. ASHLEY D, MCCAW-BINNS A, FOSTER-WILLIAMS K. The perinatal morbidity and mortality survey of Jamaica 1986-1987. Paediatr Perinat Epidemiol 1988;2z138-47. ASHLEY D, MCCAW-BINNS A, GOLDING J, et al. Perinatal mortality survey in Jamaica: aims and methodology. Paediatr Perinat Epidemiol 1994;8 Suppl 1:6-16. KLEINBAUM K, MULLER. Applied Regresion Analysis and Other Multivariable Methods, 1988. SAS INSTITUTE. Statistical Analysis System, Version 8. Gary, NC, 2000. MURRAY DM, LAWLER PG. All that wheezes is not asthma. Paradoxical vocal cord movement presenting as severe acute asthma requiring ventilatory support. Anaesthesia 1998;53:1006-1 1. BARBIERI RL, GAO X, XU H, CRAMER DW. Effects of previous use of oral contraceptives on early follicular phase follicle-stimulating hormone. Fertility and Sterility 1995;64:689-92. 124 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. MOORE JW, KEY TJ, WANG DY, BULBROOK RD, HAYWARD JL, TAKATANI 0. Blood concentrations of estradiol and sex hormone -binding globulin in relation to age at menarche in premenopausal British and Japanese women. Breast Cancer Research and Treatment 1991;18 Suppl 1:S47-50. MOVERARE R, ELFMAN L, STALENHEIM G, BJORNSSON E. Study of the Th1fTh2 balance, including IL-10 production, in cultures of peripheral blood mononuclear cells from birch-pollen-allergic patients. Allergy 2000;55:171-5. MAZZARELLA G, BIANco A, CATENA E, DE PALMA R, ABBATE GF. Th1/Th2 lymphocyte polarization in asthma. Allergy 2000;55:6-9. KUO ML, HUANG JL, YEH KW, Ll PS, HSIEH KH. Evaluation of Th1/Th2 ratio and cytokine production profile during acute exacerbation and convalescence in asthmatic children. Ann Allergy Asthma lmmunol 2001;86:272-6. PICCINNI MP, GIUOIZI MG, BIAGIOTTI R, et al. Progesterone favors the development of human T helper cells producing Th2-type cytokines and promotes both lL-4 production and membrane CD30 expression in established Th1 cell clones. Journal of Immunology (Baltimore, Md. : 1950) 1995;155:128-33. SZEKERES-BARTHO J, WEGMANN TG. A progesterone-dependent immunomodulatory protein alters the Th1fl’h2 balance. J Reprod lmmunol 1996;31:81-95. HAMANO N, TERADA N, MAESAKO K, et al. Effect of female hormones on the production of lL-4 and lL-13 from peripheral blood mononuclear cells. Acta Otolaryngol Suppl 1998;537:27-31. MICHEL FB, BOUSOUET J, GREILLIER P, ROBINET_LEVY M, COULOMB Y. Comparison of cord blood immunoglobulin E concentrations and maternal allergy for the prediction of atopic diseases in infancy. The Journal of Allergy and Clinical Immunology 1980;65:422-30. WARNER JA, JONES CA, WILLIAMS TJ, WARNER JO. Maternal programming in asthma and allergy. Clin Exp Allergy 1998;28 Suppl 5235-8; discussion 50-1. JONES CA, HOLLOWAY JA, WARNER JO. Does atopic disease start in foetal life? Allergy 2000;55:2-10. 125 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. BURKE L, SEGALL-BLANK M, LORENZO C, DYNESIUS-TRENTHAM R, TRENTHAM D, MORTOLA JF. Altered immune response in adult women exposed to diethylstilbestrol in utero. Am J Obstet Gynecol 2001;185:78-81. PETERS TJ, GOLOING J. The epidemiology of childhood eczema: ll. Statistical analyses to identify independent early predictors. Paediatric and Perinatal Epidemiology 1987;1z80-94. Xu B, JARVELIN MR, PEKKANEN J. Prenatal factors and occurrence of rhinitis and eczema among offspring. Allergy 1999;54:829-36. VIHKO R, APTER D. Endogenous steroids in the pathophysiology of breast cancer. Critical Reviews in Oncology/Hematology 1989;9:1-16. APTER D, REINILA M, VIHKO R. Some endocrine characteristics of early menarche, a risk factor for breast cancer, are preserved into adulthood. International Journal of Cancer. Journal International Du Cancer 1989;44:783-7. KIRCHENGAST S, HARTMANN B. Association between maternal age at menarche and newborn size. Soc Biol 2000;47:114-26. WESTERGAARD T, BEGTRUP K, ROSTGAARO K, KRAUSE TG, BENN CS, MELBYE M. Reproductive history and allergic rhinitis among 31145 Danish women. Clin Exp Allergy 2003;33:301-5. F RYE C, MUELLER JE, NIEDERMEIER K, WJST M, HEINRICH J. Maternal oral contraceptive use and atopic diseases in the offspring. Allergy 2003;58:229-32. NAFSTAD P, SAMUELSEN SO, IRGENS LM, BJERKEDAL T. Pregnancy complications and the risk of asthma among Norwegians born between 1967 and 1993. Eur J Epidemiol 2003;18:755-61. CALVANI M, ALESSANDRI C, SOPO SM, et al. Infectious and uterus related complications during pregnancy and development of atopic and nonatopic asthma in Children. Allergy 2004;59:99-106. STAZI MA, SAMPOGNA F, MONTAGANO G, GRANDOLFO ME, COUILLIOT MF, ANNESI-MAESANO l. Early life factors related to clinical manifestations of atopic disease but not to skin-prick test positivity in young children. Pediatr Allergy lmmun012002;13:105-12. NILSSON L, KJELLMAN NI, LOFMAN O, BJORKSTEN 8. Parity among atopic and non-atopic mothers. Pediatr Allergy Immunol 1997;8z134-6. 126 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. PICCINNI MP, MAGGIE, ROMAGNANI 8. Role of hormone-controlled T-Cell cytokines in the maintenance of pregnancy. Biochem Soc Trans 2000;28:212-5. SCHWARTZ J, GOLD D, DOCKERY DW, WEISS ST, SPEIZER FE. Predictors of asthma and persistent wheeze in a national sample of Children in the United States. Association with social Class, perinatal events, and race. Am Rev Respir Dis 1990;142:555-62. SVANES C, OMENAAS E, HEUCH JM, IRGENS LM, GULSVIK A. Birth Characteristics and asthma symptoms in young adults: results from a population-based cohort study in Nonivay. Eur Respir J 1998;12:1366-70. STEFFENSEN FH, SORENSEN HT, GILLMAN MW, et al. Low birth weight and preterm delivery as risk factors for asthma and atopic dermatitis in young adult males. Epidemiology 2000;11:185-8. GREGORY A, DOULL I, PEARCE N, et al. The relationship between anthropometric measurements at birth: asthma and atopy in childhood. Clin Exp Allergy 1999;29:330-3. OLIVETI JF, KERCSMAR CM, REDLINE 8. Pre- and perinatal risk factors for asthma in inner City African-American children. American Journal of Epidemiology 1996;143:570-7. SEARS MR, HOLDAWAY MD, FLANNERY EM, HERBISON GP, SILVA PA. Parental and neonatal risk factors for atopy, airway hyper-responsiveness, and asthma. Arch Dis Child 1996;75:392-8. KATz KA, POCOCK SJ, STRACHAN DP. Neonatal head Circumference, neonatal weight, and risk of hayfever, asthma and eczema in a large cohort of adolescents from Sheffield, England. Clin Exp Allergy 2003;33:737-45. MCCAW_BINNS A, STANDARD_GOLDSON A, ASHLEY D, WALKER G, MACGILLIVRAY I. Access to care and maternal mortality in Jamaican hospitals: 1993-1995. International Journal of Epidemiology 2001;30:796- 801. ROTHMAN KJ. Modern epidemiology, 1986. KUEHR J, FRISCHER T, KARMAUS W, MEINERT R, BARTH R, URBANEK R. Clinical atopy and associated factors in primary-school pupils. Allergy 1992;47:650-5. 127 46. 47. 48. 49. 50. FERRIS BG. Epidemiology Standardization Project (American Thoracic Society). Am Rev Respir Dis 1978;118:1-120. STRAUCH E, NEUPERT T, IHORST G, et al. Bronchial hyperresponsiveness to 4.5% hypertonic saline indicates a past history of asthma-like symptoms in Children. Pediatr Pulmonol 2001;31:44-50. CHANG AB. Cough, cough receptors, and asthma in Children. Pediatr Pulmonol 1999;28:59-70. DELL S, TERESA, T. Breastfeeding and asthma in young children. Arch of Pediatric Adolescence medicine 2001;155:1261-1265. RANGARAJ S, DOULL l. Hormones not hygeine? Birth order and atopy. Clin Exp Allergy 2003;33:277-8. mdm 3:. GE B x33 “me E mEman 5.8: Lo 305: 9230 4.4 .3 9. 3x SS :3 9. 3. - 9. ma N.N_. Nov 9. m.~v - Ego; :Em v.0 me >Eo mmSECon. odm mdv 0.2259 ucm Emmi mém NS. 2:0 685 I 9.68; mom Own 9E8 5316 8:2. N4 55:5 m>_._ m9. mg. E3503 :9: 8:9: vhm Qmm Emucoomm :9: $3 - cozmosum .3562 03 m. I. bo>=mu Co Loam. mctzu wcozmgano 5.3: _mEQNE QNN o.N_. Long 0.5.3 :96 28535 5 2392.4 No me Loom. Bonus mar Qm >ocmcmoa act—6 co_mw_Eum _ano; _mEQms. 5mm mNm 5:385 mESU 25:0th _mEofis. va v.8 55:85 9:28 wcozmo__ano _mEQmE adv Emv mm: 3:302:00 _Eo _mEoumE 83ers 68qu 292$ 3992 um new commas. E O_QEmmbsm 9.: 80: %sz Ema 0925 33.329 xmwzifi ucm _EmELmdv @ev tocoo 05.3mm 3353 09:5 ucm 958mm 5 mm_nm_._m> osmoaxo Co 83:209ch cowtquoO ... 29¢... 129 «Me M 952835 :32; xmao=m 2: 3: qt. t: is 3: QB .26 ES 5 E285 35m .0 .96“. $1 :98 mEEoE 2:8 .0 mm _..m ...? me Q? 9:329: Emscmi 0.9 «.8 men @5322, Lo nEEma. avers Scans anus Ears 321: «means sons 02 mo> oz mo> oz wm> go 965 : mEEoE 25m .0 m5~mmn>> Lo mE£w< emEmNom _Eo._. oEficmE Emzcmi mcozfimmL—EmE 062m EmEEn m5 h_o 85:38 9853366 ho mcoanoi .N 2an 130 _ I ‘ . . ‘ Ell-I... $1.11..“ xmflém 550 wdm N: wdm on. m? o. E 55 NNN odw mdm :N v.3 mEom 5 E039: 0:50 :05.— >01 3 4.0 we «.8 00 S we 2 on E :0 3 Emma :maou me on ..m E ..m we we mam on E 0.0 3. SEE E8 5 0.55:9: E0309... . . . . . . . . . . . . 3.322, N: m 3 :0. I: .0. am. me. am. .2 com me. N0. 59:59.. 381: «ENS Roars «RYE SKIS nouns gens aware 38".: fans Remus swans 0252.6 02 00> 02 00> 02 00> oz mm; oz 00> O_mEmu 22>. 