You are on page 1of 7

Critical Reviews in Toxicology, 35:713719, 2005

Copyright c Taylor and Francis Inc.


ISSN: 1040-8444 print / 1547-6898 online
DOI: 10.1080/10408440591007395

Mode of Action: Impaired Fetal Leydig Cell


FunctionEffects on Male Reproductive Development
Produced by Certain Phthalate Esters
Paul M. D. Foster
National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
Critical Reviews in Toxicology Downloaded from informahealthcare.com by University of Groningen on 04/29/14

Certain phthalate esters (di-2-ethylhexyl; di-n-butyl and butyl benzyl) have profound effects on
the developing male reproductive system when administered orally to pregnant experimental
animals during a critical window of development. These esters produce a syndrome of adverse
effects that are characteristic of a disturbance in androgen-mediated development and include
a variety of reproductive tract malformations and effects on developmental phenotypic mark-
ers. A testicular dysgenesis syndrome has been proposed to explain the secular increases in a
number of human male reproductive deficits, including decreased semen parameters, increased
incidence of cryptorchidism and hypospadias (two of the most common human birth defects),
and increased incidence of testicular (germ cell derived) cancer. The rodent phthalate data
lend support to the hypothesis. This example illustrates a number of points in the use of the
Human Relevance Framework. First, chemical agents may have more than one mode of action
(MOA): for example, phthalate-induced peroxisome proliferation leading to hepatocarcinogen-
For personal use only.

sis, compared with the induction of developmental effects via effects on androgen signaling.
Second, the case demonstrates the life-stage sensitivity of the response to these compounds.
Third, because humans may be exposed to multiple phthalate esters producing adverse effects
on reproductive development, these compounds may be useful in testing the utility of the Human
Relevance Framework (HRF) approach for evaluating cumulative and aggregate risk.

Keywords Di-n-Butyl Phthalate, Endocrine Disruptors, Human Relevance, Mechanisms

Certain phthalate esters (di-2-ethylhexyl, DEHP; di-n-butyl, vas deferens, seminal vesicles, prostate, external genitalia (hy-
DBP; and butyl benzyl, BBP) have profound effects on the de- pospadias), cryptorchidism, and testicular injury, together with
veloping male reproductive system when administered orally to permanent changes (feminization) in the retention of nipples/
experimental animals during a critical window of development areolae (sexually dimorphic structures in rodents) and demas-
(gestation days 1221 for the rat) (Mylchreest et al., 1998, 1999a, culinization of the growth of the perineum resulting in a reduced
1999b; Gray et al., 2000, 2003). These results are of interest to anogenital distance (AGD). These effects constitute a continuum
risk assessors because of the potential for human exposure from of response, with the most severe manifestations and highest in-
a wide variety of sources. Although several esters have effects, cidence of reproductive-tract malformations observed at higher
studies with DBP and, to a more limited extent, DEHP provide dose levels (500 mg DBP/kg/day), whereas changes in AGD
the basis for this article. and areolae retention are frequently seen at lower dose levels
These esters produce a syndrome of adverse effects in male (100 mg DBP/kg/day). These may be permanent phenotypic
rat offspring after in utero exposure that is characteristic of changes in affected individual animals. The general potency of
a disturbance in androgen-mediated development. The syn- response on reproductive development is DEHP > DBP > BBP.
drome is characterized by malformations of the epididymis, Interestingly, most of these adverse responses have not been ob-
served in classical (exposure on gestation days 615) teratology
studies in rats. In the case of DBP, in utero exposure has also re-
The views presented in this article do not necessarily reflect those sulted in testicular Leydig cell adenoma in 90-day-old offspring
of the National Institute of Environmental Health Sciences. (Mylchreest et al., 1999a). This tumor type is usually found at
Address correspondence to Dr. Paul M. D. Foster, National Institute around 35% in controls of the rat strain employed in the study
of Environmental Health Sciences, P.O. Box 12233, Mail Code E-
106, Research Triangle Park, NC 27709 USA. E-mail: foster2@niehs. just described and is generally considered to be a tumor of old
nih.gov age (most frequently noted at the end of a 2-year bioassay).

