Vous êtes sur la page 1sur 7

Journal of Andrology, Vol. 29, No.

5, September/October 2008
Copyright E American Society of Andrology

Endocrine Disruptors, Genital Review


Development, and Hypospadias
MING-HSIEN WANG AND LAURENCE S. BASKIN
From the UCSF Childrens Hospital, Department of Urology, University of California, San Francisco, California.

ABSTRACT: Hypospadias is one of the most common congenital hypospadias. With recent advancements in molecular biology and
anomalies in the United States, occurring in approximately 1 in 125 microarray technology, it appears that hypospadias is potentially
live male births. Embryological studies have demonstrated that, related to disrupted gene expression. Specifically, some of the
depending on where the urethral development arrests, the meatal environmental chemicals are acting as antiandrogens and interfere
opening can be anywhere along the shaft of the penis or, in more directly with the action of testosterone-related gene expression. In
severe forms, within the scrotum or in the perineum. Currently, the this paper, we briefly review the normal development of male
only available treatment is surgery. If left uncorrected, especially in external genitalia and the prevalence and environmental risk factors
its severe form, there is risk of infertility and psychological effects, related to hypospadias. In addition, we discuss some of the recent
such as avoidance of intimate relationships. The cause of laboratory findings that contribute to our current understanding of this
hypospadias is largely unknown; however, current epidemiology disease.
and laboratory studies have shed new light into the etiology of J Androl 2008;29:499505

T he term endocrine disruptor was initially coined in


1991 by Theo Colborn, one of the worlds leading
authorities on endocrine-disrupting chemicals in the
Fry 1995; Sumpter and Jobling, 1995; de Solla et al,
1998). Within the last decade, several epidemiologic
studies have suggested environmental factors as a
environment and a senior scientist with the World possible cause for the observed increased incidence of
Wildlife FundUS (Colborn et al, 1993). Endocrine abnormalities in male reproductive health. Some exam-
disruptors are exogenous substances that behave simi- ples include the increased incidence of testicular cancer,
larly to biologic hormones. They interfere with the a decline in semen quality, an increase in the frequency
physiologic functions of the endogenous hormones by of undescended testes and hypospadias, and a growing
affecting the release, binding, or metabolism of the demand for assisted reproduction (Chilvers et al, 1984;
endogenous hormones. An example is the nonsteroidal Matlai and Beral, 1985; Campbell et al, 1987; Carlsen et
estrogen diethylstilbestrol (DES). Before its ban in the al, 1992; Adami et al, 1994; Auger et al, 1995; Irvine et
early 1970s, physicians prescribed DES to pregnant al, 1996; Swan et al, 1997; Moller, 1998; Andersen et al,
women hoping to prevent spontaneous abortions and 2000). However, some of the recent studies both in
promote fetal growth. It was later noted that daughters Greenland and Denmark showed a stabilization or even
of DES-treated women have an increased incidence of decline in the incidence of hypospadias (Giwercman et
vaginal and cervical cancers. DES is just one of many al, 2006, Cortes et al, 2008).
well-known examples of endocrine disruptors in hu- In 2001, Skakkebk and others published a paper
mans. suggesting that these observations are in fact related and
Multiple animal studies have linked endocrine dis- indicate a common cause. His conclusions were based
ruptors to adverse biological effects, raising concerns on the number of male reproductive abnormalities that
that low-level exposure might cause similar effects in have risen sharply over the past 50 years, concurrent
humans (Giesy et al, 1994; Guillette et al, 1994, 1995; with the swift growth of the chemical industry and its
associated release of chemicals into the environment. He
Supported by a grant from the National Institute of Health R01
coined the term testicular dysgenesis syndromes
DK058105. (TDS), which encompasses cryptorchidism, in situ germ
Correspondence to: Laurence S. Baskin, MD, Chief, Pediatric cell carcinoma of the testis/overt testicular cancer,
Urology, Professor Urology and Pediatrics, UCSF Childrens Hospi- reduced semen quality, and hypospadias. Additional
tal, University of California, San Francisco, 400 Parnassus Avenue signs include presence of microliths in the testes, Sertoli
A640, San Francisco, CA 94143 (e-mail: lbaskin@urology.ucsf.edu).
Received for publication January 14, 2008; accepted for publication
cellonly seminiferous tubules, or immature tubules with
March 25, 2008. undifferentiated Sertoli cells (Skakkebk et al, 2001,
DOI: 10.2164/jandrol.108.004945 Skakkebk, 2004).

