You are on page 1of 7

0022-5347/02/1685-2142/0 Vol.

168, 2142–2148, November 2002


THE JOURNAL OF UROLOGY® Printed in U.S.A.
Copyright © 2002 by AMERICAN UROLOGICAL ASSOCIATION, INC.® DOI: 10.1097/01.ju.0000034424.55595.33

Review Article

ANDROGEN IMPRINTING OF THE BRAIN IN ANIMAL MODELS AND


HUMANS WITH INTERSEX DISORDERS: REVIEW AND
RECOMMENDATIONS
ZOLTAN HRABOVSZKY AND JOHN M. HUTSON
From the Surgical Department, Royal Children’s Hospital, Melbourne, Victoria, Australia

ABSTRACT

Purpose: Psychosexual development, gender assignment and surgical treatment in patients


with intersex are controversial issues in the medical literature. Some groups are of the opinion
that gender identity and sexual orientation are determined prenatally secondary to the fetal
hormonal environment causing irreversible development of the nervous system. We reviewed the
evidence in animal and human studies to determine the possible role of early postnatal androgen
production in gender development.
Materials and Methods: An extensive literature review was performed of data from animal
experiments and human studies.
Results: Many animal studies show that adding or removing hormonal stimulus in early
postnatal life can profoundly alter gender behavior of the adult animal. Human case studies show
that late intervention is unable to reverse gender orientation from male to female. Most studies
have not permitted testing of whether early gender assignment and treatment as female with
suppression/ablation of postnatal androgen production leads to improved concordance of the
gender identity and sex of rearing.
Conclusions: Animal studies support a role for postnatal androgens in brain/behavior devel-
opment with human studies neither completely supportive nor antagonistic. Therefore, gender
assignment in infants with intersex should be made with the possibility in mind that postnatal
testicular hormones at ages 1 to 6 months may affect gender identity. A case-control study is
required to test the hypothesis that postnatal androgen exposure may convert ambisexual brain
functions to committed male behavior patterns.
KEY WORDS: androgens, gender identity, testis, brain, sex differentiation disorders

