ArticlePDF AvailableLiterature Review

Inhibin B in male reproduction: Pathophysiology and clinical relevance

Authors:

Abstract and Figures

The recent availability of specific inhibin assays has demonstrated that inhibin B is the relevant circulating inhibin form in the human male. Inhibin B is a dimer of an alpha and a betaB subunit. It is produced exclusively by the testis, predominantly by the Sertoli cells in the prepubertal testis, while the site of production in the adult is still controversial. Inhibin B controls FSH secretion via a negative feedback mechanism. In the adult, inhibin B production depends both on FSH and on spermatogenic status, but it is not known in which way germ cells contribute to inhibin B production. The regulation of inhibin B production changes during life. There is an inhibin B peak in serum shortly after birth only partly correlated with an increase in serum FSH, probably reflecting the proliferating activity of the Sertoli cells during this phase of life. Afterwards, inhibin B levels decrease and remain low until puberty, when they rise again, first as a consequence of FSH stimulation and then as a result of the combined regulation by FSH and the ongoing spermatogenesis. In the adult, serum inhibin B shows a clear diurnal variation closely related to that of testosterone. The administration of FSH increases the secretion of inhibin B in normal men, but is much more pronounced in males with secondary hypogonadism. The treatment of infertile men with FSH, however, does not result in an unequivocal inhibin B increase. There is a clear inverse relationship between serum inhibin B and FSH in the adult. Serum inhibin B levels are strongly positively correlated with testicular volume and sperm counts. In infertile patients, inhibin B decreases and FSH increases. In general, there is very good correlation with the degree of spermatogenetic damage, with the arrest at the earlier stages having the lowest inhibin B levels. However, for unknown reasons, there are cases of Sertoli-cell-only syndrome with normal inhibin B levels. Inhibin B and FSH together are a more sensitive and specific marker for spermatogenesis than either one alone. However, the inhibin B concentrations are not a reliable predictor of the presence of sperm in biopsy samples for testicular sperm extraction. Suppression of spermatogenesis with testosterone and gestagens leads to a partial reduction of inhibin B in serum but it is never completely suppressed. In contrast, testicular irradiation in monkeys or humans leads to a rapid and dramatic decrease of inhibin B, which becomes undetectable when germ cells are completely absent. In summary, although inhibin B is a valuable index of spermatogenesis, the measurement of serum inhibin B levels is still of limited clinical relevance for individual patients.
Content may be subject to copyright.
INVITED REVIEW
Inhibin B in male reproduction: pathophysiology and clinical
relevance
S J Meachem, E Nieschlag and M Simoni
Institute for Reproductive Medicine of the University, MuÈnster, Germany
(Correspondence should be addressed to E Nieschlag, Institute for Reproductive Medicine of the University, Domagkstrasse 11, D-48129 MuÈnster,
Germany; Email: nieschl@uni-muenster.de)
(S J Meachem is now at Prince Henry's Institute of Medical Research, PO Box 5152, Clayton, Victoria 3168, Australia)
Abstract
The recent availability of speci®c inhibin assays has demonstrated that inhibin B is the relevant
circulating inhibin form in the human male. Inhibin B is a dimer of an a and a b
B
subunit. It is
produced exclusively by the testis, predominantly by the Sertoli cells in the prepubertal testis, while
the site of production in the adult is still controversial. Inhibin B controls FSH secretion via a negative
feedback mechanism. In the adult, inhibin B production depends both on FSH and on spermatogenic
status, but it is not known in which way germ cells contribute to inhibin B production. The regulation
of inhibin B production changes during life. There is an inhibin B peak in serum shortly after birth
only partly correlated with an increase in serum FSH, probably re¯ecting the proliferating activity of
the Sertoli cells during this phase of life. Afterwards, inhibin B levels decrease and remain low until
puberty, when they rise again, ®rst as a consequence of FSH stimulation and then as a result of the
combined regulation by FSH and the ongoing spermatogenesis. In the adult, serum inhibin B shows a
clear diurnal variation closely related to that of testosterone. The administration of FSH increases the
secretion of inhibin B in normal men, but is much more pronounced in males with secondary
hypogonadism. The treatment of infertile men with FSH, however, does not result in an unequivocal
inhibin B increase. There is a clear inverse relationship between serum inhibin B and FSH in the
adult. Serum inhibin B levels are strongly positively correlated with testicular volume and sperm
counts. In infertile patients, inhibin B decreases and FSH increases. In general, there is very good
correlation with the degree of spermatogenetic damage, with the arrest at the earlier stages having
the lowest inhibin B levels. However, for unknown reasons, there are cases of Sertoli-cell-only
syndrome with normal inhibin B levels. Inhibin B and FSH together are a more sensitive and speci®c
marker for spermatogenesis than either one alone. However, the inhibin B concentrations are not a
reliable predictor of the presence of sperm in biopsy samples for testicular sperm extraction.