6:039: 5:209: @550 mm: 02300928 gmwcmfimmfi: @550 m:o_mm_Eum mmwdwwmmmqmmficmc m:o_.mo__an0 _m:o _mEBmE 50:00 . 0 .x m _mgamo: _mEBms. .. a. ._ u _>_ _mefis. 9900... ._m: 803 E :05 9 830.9 :_ 909006 0590 .o :oquoi .m 052. 131 35:0 550 0E00 5 0.2 mew N t. 0 mm wow 08 E039: 0:50 55. >01 «.0 ms 5. ~.~_. 0.0 v. E 080~0m :mzoo mEEoE 0.4 ..0 0.0 0.0 0.0 ...v 2.00 5 0E: :5: E0309: 0.9 0.3 0.9 0.0? 53 0.9 m:_~00:>> 5 0E£0< a Bus 39".: €055 ammus 63".: 855 as 0:80.30 02 00> 02 00> 02 00> 502.00 5 5:0. 5:0_ @550 520 5:0_ 00030:. @550 0:0:00__an0 55902 259005 5 0009094 05.00.. .0: .0355: .9055: :55 9 520.9 :_ 050506 0590 5 :25qu .v 030... 132 35:0 550 0:50 5 0.0? 0.0.0. 0.0V :0 0.0.. 0.00 0.00 0.0.. E0305 00:.0 5>0._ >0... 0.0 0.3 0.0 0.0 0.0 0.0 v.0 0.0 0E0N0m. . . . . . . . . 00:00 0.:_:5E >000 N0 00 00 F0 F0 00 00 00 5085;050:0002“. 0.0? ND. 0.: 0.: 0.3 0.5 50.. 0.3 05000:; 5 0E£0< ammus Emu... fins @0010 38".: 810 sins Gowns .x. 05850 00. 9. 0.0 00. 2:0 00.0.05”. 2:0 02 00> 0.0 A 9. 00 0.0 v 00.:E5u. .0 .0005 00005 0.... 0o 9.82 £00; 55 0:000“. 0 00:: :_ 005.005 5.00: 5 000:... 0.2.00 0550.. 0.0: .0055: 55:50 :55 2 500.0: :_ 05050.0 0.0000 5 5.0002,... 8005508 .0 0.00... 133 00.0 0 029.0 . .0:00 5.00: 2 000000 0:0 5000000 .000 5:55.: 0:0 50:00 5.. >._0:00.000 0:0 0.000 05 :. 00.0009 5.. 00000.5( .. 00.0-8.0 00.0 2.0.00.0 00.0 3.0.00.0 00., 000-000 00.0 00. 0.? 0 0 P F P F 0 F .9. 0.0 I 9. 0.0 8020000 000.000 00.0 3.0-00.0 00.0 2.0-00.0 «00 00.7000 00.0 9.0.0v 000.0; 500 00. 0.000 00.0 00. 7R0 00.0 B. 30.0 00.0 00. 700.0 00.0 00:00.0 .020 00 00:52 .00_ 0o 0.8; 00:0 0.0 00. 0.000 00.0 2.0-00.0 0 2 00.300 00.0 00.0-2.0 00.0 0. 052020 5.8,. :o 0000:. 0.2.00 >5>..00 vmduomd oné 00600.0 3.; No. .3506 or... 00. Tend o _.._. 9:030 000.002.9000 50:205. . . . . :80. 00 0 S 0 00 0 0c 0 00 0 0.0 F 00 F 00 0 00 0 v0 _ 00 0 : F 05:00 0020 00050000 :0 2390.2 00. 0.000 00.0 5.0-00.0 50 00. 700.0 00.0 2.0-00.0 «I :80 08:00. 0050050 .0555; M... 000-000 00.0 000-000 00.0 00. 0-00.0 00.0 00. 30.0 00.0 5000090 00.50 000.050 1 3.0-00.0 03 8.0-00.0 03 00. 0.000 :0 05-000 0: 0.002020 0:000 5.00.800 .00.0000 5:555. 000.000 00.0 3.0-00.0 0: 00. 7:0 00._ {.300 00.0 502020 00.50 00000000. 00:90.2 >0:0:m0:0 05.000 00.0 00.0-03 00.0 5.0-00.0 00.0 00.0-8.0 .03 00.50 0808.058 00:20.2 «0.0.3.0 .000 00.0-00; .00.0 5.0-00.0 .0: 000.0: .00.? 00: 20080008 .90 .0520: 0080050 55:05.0. _0 .000 000 .0 0.00 000 _0 $00 000 .0 $00 000 920$ "2 .05 0 000% 00:00 0552: >000 5 0.0.0.5.: 50:00:”. 0500002, 5 00:50.0. 00:00 05:50: >000 5 08.0094 E00000 0:0 0500003 5 00:500. 50 050000 0.0.: 55:50 0:0 55:25 5 :00 0.0205. 00:00::00 $00 0:0 00000: 0000 .0 0.00... Ed!» 405‘. 1 Inn.“— mod w 00.0.4.0 ... .0000 5.00: 0. 000000 0:0 5000000 .000 5505:. 0:0 50:00 5. >__0:o_._000 0:0 0.00. 0:. :_ 00.:0:0> 50 00.00.00. ._- 00..-00.0 00.0 00..-00.0 00.0 00.0-00.0 .0.. 000.000 00.. 0.. 0.0A 0 _- . 0 F . . . .00. o..0l0x 0.0 00:05.50. 00.7000 .00 0.7.0.0 00.0 0.70.0 00.0 .0.-0.0 00.0 000.0v .00.00>00.0 00.0-00.0 .000 000.000 .000 00.7000 00.0 00.0-00.0 .000 00002000280000.0002 .- . . .- . . .- . . .- . . 00:50.00; 0.0000 .0. 0000.. .00. 000000 00. 00000. .0.0 0000.00.03.0000.000000005000000 00..-00.0 00.0 00.7000 0... 00.7000 00.0 00.0-.00 0... 0002.00 00.0000000000000000000.02 00.7000 00.. 00.0.00. .00.. 000.000 00.. .0700. ..00 0000.00.30 002000050000000.000.000. 00..-00.0 00.0 00.7000 .0.. 000.00 0... 00.7000 00.0 0000.88.00. 0080200 .000550. 007000 00.0 00..-00.0 00.0 00.0-00.0 00.. 000-000 .0.. 0.0000090 000000020000 . . . . . . . . . . . . .>0:0:000n. 0.7000 000 0.7000 000 000-000 00. 000-000 00. 00000 00.00.500.00000000.0.0.2 00.7000 00.. 00.7000 00.. 00.7000 00.0 007000 00.0 00000090 00000000000.0__0000002 0.0-00. ..00. 00.7000 00.. .00-000 00.. 00.0000 00.. 000000000 00.00000000000000000000.05. 00.7.0.0 .0.. 00.7000 00.0 0.0-00.0 .0.. 00.0-.00 00.. 00: 0200800080000000.00,. 00E00.0o _0.0:0.:< .0 00.00 0.00 .0 00.00 000 500.00 0.00 .0 00.00 0.00 00200“. .000n0. .000u0. >05__0 5:5 0:50 5 0:500 E0305 0.55 5 55.. >0... m. >05__0 5:5 00:00 5 E03900 000.0 5 55.. >0: 0:0 0.00000 5.. 05.000 0.00 5.00.50 0:0 50000.00 5 ..0. 05:00:. 00:00:08 00000 0:0 .0000. 0000 .0 0.000 135 COMPONENT FOUR Postnatal exposure to SHD affects biomarkers of allergy in infancy 136 1- m _. I. n'AI Immune function biomarkers in children exposed to lead and organochlorine compounds: a cross-sectional study Wilfried Karmaus”, Kevin R. Brooks“, Thomas Nebez', Jutta Witten3', Nadia Obi- Osius“, Hermann Kruses' 1 Department of Epidemiology, Michigan State University, 8601 West Fee Hall, East Lansing, MI 48824, USA 2 Central Laboratory, University Hospital Mannheim, Germany 3 Ministry of Social Welfare Hesse, Department of Health, Wiesbaden, Germany 4 Epidemiological Working Group of the Ministry of Environment and Health and the Institute for Mathematics and data management in Medicine, University Hospital Hamburg-Eppendorf and 5 Institute of Toxicology, Christian-Albrecht University, Kiel, Germany *These authors contributed equally to this work §Corresponding author Abstract Background: Different organochlorines and lead (Pb) have been shown to have immunomodulating properties. Children are at greater risk for exposure to these environmental toxicants, but very little data exist on simultaneous exposures to these substances. Methods: We investigated whether the organochlorine compounds (00) dichlorodiphenylethylene (DDE), hexachlorobenzene (HCB), hexachlorocyclohexane (y-HCH), the sum of polychlorinated biphenyls (ZPCBs) 137 and Pb were associated with immune markers such as immunoglobulin (lg) levels, white blood cell (WBC), counts of lymphocytes; eosinophils and their eosinophilic granula as well as lgE count on basophils. The investigation was part of a cross-sectional environmental study in Hesse, Germany. In 1995, exposure to OC and Pb were determined, questionnaire data collected and immune markers quantified in 331 children. For the analyses, exposure (00 and Pb) concentrations were grouped in quartiles (y-HCH into tertiles). Using linear regression, controlling for age, gender, passive smoking, serum lipids, and infections in the previous 12 months, we assessed the association between exposures and immune markers. Adjusted geometric means are provided for the different exposure levels. Results: Geometric means were: DDE 0.32pg/L, XPCBs 0.50 pg/L, HCB 0.22 pg/L, y-HCH 0.02 pg/L and Pb 26.8pg/L. The ZPCBs was significantly associated with increased lgM levels, whereas HCB was inversely related to lgM. There was a higher number of NK cells (CD56+) with increased y-HCH concentrations. At higher lead concentrations we saw increased lgE levels. DDE showed the most associations with significant increases in WBC count, in lgE count on basophils, lgE, lgG, and IgA levels. DDE was also found to significantly decrease eosinophilic granula content. Conclusion: Low-level exposures to OC and lead (Pb) in children may have immunomodulating effects. The increased lgE levels, lgE count on basophils, and the reduction of eosinophilic granula at higher DDE concentrations showed a 138 1: ..i‘yfl 1 in..." .A most consistent pattern, which could be of clinical importance in the etiology of allergic diseases. 139 fz-QHI‘ ., H ' Background Environmental toxicants such as organochlorine compounds (00) and lead (Pb) may alter immune responses. There is a paucity of studies reporting associations between organochlorine [1-4] and lead [5-8] exposures and immune function biomarkers in children. We conducted a large-scale environmental study of second-grade school children in three regions south of the Federal State of Hesse, Germany in 1995. ;Li Two of the regions are situated in the Rhine Valley with low mountains on both “1“ sides. One of these areas with several municipalities is located within a 10 km radius around an industrial waste incinerator and other industries, such as chemical plants. One plant was associated with dichlorodiphenylethylene (DDE), hexachlorobenzene (HCB), and hexachlorocyclohexane (y-HCH) pollution [9]. The other region, also industrial, is 15 km north (downwind) of the incinerator. Both Rhine valley regions are also intensively used for the production of vegetables. The third study region is located in low mountains (about 0.4 km above sea level) that separate it from the industrial area. Blood concentrations of PCBs were shown to be higher in children living close to the toxic waste incinerator [10]. Results on P085 and thyroid hormones, chromium and lymphocytes, DDE and breastfeeding and asthma have been published elsewhere [4, 11-15]. Considering infection and atopic disorder in children, we have previously shown an association between DDE blood levels; asthma and one immunoglobulin (lg), namely lgE [4]. However, the potential effects of organochlorines on other lgs 140 and cellular defense were not reported. Hence, the focus of this paper is to investigate the impact of organochlorine compounds and Pb on humoral immune markers and cell-mediated immune responses. Specifically, for immune responses