10115510730290Birth Defects Res A Clin Mol TeratolBirth Defects Res. Part A Clin. Mol. Teratol.Birth Defects Research. Part A, Clinical and Molecular Teratology1542-07521542-076022678668339383910.1002/bdra.23021NIHMS387688ArticleEnvironmental and Genetic Contributors to Hypospadias: A Review of the Epidemiologic EvidenceCarmichaelSuzan L.1ShawGary M.1LammerEdward J.2Department of Pediatrics, Division of Neonatology, Stanford University School of Medicine, Stanford, CAChildren’s Hospital Oakland Research Institute, Oakland, CACorresponding author: Suzan L. Carmichael, Ph.D., Associate Professor, Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University, 1265 Welch Road, Rm X111, Stanford, CA 94305-5415, Telephone 650.736.0735, fax 650.721.5751, scarmichael@stanford.edu22620120862012720120172013947499510

This review evaluates current knowledge related to trends in the prevalence of hypospadias, the association of hypospadias with endocrine-disrupting exposures, and the potential contribution of genetic susceptibility to its etiology. The review focuses on epidemiologic evidence. Increasing prevalence of hypospadias has been observed, but such increases tend to be localized to specific regions or time periods. Thus, generalized statements that hypospadias is increasing are unsupported. Due to limitations of study designs and inconsistent results, firm conclusions cannot be made regarding the association of endocrine-disrupting exposures with hypospadias. Studies with more rigorous study designs (e.g., larger and more detailed phenotypes) and exposure assessment that encompasses more breadth as well as depth (e.g., specific endocrine-related chemicals) will be critical to make better inferences about these important environmental exposures. Many candidate genes for hypospadias have been identified, but few of them have been examined to an extent that enables solid conclusions. Further studies are needed that include larger sample sizes, comparison groups that are more representative of the populations from which the cases were derived, phenotype-specific analyses, and more extensive exploration of variants. In conclusion, examining the associations of environmental and genetic factors with hypospadias remain important areas of inquiry, although our actual understanding of their contribution to hypospadias risk in humans is currently limited.

hypospadiasurogenitalgenesenvironment
I. Introduction

Normal urethral closure, which occurs during the 8th –14th weeks of gestation, involves a continuous process of ventral fusion in the proximal to distal direction (Kurzrock and others, 1999; Seifert and others, 2008; Van Der Werff and others, 2000). Hypospadias is a congenital malformation in which the urethral opening is on the ventral side of the penis, rather than at the tip. It is one of the most common congenital malformations, affecting about four to six males per 1,000 male births (Dolk and others, 2004; Paulozzi, 1999; Paulozzi and others, 1997). It ranges in severity from a urethral meatus that is slightly off center to a meatus in the perineal area. About 70% of cases are considered mild (the meatus is at or distal to the coronal sulcus; also referred to as ‘first degree’), while the remaining 30% are considered moderate (the meatus is on the penile shaft; ‘second degree’) or severe (the meatus is in the penoscrotal or perineal area; ‘third degree’). Some hypospadias cases can be attributed to known underlying genetic causes or syndromes (<10%), but most are idiopathic (Manson and Carr, 2003). The public health impact of hypospadias is significant, given that it usually involves surgical correction, and even after correction, individuals born with hypospadias may experience impaired sexual function and psychosocial difficulties related to sexuality and sexual activity later in life (Jugenburg and Kipikasa, 1988; Mieusset and Soulie, 2005). Hypospadias tends to be more common among babies born to women who are non-Hispanic white, have higher education, and are older and nulliparous, and babies with hypospadias are more likely to be small-for-gestational age (Manson and Carr, 2003).

Three questions that currently arise with regard to hypospadias are whether its prevalence is increasing, whether its etiology is related to exogenous exposures that influence endocrine function (endocrine disruptors), and what genes contribute to its etiology. This paper evaluates current knowledge related to these questions – clarifying dividing lines between what is known versus what is suspected. The focus is on the epidemiologic evidence; experimental evidence has been reviewed in more detail by others (Gray and others, 2004; Kalfa and others, 2011c; Kalfa and others, 2010; Yamada and others, 2006).

II. Trends in Prevalence

Several reports suggested hypospadias was increasing in Europe in the 1970s (Paulozzi, 1999). Concerns were renewed upon publication of a report that hypospadias prevalence had doubled in the Atlanta area and other parts of the United States from 1970–1993 (from about 1.5–2.0 per 1,000 total births, to about 3.5) (Paulozzi and others, 1997). This was the first report of an increase outside of Europe (Paulozzi and others, 1997). It was also the first study to report prevalence by severity of the phenotype. This was potentially an important addition given that trends in moderate to severe phenotypes may be less subject to reporting bias than mild cases, e.g., mild cases may be more completely reported over time if there is heightened awareness and therefore increased reporting of uncorrected mild cases. However, the percent of cases in the Atlanta study that had any information on severity went from about 20% to 50% during the study period, which undermines the interpretability of trends by severity (the trend among more severe cases could be attributable to improved reporting of severity).

The U.S. report was followed by data indicating that the previously reported increasing prevalences in Europe had stabilized by the 1980s and 1990s, with the exception of Denmark (Aho and others, 2000; Paulozzi, 1999). Prevalences were stable or declining in many other countries and regions during this time period (Canada, Australia, New Zealand, California, Israel, China, Mexico, South America) (Paulozzi, 1999). Prevalences were increasing only in France, Czechoslovakia and Japan.

The next wave of prevalence studies captured more recent observations, from the 1990s onward. Most reported relatively stable or declining prevalences, including Atlanta and Japan (Abdullah and others, 2007; Ahmed and others, 2004; Correa and others, 2007; Dolk and others, 2004; Elliott and others, 2011; Fisch and others, 2010; Ghirri and others, 2009; Kurahashi and others, 2004; Loane and others, 2011; Martinez-Frias and others, 2004; Porter and others, 2005). Increases were reported in Western Australia for all levels of severity (Nassar and others, 2007), and a continued increase has been confirmed in Denmark (Lund and others, 2009). The majority of registries contributing to the European Surveillance of Congenital Anomalies (EUROCAT) reported stable or declining prevalences during this time period. The report noted that any observed increasing prevalences could be attributable to changes in reporting of mild cases over time (Dolk and others, 2004; Loane and others, 2011). Increases have also been reported in China, but initial prevalences were quite low (less than one per 1,000 male births), which suggests under-ascertainment of the condition (Jin and others, 2010; Li and others, 2012).

Potential increasing prevalences in hypospadias have been observed but such increases are localized to a few specific regions or time periods. Thus, generalized statements that hypospadias is increasing are clearly unsupported. Nevertheless, the areas that reveal changes in prevalence over time would be prime candidates for study to determine whether particular exposures are responsible. Incorporation of severity of phenotype into future studies of prevalence trends would be a useful adjunct whenever feasible, given concerns that changes in reporting of mild cases over time could bias observed trends.

III. Endocrine Disruption

Urethral closure depends on fetal conversion of testosterone to dihydrotestosterone (DHT) by steroid 5α-reductase type II, binding of the ligand DHT to the nuclear androgen receptor, and proper subsequent androgen receptor (AR) action. Hypospadias can be induced experimentally by exposures that interfere with androgen and estrogen synthesis and signaling pathways during sexual differentiation, including AR antagonists, inhibitors of 5α-reductase, and inhibitors of enzymes involved in steroid hormone synthesis (Gray and others, 2004; Noriega and others, 2005; Ostby and others, 1999)105. Thus, biologic plausibility and experimental evidence exists that endocrine disruptors, defined broadly as exogenous substances that interfere with hormones, may cause hypospadias in humans.

The purported increasing prevalence of hypospadias does not necessarily provide evidence supporting a connection between hypospadias and endocrine disruptors. Given the ubiquity, variety, and dynamic mix of endocrine-disrupting exposures in our environment over time, and even assuming there existed a generalized increased prevalence, drawing an inference of such a connection is tenuous at best and ecological fallacy at worst. Determining a possible connection between endocrine-disrupting exposures and hypospadias is best carried out in carefully designed epidemiologic studies that capture more individualized exposure assessments.

Environmental chemicals

The group of chemicals that has received the most attention with respect to hypospadias is pesticides, some of which are endocrine disruptors. A recent meta-analysis of seven studies indicated that maternal agricultural work or occupational exposure to pesticides was associated with a modest increase in risk of hypospadias – reporting an odds ratio of 1.4 (95% confidence interval (CI) 1.0, 1.8) for mother’s occupation (Rocheleau and others, 2009). Three large occupation-based studies published since then, however, have not provided evidence for increased risk (Morales-Suarez-Varela and others, 2011; Nassar and others, 2010; Rocheleau and others, 2011).