499
500 Journal of Andrology N September October 2008

Figure 2. At 8 weeks of gestation, the external genitalia remain in the


indifferent stage. The urethral groove on the ventral surface of the
phallus is between the paired urethral folds (uf). The penile urethra
forms as a result of fusion of the medial edges of the endodermal
urethral folds. As development progresses, the ectodermal edges of
the urethral groove begin to fuse to form the median raphe. (A) By
1112 weeks, the coronal sulcus separates the glans from the shaft
of the penis. (B) By 16 weeks of gestation, the urethral folds have
completely fused in the midline on the ventrum of the penile shaft. (C,
D) Note the normal ventral penile curvature or chordee (vc) that
occurs during development and resolves by the 20th week. Note the
midline seam (ms).

addition, by the end of the fourth week of gestation,


both the hindgut and future urogenital system have
reached the cloacal membrane on the ventral surface of
the developing embryo. From this indifferent stage until
the end of the eighth week of gestation, the urorectal
septum continues to develop and divides the cloacal
membrane into anterior and posterior segments; with
the anterior aspect destined to be the urogenital
Figure 1. In the male, the wolffian duct develops into the rete testis, membrane and the posterior segment the future rectum.
the efferent ducts, the epididymis, the vas deferens, and the seminal
vesicles. In the female, without the mullerian-inhibiting substance, Up to this point, the male and female genitalia are
the mullerian ducts develop into the fallopian tubes, uterus, cervix, essentially indistinguishable. With the surge in luteiniz-
and upper third of the vagina. ing hormone, coupled with the influence of testosterone
and 5[alpha]-dihydrotestosterone (DHT), masculiniza-
In the following review, we focus on the role of tion of the external genitalia occurs. One of the first
environmental endocrine disruptors and penile malde- signs of masculinization is an increase in the distance
velopment in the setting of hypospadias, with a brief between the anus and the genital structures, followed by
overview of the normal embryology of male external elongation of the phallus, formation of the penile
genitalia. urethra from the urethral groove, and the development
of the prepuce. By 1112 weeks, the coronal sulcus has
Development of the Male External Urogenital System separated the glans from the shaft of the penis. At 1617
Formation of the external male genitalia is a complex weeks of gestation, the urethral folds have completely
process starting with genetic programming (ie, the fused in the midline on the ventrum of the penile shaft
presence of the Y chromosome and its associated sex- with medial fusion of the endodermal urethra and fusion
determining region Y [SRY] and its protein product, of the ectodermal edges (Figure 2; Hinman, 1993;
testis-determining factor [TDF], which are necessary for Moore and Persaud, 1998).
cell differentiation), hormonal signaling, enzyme activ- The glandular urethra, which consists of the squa-
ities, and tissue remodeling. At 3.5 weeks of gestation, mous epithelium, also completes its formation during
the wolffian system appears as 2 longitudinal ducts this period. The mechanism of the glandular urethral
connecting cranially to the mesonephros and caudally formation remains controversial. Two theories have
draining into the urogenital sinus. Meanwhile, the been proposed: endodermal cellular differentiation,
mullerian duct develops as an evagination in the wherein the glandular urethra formed by an extension
coelomic epithelium just lateral to the wolffian duct at of urogenital sinus epithelium undergoes transdifferen-
approximately the sixth week of gestation (Figure 1). In tiation, and primary intrusion of the ectodermal tissue
Wang and Baskin N Endocrine Disruptors 501