we focus on leukocytes, lymphocytes, B-cell, T-cells and their subsets. Assuming a concurrent effect of QC on immune markers, we conducted cross-sectional analyses of the data from the first of three surveys conducted in 1994/1995, 1996, and 1997. Only the first investigation included an extensive clinical assessment of immune markers. .. Methods Study population After obtaining approval from the Data Protection Agency of Hamburg, Germany, the Ministry of Cultural Affairs of Hesse, Germany, and the local school committees, we invited the parents of 1,091 second-grade school children in 18 townships to participate in our study. We obtained informed consent from all participating parents, according to the requirements of the Ethical Committee of the Board of Physicians, the Helsinki Declaration, and the Data Protection Agency of the State of Hamburg. We asked each parent to allow their child to participate in phlebotomy only when passive smoking in the private household did not exceeded 10 cigarettes per day during the previous 12 months. Questionnaires We used four self-administered parental questionnaires in the survey: one regarding the living condition and nutrition of the family, one for each parent, and 141 one regarding information on the child. Duration of breastfeeding was recorded in weeks of total and in weeks of exclusive nursing. Environmental tobacco smoke (ETS) was graded as smoking in the child‘s home in the previous 12 months (no cigarettes, 1-10 cigarettes, 11-20 cigarettes, 21-30 cigarettes, more than 30 cigarettes per day). We recorded age, gender, and the number of infections, defined as cold, coughing, and sore throat, with or without fever, in the last 12 months (none, less than 5 infections, 5-10 infections, more than 10 infections). Laboratory analyses of blood samples One parent accompanied each child in the medical examination. For blood sampling, we used the 'Vacutainer System' (Becton, Dickinson & Company, San José, California) Approximately 25 mL were drawn and separated into different aliquots. lmmunoglobulin (lg) E in serum was quantified at the Medical, Alimentary and Veterinary Institute for Research Middle Hesse, Division of Human Medicine, Dillenburg, Germany, using a florescence-immunoassay (CAP, Pharmacia, Uppsala, Sweden). To determine levels of specific lgE against inhalant allergens (aeroallergens), we incubated serum with immunocaps containing a mixture of aeroallergens and determined the reactivity using a fluorescence measurement (UNICAP Pharmacia, Uppsala, Sweden). Results from this method were provided in semi-quantitative format. We also measured lgA, G, and M by laser immunonephelometry (Dade Behring, Liederbach, Germany). The results for IgA, G and M were provided in mg/dL and for lgE in kU/L serum. Triglycerides and cholesterol were measured on a clinical chemistry analyzer according to IFCC methods (Hitachi 717, Boehringer Mannheim). 142 Leukocyte subsets We collected 8 mL of blood in tubes containing EDTA and mixed them to prevent clotting. This aliquot was transported to the Central Laboratory of the University Clinic of Mannheim and analyzed on the same day. We used 200 pL of blood for the automated differential (laser-based hematology analyzer CDSSOO, Abbott Diagnostics, Santa Clara, California), and 100 uL for each of the nine three-color test tubes analyzed by flow cytometry (FACScan, Becton, Dickinson, & Company, San José, California, equipped with a 488 nm air-cooled argon ion laser). Eosinophils were determined according to their specific depolarisation characteristics and their eosinophilic granula content by the intensity of light scatter by flow cytometry. Basophils were identified by their high lgE density on the cell surface using immunofluorescence with a Phycoerythrin (PE) labeled anti-lgE antibody. We used monoclonal antibodies directed against specific cell surface antigens to differentiate cell populations by multicolour immunofluorescence. Three antibodies were simultaneously applied with the fluorochrome combination FlTC/PE/PE-Cy5. CD4/CD8/CD3 was used to detect absolute number of lymphocytes, T-helper cells and cytotoxic T-cells; CD19/CD5/IGE was used to differentiate B-cell subsets and basophils; CD3/CD16 and CD56/CD57 were used for natural killer cells. CD45RO defines memory T-helper cells. The CD nomenclature assigns the antibodies to clusters of differentiation, according to the International Workshop on Human Leukocyte Differentiation Antigens [16]. 143 Organochlorine compounds (GO) in blood 00 including eight PCB congeners (101, 118, 138, 153, 170, 180, 183, 187), DDE, HCB, and three HCH congeners (a-, B- and y) were determined (in pg/L) at the Institute of Toxicology, University of Kiel, Germany. OC were analyzed in 5 mL samples of whole blood by high resolution gas chromatography (HRGC, Model 3400 by Varian Inc., Palo Alto, California) with a 63Ni—electron-capture- detector. The detection limit (DL) (two times the signal/low-noise ratio) was 0.02 pg/L for B- and y-HCH, DDE and each PCB congener, and 0.01 ug/L for HCB and a-HCH. For extraction and clean-up procedures, we used florisil and n- hexane for elution (9 g florisil were deactivated with 3% H20 and placed in a chromatography column 22 mm in diameter and 48 mm in length). The capillary column amounted to 30 mm in length and 0.25 mm in diameter; nitrogen was used as a carrier gas. We determined the PCB congeners by retention times on the chromatograms and identified them by comparison with known standards. Additionally, we tested reliability with gas chromatography-mass spectroscopy (GC/MS). The laboratory successfully participated in nationwide quality assessments for the determination of these OC. Lead in blood Lead (Pb) analysis was done at the Institute of Toxicology, University of Kiel, Germany. The determination in whole blood samples was by flow injection atomic absorption spectroscopy (Perkin Elmer) after adding 0.1% Triton —X-1- solution and 15 mol nitric acid to from a solution. This solution was then centrifuged at 3000 rpm. The DL for Pb was 9 pg/L (48 nmol/l; atomic weight: 144 207.19). Data analyses Since the data for leukocytes (WBC) and their subsets (lymphocytes and eosinophils), immunoglobulins, DDE, PCB congeners, HCB, y-HCH and Pb were not normally distributed, the geometric mean, 5-, 95-percentiles are provided. In order to obtain a multivariate normal distribution, we log-transformed the number of cells and immunoglobulins before testing associations with possible predictors - wuLTr WW' by multiple linear regression models. 1* All statistical analyses were performed using SAS software [17]. We calculated the sum of the PCB congeners (ZPCBs = sum of seven congeners, the congener PCB101 was not detected). For descriptive purposes, we substituted values of 00 below detection limit with one half of the detection limit. The statistical procedure (PROC RANK) was used to group exposure variables into quartiles (DDE, PCBs, HCB and Pb) or tertiles (y-HCH). All observations below the detection limit were part of the lowest level group (reference). To account for the influence of lipids on the concentration of OC, we controlled for the sum of triglycerides and cholesterol in the regression analyses. Further steps were taken to determine whether our results were different when lipids were represented as sum of triglycerides and cholesterol as opposed to triglycerides and cholesterol as individual variables. We used linear regression models (PROC GLM) with immune markers as dependent variables and all organochlorine compounds and lead as independent 145 variables in each model. We also controlled for potential confounders (age, gender, environmental tobacco smoke (ETS), number of infections during the last 12 months, and lipid concentration). Information on passive smoking (ETS) in the child’s home in the previous 12 months was divided into four categories (no cigarettes, 1-10 cigarettes, 11-20 cigarettes, 21 cigarettes per day and more). For the number of infections we considered four categories (none, less than 5, 5- 10, more than 10). Age of the child was divided into three groups; 7, 8 and 9-10 years. From the results of the regression analyses, we calculated adjusted geometric means for leukocyte subsets and immunoglobulins for increasing categories of exposure. T-tests were used to compare the statistical effect of higher exposure group to the lowest (reference). Since one major route of exposure to Child's exposure the pollutants analyzed is breast concentration feeding [18-21] and breastfeeding \ provides passive immunity [22-24], [ Breastfeeding I immune markers and pollutants could Figure 7: Diagramatic representation of be spuriously correlated if breast the breastfeeding, childhood exposures and immune markers associations feeding is not controlled for. However, this triangle (Figure 7) cannot be tested with linear regression models, as intervening variables do not qualify as confounders [25]. Controlling will 