Other groups of chemicals that include endocrine disruptors have also been evaluated. In particular, several studies have used job exposure matrices to assess probable maternal occupational exposure to specific groups of chemicals (Table 1). Use of such matrices tends to be more rigorous than simply asking mothers to recall exposure to specific types of compounds. A small questionnaire-based study in Italy found increased risks with probable exposure to several groups of chemicals (Giordano and others, 2010), but odds ratios were relatively imprecise and whether controls and cases were derived from the same underlying population is uncertain. A questionnaire-based study in England reported increased risk with phthalate exposure, but control participation was low (33%), which could undermine the study’s validity (Ormond and others, 2009). A large study in the United Kingdom based on birth defects registry data reported that hypospadias was not more likely than expected among infants born to women with any particular occupational exposures (Vrijheid and others, 2003). A large study in Australia based on occupational data from birth registration forms reported increased risk with heavy metals, but not other groups of chemicals (Nassar and others, 2010). A prospective cohort study conducted in Denmark reported increased risks with phthalates and alkylphenols (Morales-Suarez-Varela and others, 2011). Thus, some studies have suggested increased risk with certain groups of chemicals, but results are inconsistent.

Paternal exposures have also been examined

Evidence for increased hypospadias risk associated with paternal occupational exposures is modest. The recent meta-analysis of occupational exposure to pesticides included eight studies of paternal exposures; the odds ratio was 1.2 (95% CI 1.0, 1.4) (Rocheleau and others, 2009). Results from occupation-based studies of exposures to other groups of chemicals suggest increased risk with occupational exposure to biphenolic compounds, although the confidence intervals include one (Table 2). The premise underlying these studies of paternal exposures is often not stated but may include maternal exposure via seminal fluid (Rocheleau and others, 2009).

Although these studies are useful for generating clues, they must be interpreted with caution. They are limited in that they do not consider non-occupational sources of the chemicals, not all chemicals within each group have endocrine-disrupting properties, and the endocrine disruptors within each group may contribute to different (or even opposing) mechanisms of action, e.g., some endocrine disruptors within a group may have estrogenic properties while others are anti-estrogenic.

Some studies have attempted to examine exposures more directly. A study in Arkansas analyzed residential proximity to agriculture-related pesticide applications, which was estimated based on data related to phenology and average annual pesticide applications (Meyer and others, 2006). They examined many individual pesticides and also groupings of pesticides that were based on toxicologic evidence, including their specific endocrine-disrupting effects (e.g., anti-androgenic). Higher exposures were not associated with increased risk; in fact, almost all odds ratios were less than one, albeit imprecise. Several studies have measured maternal serum levels of DDE, a metabolite of DDT, a persistent pesticide used until the 1970s that has known estrogenic and anti-androgenic properties. These studies do not provide evidence for increased hypospadias risk with increased exposure (Bhatia and others, 2005; Carmichael and others, 2010; Flores-Luevano and others, 2003; Giordano and others, 2010; Longnecker and others, 2002), even during the 1960s when exposure was much higher than today (Longnecker and others, 2002). A study in India reported higher levels of DDE in cases than controls, but levels were measured up to ten years after birth (Shekharyadav and others, 2011). Cases also had significantly higher levels of HCH (hexachlorocyclohexane), but not any other organochlorine pesticide. Maternal levels of chlordane (Trabert and others, 2011) and PCBs (polychlorinated biphenyls) (McGlynn and others, 2009) were examined in a cohort of births from the 1960s, when levels were much higher than today. Chlordane was not associated with hypospadias (Trabert and others, 2011). Results suggested PCB exposure was associated with modestly increased risk (50–70% for the lowest versus higher quartiles), but the authors noted that the modest magnitude of the association, combined with lower current exposure levels, argue against PCBs being a substantial contributor to hypospadias risk. Other small studies measuring PCBs and other halogenated organic pollutants in maternal serum have not reported higher levels in mothers of hypospadias cases (Carmichael and others, 2010; Giordano and others, 2010). A study of maternal exposure to PBB (polybrominated biphenyl), a brominated flame retardant, was negative (Small and others, 2009). However, it was based on maternal PBB levels that may have been measured many years before delivery, and hypospadias was based on self-report of the sons.

Other potential chemical exposure sources have also been investigated. A study in Great Britain reported that hypospadias was not increased among infants living within 2 km of landfill sites (Elliott and others, 2001). A study in South Africa observed no differences in birth prevalence of hypospadias in villages that were, versus were not, sprayed with DDT for malaria control (Bornman and others, 2010). Two studies have reported no association of hypospadias with residential water source levels of water disinfection by-products (Iszatt and others, 2011; Luben and others, 2008).

A study in Spain observed higher placental levels of organochlorine pesticides and total effective xenoestrogen burden from organohalogenated compounds in hypospadias cases relative to controls (Fernandez and others, 2007). However, the case group of 50 infants also included isolated cryptorchidism cases, so results should be considered inconclusive. Hypospadias and cryptorchidism may have some overlapping etiologic factors, but they are mechanistically quite distinct.

One study reported that maternal self-reported use of biocides was associated with increased hypospadias risk (e.g., insect repellants, ant powder, flea and lice treatments) (Dugas and others, 2010). The exposures were not associated with increased risk when examined one at a time, with the exception of insect repellants. A higher total number of exposures was associated with increased risk. The control participation was only 33% for this study (Ormond and others, 2009), which may undermine the validity of its findings.

Vegetarian diet and phytoestrogens

Phytoestrogens are naturally occurring plant substances that have structural and functional similarities to estrogen. One small epidemiologic study suggested that a maternal vegetarian diet and high intake of legumes (which are high in lignans, a type of phytoestrogen) were associated with increased risk of hypospadias (North and Golding, 2000). Phytoestrogen intake was proposed as an explanation for the finding, and the finding has frequently been used to support the hypothesized connection between endocrine disrupting exposures and hypospadias. The study was followed by an experimental study indicating that the phytoestrogen genistein, which is particularly high in soy products, could induce hypospadias (Vilela and others, 2007). However, several studies have not confirmed an increased risk of hypospadias with a vegetarian diet or low meat or fish consumption (Brouwers and others, 2006b; Giordano and others, 2010; Giordano and others, 2008; Ormond and others, 2009; Pierik and others, 2004; Shekharyadav and others, 2011), with two exceptions (Akre and others, 2008; Samtani and others, 2011). Given limitations regarding study designs, sample sizes, and incomplete dietary assessment, however, these studies are somewhat problematic for drawing meaningful inferences.

Exogenous sex hormones and fertility

Two studies have observed at least a two-fold increased risk of hypospadias among women who took progestins (i.e., natural progesterone and synthetic progesterone and testosterone derivatives that produce biologic effects similar to those of progesterone) for the purpose of preventing pregnancy loss or other complications (Carmichael and others, 2005; Kallen and others, 1992). Progestin intake for the purpose of contraception does not appear to be associated with increased risk of hypospadias (Carmichael and others, 2005; Wogelius and others, 2006). This inconsistency is not surprising, given that the dose and types of progestins used in oral contraceptives and for pregnancy-related issues are very different, as are their mechanisms of action (Williams and Stancel, 1996). Maternal or paternal fertility problems or fertility-related procedures may also be associated with hypospadias (Bergh and others, 1999; Ericson and Kallen, 2001; Meijer and others, 2006; Silver and others, 1999; Sorensen and others, 2005b; Wennerholm and others, 1996), but it is uncertain whether the findings are due to underlying conditions, the procedures themselves or accompanying medications.

A few studies have examined whether hypospadias is associated with maternal intake of diethylstilbestrol (DES) a synthetic estrogen that was widely prescribed several decades ago. Early studies tended to be small and inconclusive (Henderson and others, 1976). However, more recent studies suggest that hypospadias risk may be increased among boys born to mothers who themselves were exposed to DES in utero (Brouwers and others, 2006a; Kalfa and others, 2011b; Klip and others, 2002; Pons and others, 2005), with one exception (Palmer and others, 2005). Each study has limitations, which is not surprising given the difficulty of studying trans-generational effects of a rare exposure. However, the findings are provocative and appear biologically plausible based on mechanistic and experimental data (Kalfa and others, 2011b).

Other medications

Certain medications may also confer endocrine-disrupting effects. Multiple studies have reported that valproic acid is associated with increased risk of hypospadias (Arpino and others, 2000; Diav-Citrin and others, 2008; Rodriguez-Pinilla and others, 2008). This association may be attributed to the gonadotropin-releasing hormone agonism of valproic acid, which has anti-androgenic effects long-term (Rodriguez-Pinilla and others, 2008). Corticosteroids, which have anti-androgenic properties, and loratadine, which has estrogenic properties, can both induce hypospadias experimentally (Ajayi and Fadiran, 1999; Lipworth, 1999; Negulescu and others, 1977; Reinisch and others, 1978; Willingham and others, 2006; Yucel and others, 2004a). They do not, however, appear to be associated with hypospadias risk in humans (Carmichael and others, 2009; CDC, 2004; Kallen and Olausson, 2006; 2007; Pedersen and others, 2008; Pedersen and others, 2006; Sorensen and others, 2005a).