713
714 P. M. D. FOSTER

A testicular dysgenesis syndrome (i.e., a failure of normal in Wolffian duct system to develop normally into the vas deferens,
utero development of the testis) has been proposed to explain epididymis, and seminal vesicles (Barlow and Foster, 2003).
the secular increases in a number of human male reproductive Lower testosterone levels also impact the dihydrotestosterone
deficits, including decreased semen parameters, increased inci- (DHT)-induced development of the prostate and external geni-
dence of cryptorchidism and hypospadias (two of the most com- talia (testosterone is converted to DHT by 5-reductase). DHT
mon human birth defects), and increased incidence of testicular is also responsible for the normal apoptosis of nipple anlagen in
(germ-cell-derived) cancer (Sharpe, 2001, 2003; Skakkebaek, males, resulting in the lack of nipple development, and also
2002). Thus far, no cause-and-effect relationship has been es- for the growth of the perineum to produce the normal male
tablished between any environmental agent and these human anogenital distance (approximately twice that of the female).
deficits. However, the rodent data lend support to the hypothesis Testicular descent into the scrotum also requires normal andro-
(Sharpe, 2001). gen levels and a failure of descent results in cryptorchidism.
This example illustrates a number of interesting points in However, the initial stages of testicular descent involve the ac-
Critical Reviews in Toxicology Downloaded from informahealthcare.com by University of Groningen on 04/29/14

the use of the human relevance framework. First, chemical tion of another Leydig cell product, insl3 (insulin-like factor 3),
agents may have more than one MOA: in this case, phthalate- the message of which is also downregulated by DEHP, DBP,
induced peroxisome proliferation leading to hepatocarcinogen- and BBP (Wilson et al., 2004). Female rat fetuses are predom-
sis, as well as induction of developmental effects. Second, inantly unaffected after treatment with these specific phthalate
the case demonstrates the life-stage sensitivity of the response esters.
to these compounds. Specifically, phthalate esters have been
known for decades to produce testicular effects in rodents Key Events
(Foster et al., 1980) with important stage-related differences: Table 1 describes the key events in the MOA of phthalate
Sertoli cells as the probable testicular target in pubertal and adult esters (based primarily on data from DBP and DEHP) in animals.
animals (Foster et al., 1982; Creasy et al., 1987), but a different A critical first step in the toxicity of phthalate diesters is the
MOA and different initial testicular target (fetal Leydig cells) in conversion, by hydrolysis, to the monoester (Lake et al., 1977).
fetal animals. Third, because humans may be exposed to mul- This can occur in the gut, in the liver, or in the blood. There is
For personal use only.

tiple phthalate esters producing adverse effects on reproductive some evidence that monoesters are preferentially absorbed from
development in test animals, these compounds may be useful in the gut. Once converted to the monoester, the entity believed
testing the utility of the Human Relevance Framework (HRF) responsible for toxicity, it then readily crosses the placenta to
approach for evaluating cumulative and aggregate risk. transfer to the fetus (Singh et al., 1975; Saillenfait et al., 1998).
One of the earliest phthalate-related fetal effects observed
I. IS THE WEIGHT OF EVIDENCE SUFFICIENT in rats was disturbance of normal fetal testicular Leydig cell
TO ESTABLISH THE MOA IN ANIMALS? function and/or development (Parks et al., 2000; Shultz et al.,
A significant decrease in the testicular production of testos- 2001; Mylchreest et al., 2002; Barlow et al., 2003). This can
terone by rat fetal Leydig cells (Parks et al., 2000; Shultz result in fetal Leydig cell hyperplasia or large aggregates
et al., 2001; Mylchreest et al., 2002) at time points critical in of fetal Leydig cells (at gestation day 21) in the developing
male reproductive system development results in failure of the testis. Downregulation of genes critical for normal steroid

TABLE 1
Key events in the animal MOA for phthalate

Key event Evidence in animals References

1. Hydrolysis to produce monoester YES. Hydrolysis measured in a number of Lake et al., 1977; Singh et al., 1975;
and transport to target animal species; passage of monoester to the Saillenfait et al., 1998
fetus measured in rats
2. Impaired Leydig cell function YES. Levels of testosterone measured in fetal Parks et al., 2000; Mylchreest et al.,
a. Decrease in fetal testicular testes; increased levels of precursors 2002; Shultz et al., 2001
androgen production indicate functional change at CYP 17
b. Decreased androgen mediated YES. Measured in fetal rat testes. insl3 Barlow et al., 2003; Adham et al.,
gene expression and insl3 knock-out mouse results in cryptorchidism 2000
expression in fetal testes
3. Androgen insufficiency syndrome/ YES. Noted in rats (and rabbits). Effects noted Gray et al., 2000; Barlow and Foster,
testicular dysgenesis on Wolffian duct development, nipples, 2003; Higuchi et al., 2003
prostate, hypospadias, cryptorchidism
IMPAIRED FETAL LEYDIG-CELL FUNCTION 715