146 VL'T "I, reduce the initial association between the risk factor and the marker, as one causal chain is split into two associations. Thus, we explored the relationship between childhood breastfeeding (total duration of breastfeeding in weeks), the concentration of OC, and immune response by path analysis [26], using the CALIS procedure SAS Institute [17]. Results The proportion of participation was 61.5 % (671 of 1091). We obtained blood samples from 350 children, conducted 00 and Pb analyses on 343 samples, and quantified immunoglobulins in 340. Overall, information (i.e., questionnaires, exposure biomarkers, and immune markers) was available for 331 children. Fewer girls than boys participated in phlebotomy; and 96 % of the children were 7 to 8 years of age (Table 1). Due to the inclusion criterion for blood sampling (passive smoking of less than 10 cigarettes in the child’s home), the prevalence of passive smoking was also lower in the group with phlebotomy than in the total group (Table 1). Nevertheless, the fact that parents were separated or divorced and shared cohabitation for their child, resulted in a re-assessment of the passive smoking status after phlebotomy. Eligibility was determined on the information provided by one parent (mother or father) for their household. In the case of separate dwellings, we re-assessed the exposure by taking the average number of cigarettes smoked in both homes. As a consequence, 26 (7.9%) children who were exposed to more than 10 cigarettes per day at home had a phlebotomy and were included in the analyses. 147 For y-HCH, 27.7 "/0 of the observations were below the detection limit, 2.9% for Pb, whereas none for DDE and HCB. At least one of seven PCB congeners was detected in each sample. Whole blood concentration for the sum of PCB congeners (118, 138, 153, 170, 180, 183, 187), HCB and of Pb showed a decline with increasing age (Table 2). DDE, PCB, and HCB concentrations were lower in children with higher passive smoking exposure. Regarding infections, lead - TIIHTV concentration was higher in children with more than 10 infections during the last 12 months, whereas DDE concentration was lower in this group (Table 2). The concentrations of DDE, EPCBs (sum of PCBs), and HCB were all correlated (Table 3). However, we used categorized levels of OC, which were then only marginally correlated; the highest rank correlation was for the PCB and HCB groups (rspearman=0.46). These correlations did not result in multicollinearity since the tolerance (variance of 00 not explained by other predictors) was at least 53%. The volume-based organochlorine concentrations were only marginally correlated with the lipid serum levels. To adjust for lipid concentrations, we included lipid concentrations as a confounder in the explanatory models for leukocyte subsets and immunoglobulins. Results derived from models using the sum of triglycerides and cholesterol compared to triglycerides and cholesterol as individual variables did not reveal any substantial difference (data not shown). We therefore reported results from models using the sum of triglycerides and cholesterol. 148 Regarding lead in whole blood, we found weak correlations with whole blood levels of OC (DDE: r= 0.15, n = 331, p < 0.01; HCB: r= 0.14, n = 331, p < 0.01; y-HCH: r= -0.02, n = 331, p < 0.70; EPCBs: r= 0.14, n = 331, p < 0.01) Increased white blood cell count (WBC; total leukocytes) was evident in the group with highest DDE level, whereas Pb, at the second, along with PCB at the highest level was associated with a reduction in WBC count. An increase in the number of eosinophils — a leukocyte subset - was identified in the highest DDE category, but not statistically significant, Table 4. However, eosinophilic granula content was significantly reduced at the upper DDE levels. In addition, lgE count on basophils was increased at higher DDE exposure, being statistically significant for the 0.3-0.43pg/L category. Regarding lymphocytes and specific lymphocyte subsets (B-cells, T-cells), the number of T-cells (CD3+), cytotoxic T-cells (CD8+) and B-cells (CD19+) were all significantly reduced in the median Pb category (Table 5). Both natural killer (NK) cells (CD56+) and a NK cells subset (CD57+) were significantly associated with y-HCH. However, these associations did not reveal dose-dependency. All four immunoglobulins were associated in a virtually dose dependent fashion to either DDE, HCB or P085 (Table 6). lgM serum levels increased with the concentration of PCBs (F-test, p<0.01) but decreased with increasing concentration of HCB (F-test, p<0.01). In the two upper quartiles of DDE exposures, IgA levels were significantly higher, but lower in the upper quartile of HCB. DDE was not associated in a dose-response mode with lgG (F-test, 149 p=0.14), however, compared to the reference, the highest DDE exposure group showed a significantly elevated lgG level (t—test, p=0.04). IgE levels more than doubled as DDE concentration increased (F-test, p=0.02). The Pb serum levels were related to a significant differences in lgE (F-test: p=0.028), but not in a dose dependent fashion (Table 6). Figure 8 shows that both 70 mew—m— __ l DDE and lead were 50 * “ “W . “‘"wj ...... ; associated with higher 50 ’ ‘ l . Q 40 - ' -_‘ serum lgE levels in 3 ' . , E 30 ii _ children. In groups wrth 9 4i ------------- a 20 , lower DDE blood __,_ _Pb <= 28 ug/L 1o ....... concentrations, Pb ' Pb >28 “91" 0 ’ . concentrations above the <=o.2 0.21 -o.29 o.3-<0.43 >043 DDElug/L) median (28IJ9/L) were Figure 8: The combined effect of increasing DDE and lead (Pb) on lgE serum levels related to increase lgE levels. In groups with higher DDE, there was no additional effect of Pb. Statistically, the combined effect of DDE and Pb on lgE was not significant. In order to determine whether breastfeeding confounded the associations identified in linear regression models (Figure 7), we repeated our analyses using structural model (path analysis) for exposures determined as significant in linear regressions. Inclusion of breast feeding did not substantially change our findings. 150 Discussion In 331 school children, age 7-10 years, we demonstrated significant relationships between 00 and Pb whole blood concentration and cellular and humoral immune markers. First, modest associations were found between NK cells (CD3- CD16+CD56+) and a subset of natural killer cells (CD3-CD16+CD56+CD57+) and y-HCH (Table 5). Second, HCB was inversely related to lgM. Third, ZPCBs were directly related to lgM. Fourth, our data showed that Pb decreased the count of T-cells (003+), cytotoxic T-cells (CD3+CD8+), and B-cells (c03+005+ ., CD19+). This reduction was most evident at the 22.1 — 28.3 pg/L Pb concentration, though not in a dose response fashion. Lastly, DDE was inversely related to all immunoglobulins, except lgM (Table 6). However, DDE was not associated with total serum protein (data not shown). The DDE effect was strongest for lgE - more than twofold increase - which also corresponded to an increased count of lgE on basophils. We did not detect a significant relationship between DDE and eosinophils, nevertheless, the number of eosinophils was positively correlated with lgE (rspeaman = 0.4, p < 0.01). However, high DDE levels were found to be significantly associated with lower eosinophilic granula content. The granula contains basic proteins which are cytotoxic and part of the inflammatory response [27]. The cross-sectional nature of the study limits conclusions on whether exposure occurred before immune responses. We can assume that organochlorine concentrations do not vary substantially in childhood, post breastfeeding. There is a decline of PCBs and HCB with age (Table 2), however the assumption of the 151 stability is supported by a follow-up of the same children and OC determined in 1997. The Spearman rank correlation between the two successive measurements were high, with the exception of y-HCH: DDE: r = 0.86, n = 274, p < 0.01; HCB: r= 0.74, n = 274, p < 0.01; y-HCH: r= 0.1, n = 270, p = 0.11; ZPCBs: r = 0.82, n = 274, p < 0.01. The reported concentrations for DC were not lipid-based. In this cohort, there is a high correlation between lipid- and non lipid-based concentrations for CC (Table 3). Thus, our findings are independent of lipid- or wet weight-based determinations. In our models we controlled for lipids instead of dividing the concentration of 00 by the lipid concentration for three reasons. First, a simple division assumes a monotonous linear relation between lipids and organochlorines. Although Phillips and co-workers reported for 20 adults that division by lipids reduces the difference between fasting and non-fasting concentration of CC [28], there is no data to justify a linear relation. Our data in children showed only weak correlations between OC and the sum of cholesterol and triglycerides (Table 3). This correlation did not increase when the sum of lipids were derived by using the 2nd formula proposed by