Other factors

Other potential risk factors for hypospadias include placental abnormalities and maternal hypertension and pre-eclampsia (Akre and others, 2008; Akre and others, 1999; Boisen and others, 2005; Fujimoto and others, 2008). It is unknown – albeit plausible, and certainly worth further exploration – whether the co-occurrence of these conditions stems from common endocrine-related pathologies.

Summary

The available observational studies that can inform the question of whether endocrine disruptors are risk factors for hypospadias have thus far not provided solid evidence in support of the question. The studies have not been sufficiently rigorous or the results sufficiently consistent to justify a firm conclusion. Studies with more rigorous study designs (e.g., larger and detailed phenotypes) and exposure assessment that encompasses more breadth as well as depth (e.g., specific endocrine-related chemicals) will be critical to make better inferences about these important environmental exposures. For example, analyses that differentiate exposures more carefully based on their actual endocrine-disrupting properties (versus lumping large, broad categories of exposures together) will be particularly informative.

IV. Genetics

Heritability of hypospadias is high, at around 65–75%, and risk is estimated to be elevated 12- to 20-fold among first-degree relatives (Harris, 1990; Schnack and others, 2008). Most evidence from family-based studies points towards multifactorial (polygenic) inheritance of hypospadias, indicating that small effects of many genes and/or environmental factors act in concert to affect risk (Harris, 1990; Opitz, 1985). The current literature regarding genes and hypospadias has largely focused on candidate genes related to genital tubercle development and sex steroid metabolism. Experimental studies and case reports have provided many informative clues regarding potential relevant pathways and candidate genes, as reviewed by others (Kalfa and others, 2010; Kojima and others, 2010; Manson and Carr, 2003). The focus here is to summarize findings from epidemiologic studies in humans that included unaffected comparison groups.

Genital tubercle outgrowth and differentiation

Homeobox genes, bone morphogenetic proteins, and fibroblast growth factors all contribute to the development of the genital tubercle, the anlage for the penis (Baskin and others, 2004; Morgan and others, 2003; Yucel and others, 2004b). WT1 also contributes to early development of the urogenital system (Kohler and others, 2007; Kohler and others, 2004; Kohler and others, 1999), and WTAP (Wilms tumor 1-associated protein) may promote WT1 function. Studies have reported a greater frequency of mutations in cases than controls for HOXA4, HOXA6, BMP4, BMP7, WT1 and WTAP among Chinese subjects (Chen and others, 2007), and FGF8 and FGFR2 but not FGF10 or BMP7 among Swedish subjects (Beleza-Meireles and others, 2007c), and one study did not observe difference in German subjects for HOXA13 or WTAP (Utsch and others, 2003). The etiologic significance of these findings remains to be elucidated, given that the functional significance of many of the mutations are uncertain and attempts to replicate the findings in other study populations have not been published.

Fetal and placental biosynthesis and biotransformation of sex steroid hormones

SRD5A2 encodes steroid 5a-reductase type II, which converts testosterone (T) to DHT in the urethral seam(Kim and others, 2002). The V89L polymorphism (rs523349, +336G>C) has been investigated by several studies (Table 3). The C allele is associated with substantial reduction in enzyme activity (Samtani and others, 2010). Three studies have reported two- to three-fold increased risk associated with heterozygosity or homozygosity for the C allele, among Chinese, Caucasian and Indian subjects (Makridakis and others, 2000; Samtani and others, 2011; Thai and others, 2005). Two of these studies conducted phenotype-specific analyses and reported that results were similar regardless of severity (Samtani and others, 2011; Thai and others, 2005). A fourth study, of Japanese subjects, reported elevated risk only for severe cases (Sata and others, 2010). A fifth study, of Caucasians, reported no association (van der Zanden and others, 2010b). It is concerning (but not easily explained) that the study reporting no association was by far the largest (van der Zanden and others, 2010b). True population differences are possible but difficult to discern given the highly variable approach to control selection across the studies.

SRD5A1 is an isoform of SRD5A2. One small study did not observe differences in mutations SRD5A1 among 10 cases and 49 controls (Tria and others, 2004).

CYP17 (p450c17) is critical to the biosynthesis and metabolism of androgens and estrogens (Sharp and others, 2004). A polymorphism in the 5′-untranslated promoter region, rs743572 (c.-34T>C), has been studied extensively (it is also referred to as the MspA1 polymorphism, with A2 signifying the variant allele). A study in India observed that relative to the T/T genotype, the OR for the T/C genotype was 1.2 (95% CI 0.6, 2.5), and for C/C it was 2.1 (95% CI 0.8, 5.1) (Samtani and others, 2010). Results were more pronounced among moderate than severe cases. Another study in India included 91 cases and 132 controls selected from pediatric outpatient clinics; the ORs were 1.2 (0.6, 2.1) for T/C and 0.9 (0.4, 2.2) for C/C (Yadav and others, 2011). Results from these two studies were imprecise, likely due to the small sample sizes. An explanation for the differences in results is uncertain, although it could be influenced by the different approach to control selection.

HSD3B2 (3beta-hydroxysteroid dehydrogenase type II) is also essential to the biosynthesis of androgens and estrogens. A sequencing study of 90 moderate/severe cases and 100 controls suggested more mutations were present in cases than controls (Codner and others, 2004).

HSD17B3 encodes 17beta-hydroxysteroid dehydrogenase type 3, which is responsible for the conversion of androstenedione to testosterone. One study examined five SNPs in HSD17B3 (Sata and others, 2010). The SNP rs2066479 (+913G>A) was associated with increased risk, regardless of severity; the OR for the GA genotype was 1.5 (95% CI 0.9, 2.4), and for the AA genotype it was 3.1 (95% CI 1.4, 6.8).

CYP1A1 contributes to the 2-hydroxylation of estrogens, which yields less estrogenic metabolites than the 4- and 16-alpha hydroxylation catalyzed by CYP3A4 (Kurahashi and others, 2005). As a Phase I biotransformation enzyme, it also contributes to the detoxification of environmental toxicants. In one study, the variant allele of the CYP1A1 m1 polymorphism (rs4646903, T3801C), which is associated with elevated enzyme activity, was protective against hypospadias, regardless of severity (Kurahashi and others, 2005). The odds ratio was 0.2 (95% CI 0.04, 0.7) for the CT genotype and 0.7 (95% CI 0.1–3.9) for the CC genotype. However, the m1 polymorphism, as well as the m2 polymorphism (rs1048943, A2455G), were not significantly associated with hypospadias in another study, although the associations with the variant alleles did tend to be in the protective direction (Shekharyadav and others, 2011). These two studies also examined GSTM1 and GSTT1, which encode glutathione S-transferases, Phase II biotransformation enzymes and which each include a well-studied null deletion. In one study, the null deletions were not associated with hypospadias (Kurahashi and others, 2005). In the other study, the presence of both deletions (versus neither) was associated with increased risk (OR 2.3, 95% CI 1.1, 4.6) (Shekharyadav and others, 2011).

Regulation of sex steroid hormone biosynthesis and action

AR (androgen receptor), which is on the X chromosome, mediates the biologic effects of gonadal androgens (Hiort and Holterhus, 2000). Longer CAG repeats, which encode a polyglutamine tract in exon 1, are associated with reduced AR expression (Tut and others, 1997). One study reported longer CAG repeats among cases than controls (Lim and others, 2000), but others have not (Aschim and others, 2004; Muroya and others, 2001). One study reported longer GGN (glycine) repeats (also in exon 1) among cases (Aschim and others, 2004). Limited investigation of variants other than the CAG repeat as well as highly variable approaches to control selection make it difficult to draw firm conclusions from these results.

Two isoforms of the human estrogen receptor, ESR1 and ESR2 (alpha and beta) function analogously to the AR. They contribute to sex steroid levels and activity, and they interact with the AR to regulate gene expression (Beleza-Meireles and others, 2007b). Several studies have suggested that polymorphisms in ESR1 and ESR2 may be associated with hypospadias (Table 4). However, there are inconsistencies in results across studies, relatively few variants have been studied, and most studies have examined different sets of variants.

FKBP4 is essential to AR activity (Yong and others, 2006). One study examined two SNPs (rs1062478 and rs3021522); differences between cases and controls were not significant (Beleza-Meireles and others, 2007a). SRY (sex determining region Y) is critical to sexual differentiation; SOX9 (SRY-box 9) is part of its downstream signaling cascade (Wang and others, 2004). One study did not observe any mutations in the exons or exon-intron boundaries of SRY or SOX9 (Wang and others, 2004).