production and transport of critical intermediates is noted prior creases in testosterone levels and significant changes in gene ex-
to observation of increased cell aggregates (Shultz et al., 2001; pression associated with Leydig cell differentiation in fetal testes
Barlow et al., 2003). at dose levels below the 50-mg/kg/day no-observed-adverse-
Recent data indicate a decrease in the expression of the effect level (NOAEL) previously reported for DBP.
Leydig cell product insl3 in rat fetal testes after DEHP (750
mg/kg/day), DBP (1 g/kg/day), or BBP (1 g/kg/day) exposure Other Information
in utero on GD 1418 (Wilson et al., 2004). This may be related Studies on the reproductive effects of specific phthalate es-
to an increased incidence of cryptorchidism after fetal exposure ters have been conducted predominantly in rats, but some sim-
to phthalates. Knockouts of this gene in mice show complete ilar events have also been noted in the rabbit (Higuchi et al.,
cryptorchidism (Nef and Parada, 1999; Adham et al., 2000; Nef 2003). No specific studies have been conducted evaluating re-
et al., 2000). The insl3 gene is associated with Leydig cell dif- productive development in mice exposed to phthalates during
ferentiation and overall levels of testosterone, with high levels gestation. However, several phthalate esters produce reproduc-
Critical Reviews in Toxicology Downloaded from informahealthcare.com by University of Groningen on 04/29/14

in fetal rat testes that decrease after birth, rise again at puberty, tive toxicity in adult male and female mice and are teratogenic
but decrease again in old animals (Paust et al., 2002). These au- to fetuses exposed in classical teratology studies when treatment
thors also review the relationship of the SF-1 promotor found is from implantation to closure of the hard palate, with fetuses
on the insl3 gene with those found on many of the genes con- examined just prior to term (see Center for the Evaluation of
trolling the steroidogenic machinery in Leydig cells. Although Risk to Human Reproduction [CERHR] reviews). Phthalate es-
human polymorphisms for insl3 have not been reported, they ter monoesters (and not the parent diesters) can impair Leydig
have been noted for the insl3 receptor (LGR8), which has been cell function in adult rat cells in vitro (Jones et al., 1993), but
shown recently to be related to cryptorchidism in humans (Ivell at dose levels significantly higher (1 mM) than those affecting
and Hartung, 2003). Sertoli cell function (10100 nM) in culture (Lloyd and Foster,
In addition to morphological changes in Leydig cells in 1988). Moreover, in vivo effects on the adult rat testis occur at
the interstitium of the fetal rat testis, the seminiferous cords significantly higher dose levels than in fetal rats (100 mg/kg/day,
also show a marked disturbance of normal gonocyte prolifera- versus 2 g/kg/day for DBP).
For personal use only.