Suppression of spermatogenesis with testosterone and gestagens leads to a partial reduction of inhibin
B in serum but it is never completely suppressed. In contrast, testicular irradiation in monkeys or
humans leads to a rapid and dramatic decrease of inhibin B, which becomes undetectable when germ
cells are completely absent. In summary, although inhibin B is a valuable index of spermatogenesis,
the measurement of serum inhibin B levels is still of limited clinical relevance for individual patients.
European Journal of Endocrinology 145 561±571
Introduction
The existence of a testicular, non-steroidal, endocrine
factor controlling pituitary function was postulated in
1932, around the time of gonadotrophin discovery and
isolation, when McCullagh (1) reported that aqueous
testicular extracts prevented the formation of `castra-
tion cells' in the rat pituitary. Moreover, McCullagh was
able to show that the selective destruction of the
seminiferous epithelium was suf®cient to cause pitui-
tary hyperfunction without inducing atrophy of the
secondary sex glands, i.e. hypoandrogenism (1). Thus,
in this early period, inhibin (latin inhibere, to suppress)
was de®ned in its essence: a non-steroidal testicular
substance, produced by the seminiferous epithelium,
speci®cally inhibiting follicle-stimulating hormone
(FSH) secretion. Decades of research have led to the
discovery and characterisation of a large family of
proteins, the inhibin family (2, 3), and to the partial
de®nition of the function of its members. However, it
was only the development of speci®c immunoassays for
biologically active, dimeric forms of inhibin that
provided new insights into our understanding of the
physiological relevance of inhibin in male reproductive
ISSN 0804-4643European Journal of Endocrinology (2001) 145 561±571
q 2001 Society of the European Journal of Endocrinology Online version via http://www.eje.org
function. Today, it is clear that inhibin B is the only
inhibin form present in male circulation. Serum inhibin
B levels re¯ect the functional state of the seminiferous
epithelium and are involved in the feedback of the
pituitary±gonadal axis, as shown by many clinical
studies investigating inhibin B in a variety of physio-
logical and pathological states. Although these studies
have provided insight into inhibin biology, many
aspects of inhibin B synthesis, secretion, function and
mechanism of action remain elusive and, sometimes,
contradictory. Perhaps not unexpectedly, inhibin B
serum levels correlate quite well with FSH concentra-
tions in the vast majority of cases and the measurement
of serum inhibin B, initially regarded as the long-
awaited tool which could render obsolete more invasive
diagnostic interventions, has not yet found unequivocal
application in clinical routine. Nevertheless, the possi-
bility of measuring serum inhibin B has enriched our
knowledge of male gonadal function. The primary aim
of this review is to discuss the recent clinical data that
have added to our understanding of inhibin patho-
physiology and the usefulness of inhibin B as a
diagnostic tool in the assessment of testicular function.
Inhibin B structure, synthesis, secretion
and action
Inhibin is a glycoprotein hormone of gonadal origin
which was ®rst identi®ed by its ability to negatively
regulate FSH (4). Inhibin consists of two covalently
linked subunits, a common a subunit and a b subunit,
the latter of which exists in two forms, A and B
(denoted b
A
and b
B
respectively). Although many
molecular forms are found in circulation, biological
activity resides only in the dimeric forms, inhibin A (a-
b
A
) and B (a-b
B
) (5).