Other candidate genes

ATF3 (activating transcription factor 3) was identified through microarray expression studies demonstrating up-regulation of ATF3 in penile tissue from boys with hypospadias (Liu and others, 2005; Wang and others, 2007) and in the genital tubercle of mice exposed to estrogen (Liu and others, 2006). Two studies have suggested a greater frequency of mutations in cases than controls (Kalfa and others, 2008b; van der Zanden and others, 2010b). A large study of Swedish subjects reported that variant alleles of three of eight studied ATF3 SNPs were independently and significantly associated with hypospadias (rs3125289, rs11119982, rs1877474, all in intron 1) (Beleza-Meireles and others, 2008). A large study of Dutch subjects examined rs11119982 (van der Zanden and others, 2010b). The study reported that the odds ratio for the C allele was 0.8 (0.7, 1.0), and results were similar regardless of phenotype. In contrast, the odds ratio for the C allele was 1.5 (95% IC 1.3, 1.9) in the Swedish study (Beleza-Meireles and others, 2008).

The contribution of MAMLD1 (mastermind-like domain containing 1, or CXorf6, on the X chromosome) to hypospadias was initially investigated because subjects who had microdeletions that involved MAMLD1 as well as MTM1, the gene responsible for myotubular myopathy, had abnormal genital development (Fukami and others, 2006). The original sequencing study, along with three others, have suggested a greater frequency of mutations among cases than controls (Chen and others, 2010; Fukami and others, 2006; Kalfa and others, 2011a; Kalfa and others, 2008a). One study examined two SNPs (rs41313406, or c.856C>T; and rs2073043, or c.1766A>G) among Swedish subjects (Chen and others, 2010). Possession of both rare alleles (the T-G haplotype) was more frequent among cases than controls. This finding was confirmed by a subsequent study (Kalfa and others, 2011a). Combining the data from the two studies resulted in an odds ratio of 1.9 (95% CI 1.2, 3.0) for the T-G haplotype.

Recently, the first genome-wide association study (GWAS) of hypospadias was published (van der Zanden and others, 2010a). The study involved a pooled GWAS of 436 Dutch cases and 494 controls, followed by individual-level genotyping of 11 SNPs in the discovery sample and 220 additional Dutch cases and 328 Swedish cases. Dutch controls were derived from a separate case-control study; Swedish controls were derived from voluntary blood donors and from placental samples. The strongest finding was for the two SNPs that were individually genotyped for DGKK (rs1934179, which is intronic, and rs7063116, which is 21kb upstream). DGKK encodes diacylglycerol kinase K and is on the X chromosome. The variant alleles were associated with more than a two-fold increased risk of mild and moderate hypospadias in the Dutch and Swedish subjects; they were associated with severe hypospadias only among the Swedish subjects. The two SNPs were in high linkage disequilibrium, and their potential functional effect is unknown.

Summary

The list of biologically plausible candidate genes for hypospadias extends well beyond those that have been studied thus far. For many of the genes that have been examined in humans, studies have not gone beyond mutational screens of small sets of subjects, nor has the potential functional significance, if any, of most of the observed variants been investigated. Few genes have been examined to an extent that enables comparisons across different study populations, but even then studies have often examined different sets of variants and had study design differences that complicate such comparisons, such as differences in racial-ethnic composition and highly variable approaches to control selection. Future studies will be better able to contribute to the current knowledge base by including larger sample sizes, comparison groups that are more representative of the populations from which the cases were derived, phenotype-specific analyses, and more extensive exploration of variants. It is also important that future endeavors include gene discovery approaches; certainly the initial findings from the recent GWAS (van der Zanden and others, 2010a), as well as a small array comparative genomic hybridization study (Tannour-Louet and others, 2010), have provided provocative findings that merit follow-up.

V. Conclusions & Recommendations

Extensive experimental evidence suggests that endocrine-disrupting exposures contribute to hypospadias etiology. The observational epidemiologic literature, however, falls short regarding whether (or which) environmental exposures contribute. There is highly publicized concern about these and other chemicals in our environment. Nevertheless, generalizations that focus on ‘positive’ findings rather than a more comprehensive view that incorporates null findings and study limitations should be avoided. It is important that future studies are designed to produce strong internal as well as external validity, as well as measurements that cover a broader range of chemicals, in a more direct way.

Extensive evidence suggests that genetic variation contributes to hypospadias, but again the observational literature does not provide firm evidence regarding which genetic variants contribute. In the area of genetic susceptibility, important challenges for future studies will be to incorporate a more in-depth inquiry of existing candidate genes and more attention to gene discovery tools. Another critical goal is to be able to interrogate interactions among the various endogenous, exogenous and genetic factors of interest. Thus far, studies have tended to be too small and/or lacking the data to make such inquiries.

Another lingering concern is whether phenotypic heterogeneity exists. Some studies suggest that heritability is somewhat higher for more severe hypospadias cases (Bauer and others, 1981; Opitz, 1985; Schnack and others, 2008; Sweet and others, 1974), while others do not (Brouwers and others, 2010; Fredell and others, 2002). Studies examining candidate genes have tended to report similar findings across the range of severity (Sata and others, 2010; Silver and Russell, 1999; Thai and others, 2005; van der Zanden and others, 2010a; Watanabe and others, 2007), as have studies examining descriptive risk factors such as age, parity and race-ethnicity(Brouwers and others, 2010; Carmichael and others, 2003). The association of hypospadias with low birthweight tends to be stronger, the more severe the phenotype (Brouwers and others, 2010; Carlson and others, 2009; Carmichael and others, 2003), but this may simply reflect the underlying severity of the condition, rather than different etiologies. Regarding embryology, more recent studies indicate that urethral closure involves a continuous process of ventral fusion in the proximal to distal direction (Kurzrock and others, 1999; Seifert and others, 2008; Van Der Werff and others, 2000). Thus, existing evidence tends to point toward similarity of etiology regardless of severity, but many studies have had sample sizes that limited the ability to detect phenotypic differences. The most prudent approach at this point is to consider phenotype-specific analyses whenever feasible.

Examining the associations of environmental and genetic factors with hypospadias remain important areas of inquiry, although our actual understanding of their contribution to hypospadias risk in humans is currently limited. A more solid understanding of their contribution to hypospadias etiology will help guide the development of appropriate messages related to public health as well as individual-level risk, and it will facilitate the elucidation of underlying mechanisms, which are all important to its eventual prevention.

This project was partially supported by NIH R01ES017060 and CDC 6U01DD000489.