A newborn with ambiguous genitalia causes severe dis- culine behavior (mounting) than control females.1 According
tress to the family and immediately raises several questions to their hypothesis testicular steroids could alter the devel-
about gender, the type and timing of treatment for the anom- oping nervous system permanently, masculinizing and de-
aly and the possible outcome. The physician is expected to feminizing sexual behavior of the animal as an adult. This
answer these questions but in many cases it is not easy. In long-lasting “organizational” effect contrasted with the “acti-
several aspects intersex disorders are not yet completely vational” effects of steroids on behavior when steroid treat-
understood and the medical literature is also divided about ment in adult animals caused only a temporary behavioral
issues of gender assignment and surgical treatment. change. The organizational influences of hormones typically
We provide an up-to-date summary of the most impor- occur during early critical periods of development, when mas-
tant data gained from animal experiments and human culine neural circuits develop or are maintained and femi-
studies on androgenic control of brain development. We nine circuits are prevented from developing or are allowed to
searched for evidence that may relate to whether postnatal atrophy. The mechanisms involved in these hormonal influ-
hormone levels have any role in masculinizing the devel- ences on neural development have been described most
oping nervous system, which could be relevant for the clearly in studies of rodents.
timing of surgery, or whether these changes occur exclu- Hormonal Influences on Reproductive Behavior: Genetically
sively prenatally. In the light of our current knowledge of female rats treated with testosterone during the first few days
this field we establish some guidelines for the clinician on of life do not show the typical female reproductive behavior of
gender assignment and the subsequent treatment of chil- lordosis in adulthood even if given activating injections of fe-
dren with ambiguous genitalia. male steroid hormones, estradiol and progesterone in adult-
hood.2 In contrast, their behavior is more like that of genetic
males. Similarly male rats castrated at birth show lordosis in
ANIMAL EXPERIMENTS
response to activating doses of estradiol and progesterone, and
Rodents. In 1959 Phoenix et al reported that adult female do not show male sexual behavior after testosterone treatment
guinea pigs exposed to androgens during gestation showed in adulthood.3 Prenatal antiandrogen treatment also prevents
less feminine copulatory behavior (lordosis) and more mas- masculinization of mating behavior in males.4 These early hor-
2142
ANDROGEN IMPRINTING OF BRAIN AND INTERSEX DISORDERS 2143
mone manipulations can permanently alter the neural poten- under the control of hypothalamic nuclei.19 Differentiation of
tial for the animal to show sex typical reproductive behavior. the cyclic or tonic pattern depends on the influence of gonadal
Ovarian hormones do not appear to be essential for the hormones early in life. Prenatal and postnatal antiandrogen
development of female reproductive behavior. Female rats treatment in male rats produces the cyclic pattern of gonad-
ovariectomized or treated with anti-estrogens after birth otropin hormone release.20
show sexual behavior similar to that of intact females.5 The Early postnatal gonadectomy in male or female rats also
absence of androgens or their metabolites appears to be suf- results in the cyclic pattern of gonadotropin hormone secre-
ficient for feminine behavioral development. tion.18, 21 Early postnatal treatment with aromatizable an-
Paradoxically estrogen treatment was even more effective drogens or high doses of estrogens in female rats produces a
than androgen treatment for masculinizing reproductive be- male (tonic) pattern of gonadotropin hormone release.18, 22, 23
havior in neonatal female rodents.6 Estradiol can be formed These observations suggest that disintegration of the cyclic
from testosterone through the action of the enzyme aro- pattern and differentiation of the tonic pattern of gonadotro-
matase. This enzyme has been localized to several brain pin hormone release are directly under estrogenic rather
regions that also are known to contain estrogen receptors than androgenic control.
and be involved in the control of reproductive behavior.7 Birds. Sexual Dimorphism of the Song System: Another
According to the estrogen hypothesis, during normal male useful model in which to study the early differentiating ef-
development testosterone enters the cells in these brain re- fects of gonadal steroids on the developing brain is the neural
gions, where it is converted to estradiol by aromatase. Estra- circuit for song in songbirds. Courtship song and the brain
diol then interacts with estrogen receptors to promote neural regions that control it show striking sexual dimorphism in
differentiation in the masculine direction. While testosterone zebra finches.24 Only males sing and the telencephalic nuclei
can enter the brain easily in males, developing female rats in the motor pathway that generate song are larger in vol-
are protected from maternal estrogens, which are bound to ume in males and contain more and larger cells than in
!-fetoprotein and unable to penetrate the blood-brain barri- females.24 Males sing more often when testosterone levels
er.8 This hypothesis has been supported by evidence that are high in adulthood but females do not sing at all, even if
treating male rats with anti-estrogens or aromatase inhibi- treated with testosterone as adults.25 Estrogens and andro-
tors prevents the normal masculine development of repro- gens given in gonadally intact, hatchling female zebra
ductive behavior despite the high level of endogenous testos- finches permanently masculinize the song system, increasing
terone.7 These drugs also block behavioral masculinization in the size of vocal control regions.26 These neonatally mascu-
testosterone treated newborn females.9 linized females also sing in adulthood.27 While the most
Sexual Differentiation of Brain Structures: Levels of go- potent virilizing agent is estradiol, nonaromatizable andro-
nadal hormones during early development also influence sex- gens have relatively little masculinizing effect in hatchling
ual differentiation of the brain structure. Several areas with females.28
cytoarchitectural sexual dimorphism have been described in However, experiments in zebra finches have yielded some
the brain and spinal cord.10 The most extensively studied contradictory results. Steroid treatment in females can effec-
structure is the preoptic area, which is involved in mating tively masculinize neural centers as well as behavior but
behavior and the control of gonadotrophin secretion.11 The castration,29 antiandrogen,30 antiestrogen31 or aromatase in-
volume of a cell dense subregion of the preoptic area called hibitor treatment32 failed to prevent male zebra finches from
the sexually dimorphic nucleus of the preoptic area is several having a masculine brain and the singing that accompanies
times larger in adult male rats than in females.11 Neurons of it. Studies measuring plasma sex steroids in the weeks after
the preoptic area in male and female rats contain more hatching did not show higher androgen levels in males than
estrogen receptors than any other brain region.12 While the in females.33 These results suggest that testicular secretions
sexually dimorphic nucleus of the preoptic area preferen- are not exclusively responsible for masculine development of
tially binds estrogen in the 2 sexes, it also accumulates the neural song system in this species. The brain of male
testosterone and dihydrotestosterone.13 Adult hormone ma- songbirds may have an alternative, steroid independent
nipulations do not alter the volume of the sexually dimorphic means of developing in the masculine direction.
nucleus in either sex, although exposure of developing female
rats to testosterone or estrogens leads to a larger, more
HUMAN STUDIES
masculine sexually dimorphic nucleus of the preoptic area in
adulthood and castration of newborn males results in a more While data from animal studies should be applied to hu-
feminine one.14 The masculinizing effect of early estrogen mans with extreme caution, the similarity of findings in
treatment is stronger than that of early androgen treatment, diverse numbers of species suggests that processes that have
suggesting that estrogens have a direct effect on stimulating evolved in lower animals may be preserved in humans.
the development of the sexually dimorphic nucleus of the The best studied sexually dimorphic brain structure in
preoptic area in male rats.14, 15 humans is the sexually dimorphic nucleus of the preoptic
It appears that testosterone has 2 major effects on the fetal area. It has been shown that males have a larger nucleus
rodent brain. Testosterone up-regulates the production of with more neurons than females but this sex difference in
androgen receptors in sexually dimorphic brain areas.16 In neuronal number is not detectable in children younger than
addition, androgen receptor activation by testosterone stim- 6 to 10 years.34
ulates aromatase synthesis, which results in increased estra- Another strategy for asking whether fetal steroids affect
diol production within the sexually dimorphic nuclei.17 the human brain is to study sex differences in human behav-
Increased levels of estradiol are then responsible for devel- ior and determine whether inadvertent exposure to excess
oping the anatomical differences of the brain in the male amount of hormones alters sexually dimorphic behavior. Sev-
direction. eral aspects of human behavior show reliable sex differences,
Hormonal Influences on Gonadotropin Regulation: Male including gender identity, sexual orientation, rough play,
and female mammals differ in their patterns of gonadotropin juvenile toy preferences, physical aggression, and certain
hormone release.18 In adult females gonadotropin hormone aspects of verbal, visual-spatial and mathematical abilities.
release is cyclic, producing fluctuations in the ovarian hor- The most characteristic differences are those in gender iden-
mones associated with estrus or menstrual cycles. In con- tity and sexual orientation.35
trast, in males gonadotropin hormone secretion is tonic, Strong evidence that hormones contribute to human be-
maintaining high levels of testicular hormones in adulthood. havioral development has come from studies of girls with
Gonadotropin hormone release from the pituitary gland is congenital adrenal hyperplasia. This disorder is caused by an
2144 ANDROGEN IMPRINTING OF BRAIN AND INTERSEX DISORDERS