tion, resulting in multinucleated cells (Mylchreest et al., 2002; In contrast to its role in the hepatic effects of phthalate es-
Barlow and Foster, 2003) and subsequent changes in the de- ters, PPAR is unlikely to be involved in these testicular and
veloping germ-cell population. In the human fetus, impaired developmental toxicities. This conclusion is based in part on
gonocyte development is an early change in the induction of the presence of testicular and developmental toxicity in PPAR
germ-cell-derived testicular cancer (Rajpert-De Meyts et al., knockout mice (Peters et al., 1997; Ward et al., 1998), and in part
1998). A marked decrease in testosterone production by fetal on data showing that the guinea pig, a nonresponder to PPAR
Leydig cells is critical to the subsequent reproductive tract mal- ligands, is as sensitive to the testicular effects of phthalates as
formations in rats exposed to specific phthalate esters (Parks are rats (Gray et al., 1982). While all the manifestations of spe-
et al., 2000; Mylchreest et al., 2002). This reduction in an- cific phthalate esters on reproductive development are similar
drogen leads to abnormal reproductive tract development. For to those seen with classical androgen receptor (AR) antagonists
example, lower AR expression in epithelial cells of fetal epi- such as flutamide, phthalate diesters and monoesters have not
didymis is followed by Wolffian duct failure to coil and de- been shown to interact with either rat or human AR using binding
velop normally into the epididymis (Mylchreest et al., 2002; or transcriptional activation assays (Foster et al., 2001).
Barlow and Foster, 2003). These androgen-dependent adverse
outcomes affecting the Wolffian duct and prostatetogether II. ARE KEY EVENTS IN THE ANIMAL MOA PLAUSIBLE
with hypospadias, cryptorchidism, retention of nipples, and a IN HUMANS?
permanently reduced anogenital distancedescribe a syndrome The action of certain phthalate esters on male reproductive
of effects characteristic of specific phthalate esters. The interre- development has been evaluated primarily in rats, with more lim-
lationship of key factors in the animal MOA is displayed dia- ited evidence obtained in rabbits (DEHP; Higuchi et al., 2003)
grammatically in Figure 1. This syndrome has parallels with the and one report on DPB effects on gonadal development in frogs
reported human testicular dysgenesis syndrome (Sharpe, 2001; (Ohtani et al., 2000). Although there are no human data relating
Fisher et al., 2003) and also shows similarities to other known exposure to phthalate esters with adverse outcomes in terms of
human genetic syndromes involving impaired androgen respon- reproductive development, the critical metabolites for reproduc-
siveness in sexual differentiation of the reproductive tract (for tive and developmental toxicity, the phthalate monoesters, are
review, see Hughes, 2001). produced in humans (Anderson et al., 2001; Barr et al., 2003)
Almost all the mechanistic studies on gene expression and and are commonly found in urine samples from the general
MOA with phthalates noted above have utilized dose lev- population (Blount et al., 2000; Barr et al., 2003) and in children
els that ensure significant prevalence of fetal reproductive (Brock et al., 2002).
abnormalitiesfor example, 500 mg/kg/day for DBP; however, The critical enzymes involved in steroidogenesis are identi-
a recent paper (Lehmann et al., 2004) reports significant de- cal in rats and humans, and all mammals are believed to have
716 P. M. D. FOSTER
Critical Reviews in Toxicology Downloaded from informahealthcare.com by University of Groningen on 04/29/14
For personal use only.

FIG. 1. Diagrammatic representation of the interaction between the fetal seminiferous cords and Leydig cells and critical gene
involvement following in utero phthalate exposure in rats. Deficits in androgen production result in a syndrome of malformations
(shown in yellow) that includes malformations of the epididymis, seminal vesicles, and prostate together with an increased incidence
of cryptorchidism and hypospadias. Male offspring also show a feminized phenotype with a permanently reduced anogenital distance
(AGD) and retention of nipples. Genes significantly down regulated by di-n-butyl phthalate include stem-cell factor and its receptor
c-kit, the high-density lipoprotein (HDL) receptor (SRB-1), steroid acute regulatory protein (StAR), and the genes coding for the
enzymes involved in the conversion of cholesterol to pregnenlone (side-chain cleavage enzyme), pregnenolone to progesterone (3
hydroxysteroid dehydrogenase), and progesterone to androstenedione (CYP 17), resulting in a lowered production of testosterone
necessary for normal reproductive development (Barlow et al., 2003). Others have shown a decrease in insl3 gene expression
following in utero exposure to DEHP (Wilson et al., 2004).

parallel activation mechanisms for androgen dependent pro- this has been seen in fetal testes from rats treated with DBP
cesses. Based on our current knowledge of comparative physi- (Barlow and Foster, 2003) and DEHP (Parks et al., 2000). Inter-
ology and endocrinology, it is thus possible for fetuses that have estingly, Leydig cell adenomas (using the rodent pathological
been sufficiently exposed to specific phthalate monoesters to definition) appear to be fairly common in humans, but they are
experience antiandrogenic effects. Table 2 illustrates the plau- termed micronodules, rather than tumors, according to a recent
sibility of the biological process occurring in humans, although publication (Holm et al., 2003). Adverse reproductive conse-
there is no direct evidence that this MOA occurs. quences have been reported in some studies that purport to re-
The testicular tumors observed in rats are Leydig cell ade- late specific phthalate exposure with adverse human outcomes
nomas that have a developmental origin (Mylchreest et al., including sperm measures (Duty et al., 2003) and premature
1999a; Barlow et al., 2004), not the germ-cell tumors most fre- breast development (Colon et al., 2000), but these limited case
quently noted in humans. However, abnormal fetal gonocyte reports should be treated with caution. Several NTP Center for
development is a precursor event to germ-cell cancer in humans the Evaluation of Risk to Human Reproduction (CERHR) mono-
(Skakkebaek et al., 1987; Rajpert-De Meyts et al., 1998), and graphs on specific phthalate esters, including DEHP (Kavlock
IMPAIRED FETAL LEYDIG-CELL FUNCTION 717