Figure 3. Theories of human penile urethral development. The ectodermal ingrowth theory postulates that the glandular urethra is formed by
ingrowth of epidermis. More recent data support the formation of the entire urethra via endodermal differentiation alone.

from the skin of the glans penis (Figure 3). Anatomical urethra), the preputial tissues do not form ventrally
and immunohistochemical studies support the new (Figure 4).
hypothesis of endodermal differentiation, which shows
that the epithelium of the entire urethra is of urogenital Prevalence of Hypospadias
sinus origin. The entire male urethra, including the The cause of hypospadias is largely unknown, although
glandular urethra, is formed by dorsal growth of the epidemiologic studies have identified some risk factors.
urethral plate into the genital tubercle and ventral Rates are highest among whites and lowest among
growth and fusion of the urethral folds. Under proper Hispanics (Centers for Disease Control, 1988). Some
mesenchymal induction, the urothelium has the ability studies have found an association between hypospadias
to differentiate into a stratified squamous phenotype and maternal factors, such as primiparity and advanced
with characteristic keratin staining, thereby explaining age (Angerpointer, 1984). Clinical studies have pointed
the cell type of the glans penis (Yamada et al, 2003).
The future prepuce is formed at the same time as the
urethra and is dependent on normal urethral develop-
ment. At about the eighth week of gestation, low
preputial folds appear on both sides of the penile shaft,
which join dorsally to form a flat ridge at the proximal
edge of the corona. The ridge does not entirely encircle
the glans because it is blocked on the ventrum by the
incomplete developed glandular urethra. The process of
preputial formation continues until it covers all of the
Figure 4. Spectrum of hypospadias. (A) Anterior, where the meatus
glans, forming a midline seam. If the genital folds fail to is on the inferior surface of the glans penis. (B) Coronal, the meatus
fuse (ie, if there is a defect in the formation of the is in the balanopenile furrow. (C) Distal, on the distal third of the shaft.
502 Journal of Andrology N September October 2008