enzymatic defect resulting in overproduction of adrenal ste- 1971 as hormonal support for at-risk pregnancy until the
roids, including androgens. Females with congenital adrenal relationship of prenatal diethylstilbestrol exposure to adeno-
hyperplasia are exposed to androgens during gestation and carcinoma of the vagina and cervix was identified.48
are typically born with some degree of genital virilization. Diethylstilbestrol exposed women showed bisexual or homo-
Postnatal exposure to androgens would also occur until ad- sexual interest more frequently than siblings and matched
renal hormone replacement therapy is begun. controls.49 They showed greater cognitive lateralization (sim-
These individuals are usually assigned the female sex and ilar to men) than their nondiethylstilbestrol treated sisters,50
raised as girls. Many have a tomboyish personality, play with which suggests that diethylstilbestrol slightly masculinized
toys that are more often favored by boys and participate in them. Diethylstilbestrol exposed females appear to be unaf-
more rough play than other girls.36, 37 Behavioral masculin- fected in regard to gender identity, childhood play and inter-
ization in congenital adrenal hyperplasia cases does not seem ests, and cognitive abilities.51 Findings in these women have
to be related to the degree of genital virilization.38 As women, special significance because they are unlikely to reflect
they are usually sexually attracted to men but are more psychosocial factors. In contrast to women with congenital
likely to have bisexual or homosexual interests than women adrenal hyperplasia, these patients are born with typical
without the condition.37, 39 Although the majority of patients female genitalia, and so the possibility that genital abnor-
assigned and reared as female have a female gender identi- malities discouraged heterosexual interest can be excluded.
ty,40 some become gender dysphoric in adolescence or adult- Further evidence supporting a contribution of the early
hood and an even smaller number choose to live as males.41 hormone environment to gender identity and behavioral de-
It has been suggested that masculinized behavior in females velopment comes from a pair of male monozygotic twins, in 1
with congenital adrenal hyperplasia could result from paren- of whom the penis was accidentally ablated during circumci-
tal doubt and self-doubt regarding sexual identity after early sion at age 7 months.52 A decision was made to reassign the
genital virilization.38 However, an androgen effect on the infant to the female sex and rear him as a girl. Surgical
developing brain, similar to that in other species, seems to be castration with initial genital reconstruction was done at age
an equally plausible explanation for the behavioral differ- 21 months. Subsequent followup showed that by early ado-
ences. lescence the patient had rejected the female identity and he
Information on patients with 5!-reductase deficiency and began to live as a male at age 14 years.52 Later the patient
17"-hydroxysteroid dehydrogenase deficiency can also be in- recalled that he had never been comfortable as a girl and he
terpreted as supporting a hormonal contribution to human had an exclusive sexual attraction to females.
psychosexual development. Individuals with these rare en- Although human sexual development is rather complicated
zymatic defects are unable to produce certain androgens because the social environment and upbringing can also sig-
needed for normal prenatal masculinization of the external nificantly contribute to the ultimate expression of human
genitalia. These newborns are often assigned the female gen- sexual behavior and gender identity, available data from
der because minimal virilization is apparent at birth and human studies appear to confirm a role for early hormone
raised as girls. However, at puberty when testicular testos- environment in shaping the developing human nervous sys-
terone secretion increases, the external genitalia virilize and tem.
in many patients male gender identity and sexual behavior
develop.42, 43 An explanation of such results is that fetal
and/or postnatal androgens had masculinized the brain, so THE CRITICAL PERIOD FOR SEXUAL DIFFERENTIATION
that social rearing notwithstanding the individuals were des-
tined to think and, therefore, behave as men. However, it is There seems to be a developmental period during which
difficult to rule out that these individuals were treated dif- the brain is more sensitive to the organizational effects of
ferently than normal girls. gonadal hormones than at any other time. This interval is
In patients with the complete androgen insensitivity syn- called the critical period for sexual differentiation and it is
drome there is a severe defect in androgen receptors, so that species specific.53 The critical period is an empirical concept
developing XY male fetuses ignore the masculinizing effect of and does not represent a clearly defined stage of develop-
androgens and show an unambiguous female exterior.44 They ment. Available data suggest that the beginning of the crit-
are accepted as female at birth and raised as girls but are ical period may follow differentiation of testicular Leydig
identified when menses fail to begin. If the testes are not cells and the onset of testosterone secretion.54 In rats typical
removed, normal female breasts develop from the increased Leydig cells first appear between days 16 and 18 after con-
conversion of testosterone to estrogen at puberty. Such indi- ception.55 Testosterone is significantly higher in male than in
viduals are feminine in gender identity and sexual orienta- females rat fetuses from day 18 after coitus through day 5
tion.44 Their cognitive abilities are also similar to that of post partum.54 The greatest differences in absolute values
normal females.45 The development of feminine characteris- were observed on days 18 and 19, when a distinct surge
tics in these patients is supported by the insensitivity of the occurred in males. Resko et al reported that the concentra-
brain to androgens and their unambiguous upbringing as tion of plasma testosterone in males decreased gradually
girls. after birth, reaching a baseline value between ages 10 and 30
There is a remarkable interspecies difference in the man- days.56 By administering testosterone to female and neona-
ifestation of androgen insensitivity in humans and lower tally castrated male rats they noted that the period during
mammals. Androgen insensitive male rats have a feminine which the volume of the sexually dimorphic nucleus of the
phenotype but a masculine nervous system and behav- preoptic area is sensitive to exogenous androgen begins