TABLE 2
Concordance analysis of key events in phthalate MOA

Key event Evidence in animals Evidence in humans References

1. Hydrolysis to produce YES. Hydrolysis measured in YES. Phthalate monoesters Schmid and Schlatter, 1985;
monoester and transport to a number of animal species; measured in human urine Anderson et al., 2001
target passage of monoester to the
fetus measured in rats.
2. Impaired Leydig cell YES. Levels of testosterone No. data. However, steroid Hughes, 2001
Function measured in fetal testes; synthetic enzymes and
a. Decrease in fetal increased levels of pathways similar/identical
testicular androgen precursors indicate in humans; androgen
Critical Reviews in Toxicology Downloaded from informahealthcare.com by University of Groningen on 04/29/14

production functional change at CYP critical for normal human


17 male development, with
syndromes reported for
genetic defects
b. Decreased androgen YES. Measured in fetal rat No. data. Insl3 exists in Ivell and Hartung, 2003
mediated gene testes; insl3 knock-out humans. Polymorphisms of
expression and insl3 mouse results in the insl3 receptor (LGR8)
expression in fetal testes cryptorchidism have been related to
cryptorchidism.
3. Androgen insufficiency YES. Noted in rats (and No. data. Human syndromes Barthold et al., 2000; Nitsche
syndrome/ testicular rabbits); effects noted on exist related to Low DHT and Hiort, 2000;
dysgenesis Wolffian duct development, levels, or impaired Imperato-McGinley, 2002;
For personal use only.

nipples, prostate, androgen receptor function; Yong et al., 2003; Toppari


hypospadias, cryptorchidism and et al., 2001
cryptorchidism hypospadias are the most
common human birth
defects

et al., 2002a) and DBP (Kavlock et al., 2002b), address some of sponse is linear for rodents and humans. Actualization of the
these issues in more detail. MOA in humans depends, then, on the likelihood of human ex-
posure levels becoming sufficient to induce these key events in
the MOA.
III. TAKING INTO ACCOUNT KINETIC AND DYNAMIC
FACTORS, IS THE ANIMAL MOA PLAUSIBLE
IN HUMANS? IV. STATEMENT OF CONFIDENCE AND IMPLICATIONS
Toxicodynamic factors critical to adverse effects on repro- Confidence is high that the evidence presented supports the
ductive outcomes from phthalate exposure during development described MOA of specific phthalate esters in rats. The com-
are similar in rats and humans. In addition, humans can indeed ponent features of the MOA are also plausible in humans.
produce the metabolites responsible for reproductive and devel- Elements of the phthalate syndrome on reproductive develop-
opmental toxicity in animals. For these reasons, key events in the ment can be noted in animals (for DEHP) at dose levels below
animal MOA are plausible in humans if the exposure level is high 30 mg/kg/day, whereas real-world human exposure in the gen-
enough. For DEHP, humans produce more of the inactive mo- eral population to individual phthalates has been reported in the
noester glucuronide in urine than rodents do for a given dose, 50100 g/kg/day range. Whether such exposure could trigger
but the placenta and fetus have active glucuronidase enzymes the key events just noted is unknown. Clearly, better information
that could release active monoester at critical targets. More- is required in humans relating the known exposure to adverse
over, humans and primates appear to have saturated metabolism health outcomes, particularly in specific groups such as pregnant
in terms of producing free phthalate monoester, with nonlinear women and infant boys.
production of the monoester from diesters administered at dose
levels above approximately 200250 mg/kg/day (for DEHP). ACKNOWLEDGMENTS
However, adverse effects on reproductive development in rats The ILSI Risk Science Institute and the Human Relevance
have been reported below this saturation level, where the re- Phase II Workgroup greatly appreciate the work of the June 2004
718 P. M. D. FOSTER