to defects in testosterone metabolism or testosterone the external genitalia and urethra, we must first look at
receptors as a potential cause (Giwercman et al, 1993), testosterone. Under normal conditions, testosterone
suggesting that exogenous hormones can potentially dissociates from its carrier proteins in the plasma and
interfere with endogenous hormonal functions and enters the cells via passive diffusion. Once intracellular,
influence the normal development of male genitalia. testosterone binds to the androgen receptor (AR) and
The most recent hypothesis suggests that widespread use forms a complex that can subsequently bind to the
of synthetic compounds, especially those with estrogenic androgen response element on DNA. With this binding,
or antiandrogenic activities, is a contributing factor multiple downstream androgen-responsive activities are
(Giwercman et al, 1993; Sharpe and Skakkebk, 1993). initiated, one of which is the development of male
In 1999, the International Clearinghouse for Birth external genitalia. Antiandrogens can directly interfere
Defects Monitoring Systems, a nongovernmental orga- with the proper functioning of testosterone in multiple
nization of the World Health Organization, reported an ways. Examples include inducing a conformational
increased rate of hypospadias in 7 European countries, change within AR, increasing AR degradation, or
including Norway, Sweden, England and Wales, Hun- blocking the release of heat shock proteins from AR.
gary, Denmark, Italy, and France in the 1960s, 1970s, A well-known example of an antiandrogen is DDT
and 1980s (Paulozzi, 1999). Independently, the Centers (dichlorodiphenyltrichloroethane). DDT was widely
for Disease Control and Prevention (CDC) reported used to combat mosquitoes spreading malaria, typhus,
their findings of a doubling of hypospadias from 1968 to and other insectborne diseases. In 1962, Rachel Carson
1993 in the United States (Paulozzi et al, 1997). The data published the book Silent Spring, questioning the
were based on collective analysis from 2 independent environmental effects of the indiscriminate use of
surveillance systems. Specifically, data from the Metro- DDT and its effect on ecology and human health
politan Atlanta Congenital Defects Program (MoD/ (Carson, 1962). A study in the male rat fetus found that
MACDP/CBDMP, 2001), a population-based registry in utero exposure to p,p9-DDE, a lipophilic metabolite
that uses active case studies in 22 hospitals and clinics in of DDT, leads to feminization of the genital urinary
the Atlanta, Georgia, area, indicate that the rate of total tracts; reduced anogenital distance, hypospadias, and
hypospadias almost doubled from 1968 to 1993 at an cryptorchidism (Kelce et al, 1995). Perhaps the most
annual rate of 2.9%. Concurrently, the Birth Defects alarming aspect of the study is that the experimental
dose used was within the range of human exposure.
Monitoring Program, a program that gathered dis-
Normal urogenital differentiation also relies on the
charge diagnoses of newborns across the country, also
interaction between testosterone and epidermal growth
reports an increase in hypospadias: from 20.2 per 10 000
factors (EGFs). In vivo experiments show that EGF
live births in 1970 to 39.7 per 10 000 live births in 1993.
alone can induce partial virilization of the external
Overall, these longitudinal studies suggest an almost
genitalia and that sexual differentiation is inhibited
doubling of hypospadias in the United States over a 14-
when EGF is depleted (Gupta et al, 1996). Dioxin
year period.
(TCDD 2,3,7,8-tetrachlorodibenzo-p-dioxin), a compo-
Environmental Factors nent of agent orange, a herbicide and defoliant used by
the US Armed Forces in the Vietnam War, has gained
In the past, environmental effects were generally ruled out popular attention because of its association with various
as causes for hypospadias. However, in light of the cancers. It has been shown in rats to induce c-Src kinase
growing number of endocrine disruptors reported in activity and reduces EGF receptor binding during
human tissue and the probability of shared exposure, testicular development (El-Sabeawy et al, 1998). How-
environmental contaminants are now being evaluated in ever, there are no definitive studies that have demon-
familial cluster studies. Reports of increase rates of strated an association between agent orange and human
hypospadias have paralleled reports of other adverse cancers. Chlorinated pesticides, such as dioxin, as well
effects observed in male reproductive health, include as furans and polychlorinated biphenyls are known
increased rates of testicular cancer, cryptorchidism, and cytochrome P450 (CYP450) agonists that interfere with
decreasing semen and sperm quality (Bergstrom et al, 1995; the normal aromatization of androgens (Haake et al,
Carlsen et al, 1995). Cheng et al (1996) found that 8% of 1987; Paolini et al, 1996; Sanderson et al, 1997).
patients (n 5 252) with undescended testes also had other Several synthetic chemicals have consistently been
urogenital anomalies, including hypospadias. A separate shown to induce hypospadias in laboratory animals.
study reports an increase in testicular cancer risk with Vinclozolin, a commonly used fungicide, induces female-
undescended testicles and hypospadias (Prener et al, 1996). like AGD (anal-genital distance), retained nipples,
To understand the biology of antiandrogens, and reduced sperm count and fertility, and hypospadias in
their roles as endocrine disruptors in the development of 100% of male offspring that were exposed to the chemical
Wang and Baskin N Endocrine Disruptors 503