ior,46, 47 which underline the importance of estrogen receptors strictly on day 18 of gestation and extends through postnatal
for the sexual differentiation of behavior in this species. day 5.57 Interestingly it was recently noted that castration
Conversely the feminine behavior of androgen insensitive can decrease the volume of the sexually dimorphic nucleus of
humans indicates that sexual steroids act exclusively the preoptic area in male rats even as late as postnatal day
through androgen receptors to masculinize the nervous sys- 29.58 A possible explanation for this paradox could be the
tem or androgen receptors must be functional for estrogen dependence of sexually dimorphic nucleus of the preoptic
receptor activation to be effective or occur at all.10 area cells on gonadal hormones for survival during develop-
Studies of women who had been exposed to diethylstilbes- ment. Administering exogenous testosterone may be effective
trol during gestation suggest that fetal estrogens are also able for a shorter period because cells of the sexually dimorphic
to masculinize human sexual behavior. Diethylstilbestrol nucleus of the preoptic area may only survive for a limited
was extensively used in the United States between 1947 and time after neonatal castration. However, in intact males
ANDROGEN IMPRINTING OF BRAIN AND INTERSEX DISORDERS 2145
these cells remain alive, but dependent on exposure to tes- in lower animals, testosterone in humans also masculinizes
tosterone for survival.58 the central nervous system, we must suppose that these 2
It has also been observed that there are different times peaks of serum testosterone in males have special signifi-
when specific neural and behavioral characteristics are max- cance for sexual development in the male direction.
imally sensitive to steroid hormone influences. For example,
the effect of castration has different time courses on the HYPOTHESIS
luteinizing hormone surge and lordosis behavior in rats.58
Gender identity depends on stepwise exposure to androgen
Only females and males castrated on postnatal day 1 and not
secretion prenatally, postnatally and at puberty. Although
later showed luteinizing hormone surges after estrogen and
the exact role of postnatal androgen production is unknown,
progesterone priming in adulthood. There was a stepwise
we propose that it is a crucial adjunct for masculine devel-
decrease in adult lordosis behavior in male rats castrated
opment of the brain in animals and humans. Furthermore,
postnatally and the response decreased with increasing age
when treating intersex conditions in infants, we should as-
at castration until day 7. These data suggest the existence of
sume that postnatal androgens may masculinize behavior at
several specific critical periods of different lengths that are
least until definitive case-control studies prove otherwise. If
involved in discrete developmental processes of sexual differ-
postnatal androgens have a key role as we suspect, infants
entiation of the central nervous system, in contrast to a
with early female gender assignment and suppression or
single critical period for all developmental mechanisms.58
ablation of androgen production should have better long-
Another important aspect is that hormonal influences on
term outcome for psychosexual development. Moreover, if
sexual differentiation of the brain and behavior are seem-
postnatal sexual steroid imprinting occurs, reassigning in-
ingly not all or nothing. Each masculine and feminine char-
fants with intersex to the female gender should be avoided
acteristic exists on a continuum and the amount, duration or
after ages 4 to 6 months.
timing of hormone exposure determines its position on the
continuum.35
GENDER ASSIGNMENT
In humans to our knowledge the exact timing of the critical
period for sexual differentiation of the central nervous sys- Gender assignment in an individual with intersex is a
tem is unknown. However, we can assume that during a serious and sometimes difficult decision with life-long impli-
critical period testosterone levels in the circulation are sig- cations. Not only do patients need a gender that they identify
nificantly higher in males than in females, similar to the with psychosexually, but also it is equally important to pro-
results of animal experiments. Fetal testis incubation in vitro duce functional genitalia consistent with the gender as-
and perfusion studies in vivo have shown that testicular signed. Money et al first emphasized the role of upbringing in
testosterone production occurs as early as 7 to 8 weeks of gender identity development.66 Their studies in the 1950s of
gestation.59 By 14 to 18 weeks Leydig cells occupy half of the patients with hermaphroditism led to the belief that individ-
volume of the fetal testes, after which they gradually involute uals are psychosexually neutral at birth and become differ-
until term.60 Serum testosterone in the male fetus attains a entiated as male or female during the course of various
peak at 14 to 16 weeks.61 At this time values are in the adult growth experiences. They believed that unambiguously rais-
male range, which is essential for male genital differentia- ing a child with a physical intersex condition as a member of
tion.61 After 24 weeks there is no significant sex difference in the assigned sex would be more important for gender identity
umbilical arterial serum testosterone,62 although the obser- development than chromosomal, gonadal or hormonal sex.67
vation that amniotic fluid levels even in late pregnancy are Money et al recommended early gender assignment with
higher in males than in females61 suggests continued testic- early corrective surgery in an attempt to identify unambig-
ular secretion. uously the child with the chosen sex of rearing. The most
While some investigators noted higher testosterone at important factor for making the decision was “the morphol-
term in the umbilical cord and peripheral venous blood sam- ogy of the external genitals and the ease with which these
ples in males than in females,63 others failed to detect a organs can be surgically reconstructed to be consistent with
significant sex difference.64 In female infants testosterone the assigned sex.”67 The assumption that individuals are
concentration remains constantly low during year 1 of life.65 psychosexually undifferentiated at birth was gradually repu-
In males the level increases from birth to a peak at 1 to 3 diated as further research demonstrated that prenatal hor-
months of life and thereafter it decreases to prepubertal mones can influence certain aspects of human sex dimorphic
levels by ages 4 to 6 months (see figure). If we accept that as behavior in the same direction as in other mammals.