Peer Review Panel and Dr. John Moores skilled facilitation as Foster, P.M.D., Mylchreest, E., Gaido, K.W., and Sar, M. (2001). Effects
chairman. This ILSI Risk Science Institute project was supported of phthalate esters on the developing reproductive tract of male rats.
by funding from the Office of Pesticide Programs and the Office Human Reprod. Update 7:231235.
of Pollution Prevention and Toxics of the U.S. Environmental Gray, L., Barlow, N., Furr, J., Brock, J., Silva, M., Barr, D., and Ostby,
Protection Agency and by the Existing Substances Division of J. (2003). Transgenerational effects of di(2-ethylhexyl) phthalate in
the male rat. Toxicologist 72:283.
Health Canada.
Gray, L.E., Jr., Ostby, J., Furr, J., Price, M., Veeramachaneni, D.N., and
In addition to the authors of the overview for this special Parks, L. (2000). Perinatal exposure to the phthalates DEHP, BBP,
issue, members of the Phase II Workgroup included Dr. Elaine and DINP, but not DEP, DMP, or DOTP, alters sexual differentiation
Faustman, Dr. Robert Snyder, Dr. Vicki Dellarco, and Dr. Karl of the male rat. Toxicol. Sci. 58:350365.
Baetcke. Dr. James Klaunig chaired the workgroup. Gray, T.J.B., Rowland, I.R., Foster, P.M.D., and Gangolli, S.D. (1982).
Species differences in the testicular toxicity of phthalate esters. Tox-
REFERENCES icol. Lett. 11:141147.
Critical Reviews in Toxicology Downloaded from informahealthcare.com by University of Groningen on 04/29/14

Adham, I.M., Emmen, J.M., and Engel, W. (2000). The role of the Higuchi, T.T., Palmer, J.S., Gray, L.E., Jr., and Veeramachaneni, D.N.
testicular factor INSL3 in establishing the gonadal position. Mol. (2003). Effects of dibutyl phthalate in male rabbits following in
Cell Endocrinol. 160:1116. utero, adolescent, or postpubertal exposure. Toxicol. Sci. 72:301
Anderson, W.A., Castle, L., Scotter, M.J., Massey, R.C., and Springall, 313.
C. (2001). A biomarker approach to measuring human dietary ex- Hughes, I.A. (2001). Minireview: Sex differentiation. Endocrinology
posure to certain phthalate diesters. Food Addit. Contam. 18:1068 142:32813287.
1074. Imperato-McGinley, J. (2002). 5-Alpha-reductase-2 deficiency and
Barlow, N.J., and Foster, P.M.D. (2003). Pathogenesis of male repro- complete androgen insensitivity: Lessons from nature. Adv. Exp.
ductive tract lesions from gestation through adulthood following in Med. Biol 511:121131; discussion 131124.
utero exposure to di(n-butyl) phthalate. Toxicol. Pathol. 31:397 Ivell, R., and Hartung, S. (2003). The molecular basis of cryp-
410. torchidism. Mol. Hum. Reprod. 9:175181.
Barlow, N.J., Phillips, S.L., Wallace, D.G., Sar, M., Gaido, K.W., and Jones, H.B., Garside, D.A., Liu, R., and Roberts, J.C. (1993). The in-
Foster, P.M. (2003). Quantitative changes in gene expression in fetal fluence of phthalate esters on Leydig cell structure and function in
rat testes following exposure to di(n-butyl) phthalate. Toxicol. Sci.
For personal use only.

vitro and in vivo. Exp. Mol. Pathol. 58:179193.