in utero (Gray et al, 1999a; Kelce et al, 1994). estrogen showed an increase in staining for ATF-3
Procymidone, another antiandrogenic fungicide, induces within 2 hours of treatment (Liu et al, 2006). This was
hypospadias in all male rat offspring when mothers are further substantiated by an increase in protein expres-
exposed during gestation (Ostby et al, 1999). It has been sion, and ATF-3 promoter activity (Liu et al, 2007).
shown that procymidone exerts its effects by inhibiting Similar results were also noted in the mouse genital
DHT-induced transcriptional activities (Gray et al, tubercle, where quantitative reverse transcriptase poly-
1999b). Mice exposed to vinclozolin have less robust merase chain reaction showed that ATF-3 mRNA is up-
and atrophic urethras when compare with controls (Kim regulated in estrogen-exposed specimen compared with
et al, 2004; Buckley et al, 2006). Phthalates, ubiquitous controls. A study of human tissues from 28 children who
chemicals found in plastics, have gained attention underwent repair of hypospadias vs 20 tissue samples
because of their hormonal effects in laboratory animals. from children who underwent elective circumcision,
Male rodents exposed to dibutyl phthalate and diethyl- demonstrated a significant increase in immunohisto-
hexyl phthalate have reduced AGD, retained nipples, chemical staining for ATF-3 in the hypospadias
epididymal agenesis, undescended testes, and hypospadias population (86% vs 13%), (Liu et al, 2005). Studies
(Gray, 1998). have shown that ATF-3 protein suppresses cellular
As mentioned previously, DES provides an excellent growth, we can hypothesize that its up-regulation
model for studying interrupted genital development induces an arrest in urethral development as a potential
because of exogeneous hormones in humans. Longitu- cause of hypospadias. Further studies of ATF-3 and
dinal studies have shown that DES daughters have a other estrogen-related genes would perhaps provide a
2.5-fold increase in breast cancer after age 40, in link between exogenous hormones and the development
addition to an increased risk for vaginal and cervical of hypospadias.
cancers (Herbst et al, 1971). Sons of DES-exposed With the increasing incidence of hypospadias paral-
mothers are at an increased risk for feminization of the leling the rate of increase in environmental pollutants, it
male fetus; a study noted a 20-fold increase in the is imperative that we consider endocrine disruptors as a
development of hypospadias (Brouwers et al, 2006). A potential cause for this anomaly. Additional molecular
separate study confirmed this risk: 10 of 225 DES- studies will help us elucidate the role of these exogenous
exposed male offspring developed penile malformations, hormones and offer insights into other male develop-
including strictures/stenoses and hypospadias, vs 0 in mental disorders.
patients who were unexposed (Henderson et al, 1976).

Molecular Mechanism of Hypospadias and References


Endocrine Disruptors
The exact molecular events that lead to normal urethral Adami H, Bergstrom R, Mohner M. Testicular cancer in nine northern
development and hypospadias remain largely unknown; European countries. Int J Cancer. 1994;59:3338.
Andersen AG, Jensen TK, Carlsen E. High frequency of sub-optimal
however, recent studies have revealed possible involve- semen quality in an unselected population of young men. Hum
ment of sonic hedgehog (SHH), fibroblast growth Reprod. 2000;15:366372.
factors 8 and 10 (FGF8 and FGF10), and homeobox Angerpointer TA. Hypospadiasgenetics, epidemiology and other
A13 and D13 (HOXA13 and HOXD13) genes in early possible aetiological influences. Z Kinderchir. 1984;39:112118, 27.
genital tubercle outgrowth and patterning (Haraguchi et Auger J, Kunstmann JM, Czyglik F, Jouannet P. Decline in semen
quality among fertile men in Paris during the past 20 years. New
al, 2001; Perriton et al, 2002; Morgan et al, 2003). As
Engl J Med. 1995;332:281285.
discussed earlier, antiandrogen might play a role in the Bergstrom R, Adami HO, Mohner M, Zatonski W, Storm H, Ekbom
development of hypospadias. It has been shown that A, Tretli S, Teppo L, Birnbaum LS. Developmental effects of
pregnant female mice exposed to synthetic estrogen dioxin. Environ Health Perspect. 1995;103:8994.
compounds gave birth to offspring with hypospadias Brouwers MM, Feitz WF, Roelofs LA, Kiemeney LA, de Gier RP,
Roeleveld N. Hypospadias: a transgenerational effect of diethyl-
(Kim et al, 2004; Willingham et al, 2006). Microarray
stilbestrol? Hum Reprod. 2006;21(3):666669.
analysis of tissue samples from hypospadias have Buckley J, Willingham E, Agras K, Baskin LS. Embryonic exposure to
identified additional genes that are both estrogen the fungicide vinclozolin causes virilization of females and
responsive and up-regulated (Wang et al, 2007). One alteration of progesterone receptor expression in vivo: an
such gene found in the tissues/foreskin of human experimental study in mice. Environ Health. 2006;21(5):4.
Campbell DM, Webb JA, Hargreave TB. Cryptorchidism in Scotland.
hypospadias, which also demonstrated significant up-
Br Med J. 1987;295:12351236.
regulation in the presence of estrogen, is activating Carlsen E, Giwercman A, Keiding N, Skakkebk NE. Evidence for
transcription factor-3 (ATF-3). A recently published decreasing quality of semen during past 50 years. Br Med J.
study utilizing human foreskin fibroblast treated with 1992;305:609613.
504 Journal of Andrology N September October 2008