Phillips et al. [28]. Second, there is no standard approach to adjust concentrations below the limit of detection for lipids. In particular, the probability of detection may be influenced by the individual lipid concentration of a child. Third, division by lipids does not take into account that they may confound the organochlorine - immune response relationships. Confounding is likely since lipids and DC are correlated, plus lipids 152 are, for example, associated with the count of lymphocytes [29, 30]. There is evidence that breast milk is a significant source of OC, Pb [18-21], and passive immunity [22-24]. Path analytical techniques (Figure 7) were used to verify whether breastfeeding as an intervening variable confounded our associations. The inclusion of breastfeeding in the path analysis did not reveal results different from the linear regression models. Hence, the associations between pollutants and immune markers were independent of breastfeeding. We found whole blood concentrations of OC in our cohort comparable to similar children in Germany [31]. Compared to children in the United States, age 12-19 years (NHANES - 1999-2000) [32], our DDE values were lower though still within the 95% confidence interval. However, when comparing our results (in whole blood) with those of NHANES (in serum), we have to consider differences between serum and whole blood concentrations. Mes et al. reported that DDE was higher in sera and plasma than in whole blood samples [33]. Conversely, PCBs were higher in whole blood samples. No other comparison with NHANES data was possible as the values for PCB congeners and other OC were below the limit of detection. Regarding lead (Pb), the geometric mean of 27ug/L in our investigation was similar to the 33pg/L found in a study of 797 East-German children 5-14 years of age [34]. Against that, the 1999-2000 NHANES study showed a lower geometric mean (15.1 pg/L) in 905 children 6-11 years of age [32]. However other studies in areas of higher exposure, reported average concentrations above the NHANES 153 value: 40 pg/L for children, 6 to 15 years of age in four communities with mining and smelting operations and two control groups in the United States [6], and 95pg/L in Chinese children 3-6 years old [8]. We selected a subgroup for blood analyses due to budget constraints. The group having a lower ETS exposure in their homes was selected to reduce the potentially confounding effect of ETS. This group did not significantly differ from other participating children (Table 1). Parents did not know the individual results m— of the blood analyses, when they provided information on their children, thus reducing recall bias. The inverse association between DDE and the number of infections 12 months prior to the interview is surprising (Table 2). However, in a logistic regression model the number of infections reported did not show a significant protective effect of DDE. Additionally, when infection was eliminated from the models, there were no major changes in the OC - immune markers association. The few existing studies estimating the immunotoxicity of lead (Pb) in children, measured by immune markers, are inconsistent in their findings. Regarding immunoglobulins, our positive relation between Pb and lgE was consistent with that of Lutz et al. [7]. However, Sun and co-workers had different results [8]. Concerning lymphocytes, we found that the number of B-cells was significantly reduced with increased Pb concentration. Conversely, Sarasua et al. reported an increase in the number of B-cells for children less than 3 years old [6]. 154 Studies assessing the relation between organochlorine and immune markers, determined in our study, also showed conflicting results and focused mostly on adults [35-37]. In comparison with these adult studies, Vine et al. reported similar modest findings for immunoglobulins and DDE. However, only results for IgA showed statistical significance [35]. Our findings regarding lgE and eosinophilic granula suggest that DDE shifts the immune response into a T helper (Th) 2 F direction [38]. Mechanistically, immune responses have been polarized into Th1 9 A and Th2 reactions. Th1 responses lead to the secretion of immunoglobulin G (IgG) and removal of the allergen. The allergic Th2 phenotype is characterized by secretion of cytokines that promote immunoglobulin E (lgE) production resulting in allergies. This suggestion is in agreement with findings of Daniel and co- workers, who reported an association between DDE and interleukin-4, a Th2 cytokine [39]. In addition, our interpretation that DDE may be associated with an allergy-like response is supported by the distribution of aeroallergen-specific lgE results over the four DDE exposure levels. In the lowest DDE exposure group 11.3% of the children showed a positive specific lgE, 10.9% and 12.2% in the two intermediate groups, but 23.0% in the highest DDE exposure group (p=0.03). Interestingly, the effects of lead (Pb) and DDE on lgE seems to be competitive. At lower DDE exposure, Pb seems to increase lgE concentrations (Figure 8). There was no additional effect of the other pollutant if one is high; therefore it is possible that both pollutants are involved in the same mechanism. Indeed, studies have surmised that Pb may also shift the immune responses in a Th2 155 direction [40-42]. There are only few studies on OC blood/serum concentration and immune responses in children. Weisglas-Kuperus et al. reported that prenatal PCB exposure was associated with an increase in the T-cell markers CDBCD8+ and CD4+CD45RO+ [2]. Our data did not support these findings. In another study with prenatal exposures to PCBs, HCB, and DDE, Dewailly et al. did not identify significant associations with immune markers including CD3+, CD4+, CD8+ lymphocytes nor with IgA, IgG, and lgM [3]. However, we found significant relationships between PCBs and HCB with lgM (Table 6). Reichrtova et al. have shown that in utero exposure to DDE is positively correlated with cord serum lgE [43]. No other study of children has investigated the relationship between DDE determined postnatally and Th2 markers such as lgE and eosinophilic granula. This is the second publication showing an association between DDE and serum lgE [4] and the first to report associations between Pb, and DDE and lgE count on basophils and eosinophilic granula. Conclusion In conclusion, our study suggests a non-linear association between lgE and Pb concentration. Regarding OC, our data indicated an increase of lgE related to DDE serum concentrations. A parallel association between DDE, lgE count on basophils, and reduction of eosinophilic granula contents further supports a potential stimulation of a Th2 response related to DDE exposure. 156 Prospective studies should determine more than one DC in a scenario with multiple exposures in order to prevent spurious correlations and include repeated determinations of immune responses to determine changes in immune development during childhood. Furthermore, studies are warranted that determine allergic susceptibilities following DDE and Pb exposure in children. Acknowledgements i This study was supported by the Ministry of Environment, Energy, Youth, Family and Health Hesse, Germany. We acknowledge Dr. Rauterberg for the analyses to of immunoglobulins. The analyses were conducted in preparation of an EPA STAR grant (R830825). KRB was a graduate assistant in the EPA grant while preparing the final manuscript. 157 References 1. 10. 11. Marth E, Sixl W, Bencko V, Medwed M, Lapajne S, Voncina E, Brumen S: People on the garbage dumps of Cairo: a toxicological in vivo model? Cent EurJ Public Health 1995, 3:154-157. Weisglas-Kuperus N, Patandin S, Berbers GA, Sas TC, Mulder PG, Sauer PJ, Hooijkaas H: Immunologic effects of background exposure to polychlorinated biphenyls and dioxins in Dutch preschool children. Environ Health Perspect 2000, 108:1203-1207. Dewailly E, Ayotte P, Bruneau S, Gingras S, Belles-Isles M, Roy R: Susceptibility to infections and immune status in Inuit infants exposed to organochlorines. Environ Health Perspect 2000, 108:205-211. Karmaus W, Kuehr J, Kruse H: Infections and atopic disorders in childhood and organochlorine exposure. Arch Environ Health 2001, 56:485-492. Rabinowitz MB, Allred EN, Bellinger DC, Leviton A, Needleman HL: Lead and childhood propensity to infectious and allergic disorders: is there an association? Bull Environ Contam Toxicol 1990, 44:657-660. Sarasua SM, Vogt RF, Henderson LO, Jones PA, Lybarger JA: Serum immunoglobulins and lymphocyte subset distributions in children and adults living in communities assessed for lead and cadmium exposure. J Toxicol Environ Health A 2000, 60:1-15. Lutz PM, Wilson TJ, Ireland J, Jones AL, Gorman JS, Gale NL, Johnson JC, Hewett JE: Elevated immunoglobulin E (lgE) levels in children with exposure to environmental lead. Toxicology 1999, 134:63-78. Sun L, Hu J, Zhao Z, Li L, Cheng H: Influence of exposure to environmental lead on serum immunoglobulin in preschool children. Environ Res 2003, 92:124-128. Forth W: Hexachlorcyclohexan - Gift in den Lebensmitteln? Deutsches Aerzteblatt 1980, 83:2169-2176. Holdke B, Karmaus W, Kruse H: [Body burden of PCB in whole human blood of 7-10-year—old children in the vicinity of a hazardous waste incinerator]. Gesundheitwesen 1998, 60:505-512. Osius N, Karmaus W, Kruse H, Witten J: Exposure to polychlorinated biphenyls and levels of thyroid hormones in children. Environ Health Perspect 1999, 107:843-849. 