73:431441. Kavlock, R., Boekelheide, K., Chapin, R., Cunningham, M., Faustman,
Barr, D.B., Silva, M.J., Kato, K., Reidy, J.A., Malek, N.A., Hurtz, D., E., Foster, P., Golub, M., Henderson, R., Hinberg, I., Little, R., Seed,
Sadowski, M., Needham, L.L., and Calafat, A.M. (2003). Assessing J., Shea, K., Tabacova, S., Tyl, R., Williams, P., and Zacharewski,
human exposure to phthalates using monoesters and their oxidized T. (2002a). NTP Center for the Evaluation of Risks to Human Re-
metabolites as biomarkers. Environ. Health Perspect. 111:1148 production: Phthalates expert panel report on the reproductive and
1151. developmental toxicity of di(2-ethylhexyl) phthalate. Reprod. Toxi-
Barthold, J.S., Kumasi-Rivers, K., Upadhyay, J., Shekarriz, B., and col. 16:529653.
Imperato-McGinley, J. (2000). Testicular position in the androgen Kavlock, R., Boekelheide, K., Chapin, R., Cunningham, M., Faustman,
insensitivity syndrome: Implications for the role of androgens in E., Foster, P., Golub, M., Henderson, R., Hinberg, I., Little, R., Seed,
testicular descent. J. Urol. 164:497501. J., Shea, K., Tabacova, S., Tyl, R., Williams, P., and Zacharewski, T.
Blount, B.C., Silva, M.J., Caudill, S.P., Needham, L.L., Pirkle, J.L., (2002b). NTP Center for the Evaluation of Risks to Human Repro-
Sampson, E.J., Lucier, G.W., Jackson, R.J., and Brock, J.W. (2000). duction: Phthalates expert panel report on the reproductive and devel-
Levels of seven urinary phthalate metabolites in a human reference opmental toxicity of di-n-butyl phthalate. Reprod. Toxicol. 16:489
population. Environ. Health Perspect. 108:979982. 527.
Brock, J.W., Caudill, S.P., Silva, M.J., Needham, L.L., and Hilborn, Lake, B.G., Phillips, J.C., Linnell, J.C., and Gangolli, S.D. (1977). The
E.D. (2002). Phthalate monoesters levels in the urine of young chil- in vitro hydrolysis of some phthalate diesters by hepatic and intesti-
dren. Bull. Environ. Contam. Toxicol 68:309314. nal preparations from various species. Toxicol. Appl. Pharmacol.
Colon, I., Caro, D., Bourdony, C.J., and Rosario, O. (2000). Identifica- 39:239248.
tion of phthalate esters in the serum of young Puerto Rican girls with Lehmann, K.P., Phillips, S., Sar, M., Foster, P.M.D., and Gaido, K.W.
premature breast development. Environ. Health Perspect. 108:895 (2004). Dose-dependent alterations in gene expression and testos-
900. terone synthesis in the fetal testes of male rats exposed to di (n-butyl)
Creasy, D.M., Beech, L.M., Gray, T.J.B., and Butler, W.H. (1987). The phthalate. Toxicol. Sci. 81:6068.
ultrastructural effects of di-n-pentyl phthalate on the testis of the Lloyd, S.C., and Foster, P.M.D. (1988). Effect of mono-(2-
mature rat. Exp. Mol. Pathol. 46:357371. ethylhexyl)phthalate on follicle-stimulating hormone responsiveness
Duty, S.M., Silva, M.J., Barr, D.B., Brock, J.W., Ryan, L., Chen, Z., of cultured rat Sertoli cells. Toxicol. Appl. Pharmacol. 95:484489.
Herrick, R.F., Christiani, D.C., and Hauser, R. (2003). Phthalate ex- Mylchreest, E., Cattley, R.C., and Foster, P.M.D. (1998). Male repro-
posure and human semen parameters. Epidemiology 14:269277. ductive tract malformations in rats following gestational and lacta-
Fisher, J.S., Macpherson, S., Marchetti, N., and Sharpe, R.M. (2003). tional exposure to di(n-butyl) phthalate: An antiandrogenic mecha-
Human testicular dysgenesis syndrome: A possible model using in- nism? Toxicol. Sci. 43:4760.
utero exposure of the rat to dibutyl phthalate. Hum. Reprod. 18:1383 Mylchreest, E., Sar, M., Cattley, R.C., and Foster, P.M.D. (1999a). Dis-
1394. ruption of androgen-regulated male reproductive development by
IMPAIRED FETAL LEYDIG-CELL FUNCTION 719