Carlsen E, Giwercman A, Keiding N, Skakkebk NE. Declining contaminated and control lakes in Florida. Environ Health
semen quality and increasing incidence of testicular cancer: is there Perspect. 1994;102:680688.
a common cause? Environ Health Perspect. 1995;103(suppl Gupta C, Chandorkar A, Nguyen AP. Activation of androgen
7):137139. receptor in epidermal growth factor modulation of fetal mouse
Carson R. Silent Spring. Boston, MA: Houghton Mifflin; 1962. sexual differentiation. Mol Cell Endocrinol. 1996;123(1):8995.
Centers for Disease Control. Leading major congenital malformations Haake J, Kelley M, Keys B, Safe S. The effects of organochlorine
among minority groups in the United States, 19811986. Morb pesticides as inducers of testosterone and benzo[a]pyrene hydrox-
Mortal Wkly Rep. 1988;37:1724. ylases. Gen Pharmacol. 1987;18(2):165169.
Cheng W, Mya GH, Saing H. Associated anomalies in patients with Haraguchi R, Mo R, Hui C, Motoyama J, Makino S, Shiroishi T,
undescended testes. J Trop Pediatr. 1996;42(4):204206. Gaffield W, Yamada G. Unique functions of sonic hedgehog
Chilvers C, Pike MC, Forman D. Apparent doubling of frequency of signaling during external genitalia development. Development.
undescended testis in England and Wales in 196281. Lancet, 2001;128:42414250.
1984;330332. Henderson BE, Benton B, Cosgrove M, Baptista J, Aldrich J,
Colborn T, Vom Saal FS, Soto AM. Developmental effects of Townsend D, Hart W, Mack TM. Urogenital tract abnormalities
endocrine-disrupting chemicals in wildlife and humans. Environ in sons of women treated with diethylstilbestrol. Pediatrics. 1976;
Health Perspect. 1993;101:378384. 58(4):505507.
Cortes D, Kjellberg EM, Breddam M, Thorup J. The true incidence of Herbst AL, Ulfelder H, Poskanzer DC. Adenocarcinoma of the
cryptorchidism in Denmark. J Urol. 2008;179:314318. vagina. Association of maternal stilbestrol therapy with tumor
de Solla SR, Bishop CA, Van der Kraak G, Brooks RJ. Impact of appearance in young women. N Engl J Med. 1971;284(15):878881.
organochlorine contamination on levels of sex hormones and Hinman FJ. Penis and Male Urethra. Atlas of Urosurgical Anatomy.
external morphology of common snapping turtles (Chelydra Philadelphia, Pa: WB Saunders, 1993:417470.
serpentina serpentina) in Ontario, Canada. Environ Health Perspect. Irvine S, Cawood E, Richardson D. Evidence of deteriorating semen
1998;106:253260. quality in the United Kingdom: birth cohort study in 577 men in
El-Sabeawy F, Wang S, Overstreet J, Miller M, Lasley B, Enan E. Scotland over 11 years. Br Med J. 1996;312:467471.
Treatment of rats during pubertal development with 2,3,7,8- Kelce WR, Monosson E, Gamcsik MP, Laws SC, Gray LE Jr.
tetrachlorodibenzo-p-dioxin alters both signaling kinase activities Environmental hormone disruptors: evidence that vinclozolin
and epidermal growth factor receptor binding in the testis and the developmental toxicity is mediated by antiandrogenic metabolites.
motility and acrosomal reaction of sperm. Toxicol Appl Pharmacol. Toxicol Appl Pharmacol. 1994;126(2):276285.
1998;150(2):427442. Kelce WR, Stone CR, Laws SC, Gray LE, Kemppainen JA, Wilson
Fry DM. Reproductive effects in birds exposed to pesticides and EM. Persistent DDT metabolite p,p9-DDE is a potent androgen
industrial chemicals. Environ Health Perspect. 1995;103(suppl receptor antagonist. Nature. 1995;35(6532):581585.
7):165171. Kim KS, Torres CR Jr, Yucel S, Raimondo K, Cunha GR, Baskin LS.