158 .I’F .— d 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Karmaus W, Huang S, Osius N, Nebe T: Chromium urine concentration and effects on lymphocyte subpopulations in children. J Environ Med 1999,11153-161. Karmaus W, DeKoning EP, Kruse H, Witten J, Osius N: Early childhood determinants of organochlorine concentrations in school-aged children. Pediatr Res 2001, 50:331-336. Karmaus W, Davis S, Chen Q, Kuehr J, Kruse H: Atopic manifestations, breast-feeding protection and the adverse effect of DDE. Paediatr Perinat Epidemiol 2003, 17:212-220. Obi-Osius N, Misselwitz B, Karmaus W, Witten J: Twin frequency and industrial pollution in different regions of Hesse, Germany. Occup Environ Med 2004, 61:482-487. International workshop on human leukocyte differentiation antigens [http:l/www.ncbi.nlm.nih.gov/prow/guide/45277084.html SAS Institute. Statistical Analysis System, Version 9.1. Gary, NC, 2002- 2003. Abraham K, Papke 0, Gross A, Kordonouri O, Wiegand S, Wahn U, Helge H: Time course of PCDD/PCDF/PCB concentrations in breast-feeding mothers and their infants. Chemosphere 1998, 37:1731-1741. Lackmann GM, Schaller KH, Angerer J: Organochlorine compounds in breast-fed vs. bottle-fed infants: preliminary results at six weeks of age. Sci Total Environ 2004, 329:289-293. Rabinowitz M, Leviton A, Needleman H: Lead in milk and infant blood: a dose-response model. Arch Environ Health 1985, 40:283-286. Ettinger AS, Tellez-Rojo MM, Amarasiriwardena C, Bellinger D, Peterson K, Schwartz J, Hu H, Hernandez-Avila M: Effect of breast milk lead on infant blood lead levels at 1 month of age. Environ Health Perspect 2004, 112:1381-1385. Hanson LA: Breastfeeding provides passive and likely long-lasting active immunity. Ann Allergy Asthma Immunol 1998, 81 :523-533; quiz 533-524, 537. Hanson LA, Korotkova M, Lundin S, Haversen L, Silfverdal SA, Mattsby- Baltzer l, Strandvik B, Telemo E: The transfer of immunity from mother to child. Ann N YAcad Sci 2003, 987:199-206. 159 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. Van de Perre P: Transfer of antibody via mother's milk. Vaccine 2003, 21 :3374-3376. Rothman KJ: Modern Epidemiology. Boston/Toronto: Little Brown and company; 1986. Hatcher L: A step-by-step Approach to Using SAS for Factor Analysis and Structural Equation Modeling. 6 edn. Cary, NC: SAS Institute Inc.; 1994. Leung DY: Molecular basis of allergic diseases. Mol Genet Metab 1998, 63:157-167. Phillips DL, Pirkle JL, Burse VW, Bernert JT, Jr., Henderson LO, IE... Needham LL: Chlorinated hydrocarbon levels in human serum: effects of fasting and feeding. Arch Environ Contam Toxicol 1989, 18:495-500. Moreno LA, Sarria A, Lazaro A, Lasierra MP, Larrad L, Bueno M: Lymphocyte T subset counts in children with hypercholesterolemia receiving dietary therapy. Ann Nutr Metab 1998, 42:261-265. Ogawa Y, lmaki M, Yoshida Y, Shibakawa M, Tanada S: An epidemiological study on the association between the total leukocyte and neutrophil counts, and risk factors of ischemic heart disease by smoking status in Japanese factory workers. Appl Human Sci 1998, 17:239-247. Landesgesundheitsamt Baden-Wuerttemberg: Projekt Beobachtungsgesundheitsaemter. Belastungs- und Wirkungsmonitoring. Stuttgart; 2000. Second national report on human exposure to environmental chemicals lhttp:l/www.cdc.qov/exposurereport/Znd/pdf/secondner.pdfl Mes J, Marchand L, Turton D, Lau P, Ganz P: The determination of polychlorinated biphenyls congeners and other chlorinated hydrocarbon residues in human blood, serum and plasma. A comparative study. Intem J Environ Anal Chem 1992, 48: 1 75-1 86. Jacob B, Ritz B, Heinrich J, Hoelscher B, Wichmann HE: The effect of low-level blood lead on hematologic parameters in children. Environ Res 2000, 82:150-159. Vine MF, Stein L, Weigle K, Schroeder J, Degnan D, Tse CK, Backer L: Plasma 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE) levels and immune response. Am J Epidemiol 2001, 153:53-63. 160 36. 37. 38. 39. 40. 41. 42. 43. Daniel V, Huber W, Bauer K, Suesal C, Conradt C, Opelz G: Associations of dichlorodiphenyltrichloroethane (DDT) 4.4 and dichlorodiphenyldichloroethylene (DDE) 4.4 blood levels with plasma |L-4. Arch Environ Health 2002, 57:541-547. Cooper GS, Martin SA, Longnecker MP, Sandler DP, Germolec DR: Associations between plasma DDE levels and immunologic measures in African-American farmers in North Carolina. Environ Health Perspect 2004, 112:1080-1084. Mosmann TR, Sad S: The expanding universe of T-cell subsets: Th1, Th2 and more. lmmunol Today 1996, 17:138-146. Daniel V, Huber W, Bauer K, Suesal C, Conradt C, Opelz G: Associations of blood levels of PCB, HCHs, and HCB with numbers of lymphocyte subpopulations, in vitro lymphocyte response, plasma cytokine levels, and immunoglobulin autoantibodies. Environ Health Perspect 2001, 1092173- 178. McCabe MJ, Jr., Lawrence DA: Lead, a major environmental pollutant, is immunomodulatory by its differential effects on CD4+ T cells subsets. Toxicol Appl Pharmacol 1991 , 1 1 1:13-23. Heo Y, Lee BK, Ahn KD, Lawrence DA: Serum lgE elevation correlates with blood lead levels in battery manufacturing workers. Hum Exp Toxicol 2004, 23:209-213. Heo Y, Lee WT, Lawrence DA: In vivo the environmental pollutants lead and mercury induce oligoclonal T cell responses skewed toward type-2 reactivities. Cell lmmunol 1997, 179:185-195. Reichrtova E, Ciznar P, Prachar V, Palkovicova L, Veningerova M: Cord serum immunoglobulin E related to the environmental contamination of human placentas with organochlorine compounds. Environ Health Perspect 1999, 107:895-899. 161 Table 1: Descriptive characteristics of the study cohort. Total group Subgroup with CO and immune markers (N=671) (n=331) % % Boys 53.1 56.8 Age 7 years 45.8 46.2 8 years 50.2 50.2 9-10 years 4.1 3.6 Passive smoking in the child’s home during the last 12 months(cigarettes per day) None 52.2 66.5 1-10 23.4 24.8 11-20 14.3 5.3 more than 30 10.1 2.4 Number of infections during the last 12 months None 6.0 5.7 Less than 5 74.7 74.8 5 to 10 17.2 17.4 more than 10 2.1 2.1 Duration of total breastfeeding (weeks) 0 19.1 15.1 less than 5 17.9 15.4 5 — 8 12.5 12.1 9- 12 10.6 11.8 more than 12 34.7 41.1 Missing 5.2 4.5 Serum cholesterol concentration (mean, 5-95%-value, mg/dL) Triglyceride concentration imean, 5-95%-value, mg/dL) 162 186 (143-235) 130 (53-262) $.27 38 v.2“ 8.213: new :2: I 3: New 22% Isa: 33 8:14.28 .3 2:: $2802 8.? I cs: 0.3 2.: I 3: new 3.3 $284.2 8.: I no: new $.27 e28 new 3.3 I 3: EN :2 «If :28 $329888 23829888 3328888 83.329888 $320888 23829888 $349888 6329888 3329888 288228.888 832983.: $329888 33.323888 Charm—.8 ad afiolfi _dv mmd 23.0.; :8 mmd Smdlo— .8 $6 :2: 2.828 aseI882 .o GE: 288.0 32; :33 84888828 :25; _8 .3 627 2 _8 3o comer _ 18mg 223; _ .8 :8 EST :8 one :N.NI 8.8 28 a? I 2.8 83 2&7 8.8 as $.de 2.8 Re 8.2 I 2.8 83 E: I 2.8 one 8: I :8 SB 327 88 Rd 33 I 2.8 :8 so; I 2.8 who 33 I 2.8 and 33 I28 28 5.8 I 2.8 «we a; I 2.8 one :23 I 2.8 :3 a: I 2.8 88 2 I I 2.8 23 33 I 8.8 3o :23 I 2.8 e3 22: I 3.8 28 is I 8.8 :8 23.9 I 2.8 is 28.: I 2.8 NS See I 2.8 23 E: I 2.8 22. 5 8n :8 EA :1me v 3: 9:02 A287: A237: :8 8 I _ 528 252 a: as; 8 d Go: 28% w an: as; e 828 mom So: 25 23:92 N. 22s. 2: wztsn 2.2585 mo $3552 :8 com 329833 mfizoE 2 a2 2: mats—o 0E0: PEEU 2: E wExcEm 332$ masocwIQw/x Sacco 338 ca 388 :81? 3&3 mu: 3&8 emote N 388 man A5 @0350 828.50 .mouatgoo .3 am can DO coo—a 9.2—3 .5.— mo=_a> Roma ..m ...—a 58:. 2.52.806 "N 235. 163 n3 .30 .03 6: 69. d? _w: .2: mmeomcoo mom ho Ezm mmOQN .Gmdnseaam: mm; 00.3808 500 So: 020: .29 0858 00:90:00 x02 .82....“ so mezzo 20 N 0.05.2 0500 02.0.8.8 022. .002 0 002.8sz ucm 602.0205 6 E30 mm 62200.8 028. .99. 3 $08 3 082.20: 00.0 .10: 0.00 20208 a 00000-20: 2.2 02.0 .00... 00.00 8.00 20208 3 0008-20: 00.3: 3.0 5.0 08% 59¢ 800 20.00 608 a 808.20: 00.02 00.0 00.0 00.0 .moo 20.00 00.00 00.00 N000 00.0V0 0.08 «0.0 00.0 00.0 00.0 2.0 :0 2.0 :08 10:-» :00 20.00 20.00 000 $00 0000 200 ~00 00.0 00.0 E0 00.0 00.0 00.0 8.0 :03 mo: :00 S00 5.00 5.03 «0.0...0 00.00 tone 5.00 00.0 00.0 05.0 00.0 00.0 00.0 00.0 00.0 050 :08 000de E00 S00 000 S00 00.00 800 20.00 20.00 5.0%. 00.0 3.0 020 5.0 00.0 00.0 00.0 9.0 00.0 20.0 :08 moo cmmE 050580 05:80:-» 29:00: 29.3092 055000 0020: 9020: 10:-» m0: mmOQN .mcuoE o_.