AbdullahNPearceMParkerLWilkinsonJJaffrayBMcNallyRJ2007Birth prevalence of cryptorchidism and hypospadias in northern England, 1993–2000Arch Dis Child92757657917142312AhmedSFDobbieRFinlaysonARGilbertJYoungsonGChalmersJStoneD2004Prevalence of hypospadias and other genital anomalies among singleton births, 1988–1997, in ScotlandArch Dis Child Fetal Neonatal Ed892F149F15114977900AhoMKoivistoAMTammelaTLJAuvinenA2000Is the incidence of hypospadias increasing? Analysis of Finnish hospital discharge data 1970–1994Environ Health Perspect108546346510811575AjayiGOFadiranEO1999The effect of combined iron therapy (Chemiron) and single iron therapy on the dexamethasone-estriol reaction test for placenta insufficiency during normal pregnancyClin Exp Obstet Gynecol261273010412619AkreOBoydHAAhlgrenMWilbrandKWestergaardTHjalgrimHNordenskjoldAEkbomAMelbyeM2008Maternal and gestational risk factors for hypospadiasEnviron Health Perspect11681071107618709149AkreOLipworthLCnattingiusSSparCnPEkbomA1999Risk factor patterns for cryptorchidism and hypospadiasEpidemiol104364369ArpinoCBrescianiniSRobertECastillaEECocchiGCornelMCde ViganCLancasterPALMerlobPSumiyoshiYZampinoGRenziCRosanoAPastroiacovoP2000Teratogenic effects of antiepileptic drugs: use of an International Database on Malformations and Drug Exposure (MADRE)Epilepsia411436144311077457AschimELNordenskjoldAGiwercmanALundinKBRuhayelYHaugenTBGrotmolTGiwercmanYL2004Linkage between cryptorchidism, hypospadias, and GGN repeat length in the androgen receptor geneJ Clin Endocrinol Metab89105105510915472213BaskinLSLiuWBastackyJYucelSBaskinLS2004Anatomical studies of the mouse genital tubercleHypospadias and Genital DevelopmentNew YorkKluwer Academic/Plenum Publishers103121BauerSBRetikABColodnyAH1981Genetic aspects of hypospadiasUrol Clin North Am835595647324321Beleza-MeirelesABarbaroMWedellATohonenVNordenskjoldA2007aStudies of a co-chaperone of the androgen receptor, FKBP52, as candidate for hypospadiasReprod Biol Endocrinol5817343741Beleza-MeirelesAKockumILundbergFSoderhallCNordenskjoldA2007bRisk factors for hypospadias in the estrogen receptor 2 geneJ Clin Endocrinol Metab9293712371817579196Beleza-MeirelesALundbergFLagerstedtKZhouXOmraniDFrisenLNordenskjoldA2007cFGFR2, FGF8, FGF10 and BMP7 as candidate genes for hypospadiasEur J Hum Genet15440541017264867Beleza-MeirelesATohonenVSoderhallCSchwentnerCRadmayrCKockumINordenskjoldA2008Activating transcription factor 3: a hormone responsive gene in the etiology of hypospadiasEur J Endocrinol158572973918426833BerghTEricsonAHillensjoTNygrenKGWennerholmUB1999Deliveries and children born after in-vitro fertilisation in Sweden 1982–95: a retrospective cohort studyLancet35491901579158510560671BhatiaRShiauRPetreasMWeintraubJMFarhangLEskenaziB2005Organochlorine pesticides and male genital anomalies in the child health and development studiesEnviron Health Perspect113222022415687061BoisenKAChellakootyMSchmidtIMKaiCMDamgaardINSuomiAMToppariJSkakkebaekNEMainKM2005Hypospadias in a cohort of 1072 Danish newborn boys: prevalence and relationship to placental weight, anthropometrical measurements at birth, and reproductive hormone levels at three months of ageJ Clin Endocrinol Metab9074041404615870122BornmanRde JagerCWorkuZFariasPReifS2010DDT and urogenital malformations in newborn boys in a malarial areaBJU Int106340541119849691BrouwersMMFeitzWFRoelofsLAKiemeneyLAde GierRPRoeleveldN2006aHypospadias: a transgenerational effect of diethylstilbestrol?Hum Reprod21366666916293648BrouwersMMFeitzWFRoelofsLAKiemeneyLAde GierRPRoeleveldN2006bRisk factors for hypospadiasEur J Pediatr16667167817103190BrouwersMMvan der ZandenLFde GierRPBartenEJZielhuisGAFeitzWFRoeleveldN2010Hypospadias: risk factor patterns and different phenotypesBJU Int105225426219751252CarlsonWHKiselySRMacLellanDL2009Maternal and fetal risk factors associated with severity of hypospadias: a comparison of mild and severe casesJ Pediatr Urol5428328619131278CarmichaelSLHerringAHSjodinAJonesRNeedhamLMaCDingKShawGM2010Hypospadias and halogenated organic pollutant levels in maternal mid-pregnancy serum samplesChemosphere80664164620494400CarmichaelSLMaCWerlerMMOlneyRSShawGM2009Maternal corticosteroid use and hypospadiasJ Pediatr1551394419394038CarmichaelSLShawGMLaurentCCroughanMSOlneyRSLammerEJThe National Birth Defects Prevention S2005Maternal progestin intake and risk of hypospadiasArch Pediatr Adolesc Med15995796216203941CarmichaelSLShawGMNelsonVSelvinSTorfsCPCurryCJ2003Hypospadias: trends and descriptive epidemiology in a California populationEpidemiol14701706CDC2004Evaluation of an association between loratadine and hypospadias—United States, 1997–2001MMWR Morb Mortal Wkly Rep531021922115029117ChenTLiQXuJDingKWangYWangWLiSShenY2007Mutation screening of BMP4, BMP7, HOXA4 and HOXB6 genes in Chinese patients with hypospadiasEur J Hum Genet151232817003840ChenYThaiHTLundinJLagerstedt-RobinsonKZhaoSMarkljungENordenskjoldA2010Mutational study of the MAMLD1-gene in hypospadiasEur J Med Genet53312212620347055CodnerEOkumaCIniguezGBoricMAAvilaAJohnsonMCCassorlaFG2004Molecular study of the 3 beta-hydroxysteroid dehydrogenase gene type II in patients with hypospadiasJ Clin Endocrinol Metab89295796414764821CorreaACraganJDKucikJEAlvrsonCJGilboaSM2007The Metropolitan Atlanta Congenital Defects Program 40th anniversary edition surveillance reportBirth Defects Res A Clin Mol Teratol79118617177279Diav-CitrinOShechtmanSBar-OzBCantrellDArnonJOrnoyA2008Pregnancy outcome after in utero exposure to valproate : evidence of dose relationship in teratogenic effectCNS Drugs22432533418336060DolkHVrijheidMScottJEAddorMCBottingBde ViganCde WalleHGarneELoaneMPieriniAGarcia-MinaurSPhysickNTenconiRWieselACalzolariEStoneD2004Toward the effective surveillance of hypospadiasEnviron Health Perspect112339840214998760DugasJNieuwenhuijsenMJMartinezDIszattNNelsonPElliottP2010Use of biocides and insect repellents and risk of hypospadiasOccup Environ Med67319620019951933ElliottCSHalpernMSPaikJMaldonadoYShortliffeLD2011Epidemiologic trends in penile anomalies and hypospadias in the state of California, 1985–2006J Pediatr Urol7329429821527236ElliottPBriggsDMorrisSde HooghCHurtCJensenTKMaitlandIRichardsonSWakefieldJJarupL2001Risk of adverse birth outcomes in populations living near landfill sitesBMJ323730936336811509424EricsonAKallenB2001Congenital malformations in infants born after IVF: a population-based studyHum Reprod16350450911228220FernandezMFOlmosBGranadaALopez-EspinosaMJMolina-MolinaJMFernandezJMCruzMOlea-SerranoFOleaN2007Human exposure to endocrine-disrupting chemicals and prenatal risk factors for cryptorchidism and hypospadias: a nested case-control studyEnviron Health Perspect115Suppl 181418174944FischHHyunGHensleTW2010Rising hypospadias rates: disproving a mythJ Pediatr Urol61373920022563Flores-LuevanoSFariasPHernandezMRomano-RiquerPWeberJPDewaillyECuevas-AlpucheJRomieuI2003DDT/DDE concentrations and risk of hypospadias. Pilot case-control studySalud Publica Mex4543143814974286FredellLKockumIHanssonEHolmnerSLundquistLLackgrenGPedersenJStenburgAWestbackeGNordenskjoldA2002Heredity of hypospadias and the significance of low birth weightJ Urol16731423142711832761FujimotoTSuwaTKabeKAdachiTNakabayashiMAmamiyaT2008Placental insufficiency in early gestation is associated with hypospadiasJ Pediatr Surg43235836118280290FukamiMWadaYMiyabayashiKNishinoIHasegawaTCamerinoGKretzCBuj-BelloALaporteJYamadaGMorohashiKOgataT2006CXorf6 is a causative gene for hypospadiasNat Genet38121369137117086185GhirriPScaramuzzoRTBertelloniSPardiDCelandroniACocchiGDanieliRDe SantisLDi StefanoMCGerolaOGiuffreMGragnaniGSMagnaniCMeossiCMerusiISabatinoGTuminiSCorselloGBoldriniA2009Prevalence of hypospadias in Italy according to severity, gestational age and birthweight: an epidemiological studyItal J Pediatr351819558700GiordanoFAbballeADe FelipEdi DomenicoAFerroFGrammaticoPIngelidoAMMarraVMarroccoGVallascianiSFiga-TalamancaI2010Maternal exposures to endocrine disrupting chemicals and hypospadias in offspringBirth Defects Res A Clin Mol Teratol88424125020196143GiordanoFCarbonePNoriFMantovaniATaruscioDFiga-TalamancaI2008Maternal diet and the risk of hypospadias and cryptorchidism in the offspringPaediatr Perinat Epidemiol2224926018426520GrayLEJrOstbyJFurrJWolfCLambrightCWilsonVNoriegaN2004Toxicant-induced hypospadias in the male ratAdv Exp Med Biol54521724115086030HarrisEL1990Genetic epidemiology of hypospadiasEpidemiol Rev1229402286224HendersonBEBentonBCosgroveMBaptistaJAldrichJTownsendDHartWMackTM1976Urogenital tract abnormalities in sons of women treated with diethylstilbestrolPediatrics584505507972792HiortOHolterhusPM2000The molecular basis of male sexual differentiationEur J Endocrinol142210111010664515IszattNNieuwenhuijsenMJNelsonPElliottPToledanoMB2011Water consumption and use, trihalomethane exposure, and the risk of hypospadiasPediatrics1272e38939721220402JinLYeRZhengJHongSRenA2010Secular trends of hypospadias prevalence and factors associated with it in southeast China during 1993–2005Birth Defects Res A Clin Mol Teratol88645846520589915JugenburgIKipikasaA1988Fertility in patients with hypospadiasActa Chir Plast30286932470226KalfaNCassorlaFAudranFOulad AbdennabiIPhilibertPBeroudCGuysJMReynaudRAlessandriniPWagnerKBreaudJVallaJSMorisson LacombeGDauresJPBaskinLFukamiMOgataTSultanC2011aPolymorphisms of MAMLD1 gene in hypospadiasJ Pediatr Urol7658559122030455KalfaNLiuBKleinOAudranFWangMHMeiCSultanCBaskinLS2008aMutations of CXorf6 are associated with a range of severities of hypospadiasEur J Endocrinol159445345818635673KalfaNLiuBKleinOWangMHCaoMBaskinLS2008bGenomic variants of ATF3 in patients with hypospadiasJ Urol18052183218818804813KalfaNParisFSoyer-GobillardMODauresJPSultanC2011bPrevalence of hypospadias in grandsons of women exposed to diethylstilbestrol during pregnancy: a multigenerational national cohort studyFertil Steril9582574257721458804KalfaNPhilibertPBaskinLSSultanC2011cHypospadias: interactions between environment and geneticsMol Cell EndocrinolKalfaNSultanCBaskinLS2010Hypospadias: etiology and current researchUrol Clin North Am37215916620569795KallenBMartinez-FriasMLCastillaEERobertELancasterPAL1992Hormone therapy during pregnancy and isolated hypospadias: an international case-control studyInt J Risk Safety Med3183198KallenBOlaussonPO2006No increased risk of infant hypospadias after maternal use of loratadine in early pregnancyInt J Med Sci3310610716761079KallenBOlaussonPO2007Use of anti-asthmatic drugs during pregnancy. 3. Congenital malformations in the infantsEur J Clin Pharmacol6338338817279357KimKSLiuWCunhaGRRussellDWHuangHShapiroEBaskinLS2002Expression of the androgen receptor and 5 alpha-reductase type 2 in the developing human fetal penis and urethraCell Tissue Res307214515311845321KlipHVerloopJvan GoolJDKosterMEBurgerCWvan LeeuwenFE2002Hypospadias in sons of women exposed to diethylstilbestrol in utero: a cohort studyLancet35993121102110711943257KohlerBDelezoideALBoizet-BonhoureBMcPhaulMJSultanCLumbrosoS2007Coexpression of Wilms’ tumor suppressor 1 (WT1) and androgen receptor (AR) in the genital tract of human male embryos and regulation of AR promoter activity by WT1J Mol Endocrinol38554755417496156KohlerBPienkowskiCAudranFDelsolMTauberMParisFSultanCLumbrosoS2004An N-terminal WT1 mutation (P181S) in an XY patient with ambiguous genitalia, normal testosterone production, absence of kidney disease and associated heart defect: enlarging the phenotypic spectrum of WT1 defectsEur J Endocrinol150682583015191353KohlerBSchumacherVSchulte-OverbergUBiewaldWLennertTl’AllemandDRoyer-PokoraBGrutersA1999Bilateral Wilms tumor in a boy with severe hypospadias and cryptochidism due to a heterozygous mutation in the WT1 genePediatr Res45218719010022588KojimaYKohriKHayashiY2010Genetic pathway of external genitalia formation and molecular etiology of hypospadiasJ Pediatr Urol6434635419995686KurahashiNMurakumoMKakizakiHNonomuraKKoyanagiTKasaiSSataFKishiR2004The estimated prevalence of hypospadias in Hokkaido, JapanJ Epidemiol143737715242062KurahashiNSataFKasaiSShibataTMoriyaKYamadaHKakizakiHMinakamiHNonomuraKKishiR2005Maternal genetic polymorphisms in CYP1A1, GSTM1 and GSTT1 and the risk of hypospadiasMol Hum Reprod112939815579657KurzrockEABaskinLSCunhaGR1999Ontogeny of the male urethra: theory of endodermal differentiationDifferentiation6411512210234808LiYMaoMDaiLLiKLiXZhouGWangYLiQHeCLiangJZhuJ2012Time trends and geographic variations in the prevalence of hypospadias in ChinaBirth Defects Res A Clin Mol Teratol941364121960504LimHNChenHMcBrideSDunningAMNixonRMHughesIAHawkinsJR2000Longer polyglutamine tracts in the androgen receptor are associated with moderate to severe undermasculinized genitalia in XY malesHum Mol Genet9582983410749991LipworthBJ1999Systemic