Over the last three decades there have been
numerous studies on inhibin synthesis and secretion
in a number of in vivo and in vitro models; however,
such studies failed to demonstrate an unequivocal or
systematic association between serum inhibin levels
and spermatogenesis (6, 7). One reason for this was the
lack of speci®c and sensitive assays to measure the
variously sized inhibin forms. Levels of inhibin in blood
were measured using heterologous assays, e.g. the
Monash radioimmunoassay (6) which could not
distinguish between the bioactive, dimeric inhibin
form (a-b
A
and a-b
B
) and the inactive forms such as
free a subunit (pro-aN-aC) and its precursor (pro-aC)-
related immunoactive peptides (8, 9). However, the
production of highly speci®c antibodies and the advent
of a new two-site, enzyme-linked immunoabsorbent
assay (ELISA) for speci®c measurement of the bioactive
inhibin A and B (10) has overcome this problem and
has led to the realisation that inhibin B is the major
form produced in the foetal and adult human male,
where inhibin A is undetectable (8, 11±13). This is in
agreement with animal studies including the rhesus
monkey (14) and the rat (15).
The Sertoli cell is considered the predominant source
of inhibin B (16, 17). This notion is currently
challenged and confounded by a mounting number of
sometimes contradictory studies, based on immuno-
localisation of inhibin subunits and on the in vitro and/
or in vivo production of inhibin B in various experi-
mental settings. These studies indicate that germ cells
and possibly even Leydig cells would produce inhibin as
well (18, 19). The human male inhibin subunits seem
to be differentially localised and secreted depending on
age and cell type. For example, in the foetal testis, a-
and b
B
subunits, but not b
A
were immunolocalised in
the Sertoli and Leydig cells (20). In cultured testicular
cells from prepubertal boys, both highly puri®ed
luteinizing hormone (LH) and recombinant FSH
stimulated inhibin B secretion (21). In the adult
human testis, b
A
and b
B
subunits were found in Sertoli
and Leydig cells but not in germ cells (22). In a recent
intriguing study, positive staining for the inhibin a
subunit was evident in Sertoli cells, while b
B
subunit
was evident in germ cells from pachytene spermato-
cytes to early spermatids but not in Sertoli cells (19),
suggesting that the two subunits that constitute inhibin
B could be contributed by different cell types in adult
men. Electron microscopic evidence reports the transfer
of inhibin a subunit from Sertoli cells to spermatocytes
in the human testis (23), suggesting that inhibin
subunit dimerisation could occur in the spermatocytes.
Extracellular dimerisation may also take place (24). In
any case, it should be considered that the different
antibodies used in these studies are recognising
different epitopes which could be shared by intermedi-
ate products of the inhibin family and/or they could
identify b subunits destined to become part of activins.
The development of an activin B immunoassay, not
available at the moment, should help in resolving this
question.
If the a and the b
B
subunits localised in Leydig cells
are assembled into mature inhibin B, Leydig cells
should contribute to the levels of inhibin B in
circulation. However, administration of recombinant
LH to hypogonadal men or human chorionic gonado-
trophin to normal men is unable to raise serum inhibin
B levels, demonstrating that, in man, Leydig cells do
not contribute to the pool of circulating inhibin B (25,
26). In summary, although the contribution of germ
cells in the regulation of inhibin B secretion is no longer
subject to question (see below), Sertoli cells should still
be considered to be the main source of inhibin B in the
human male, until its assembly and secretion by other
cell types is unequivocally proved.
The route of secretion of inhibin B in the bloodstream
is still poorly understood. Inhibin B is secreted in
seminal plasma and it has been shown that Sertoli cells
secrete inhibin in a highly vectorial manner. In
cultured primate Sertoli cells .90% of inhibin is
562
S J Meachem and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
secreted into the adluminal compartment in the
presence of FSH (17). Studies in the rat have shown
that the major route via which inhibin reaches the
bloodstream is through secretion into the seminiferous
tubular ¯uid, thus supporting the idea of intratubular
production of inhibin (27). The presence of bioactive
inhibin in seminal plasma has been long recognised
(28). Indeed, one of the ®rst studies supporting the
inhibin hypothesis showed that the subcutaneous
injection of seminal plasma from normal men into
castrated rats signi®cantly inhibited serum FSH, while
seminal plasma from azoospermic men did not (29).