=oEoom :2: new :3": €803-23. can com—3.35 mucsonEoo 8500200590 2353 3:22:30 cozflotoo cant—03m “a min... 164 Table 4: White blood cell, eosinophilic characteristics, and basophilic surface lgE by CC and Pb (geometric mean} HCB (pg/L) DDE (pg/L) Sum of PCBs (pg/L) y-HCH (pg/L) Pb (pg/L) Outcome 3 0.2 0.21—0.29 0.30- 0.43 >043 50.30 0.31—0.48 0.49— 0.75 >075 s 0.15 0.16 —02 0.21—0.27 >027 0.01 0.02 >002 <22.0 22.1—28.3 28.4— 34.1 >34.1 N 78 89 79 85 80 86 82 83 84 77 86 84 91 130 110 82 81 86 82 White blood cells, x1 03/10 Total: crude 8136 8579 8131 8555 8373 8456 8528 8074 8456 8354 8383 8233 8062 8512 8421 8612 7907 8376 8552 Adjusted § 7782 8275 7970 8584’ 8354 8318 8447 7488‘ 8280 8037 8141 8131 7920 8284 8247 8397 7730' 8167 8290 F-test: p=0.09 F-test: p=0.02 F-test: p=0.92 F—test: p=0.09 F-test: p=0.33 Eosinophil cell count, x103/pL Total: crude 182 206 188 207 178 182 197 210 192 187 193 190 186 206 177 176 166 210 214 Adjusted § 176 220 208 223 181 194 217 235 245 217 196 172 199 225 194 190 185 233 218 F-test: p=0.37 F-test: p=0.61 F-test: p=0.31 F-test: p=0.32 F-test: p=0.22 Eosinophilic granula Total; crude 902 903 901 893 897 901 896 904 899 897 905 897 906 896 898 890 904 900 904 AdeSted § 919 913 910 895' 902 907 905 923 908 907 916 906 915 906 906 898 913 911 914 F-test: p=0.22 F-test: p=0.37 F-test: p=0.65 F-test: p=0.47 F-test: p=0.27 lgE count on basophils Total: CTUde 810 822 863 859 807 842 838 866 808 841 839 867 845 838 833 836 811 845 860 Adjusted § 851 851 896' 884 860 884 861 877 852 879 866 886 880 868 863 871 845 878 888 F-test: p=0.67 F-test: p=0.50 F-test: p=0.41 F-test: p=0.60 F-test: p=0.10 '1 absolute number of cells/0L. p s 0.05 based on a t-test compared with the lowest exposure category as the reference. § Adjusted for all exposures in the table (OC & Pb) in addition to gender, age, number of infections in the last 12 months, passive smoke exposure in the child's home in the last 12 months, and lipids (sum of cholesterol and triglycerides) 165 Table 5: Lymphocyte phenotypes by whole blood DDE, PCBs, HCB, y-HCH and Pb concentration (geometric mean) DDE (pg/L) Sum of PCBs (pg/L) HCB (119/L) y—HCH (pg/L) Pb (pg/L) <07 071-079 030 04‘ >043 <030 031048 049- > < Cells _ .- .- .2 . - . 3 . _ . . - . 0.75 0.75 _0.15 0.16 - 0.2 0.21- 0.27 >027 0_01 002 >002 <22.0 22.1-28.3 28.4-34.1 >34.1 N 78 89 79 85 80 86 82 83 84 77 86 84 91 130 110 82 81 86 82 T—cells (CD3+) Total: 7 crude -193 2183 2139 2242 2286 2179 2144 2156 2289 2170 2155 2146 2131 2212 2213 2318 2101 2160 2184 Adjusted § 1950 2005 1980 2076 2092 1998 1990 1932 2082 1975 1986 1968 1967 2020 2021 2118 1919' 1979 1999 F-test: p=0.71 F-test: p=0.74 F-test: p=0.76 F-test: p=0.77 F-test: p=0.17 T-helper cells CD3+CD4+) Total crude: 1204 1209 1205 1251 1276 1212 1198 1189 1293 1190 1209 1180 1155 1232 1255 1297 1182 1203 1192 AdeSted§ 1087 1138 1147 1200 1183 1145 1141 1100 1220 1113 1139 1100 1101 1150 1176 1214 1106 1128 1123 F-test: p=0.51 F-test: p:0.83 F-test: p=0.37 F-test: p20.37 F-test: p=0.24 Cytotoxic T-cells (CD3+CD8+) Totali crude 753 743 727 778 773 740 739 752 769 744 740 751 750 753 748 799 711 746 750 AdeSted § 660 665 649 692 697 660 668 642 678 658 662 669 671 669 659 712 634‘ 661 662 F-test: p=0.78 F-test: p:0.80 F--test: p=0.97 F-test: p=0.93 F—test: p=0.23 Memory T-helper cells (CD4+CD45RO+) Total crud: 315 249 352 325 316 277 308 325 265 349 311 307 331 274 325 317 304 327 341 Adjusted § 332 323 363 362 343 342 347 347 354 363 345 317 348 341 345 358 321 348 351 F-test: p=0.18 F -test: p=1.00 F-test: p=0.34 F—test: p=0.91 F-test: p=0.22 Natural killer cells (CD16+CD56+) Total . crud: 384 367 350 372 382 378 350 364 369 362 394 348 328 395 373 371 348 365 389 Adjusted § 371 338 334 362 378 366 317 345 326 348 390 342 322 368' 305' 350 322 354 378 F-test: p:0.48 F-test: p=0.24 F-test: p=0.16 F-test: p<0.01 F-test: p=0.20 > Natural killer cells subset (CD16+CD56+CD57+) 3 Tot cruzl: 156 153 148 148 159 156 142 147 155 144 164 142 137 162 151 154 148 148 156 Adjusted§ 168 157 158 162 177 171 142 156 148 153 183 162 143 175‘ 167 162 152 161 169 F-test: p=0.89 F-test: p=0.21 F-test: p=0.18 F-test: p=0.03 F-test: p=0.69 B-cells (CD3+CD5+CD19+) T c352]: 457 474 456 468 463 461 464 469 473 482 45 8 446 445 474 469 505 423 462 469 Adjusted§ 378 398 382 393 369 372 395 416 420 411 375 349 371 395 397 418 353' 389 393 F-test: p=1.00 F-test: p=0.67 F ~test: p=0.26 F-test: p=0.53 F-test: p=0.10 I absolute number of subtype cells/uL based on percent of lymphocytes x total lymphocyte count. p- < 0. 05 based on a t-test compared with the lowest exposure category as the refe § Adjusted for all exposures in the table (0C & Pb) in addition to gender, age, num and triglycerides) 166 rence. ber of infections in the last 12 months, passive smoke exposure in the child's home in the last 12 months and lipids (sum of cholesterol 0002.33wc. .050 300.00.00.93 8:0. 020: “.50 0500:. N. .00_ on. E 0:50. 0.2.0.0 2.. ... 0.000000 3.60:0 0.0.0000. .0502... N. .00. 2.. :. 0:0..00..E ..o .5953: 60.0 doucow o. 02.6.00 ... 30 0% 00. 0.0.0. 2.. E 00.200008 :0 81. 8.00.030 a 095.0%. 0... 0.0 0.00.0.8 0.00096 .0030. 2.. 0...? 3.00.0.3 .00.-. .0 so .0003 3.3 w 00 . 00.010 009.0 00.010 003.0 00.010 002.0 00010380-“. 00.010 080-0 00 00 00 00 .0 00 .0 00 00 .0 00 00 .0 00 00 .00 00 00 00 08.00.00. 00 00 00 .0 00 00 00 00 .0 00 00 00 00 00 00 .0 00 00 00 0.02.3 .0:..00. 00010 08.-.. 00.010 00.2-.. .000 08.-.. .000 002-0 00.010 009-0 00. 00. 00. 00. 00. 00. 00. .00. .0. 00. 00. .00. 00. 00. 00. 00. 00. 00. 00.000.00.03 00. 00. 0.. 00. 00. 0.. 00. 0.. 00. 00. 00. .0. 0.. 00. 00. 00. 00. .0. 00. 00.5 30.08.20. 00.010 08.-.. .0010 000... 00.010 080-0 00.010 080-... 00.010 080-0 00. 00. .0. 00. 00. .0. 00. .... 00. 00. 00. 00. 0.. 00. .0. ..0. .00. 00. 0:03.903. 005.0 00. 00. 00. 0.0. .0. 00. 00. 00. 00. 00. 00.1mm.-. 00. 00. 00. 00. 00. 00. 00. 30.08. <0. 00.010 003-... 00.010 0000.0 00010 009.0 00.010 08.-.. 0.010 0000.0 .00. .00. 0.0.. 0.0. 000. 000. .00. 00: 00: 80. 000. 000. 0.0. 00: 000. .000. 00.. 00.. E. 0.8.9.3. 203.0 0.: .0.. 0:. 00.. 00.. .... .0.. 00: 000. 00.. 00: 00.. 00.. 00.. 00.. 00.. 00.. 0... .0: 30.08.00. 00 00 .0 00 0.. 00. .0 00 00 00 00 00 00 00 00 00 00 00 00 z ..00 0.00 00.0 0.0 00.0 00.0 00.0 00.0 ..00A -000 -..00 0.00v 00.0A 00.0 .00 00.0A -.00 0.0 0.00 00.0A -000 -.00 00.00 00.0A -000 -.00 0.00 088.00 3.00. 00 3.00. 0.0:-.. 30.... mo: 3.00. 0.0.00.0 800 3.00. .00.0. Enos. £50582 00.2.8 ... 00.02.0808 00 0:... :01... do... 8.000 Mon 08... 0.2.; .3 05.000.020.08. ”0 0.000 167 CHAPTER 4 DISCUSSION 168 The pieces of the sex hormone disrupters — childhood allergy puzzle presented in this monograph suggest an immunomodulating role of sex hormone disrupters in the etiology of childhood allergic disorders. First, evidence was presented in support of DDE playing a role in the prenatal priming of allergic diseases through the alteration of cord plasma cytokines. Second, an association between prenatal exposure to exogenous sex steroid hormones and biomarkers of allergic susceptibility was demonstrated. Third, prenatal exposure to exogenous sex steroid hormones appears to be associated with allergic phenotypes such as asthma; and fourth, postnatal organochlorine exposure seems to alter serum biomarkers of allergy. The motivation for this work stems from my curiosity to investigate the speculation that sex steroid hormones disrupters may be important in the etiology of childhood allergy. This effort was represented by the use of both questionnaire and lab data for various exposures and outcomes. In addition, the data used were from cohorts of varying backgrounds (Caucasian and African-Caribbean) and study designs (cross-sectional and follow-up). The pathways explored in the first three manuscripts highlights the importance of in utero exposure by ‘connecting the dots' showing a link between: 1) placental p,p’ DDE and cord plasma lL-4, |L-13, and INF-y (cytokines suggesting allergy susceptibility); 2) Maternal oral contraceptive use before pregnancy (a proxy for in utero progesterone/estrogen exposure) and serum lgE, IgA, and basophilic surface lgE counts; 3) Maternal contraceptive use and asthma and hay fever in 169 offspring. These three pieces of work suggest that in utero exposure to sex steroid hormones may alter the immune status at birth, which may result in childhood allergic disorders. This suggests that the in utero environment may play a major role in the pathogenesis of childhood allergy. To the best of our knowledge, there is no report of an association between placental p,p' DDE and cord plasma cytokines. Noakes and co-workers 1 were unable to detect placental p,p’-DDE and used maternal levels instead. Though their results for cord lL-13 and INF-y were not significant, they tended in the same direction (increased |L-13, decreased INF-y) as our findings for these cytokines. This overall sex hormone — childhood allergy puzzle was made clearer with findings from the fourth manuscript. In children ages 7 to 10 years, serum DDE was shown to significantly increase humoral WBC count, lgE levels, lgE count on basophils, 196, and IgA: all considered important immune markers of allergy. This work would