di(n-butyl) phthalate during late gestation in rats is different from velopmental toxicity, metabolism, and placental transfer of di-n-
flutamide. Toxicol. Appl. Pharmacol. 156:8195. butyl phthalate administered to pregnant rats. Toxicol. Sci. 45:212
Mylchreest, E., Wallace, D.G., Cattley, R.C., and Foster, P.M.D. 224.
(1999b). Dose-response for altered male reproductive development Schmid, P., and Schlatter, C. (1985). Excretion and metabolism of di(2-
and function induced by di(n-butyl) phthalate. Toxicologist 48:147. ethylhexyl) phthalate in man. Xenobiotica 15:251256.
Mylchreest, E., Sar, M., Wallace, D.G., and Foster, P.M.D. (2002). Sharpe, R.M. (2001). Hormones and testis development and the possible
Fetal testosterone insufficiency and abnormal proliferation of Leydig adverse effects of environmental chemicals. Toxicol. Lett. 120:221
cells and gonocytes in rats exposed to di(n-butyl) phthalate. Reprod. 232.
Toxicol. 16:1928. Sharpe, R.M. (2003). The oestrogen hypothesiswhere do we stand
Nef, S., and Parada, L.F. (1999). Cryptorchidism in mice mutant for now? Int. J. Androl. 26:215.
Insl3. Nat. Genet. 22:295299. Shultz, V.D., Phillips, S., Sar, M., Foster, P.M., and Gaido, K.W. (2001).
Nef, S., Shipman, T., and Parada, L.F. (2000). A molecular ba- Altered gene profiles in fetal rat testes after in utero exposure to di(n-
sis for estrogen-induced cryptorchidism. Dev. Biol. 224:354 butyl) phthalate. Toxicol. Sci. 64:233242.
Critical Reviews in Toxicology Downloaded from informahealthcare.com by University of Groningen on 04/29/14

361. Singh, A., Lawrence, W., and Autian, J. (1975). Maternal-fetal transfer
Nitsche, E.M., and Hiort, O. (2000). The molecular basis of androgen of 14 C-di-2-ethylhexyl phthalate and 14 C-diethyl phthalate in rats. J.
insensitivity. Horm. Res. 54:327333. Pharm. Sci. 64:13471350.
Ohtani, H., Miura, I., and Ichikawa, Y. (2000). Effects of dibutyl phtha- Skakkebaek, N.E. (2002). Endocrine disrupters and testicular dysgen-
late as an environmental endocrine disrupter on gonadal sex differ- esis syndrome. Horm. Res. 57(suppl. S2):43.
entiation of genetic males of the frog Rana rugosa. Environ. Health Skakkebaek, N.E., Berthelsen, J.G., Giwercman, A., and Muller, J.
Perspect. 108:11891193. (1987). Carcinoma-in-situ of the testis: Possible origin from gono-
Parks, L.G., Ostby, J.S., Lambright, C.R., Abbott, B.D., Klinefelter, cytes and precursor of all types of germ cell tumours except sperma-
G.R., Barlow, N.J., and Gray, L.E., Jr. (2000). The plasticizer diethyl- tocytoma. Int. J. Androl. 10:1928.
hexyl phthalate induces malformations by decreasing fetal testos- Toppari, J., Kaleva, M., and Virtanen, H.E. (2001). Trends in the inci-
terone synthesis during sexual differentiation in the male rat. Toxicol. dence of cryptorchidism and hypospadias, and methodological lim-
Sci. 58:339349. itations of registry-based data. Hum. Reprod. Update 7:282286.
Peters, J.M., Taubeneck, M.W., Keen, C.L., and Gonzalez, F.J. (1997). Ward, J.M., Peters, J.M., Perella, C.M., and Gonzalez, F.J. (1998).
For personal use only.

Di(2-ethylhexyl) phthalate induces a functional zinc deficiency dur- Receptor and nonreceptor-mediated organ-specific toxicity of di(2-
ing pregnancy and teratogenesis that is independent of peroxisome ethylhexyl)phthalate (DEHP) in peroxisome proliferator-activated
proliferator-activated receptor-alpha. Teratology 56:311316. receptor alpha-null mice. Toxicol. Pathol. 26:240246.
Rajpert-De Meyts, E., Jorgensen, N., Brondum-Nielsen, K., Muller, J., Wilson, V. S., Lambright, C., Furr, J., Ostby, J., Wood, C., Held, G., and
and Skakkebaek, N.E. (1998). Developmental arrest of germ cells in Gray, L.E., Jr. (2004). Phthalate ester-induced gubernacular lesions
the pathogenesis of germ cell neoplasia. Apmis 106:198204; dis- are associated with reduced insl3 gene expression in the fetal rat
cussion 204196. testis. Toxicol. Lett. 146:207215.
Saillenfait, A.M., Payan, J.P., Fabry, J.P., Beydon, D., Langonne, Yong, E.L., Loy, C.J., and Sim, K.S. (2003). Androgen receptor gene
I., Gallissot, F., and Sabate, J.P. (1998). Assessment of the de- and male infertility. Hum. Reprod. Update 9:17.

You might also like