adverse effects of inhaled corticosteroid therapy: A systematic review and meta-analysisArch InternMed1599941955LiuBAgrasKWillinghamEVilelaMLBaskinLS2006Activating transcription factor 3 is estrogen-responsive in utero and upregulated during sexual differentiationHorm Res65521722216569931LiuBWangZLinGAgrasKEbbersMWillinghamEBaskinLS2005Activating transcription factor 3 is up-regulated in patients with hypospadiasPediatr Res5861280128316306208LoaneMDolkHKellyATeljeurCGreenleesRDensemJ2011Paper 4: EUROCAT statistical monitoring: identification and investigation of ten year trends of congenital anomalies in EuropeBirth Defects Res A Clin Mol Teratol91Suppl 1S314321381187LongneckerMPKlebanoffMABrockJWZhouHGrayKANeedhamLLWilcoxAJ2002Maternal serum level of 1,1-Dichloro-2,2-bis( p -chlorophenyl)ethylene and risk of cryptorchism, hypospadias, and polythelia among male offspringAm J Epidemiol155431332211836195LubenTJNuckolsJRMosleyBSHobbsCReifJS2008Maternal exposure to water disinfection by-products during gestation and risk of hypospadiasOccup Environ Med65642042918032532LundLEngebjergMCPedersenLEhrensteinVNorgaardMSorensenHT2009Prevalence of hypospadias in Danish boys: a longitudinal study, 1977–2005Eur Urol5551022102619155122MakridakisNMdi SalleEReichardtJK2000Biochemical and pharmacogenetic dissection of human steroid 5 alpha-reductase type IIPharmacogenetics10540741310898110MansonJMCarrMC2003Molecular epidemiology of hypospadias: review of genetic and environmental risk factorsBirth Defects ResA ClinMolTeratol6710825836Martinez-FriasMLPrietoDPrietoLBermejoERodriguez-PinillaECuevasL2004Secular decreasing trend of the frequency of hypospadias among newborn male infants in SpainBirth Defects Res A Clin Mol Teratol702758114991914McGlynnKAGuoXGraubardBIBrockJWKlebanoffMALongneckerMP2009Maternal pregnancy levels of polychlorinated biphenyls and risk of hypospadias and cryptorchidism in male offspringEnviron Health Perspec11714721476MeijerWMde Jong-Van den BergLTvan den BergMDVerheijJBde WalleHE2006Clomiphene and hypospadias on a detailed level: signal or chance?Birth Defects Res A Clin Mol Teratol76424925216586447MeyerKJReifJSVeeramachaneniDNLubenTJMosleyBSNuckolsJR2006Agricultural pesticide use and hypospadias in eastern ArkansasEnviron Health Perspect114101589159517035148MieussetRSoulieM2005Hypospadias: psychosocial, sexual, and reproductive consequences in adult lifeJ Androl26216316815713818Morales-Suarez-VarelaMMToftGVJensenMSRamlau-HansenCKaerlevLThulstrupAMLlopis-GonzalezAOlsenJBondeJP2011Parental occupational exposure to endocrine disrupting chemicals and male genital malformations: A study in the Danish National Birth Cohort studyEnviron Health101321235764MorganEANguyenSBScottVStadlerHS2003Loss of Bmp7 and Fgf8 signaling in Hoxa13-mutant mice causes hypospadiaDevelopment130143095310912783783MuroyaKSasagawaISuzukiYNakadaTIshiiTOgataT2001Hypospadias and the androgen receptor gene: mutation screening and CAG repeat length analysisMol Hum Reprod7540941311331662NassarNAbeywardanaPBarkerABowerC2010Parental occupational exposure to potential endocrine disrupting chemicals and risk of hypospadias in infantsOccup Environ Med67958558919939854NassarNBowerCBarkerA2007Increasing prevalence of hypospadias in Western Australia, 1980–2000Arch Dis Child92758058417405856NegulescuRIStreckerJRLauritzenCPalS1977The influence of betamethasone on the feto-placental unit: a preliminary reportJ Perinat Med53120132578813NoriegaNCOstbyJLambrightCWilsonVSGrayLEJr2005Late gestational exposure to the fungicide prochloraz delays the onset of parturition and causes reproductive malformations in male but not female rat offspringBiol Reprod7261324133515673607NorthKGoldingJ2000A maternal vegetarian diet in pregnancy is associated with hypospadias. The ALSPAC Study Team. Avon Longitudinal Study of Pregnancy and ChildhoodBJU Int8510711310619956OpitzJM1985Hypospadias [editorial comment]Am J Med Genet215760OrmondGNieuwenhuijsenMJNelsonPToledanoMBIszattNGenelettiSElliottP2009Endocrine disruptors in the workplace, hair spray, folate supplementation, and risk of hypospadias: case-control studyEnviron Health Perspec117303307OstbyJMonossonEKelceWRGrayLEJr1999Environmental antiandrogens: low doses of the fungicide vinclozolin alter sexual differentiation of the male ratToxicol Ind Health151–2486410188191PalmerJRWiseLARobboySJTitus-ErnstoffLNollerKLHerbstALTroisiRHooverRN2005Hypospadias in sons of women exposed to diethylstilbestrol in uteroEpidemiology16458358615951681PaulozziLJ1999International trends in rates of hypospadias and cryptorchidismEnviron Health Perspect10729730210090709PaulozziLJEricksonJDJacksonRJ1997Hypospadias trends in two US surveillance systemsPediatrics1008318349346983PedersenLNorgaardMRothmanKJSorensenHT2008Loratadine during pregnancy and hypospadiasEpidemiology19235936018277174PedersenLSkriverMVNorgaardMSorensenHT2006Maternal use of Loratadine during pregnancy and risk of hypospadias in offspringInt J Med Sci31212516575420PierikFHBurdorfADeddensJAJuttmannREWeberRF2004Maternal and paternal risk factors for cryptorchidism and hypospadias: a case-control study in newborn boysEnviron Health Perspect112151570157615531444PonsJCPapiernikEBillonAHessabiMDuymeM2005Hypospadias in sons of women exposed to diethylstilbestrol in uteroPrenat Diagn25541841915906411PorterMPFaizanMKGradyRWMuellerBA2005Hypospadias in Washington State: maternal risk factors and prevalence trendsPediatrics1154e495e49915741350ReinischJMSimonNGKarowWGGandelmanR1978Prenatal exposure to prednisone in humans and animals retards intrauterine growthScience2024366436438705336RocheleauCMRomittiPADennisLK2009Pesticides and hypospadias: a meta-analysisJ Pediatr Urol51172418848807RocheleauCMRomittiPASandersonWTSunLLawsonCCWatersMAStewartPAOlneyRSReefhuisJ2011Maternal occupational pesticide exposure and risk of hypospadias in the national birth defects prevention studyBirth Defects Res A Clin Mol Teratol911192793621954192Rodriguez-PinillaEMejiasCPrieto-MerinoDFernandezPMartinez-FriasML2008Risk of hypospadias in newborn infants exposed to valproic acid during the first trimester of pregnancy: a case-control study in SpainDrug Saf31653754318484787SamtaniRBajpaiMGhoshPKSaraswathyKN2010SRD5A2 gene mutations--a population-based reviewPediatr Endocrinol Rev81344021037542SamtaniRBajpaiMVashishtKGhoshPKSaraswathyKN2011Hypospadias risk and polymorphism in SRD5A2 and CYP17 genes: case-control study among Indian childrenJ Urol18562334233921511288SataFKurahashiNBanSMoriyaKTanakaKDIshizukaMNakaoHYahataYImaiHKakizakiHNonomuraKKishiR2010Genetic polymorphisms of 17beta-hydroxysteroid dehydrogenase 3 and the risk of hypospadiasJ Sex Med782729273820059664SchnackTHZdravkovicSMyrupCWestergaardTChristensenKWohlfahrtJMelbyeM2008Familial aggregation of hypospadias: a cohort studyAm J Epidemiol167325125618042671SeifertAWHarfeBDCohnMJ2008Cell lineage analysis demonstrates an endodermal origin of the distal urethra and perineumDev Biol318114315218439576SharpLCardyAHCottonSCLittleJ2004CYP17 gene polymorphisms: prevalence and associations with hormone levels and related factors. a HuGE reviewAm J Epidemiol160872974015466495ShekharyadavCBajpaiMKumarVAhmedRSGuptaPBanerjeeBD2011Polymorphism in CYP1A1, GSTM 1, GSTT1 genes and organochlorine pesticides in the etiology of hypospadiasHum Exp Toxicol30101464147421300689SilverRIRodriguezRChangTSGearhartJP1999In vitro fertilization is associated with an increased risk of hypospadiasJ Urol16161954195710332480SilverRIRussellDW19995alpha-reductase type 2 mutations are present in some boys with isolated hypospadiasJ Urol1623 Pt 21142114510458450SmallCMDeCaroJJTerrellMLDominguezCCameronLLWirthJMarcusM2009Maternal exposure to a brominated flame retardant and genitourinary conditions in male offspringEnviron Health Perspec11711751179SorensenHTPedersenLNorgaardMWogeliusPRothmanKJ2005aMaternal asthma, preeclampsia and risk of hypospadiasEpidemiology16680680716222172SorensenHTPedersenLSkriverMVNorgaardMNorgardBHatchEE2005bUse of clomifene during early pregnancy and risk of hypospadias: population based case-control studyBMJ330748312612715613365SweetRASchrottHGKurlandRCulpOS1974Study of the incidence of hypospadias in Rochester, Minnesota, 1940–1970, and a case-control comparison of possible etiologic factorsMayo Clin Proc49152584855617Tannour-LouetMHanSCorbettSTLouetJFYatsenkoSMeyersLShawCAKangSHCheungSWLambDJ2010Identification of de novo copy number variants associated with human disorders of sexual developmentPLoS One510e1539221048976ThaiHTKalbasiMLagerstedtKFrisenLKockumINordenskjoldA2005The valine allele of the V89L polymorphism in the 5-alpha-reductase gene confers a reduced risk for hypospadiasJ Clin Endocrinol Metab90126695669816174723TrabertBLongneckerMPBrockJWKlebanoffMAMcGlynnKA2011Maternal pregnancy levels of rans-nonachlor and oxychlordane and [revalence of cryptorchidism and hypospadias in male offspringEnviron Health PerspectEpub ahead of printTriaAHiortOSinneckerGH2004Steroid 5alpha-reductase 1 polymorphisms and testosterone/dihydrotestosterone ratio in male patients with hypospadiasHormRes614180183TutTGGhadessyFJTrifiroMAPinskyLYongEL1997Long polyglutamine tracts in the androgen receptor are associated with reduced trans-activation, impaired sperm production, and male infertilityJ Clin Endocrinol Metab8211377737829360540UtschBKayaAOzburunALentzeMJAlbersNLudwigM2003Exclusion of WTAP and HOXA13 as candidate genes for isolated hypospadiasScand J Urol Nephrol37649850114675924Van Der WerffJFANievelsteinRAJBrandsELuijsterburgAJMVermeij-KeersC2000Normal development of the male anterior urethraTeratology6117218310661906van der ZandenLFvan RooijIAFeitzWFKnightJDondersARRenkemaKYBongersEMVermeulenSHKiemeneyLAVeltmanJAArias-VasquezAZhangXMarkljungEQiaoLBaskinLSNordenskjoldARoeleveldNFrankeBKnoersNV2010aCommon variants in DGKK are strongly associated with risk of hypospadiasNat Genet431485021113153van der ZandenLFvan RooijIAFeitzWFVermeulenSHKiemeneyLAKnoersNVRoeleveldNFrankeB2010bGenetics of hypospadias: are single-nucleotide polymorphisms in SRD5A2, ESR1, ESR2, and ATF3 really associated with the malformation?J Clin Endocrinol Metab9552384239020215396VilelaMLWillinghamEBuckleyJLiuBCAgrasKShiroyanagiYBaskinLS2007Endocrine disruptors and hypospadias: role of genistein and the fungicide vinclozolinUrology70361862117905137VrijheidMArmstrongBDolkHvan TongerenMBottingB2003Risk of hypospadias in relation to maternal occupational exposure to potential endocrine disrupting chemicalsOccup Environ Med60854355012883014WangYLiQXuJLiuQWangWLinYMaFChenTLiSShenY2004Mutation analysis of five candidate genes in Chinese patients with hypospadiasEurJ HumGenet129706712WangZLiuBCLinGTLinCSLueTFWillinghamEBaskinLS2007Up-regulation of estrogen responsive genes in hypospadias: microarray analysisJ Urol17751939194617437852WatanabeMYoshidaRUeokaKAokiKSasagawaIHasegawaTSueokaKKamataniNYoshimuraYOgataT2007Haplotype analysis of the estrogen receptor 1 gene in male genital and reproductive abnormalitiesHumReprod22512791284WennerholmUBBerghCHambergerLNilssonLReismerEWennergrenMWiklandM1996Obstetric and perinatal outcome of pregnancies following intracytoplasmic sperm injectionHum Reprod115111311198671401WilliamsCLStancelGMHardmanJGLimbirdLEGoodman GilmanA1996Estrogens and progestinsGoodman & Gilman’s The pharmacologic basis of therapeuticsNew YorkMcGraw Hill14111440WillinghamEAgrasKVilelaMBaskinLS2006Loratadine exerts estrogen-like effects and disrupts penile development in the mouseJ Urol175272372616407038WogeliusPHorvath-PuhoEPedersenLNorgaardMCzeizelAESorensenHT2006Maternal use of oral contraceptives and risk of hypospadias - a population-based case-control studyEur J Epidemiol211077778117077991YadavCSBajpaiMKumarVKumar DattaSGuptaPAhmedRSDev BanerjeeB2011Polymorphisms in the P450 c17 (17-Hydroxylase/17, 20-Lyase) gene: Association with estradiol and testosterone concentration in hypospadiasUrology78490290721676446YamadaGSuzukiKHaraguchiRMiyagawaSSatohYKamimuraMNakagataNKataokaHKuroiwaAChenY2006Molecular genetic cascades for external genitalia formation: an emerging organogenesis programDev Dyn23571738175216598715YongWYangZPeriyasamySChenHYucelSLiWLinLYWolfIMCohnMJBaskinLSSanchezERShouW2006Essential role for Co-chaperone FKBP52 but not FKBP51 in androgen receptor-mediated signaling and physiologyJ Biol ChemYucelSDesouzaABaskinLS2004aIn utero prednisone exposure affects genital developmentJ Urol1724 Pt 21725173015371800YucelSLiuWCorderoDDonjacourGCBaskinLSBaskinLS2004bAnatomical studies of the fibroblast growth factor-10 mutant, sonic hedge hog mutant and androgen receptor mutant mouse genital tubercleHypospadias and Genital DevelopmentNew YorkKluwer Academic/Plenum Publishers123148