Mean serum testosterone in male fetus and infant61


2146 ANDROGEN IMPRINTING OF BRAIN AND INTERSEX DISORDERS

Currently there are 2 opposing viewpoints in the medical culinize further the nervous system.41 We perform feminiz-
literature regarding gender assignment and the treatment of ing genitoplasty within the first 2 months of life to ensure
individuals with ambiguous genitalia. Traditionally gender that the appearance of the external genitalia is consistent
assignment and genital reconstruction have been quite uni- with the female sex of rearing and relieve parental anxiety
formly done shortly after birth. This practice helps the indi- about the child regarding the concerns of relatives and
vidual to avoid internal conflicts and stigmatization by the friends. When the patient presents after 4 to 6 months and
family or peer environment and facilitates parental accep- has been erroneously raised as a male because of almost
tance of the child as male or female. However, in recent years complete virilization of the external genitalia, we recommend
some intersex support groups, such as the Intersex Society of the maintenance of the originally assigned gender and per-
North America, comprising many adults born with ambigu- form phallic surgery to enable the patient to function as a
ous genitalia who are now dissatisfied with the outcome, male.
have raised their voices against early genital reconstruc- Individuals with complete androgen resistance have an
tion.68 Diamond and Sigmundson have the opinion that the unambiguous female phenotype, gender identity and sexual
internal and external genitals should be left alone until after orientation. Rearing the child as female is the right decision
puberty, when the patient, who is the ultimate decision in gender assignment. The testes can be removed at diagno-
maker, is able to express individual needs and wants as a sis or preserved until puberty to induce spontaneous femini-
sexual being and provide informed consent to corrective sur- zation. After puberty they should be removed because of the
gery.69 They argue that gender identity, sexual orientation increased risk of malignant change.70
and sexual preferences are personal choices, and neither the The most difficult decision is assigning the appropriate
family nor physician can predict how an individual with gender in under virilized genetic males with ambiguous gen-
intersex would choose to live or with what type of genitals. italia. A wide spectrum of anomalies can produce under vir-
According to their principles the patient should undergo gen- ilization in males, such as defects in testosterone synthesis or
der assignment at birth depending on the nature of the metabolism and poor tissue response to androgens. Special
diagnosis, although no corrective surgery on the external or attention should be given to the appearance of the external
internal genitalia should be performed.69 The parents must genitalia and their ability to respond to androgen or human
be consistent in rearing the child as a boy or girl. However, chorionic gonadotropin stimulation. Strikingly masculinized
after puberty it is the patient who eventually decides patients should be assigned the male gender, particularly
whether to live as male, female or even intersexual depend- when phallic enlargement is noted in response to exogenous
ing on how the central nervous system dictates sexual iden- androgens. On the other hand, when the phallus is inade-
tity in the individual. Opponents of this view argue that the quate and does not increase in size with stimulation, female
ambiguous appearance of the genitalia may hinder gender gender assignment is most prudent when the patient pre-
identity development and unambiguous rearing by the par- sents in the neonatal period. To make the decision we must
ents is virtually impossible when the genitals are not consis- also consider the expected course and outcome of that partic-
tent with the sex chosen. While corrective surgery with cur- ular anomaly. Patients with 5!-reductase deficiency and
rent techniques can offer good results in cases of severe 17"-hydroxysteroid dehydrogenase deficiency require spe-
hypospadias, not performing the operation imposes obvious cialized laboratory evaluation to make the diagnosis but they
practical difficulties in genotypically male patients. In cer- are good candidates for male gender assignment because
tain intersex disorders retaining dysgenetic or streak gonads marked spontaneous virilization can be expected at puber-
with a potential for tumor development can be a hazardous ty.42, 43 Should the female gender be elected the patient needs
practice. Furthermore, we do not know what role the physi- early orchiectomy and estrogen replacement therapy at pu-
ologically increased level of postnatal testosterone in males berty.
has on later psychosexual development. If it has a role, it In true hermaphrodites when male gender assignment is
would seem wise in any patient with intersex assigned the being considered, serum testosterone levels before and after
female gender to remove the testes in the first few weeks of human chorionic gonadotropin stimulation are said to have a
life well before the serum testosterone peak. predictive value. If levels are consistently low despite stim-
ulation, poor masculinization at puberty can be expected.71
While in individuals raised as males spermatogenesis and
RECOMMENDATIONS
testicular hormone production are characteristically defi-