Giesy JP, Ludwig JP, Tillitt DE. Deformities of birds in the Great Induction of hypospadias in a murine model by maternal exposure
Lakes region: assigning causality. Environ Sci Technol. 1994;28: to synthetic estrogens. Environ Res. 2004;94(3):267275.
128A135A. Liu B, Agras K, Willingham E, Vilela ML, Baskin LS. Activating
Giwercman A, Carlsen E, Keiding N, Skakkebk NE. Evidence for transcription factor 3 is estrogen-responsive in utero and upregu-
increasing incidence of abnormalities of the human testis: a review. lated during sexual differentiation. Horm Res. 2006;65:217
Environ Health Perspect. 1993;101(suppl 2):6571. 222.
Giwercman Y, Kleist Ke, Giwercman A, Giwercman C, Toft G, Bonde Liu B, Lin G, Willingham E, Ning H, Lin CS, Lue TF, Baskin LS.
JP, Pedersen HS. Remarkably low incidence of hypospadias in Estradiol upregulates ATF3: a candidate gene in the etiology of
Greenland despite high exposure to endocrine disruptors; possible hypospadias. Pediatr Dev Pathol. 2007;10(6):446454.
protective effect of androgen receptor genotype. Pharmaco Liu B, Wang Z, Lin G, Agras K, Ebbers M, Willingham E, Baskin LS.
Genomics. 2006;16:375377. Activating transcription factor 3 is up-regulated in patients with
Gray LE Jr. Xenoendocrine disrupters: laboratory studies on male hypospadias. Pediatr Res. 2005;58:12801283.
reproductive effects. Toxicol Lett. 1998;102103:331335. March of Dimes, Metropolitan Atlanta Congenital Defects Program,
Gray LE Jr, Ostby J, Monosson E, Kelce WR. Environmental and California Birth Defects Monitoring Program, MoD/MACDP/
antiandrogens: low doses of the fungicide vinclozolin alter sexual CBDMP. Leading Categories of Birth Defects. March 2001.
differentiation of the male rat. Toxicol Ind Health. 1999a;15(1 Matlai P, Beral V. Trends in congenital malformations of external
2):4864. genitalia. Lancet. 1985;108.
Gray LE Jr, Wolf C, Lambright C, Mann P, Price M, Cooper RL, Mller H. Trends in sex-ratio, testicular cancer and male reproductive
Ostby J. Administration of potentially antiandrogenic pesticides hazards: are they connected? APMIS. 1998;106:232239.
(procymidone, linuron, iprodione, chlozolinate, p,p9-DDE, and Moore KL, Persaud TV. The Developing Human: Clinically Oriented
ketoconazole) and toxic substances (dibutyl and diethylhexyl Embryology. 6th ed. Philadelphia, PA: WB Saunders; 1998.
phthalate, PCB 169, and ethane dimethane sulphonate) during Morgan EA, Nguyen SB, Scott V, Stadler HS. Loss of Bmp7 and Fgf8
sexual differentiation produces diverse profiles of reproductive signaling in Hoxa13-mutant mice causes hypospadias. Develop-
malformations in the male rat. Toxicol Ind Health. 1999b;15(1 ment. 2003;30:30953109.
2):94118. Ostby J, Kelce WR, Lambright C, Wolf CJ, Mann P, Gray LE Jr. The
Guillette LJ Jr, Crain DA, Rooney AA, Pickford DB. Organization fungicide procymidone alters sexual differentiation in the male rat
versus activation: the role of endocrine-disrupting contaminants by acting as an androgen-receptor antagonist in vivo and in vitro.
(EDCs) during embryonic development in wildlife. Environ Health Toxicol Ind Health. 1999;15(12):8093.
Perspect. 1995;103(suppl 7):157164. Paolini M, Pozzetti L, Mesirca R, Sapone A, Cantelli-Forti G.
Guillette LJ Jr, Gross TS, Masson GR, Matter JM, Percival HF, Testosterone hydroxylase in evaluating induction and suppression
Woodward AR. Developmental abnormalities of the gonad and of murine CYP isoenzymes by fenarimol. Arch Toxicol.
abnormal sex hormone concentrations in juvenile alligators from 1996;70(7):451456.
Wang and Baskin N Endocrine Disruptors 505