Odds ratios (95 percent confidence intervals) for associations of hypospadias with probable maternal occupational exposure to selected classes of chemicals that include endocrine disruptors, from studies using job exposure matrices.*

StudyBirth years and location of study populationSubject selection (case:control)PesticidesPhthalatesOrgano-chlorinesBiphenolsAlkyl-phenolsHeavy metalsAny Exposure
Vrijheid et al. ’031980-96, England and WalesPassive registry; malformed comparison group; (3,471:35,962)0.8 (0.5–1.4)0.9 (0.7–1.1)1.0 (0.2–4.1)0.7 (0.3, 1.9)0.8 (0.7–1.1)0.7 (0.4–1.2)0.9 (0.7–1.0)
Ormond et al. ’081997-98, Southeast EnglandCase-control study with interview (471:490)n.a.3.1 (1.0–11)n.a.n.a.n.a.n.a.n.a.
Nassar et al. ’091980–2000, Western AustraliaRegistry-based case-control study (1,202:2,583)0.9 (0.5–1.7)1.2 (0.8–1.7)1.0 (0.2–4.9)1.0 (0.4–2.5)1.1 (0.8–1.5)2.6 (1.3–5.2)1.3 (1.0–1.7)
Giordano et al. ’10Recruited 2005-07 (age 0–24 months), ItalyCase-control study with interview (80:80)n.a.3.6 (0.9–14.0)n.a.3.3 (0.3–33.6)1.9 (0.9–4.0)2.0 (0.4–9.0)2.4 (1.1–5.6)#
Varela et al. ’111997–2009, DenmarkCohort study with interview (262:45,341)1.1 (0.9–1.3)2.3 (0.9–3.7)n.a.0.8 (0.2–1.4)2.3 (1.0–3.6)1.2 (0.4–2.0)1.8 (1.0–2.6)