Rapid detailed investigations must be performed in new- cient,71 others raised as females may ovulate, become preg-
borns with genital ambiguity to evaluate the chromosomal nant and give birth to normal offspring.72 After gender as-
gonadal sex, hormonal environment, and anatomy of the signment gonads and internal organs discordant for the sex
external and internal genital organs to clarify the type of of rearing should be removed. Males need regular evaluation
anomaly. The patient needs gender assignment as early as because tumor may form in testicular tissue.73
possible, preferentially within the first few weeks of life. In 46 XY pure and in mixed gonadal dysgenesis the gonads
Fortunately gender assignment is relatively straightforward have a propensity toward malignant tumors.74 All dysgenetic
in the majority of individuals with intersex. testes and streak gonads should be removed regardless of the
In newborns with ambiguous genitalia secondary to con- gender assigned. The preferred sex of rearing is female.
genital adrenal hyperplasia or intrauterine exposure to high However, markedly virilized individuals can be raised as
levels of androgens regardless of the degree of virilization male and in selected situations the testis can be retained if it
female gender assignment is warranted for several reasons. is in the scrotal position.75
Because the internal organs in these patients are essentially
intact, they have the potential for normal sexual function and
CONCLUSIONS
fertility. Although there is more rough play than may be
expected of girls, and a higher incidence of bisexuality and Animal experiments in songbirds and mammals have pro-
homosexuality than in the general population, these individ- vided evidence that prenatal and early postnatal sexual hor-
uals almost always identify as female if gender assignment is mone secretions have an organizational effect on the devel-
made early in life.40 Most often individuals with congenital oping nervous system, which results in sexual dimorphism of
adrenal hyperplasia who want the gender changed to male brain structure and behavior. Studies in individuals with
have been discovered in late infancy or childhood, or have not intersex and traumatized genitalia who underwent sex reas-
been receiving adrenal suppressive medication consistently signment suggest that a mechanism similar to that in ani-
since birth, allowing androgen levels to remain high to mas- mals may also exist in our species. However, sexual develop-
ANDROGEN IMPRINTING OF BRAIN AND INTERSEX DISORDERS 2147
ment of the human brain is much more complicated since estrogen antagonist and an androgen antagonist on differen-
social factors also contribute to the ultimate expression of tiation of the sexually dimorphic nucleus of the preoptic area
human sexual behavior and gender identity. in male and female rats. Neuroendocrinology, 42: 443, 1986
The 2 periods during early human development when an- 16. Lu, S. F., McKenna, S. E., Cologer-Clifford, A., Nau, E. A. and
drogens are elevated in males compared with females are the Simon, N. G.: Androgen receptor in mouse brain: sex differ-
ences and similarities in autoregulation. Endocrinology, 139:
prenatal period from about weeks 8 to 24 of gestation and the
1594, 1998
postnatal period from about months 1 to 6 of life (see figure). 17. McCarthy, M. M.: Molecular aspects of sexual differentiation of
We propose that hormone induced masculinization on the the rodent brain. Psychoneuroendocrinology, 19: 415, 1994
developing brain occurs during these 2 periods of hormone 18. Pfeiffer, C. A.: Sexual differences of the hypophyses and their
elevation. Moreover, when decisions about gender assign- determination by the gonads. Am J Anat, 58: 195, 1936
ment and treatment are made early, it should allow for 19. Gorski, R. A.: Localization and sexual differentiation of the ner-
possible postnatal virilization. vous structures which regulate ovulation. J Reprod Fertil, 1:
Early gender assignment followed by early removal of the 67, 1966
source of excess androgens when the female gender is chosen 20. Neumann, F. and Elger, W.: Permanent changes in gonadal
may prevent the brain from further masculinization after function and sexual behavior as a result of early feminization
of male rats by treatment with an antiandrogenic steroid.
birth. This result requires prompt administration of hydro-
Endokrinologie, 50: 209, 1966
cortisone in patients with congenital adrenal hyperplasia 21. Takewaki, K.: Some experiments on the control of hypophyseal-
and removal of all testicular tissue in genotypically male gonadal system in the rat. Gen Comp Endocrinol, suppl., 1:
individuals before postnatal androgen production causes fur- 309, 1962
ther masculine development of the nervous system. The best 22. Gorski, R. A. and Barraclough, C. A.: Effects of low dosages of
outcome may be expected when early gender assignment and androgen on the differentiation of hypothalamic regulatory
genitoplasty are combined with unambiguous upbringing in control of ovulation in the rat. Endocrinology, 73: 210, 1963
the sex chosen for the patient with the intersex anomaly. 23. Greene, R. R., Burrill, M. W. and Ivy, A. C.: Experimental inter-
Infants diagnosed late and treated beyond ages 4 to 6 months sexuality: the paradoxical effects of estrogens on the sexual
should not undergo sex reversal to the female gender since development of the female rat. Anat Rec, 74: 429, 1939
24. Nottebohm, F. and Arnold, A. P.: Sexual dimorphism in vocal