Paulozzi LJ. International trends in rates of hypospadias and Skakkebk NE, Rajpert-De Meyts E, Main KM. Testicular dysgenesis
cryptorchidism. Environ Health Perspect. 1999;107:297302. syndrome: an increasingly common developmental disorder with
Paulozzi LJ, Erickson JD, Jackson RJ. Hypospadias trends in two environmental aspects. Hum Reprod. 2001;16:972978.
U.S. surveillance systems. Pediatrics. 1997;100(5):831834. Sumpter JP, Jobling S. Vitellogenesis as a biomarker for estrogenic
Perriton CL, Powles N, Chiang C, Maconochie MK, Cohn MJ. Sonic contamination of the aquatic environment. Environ Health
hedgehog signaling from the urethral epithelium controls external Perspect. 1995;103(suppl 7):173178.
genital development. Dev Biol. 2002;247:2646. Swan SH, Elkin EP Fenster L. Have sperm densities declined? A re-
Prener A, Engholm G, Jensen OM. Genital anomalies and risk for analysis of global trend data. Environ Health Perspect.
testicular cancer in Danish men. Epidemiology. 1996;7(1):1419. 1997;105:12281232.
Sanderson JT, Janz DM, Bellward GD, Giesy JP. Effects of embryonic Wang Z, Chun B, Lina GT, Lina CS, Lue TF, Willingham E, Baskin
and adult exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin on hepatic LS. Upregulation of estrogen-responsive genes in hypospadias, a
microsomal testosterone hydroxylase activities in great blue herons urethral abnormality. J Urol. 2007;177:19391946.
(Ardea herodias). Environ Toxicol Chem. 1997;16(6):13041310. Willingham E, Agras K, Vilela M, Baskin LS. Loratadine exerts
Sharpe RM, Skakkebk NE. Are oestrogens involved in falling sperm estrogen-like effects and disrupts penile development in the mouse.
counts and disorders of the male reproductive tract? Lancet. J Urol. 2006;175:723726.
1993;341:13921395. Yamada G, Baskin LS, Satoh Y, Cunha G. Cellular and molecular
Skakkebk NE. Testicular dysgenesis syndrome: new epidemiological mechanisms of development of the external genitalia. Differentia-
evidence. Int J Andrology. 2004;27:189191. tion. 2003;71(8):445460.

Vous aimerez peut-être aussi