Odds ratios were adjusted for various covariates.

Odds ratio for one versus zero exposure; odds ratio for more than one versus zero exposure was 4.1 (95% CI 1.3, 12.6).

Odds ratios (95 percent confidence intervals) for associations of hypospadias with probable paternal occupational exposure to selected classes of chemicals that include endocrine disruptors, from studies using job exposure matrices.*

StudyBirth years and location of study populationSubject selection (case:control)PesticidesPhthalatesOrgano-chlorinesBiphenolsAlkylphenolsHeavy metalsAny Exposure
Giordano et al. ’10Recruited 2005-07 (age 0–24 months), ItalyCase-control study with interview (80:80)n.a.1.0 (0.5–2.3)0.9 (0.3–2.7)1.7 (0.3–10.7)1.6 (0.7–3.7)1.0 (0.3–3.6)1.4 (0.6–3.2)#
Varela et al. ’111997–2009, DenmarkCohort study with interview (262:45,341)1.6 (0.8–2.4)1.7 (0.9–2.5)1.1 (0.4–1.8)1.8 (0.8–2.8)0.7 (0.3–1.1)0.7 (0.2–1.2)1.3 (0.7–1.9)

Odds ratios were adjusted for various covariates.

Odds ratio for one versus zero exposure; odds ratio for more than one versus zero exposure was 1.5 (95% CI 0.6–3.2)

Association of hypospadias with the SRD5A2 V89L (rs 523349, +336G>C) polymorphism.

StudySubjectsResults overallResults by phenotype
Samtani et al. 201180 cases, 100 controls from house-to-house surveys; IndianCC: 3.6 (1.5, 8.8)CG: 2.1 (1.0, 4.1)GG: ReferenceSimilar for moderate and severe (too few mild to analyze separately)
Sata et al. 201089 cases, 291 controls born in Hokkaido; JapaneseCC: 1.2 (0.6, 2.5)CG: 1.3 (0.7, 2.3)GG: ReferenceMild/moderate: CC: 0.8 (0.3, 1.9) CG: 0.9 (0.5, 1.7)Severe: CC: 3.3 (1.0, 11.5) CG: 3.2 (01.1, 9.5)
Thai et al. 2005158 cases, 96 ‘controls’; Caucasian (Sweden)CC: 3.5 (1.8, 6.8)CG: 4.8 (2.5, 9.2)GG: ReferenceSimilar for mild, moderate and severe
Van der Zanden et al. 2010609 cases, 596 controls from an unrelated case-control study; Caucasian (the Netherlands)CC: 0.9 (0.6, 1.4)CG: 1.0 (0.6, 1.5)GG: ReferenceSimilar by severity (data not shown)
Wang et al. 200490 cases, 276 controls (“unrelated normal males”); ChineseCC: 3.6 (1.4, 9.0)CG: 2.3 (1.1, 4.7)GG: Referencen.a.

n.a. = not available

Association of hypospadias with polymorphisms in ESR1 and ESR2.

StudySubjectsESR1ESR2
Aschim et al. 200551 cases, 186 controls who were military conscripts; Caucasian (Sweden)n.a.rs1256049 (1082G>A, a.k.a. RsaI) AA: n.a. AG: 1.4 (0.4, 5.4) GG: Referencers4986938 (1730G>A, a.k.a. AluI) AA: 1.4 (0.6, 3.5) AG: 1.0 (0.5, 2.0) GG: Reference
Ban et al. 200859 cases, 286 controls born in Hokkaido; Japanesers2234693 (c.454– 397T>C, a.k.a. PvuII) & rs9340799 (c.454- 351A>G, a.k.a. XbaI) – CC/AA genotype vs. others: 3.1 (1.2, 8.2)rs944050 (2681-4A>G) GG: 0.8 (0.3, 1.8) GA: 0.4 (0.2, 0.8) AA: Reference
Beleza-Meireles et al. 200690 cases, 94 controls who were “voluntary blood donors”; primarily Caucasian (Sweden)TA repeat n.s. (p>0.05)Longer CA repeat (p<0.05)rs944050 (2681-4A>G) AG: 6.6 (0.8, 56.3) AA: Reference
Beleza-Meireles et al. 2007354 cases, 380 controls who were “voluntary blood donors”; primarily Caucasian (Sweden)n.a.Studied CA repeat and 6 SNPs (excluding 944050, including rs2987983); only CA repeat was independently associated with increased risk
Van der Zanden et al. 2010620 cases, 596 controls from an unrelated case- control study; Caucasian (the Netherlands)rs6932902 (c.1236- 5602G>A) AA: 1.8 (0.8, 4.2) AG: 1.2 (0.9, 1.6) GG: Referencers2987983 (-13950T>C) GG: 0.8 (0.6, 1.3) GA: 0.8 (0.6, 1.0) AA: Reference
Watanabe et al. 200743 cases, 135 controls, including 82 boys seen for short stature and 53 fertile adult males; JapaneseStudied 8 SNPs; focused on 5 SNPs composing “AGATA” haplotype (includes rs6932902) – associated with increased risk (p<0.05)n.a.s

n.s. = not significant; n.a. = not available