have benefited from the use of actual blood concentrations of estrogen and progesterone since previous findings on DC use and hormone levels are not consistent 2’3. However, since DDE is known to have weak estrogenic properties, the association between placental DDE and cord cytokines provides useful clues of possible results. Another limitation is the fact that, the duration between 0C use and pregnancy was not taken into consideration. The 170 effect of 00 use on childhood allergy may be minuscule if the period between 00 use and pregnancy is long. The fact that data from studies of different ethnic groups and study designs was used is noteworthy. Data from an African-Caribbean cohort (component 3) provided consistent findings to those from Caucassian cohorts. Another strength of this work is the different time-windows in which exposures and outcomes were assessed. On a whole, the effects of sex hormone disrupters on childhood allergic disorders are pervasive but unexplained. This compilation of four studies from three diverse cohorts suggests possible explanations. The first component (In utero exposure to SHD affects perinatal biomarkers of allergy) implies that placental DDE (a SHD) concentration may influence the priming of cord blood immune marker towards an allergic state. Whilst important, on its own this component may not be sufficient to make the leap from placental DDE to childhood allergy. This gap was narrowed by findings from component two (In utero SHD affects postnatal biomarkers of allergy), which indicate a possible relation between maternal oral contraceptive use before pregnancy and serum immune markers of allery in offspring. Furthermore, the data suggest that maternal 00 use may have sex-related differences of the immunomodulating effects in offspring. The first two components breached the gap between the in utero and postnatal environment. However, though immune markers (especially lgE levels) are good predictors of disease outcome they do not always provide consistent results 4'8. 171 The investigation was therefore taken to another level with component three (In utero exposure to SHD affects allergic outcomes in infancy) providing evidence of a possible involvement of maternal oral comtraceptive use in the etiology of childhood allergic diseases including asthma. By connecting the clues embedded in these three studies, a picture is painted that implies that in utero exposure to SHD may be a risk factor for childhood asthma (for example) by first primining the fetal immune response. This priming may be sustained into childhood, evident by altered serum allergic markers, and ultimately clinical allergic manesfestations. The findings of component four (Postnatal exposure to SHD affects biomarkers of allergy in infancy) was consistent with previous studies that inferred a link between postnatal HOC exposure and serum immune markers. These results should be interpreted with caution since HOC concentrations were measured once at 7-10 years of age. Hence we do not know whether the concentrations were higher before this age and are therefore responsible for the observed immune response. For example, it is possible that the increased serum immune levels may be the result of in utero and not postnatal exposures (see component 2). it would be advantageous to test all four components in one study; collecting all relevant biologic samples and questionnaire data at the appropriate time. However, since no such study exist, and may not be realistic, the present work provides important stepping stones along the path of better understanding the role of SHD (sex hormone disrupters) in the etiology of childhood allergy. 172 Future work The mechanism through which these compounds (organochlorines, exogenous estrogen and progesterone) disrupt normal sex hormone function is unclear. The epigenetic concept has been proposed as a credible explanation. This concept posits that adverse effects can result from enduring changes in gene expression 9. This means that the active portion of the genetic information is altered without a change in the genetic blueprint (epigenesis). The expression or silencing of genes then results in cells/tissue with permanent (irreversible) committed activities. Epigenetic mechanisms have been suggested as the underlying reason for the observed increased risk of several chronic diseases in adulthood associated with . . . . 1 -12 xenobrotrc exposures early in life 0 . For example, Fukuzawa et al. reported that (coplanar) PCB affects the expression of steroidogenic enzyme genes 13. In addition, Goldman et al. found that PCBs (Aroclor 1254) inhibit the cytochrome P—450 21-hydroxylase activity, thus blocking the branch from progesterone to glucocorticoid production 14. Furthermore, Chen et al. reported that DDT and its metabolites (including DDE) down-regulate the gene expression of the estrogen 1 5 receptor Future studies aimed at explicating the mechanism through which sex hormones are disrupted should embrace the epigenetic concept. Since gene expression can be detected as early as in fetal tissues of the placenta, it may be prudent to 173 assess the variations in gene expression in neonates with varying concentrations of exposure to DDE, PCB, exogenous estrogen, and progesterone. The effect of sex hormone disrupters on allergy may be from their direct impact on the gene expression of allergy related cytokines such as lL—4, lL-13, and INF-y. In summary, it is of personal and public health importance to consider the potential impact of sex hormone disrupters on the pathogenesis of allergy. The prenatal period is of particular importance since the fragile fetal immune system is more vulnerable to exogenous exposures. Therefore, intervention through education regarding exposure prevention should be implemented before pregnancy. Further exploration of the epigenetic concept is a fertile area for future research. 174 References 1. 10. Noakes PS, Taylor P, Wilkinson S, Prescott SL. The relationship between persistent organic pollutants in maternal and neonatal tissues and immune responses to allergens: A novel exploratory study. Chemosphere 2006;63(8):1304-1311. Barbieri RL, Gao X, Xu H, Cramer DW. Effects of previous use of oral contraceptives on early follicular phase follicle-stimulating hormone. Fertility and Sterility 1995;64(4):689-92. Moore JW, Key TJ, Wang DY, Bulbrook RD, Hayward JL, Takatani 0. Blood concentrations of estradiol and sex hormone -binding globulin in relation to age at menarche in premenopausal British and Japanese women. Breast Cancer Research and Treatment 1991;18 Suppl 12847-50. Eiriksson TH, Sigurgeirsson B, Ardal B, Sigfusson A, Valdimarsson H. Cord blood lgE levels are influenced by gestational age but do not predict allergic manifestations in infants. pediatric allergy and immunology 1994;5(1):5-10. Klink M, Cline MG, Halonen M, Burrows B. Problems in defining normal limits for serum lgE. J Allergy Clin lmmunol 1990;85(2):440-4. Bergmann RL, Edenharter G, Bergmann KE, Guggenmoos-Holzmann I, Forster J, Bauer CP, Wahn V, Zepp F, Wahn U. Predictability of early atopy by cord blood-lgE and parental history. Clin Exp Allergy 1997;27(7):752-60. Perkin MR, Strachan DP, Hc W, Lack G, Golding J. The predictive value of early life total immunoglobulin E measurement in identifying atopic children in a population-based birth cohort study. Pediatr Allergy lmmunol 2006;17(2):118-24. Croner S, Kjellman Nl, Eriksson B, Roth A. lgE screening in 1701 newborn infants and the development of atopic disease during infancy. Arch Dis Child 1982;57(5):364-8. Tchernitchin AN, Tchernitchin NN, Mena MA, Unda C, Soto J. Imprinting: perinatal exposures cause the development of diseases during the adult age. Acta Biol Hung 1999;50(4):425-40. Jirtle RL, Sander M, Barrett JC. Genomic imprinting and environmental disease susceptibility. Environ Health Perspect 2000;108(3):271-8. 175 11. 12. 13. 14. 15. Li S, Hursting SD, Davis BJ, McLachlan JA, Barrett JC. Environmental exposure, DNA methylation, and gene regulation: lessons from diethylstilbesterol-induced cancers. Ann N Y Acad Sci 2003;983:161-9. Li S, Washburn KA, Moore R, Uno T, Teng C, Newbold RR, McLachlan JA, Negishi M. Developmental exposure to diethylstilbestrol elicits demethylation of estrogen-responsive lactoferrin gene in mouse uterus. cancer research 1997;57(19):4356-9. Fukuzawa NH, Ohsako S, Nagano R, Sakaue M, Baba T, Aoki Y, Tohyama C. Effects of 3,3',4,4',5-pentachlorobiphenyl, a coplanar polychlorinated biphenyl congener, on cultured neonatal mouse testis. Toxicol In Vitro 2003;17(3):259-69. Goldman D, Yawetz A. The interference of aroclor 1254 with progesterone metabolism in guinea pig adrenal and testes microsomes. Journal of Biochemical Toxicology 1990;5(2):99-107. Chen CW, Hurd C, Vorojeikina DP, Arnold SF, Notides AC. Transcriptional activation of the human estrogen receptor by DDT isomers and metabolites in yeast and MCF-7 cells. Biochem Pharmacol 1997;53(8):1161-72. 176