prenatal and postnatal imprinting can be expected.
control areas of the songbird brain. Science, 194: 211, 1976
25. Arnold, A. P.: Behavioral effects of androgen in zebra finches
REFERENCES (Poephila guttata) and a search for its action. Ph. D. Thesis,
Rockefeller University, New York, 1974
1. Phoenix, C. H., Goy, R. W., Gerall, A. A. and Young, W. C.:
26. Gurney, M. E.: Hormonal control of cell form and number in the
Organizing action of prenatally administered testosterone pro-
pionate on the tissues mediating mating behavior in the fe- zebra finch song system. J Neurosci, 1: 658, 1981
male guinea pig. Endocrinology, 65: 369, 1959 27. Pohl-Apel, G.: The correlation between the degree of brain mas-
2. Ward, I. L.: Differential effect of pre- and postnatal androgen on culinization and song quality in estradiol treated female zebra
the sexual behavior of intact and spayed female rats. Horm finches. Brain Res, 336: 381, 1985
Behav, 1: 25, 1969 28. Grisham, W. and Arnold, A. P.: A direct comparison of the
3. Gerall, A. A., Hendricks, S. E., Johnson, L. L. and Bounds, T. W.: masculinizing effects of testosterone, androstenedione, estro-
Effects of early castration in male rats adult sexual behavior. gen, and progesterone on the development of the zebra finch
J Comp Physiol Psychol, 64: 206, 1967 song system. J Neurobiol, 26: 163, 1995
4. Clemens, L. G., Gladue, B. A. and Coniglio, L. P.: Prenatal 29. Arnold, A. P.: The effects of castration and androgen replace-
endogenous androgenic influences on masculine sexual behav- ment on song, courtship, and aggression in zebra finches
ior and genital morphology in male and female rats. Horm (Poephila guttata). J Exp Zool, 191: 309, 1975
Behav, 10: 40, 1978 30. Schlinger, B. A. and Arnold, A. P.: Androgen effects on the
5. Young, W. C., Goy, R. W. and Phoenix, C. H.: Hormones and development of the zebra finch song system. Brain Res, 561:
sexual behavior. Science, 143: 212, 1964 99, 1991
6. Whalen, R. E. and Nadler, R. D.: Suppression of the development 31. Mathews, G. A., Brenowitz, E. A. and Arnold, A. P.: Paradoxical
of female mating behavior by estrogen administered in in- hypermasculinization of the zebra finch song system by an
fancy. Science, 141: 273, 1963 antiestrogen. Horm Behav, 22: 540, 1988
7. McEwen, B. S., Lieberburg, I., Chaptal, C. and Krey, L. C.: 32. Wade, J., Schlinger, B., Hodges, L. and Arnold, A. P.: Fadrozole,
Aromatization: important for sexual differentiation of the neo- a potent and specific inhibitor of aromatase in the zebra finch
natal rat brain. Horm Behav, 9: 249, 1977 brain. Gen Comp Endocrinol, 94: 53, 1994
8. Vermeulen, A. and Verdonck, L.: Studies on the binding of tes- 33. Hutchison, J. B., Wingfield, J. C. and Hutchison, R. E.: Sex
tosterone to human plasma. Steroids, 11: 609, 1968 differences in plasma concentrations of steroids during the
9. Fadem, B. H. and Barfield, R. J.: Neonatal hormonal influences sensitive period for brain differentiation in the zebra finch.
on the development of proceptive and receptive feminine sex- J Endocrinol, 103: 363, 1984
ual behavior in rats. Horm Behav, 15: 282, 1981 34. Hofman, M. A. and Swaab, D. F.: The sexually dimorphic nu-
10. Cooke, B., Hegstrom, C. D., Villeneuve, L. S. and Breedlove, cleus of the preoptic area in the human brain: a comparative
S. M.: Sexual differentiation of the vertebrate brain: principles morphometric study. J Anat, 164: 55, 1989
and mechanisms. Front Neuroendocrinol, 19: 323, 1998 35. Hines, M.: Abnormal sexual development and psychosexual is-
11. Gorski, R. A., Gordon, J. H., Shryne, J. E. and Southam, A. M.: sues. Baillieres Clin Endocrinol Metab, 12: 173, 1998
Evidence for a morphological sex difference within the medial 36. Berenbaum, S. A. and Hines, M.: Early androgens are related to
preoptic area of the rat brain. Brain Res, 148: 333, 1978 childhood sex-typed toy preferences. Psychol Sci, 3: 203, 1992
12. Herbison, A. E. and Theodosis, D. T.: Immunocytochemical iden- 37. Ehrhardt, A. A. and Meyer-Bahlburg, H. F.: Effects of prenatal
tification of oestrogen receptors in preoptic neurones contain- sex hormones on gender-related behavior. Science, 211: 1312,
ing calcitonin gene-related peptide in the male and female rat. 1981
Neuroendocrinology, 56: 761, 1992 38. Slijper, F. M. E.: Androgens and gender role behaviour in girls
13. Jacobson, C. D., Arnold, A. P. and Gorski, R. A.: Steroid autora- with congenital adrenal hyperplasia (CAH). Prog Brain Res,
diography of the sexually dimorphic nucleus of the preoptic 61: 417, 1984
area. Brain Res, 414: 349, 1987 39. Money, J., Schwartz, M. and Lewis, V. G.: Adult erotosexual
14. Dohler, K. D., Coquelin, A., Davis, F., Hines, M., Shryne, J. E. status and fetal hormonal masculinization and demasculiniza-
and Gorski, R. A.: Pre- and postnatal influence of testosterone tion: 46,XX congenital virilizing adrenal hyperplasia and
propionate and diethylstilbestrol on differentiation of the sex- 46,XY androgen-insensitivity syndrome compared.
ually dimorphic nucleus of the preoptic area in male and Psychoneuroendocrinology, 9: 405, 1984
female rats. Brain Res, 302: 291, 1984 40. Money, J. and Ehrhardt, A. A.: Gender dimorphic behavior and
15. Dohler, K. D., Coquelin, A., Davis, F., Hines, M., Shryne, J. E., fetal sex hormones. Rec Prog Horm Res, 28: 735, 1972
Sickmoller, P. M. et al: Pre- and postnatal influence of an 41. Meyer-Bahlburg, H. F. L., Gruen, R. S., New, M. I., Bell, J. J.,
2148 ANDROGEN IMPRINTING OF BRAIN AND INTERSEX DISORDERS

Morishima, A., Shimshi, M. et al: Gender change from female period for the sexual differentiation of the sexually dimorphic
to male in classical adrenal hyperplasia. Horm Behav, 30: 319, nucleus of the preoptic area in the rat. Neuroendocrinology,
1996 62: 579, 1995
42. Imperato-McGinley, J., Guerrero, L., Gautier, T. and Peterson, 59. Siiteri, P. K. and Wilson, J. D.: Testosterone formation and
R. E.: Steroid 5alpha-reductase deficiency in man: imprinting metabolism during male sexual differentiation in the human
and the establishment of gender role. AMA Arch Neurol Psych, embryo. J Clin Endocrinol Metab, 38: 113, 1974
77: 333, 1957 60. Mancini, R. E., Vilar, O. and Lavieri, J. C.: Development of
43. Farkas, A. and Rosler, A.: Ten years experience with masculin- Leydig cells in the normal human testis. Am J Anat, 112: 203,
izing genitoplasty in male pseudohermaphroditism due to 17 1963
beta-hydroxysteroid dehydrogenase deficiency. Eur J Pediatr, 61. Smail, P. J., Reyes, F. I., Winter, J. S. D. and Faiman, C.: The
152: S88, 1993 fetal hormonal environment and its effect on the morphogen-
44. Wilson, J. D., George, F. W. and Griffin, J. E.: The hormonal esis of the genital system. In: Pediatric Andrology. Edited by
control of sexual development. Science, 211: 1278, 1981 S. J. Kogan and E. S. E. Hafez. The Hague: Martinus Nijhoff,
45. Masica, D. N., Money, J., Ehrhardt, A. A. and Lewis, V. G.: IQ, 1981
fetal sex hormones and cognitive patterns: studies in the tes- 62. Takagi, S., Yoshida, T., Tsubata, K., Ozaki, H., Fujii, T. K.,
ticular feminizing syndrome of androgen insensitivity. Johns Nomura, Y. et al: Sex differences in fetal gonadotropins and
Hopkins Med J, 124: 34, 1969 androgens. J Steroid Biochem, 8: 609, 1977
46. Jacobson, C. D.: The characterization, ontogeny and influence of 63. Herruzo, A. J., Mozas, J., Alarcon, J. L., Lopez, J. M., Molina, R.,
androgen on the sexually dimorphic nucleus of the preoptic Molto, L. et al: Sex differences in serum hormone levels in
area. Ph. D. Thesis, University of California, Los Angeles, umbilical vein blood. Int J Gynaecol Obstet, 41: 37, 1993
California, 1980 64. Rivarola, M. A., Forest, M. G. and Migeon, C. J.: Testosterone,
47. Olsen, K. L.: Androgen-insensitive rats are defeminised by their androstendione and dehydroepiandrosterone in plasma during
testes. Nature, 279: 238, 1979 pregnancy and at delivery: concentration and protein binding.
48. Herbst, A. L., Ulfelder, H. and Poskanzer, D. C.: Adenocarcinoma J Clin Endocrinol Metab, 28: 34, 1968
of the vagina. Association of maternal stilbestrol therapy with 65. Forest, M. G., Sizonenko, P. C., Cathiard, A. M. and Bertrand, J.:
tumor appearance in young women. N Engl J Med, 284: 878,
Hypophyso-gonadal function in humans during the first year
1971
of life. I. Evidence for testicular activity in early infancy. J Clin
49. Meyer-Bahlburg, H. F. L., Ehrhardt, A. A., Rosen, L. R., Gruen,
Invest, 53: 819, 1974
R. S., Veridiano, N. P., Vann, F. H. et al: Prenatal estrogens
66. Money, J., Hampson, J. G. and Hampson, J. L.: An examination
and the development of homosexual orientation. Dev Psychol,
of some basic sexual concepts: the evidence of human her-
31: 12, 1995
maphroditism. Bull Johns Hopkins Hospital, 97: 301, 1955
50. Hines, M. and Shipley, C.: Prenatal exposure to diethylstilbes-
trol (DES) and the development of sexually dimorphic cogni- 67. Money, J., Hampson, J. G. and Hampson, J. L.: Imprinting and
tive abilities and cerebral lateralization. Dev Psychol, 20: 81, the establishment of gender role. AMA Arch Neurol Psych, 77:
1984 333, 1957
51. Lish, J. D., Meyer-Bahlburg, H. F., Ehrhardt, A. A., Travis, B. G. 68. Recommendations for Treatment: Intersex Infants and Children.
and Veridiano, N. P.: Prenatal exposure to diethylstilbestrol San Francisco: Intersex Society of North America, 1995
(DES): childhood play behavior and adult gender-role behavior 69. Diamond, M. and Sigmundson, H. K.: Management of intersex-
in women. Arch Sex Behav, 21: 423, 1992 uality: guidelines for dealing with persons with ambiguous
52. Bradley, S. J., Oliver, G. D., Chernick, A. B. and Zucker, K. J.: genitalia. Arch Pediatr Adolesc Med, 151: 1046, 1997
Experiment of nurture: ablatio penis at 2 months, sex reas- 70. Snyder, H. McC., III: Management of ambiguous genitalia in the
signment at 7 months, and a psychosexual follow-up in young neonate. In: Urologic Surgery in Neonates and Young Infants.
adulthood. Pediatrics, 102: e9, 1998 Edited by L. R. King. Philadelphia: W. B. Saunders Co., pp.
53. MacLusky, N. J. and Naftolin, F.: Sexual differentiation of the 346 –385, 1988
central nervous system. Science, 211: 1294, 1981 71. Aaronson, I. A.: True hermaphroditism. A review of 41 cases
54. Weisz, J. and Ward, I. L.: Plasma testosterone and progesterone with observations on testicular histology and function. Br J
titers of pregnant rats, their male and female fetuses and Urol, 57: 775, 1985
neonatal offspring. Endocrinology, 106: 306, 1980 72. Kim, M. H., Gumpel, J. A. and Graff, P.: Pregnancy in a true
55. Merchant-Larios, H.: The onset of testicular differentiation in hermaphrodite. Obstet Gynecol, 53: 40S, 1979
the rat: an ultrastructural study. Am J Anat, 145: 319, 1976 73. Guaschino, S., Stola, E., Spinillo, A., Marchetti, M. and Aguzzi,
56. Resko, J. A., Feder, H. H. and Goy, R. W.: Androgen concentra- A.: True hermaphroditism: diagnosis and surgical treatment.
tions in plasma and testis of developing rats. J Endocrinol, 40: Clin Exp Obstet Gynecol, 15: 74, 1988
485, 1968 74. Amice, V., Amice, J., Bercovici, J. P., Riviere, D. and Corolleur,
57. Rhees, R. W., Shryne, J. E. and Gorski, R. A.: Termination of the M. J.: Gonadal tumor and H-Y antigen in 46,XY pure gonadal
hormone-sensitive period for differentiation of the sexually dysgenesis. Cancer, 57: 1313, 1986
dimorphic nucleus of the preoptic area in male and female 75. Giwercman, A., von der Maase, H. and Skakkebaek, N. E.:
rats. Brain Res Dev Brain Res, 52: 17, 1990 Epidemiological and clinical aspects of carcinoma in situ of the
58. Davis, E. C., Shryne, J. E. and Gorski, R. A.: A revised critical testis. Eur Urol, 23: 104, 1993

You might also like