Seminal plasma of normozoospermic men contains
high but extremely variable levels of inhibin B (from
undetectable up to .50 000 pg/ml) while inhibin is
undetectable in vasectomised patients (13, 30). The
reason for this large variability in seminal plasma
concentrations in the presence of rather constant
serum levels is unknown, but suggests that the
transport of inhibin B into the blood might involve
some active, yet unknown mechanism.
Serum inhibin B shows a clear diurnal rhythm
parallel to that of testosterone. Individual diurnal
pro®les of serum inhibin B in 13 subjects, in whom
frequent serum samples were withdrawn continuously
for 24 h, suggested that testosterone and oestradiol
might play a role in the diurnal rhythm of inhibin B,
which is independent of FSH (31). In fact, administra-
tion of a high FSH dose (3000 IU) to healthy volunteers
results in a signi®cant increase of serum inhibin B
which, however, maintains its typical diurnal rhythm
with the lowest concentrations in the evening/night
hours (32). Since circadian testosterone ¯uctuations
are independent of LH as well, both inhibin B and
testosterone rhythms could be regulated by the same,
gonadotrophin-independent, local mechanism.
The main function of dimeric inhibin (both A and B) is
the negative control of FSH secretion (33). In the mouse,
inhibin might have an intratesticular action, e.g. in the
control of Sertoli and Leydig cell neoplastic proliferation
(34), but there is no evidence of any clinically relevant,
intratesticular inhibin action so far. The mechanism by
which inhibin B exerts its biological effect is still the
subject of intensive research. Inhibin has been shown to
bind to the activin type II receptor (ActRII) and thereby
inhibit activin action. However, inhibin does not antag-
onise activin action in all the tissues in which this
hormone acts, suggesting the existence of a speci®c
binding factor required for inhibin action. In addition,
evidence has accumulated that, besides being an activin
antagonist, inhibin has speci®c binding sites in tissues
where it is thought to act, such as the pituitary (35).
Indeed, two putative, speci®c inhibin receptors have been
isolated and molecularly characterised last year: beta-
glycan and p120 (36,37). Betaglycan, also known as type
III transforming growth factor-b receptor, functions as
inhibin co-receptor in the presence of ActRII, resulting in
enhanced antagonism of activin action (36). p120,
isolated with a classical biochemical approach from
bovine pituitaries based on its capacity to bind inhibin
A, is localised to the gonadotropes of the pituitary gland
and Leydigand Sertoli cells (37). p120,alsocalled inhibin-
binding protein, is a speci®c inhibin B receptor and, via
association with the type IB activin receptor, appears to be
necessary for inhibin B-induced antagonistic effect on
activin-stimulated signal transduction (38).
Control of inhibin B secretion
The ®rst speci®c assay for the measurement of serum
inhibin B was available in 1996 (39). After the ®rst
demonstration that inhibin B is the physiologically
relevant inhibin form in man (8), this assay was used to
study the changes in serum inhibin B levels in a
number of pathophysiological conditions, such as
maturation and senescence, infertility, hypogonadism,
gonadotrophin administration, hormonal contracep-
tion and testicular damage resulting from radiation or
chemotherapy. All these studies provided very relevant,
although sometimes contradictory, information about
inhibin physiology, explaining in part the complex
interplay between FSH, inhibin B and spermatogenesis.
It should be considered that all the data published so far
have been obtained using only one assay, which still
has a rather poor interassay precision (Fig. 1) and
cannot be properly evaluated in terms of accuracy since
internationally recognised reference preparations are
not available. Considering the extreme heterogeneity of
immunoreactive species of inhibin in the circulation,
appropriate standardisation is crucial and the avail-
ability of alternative assays would be very valuable.
Childhood and pubertal maturation
Serum inhibin B levels as well as the relationship
between FSH and inhibin B vary throughout life in the
human male, with a major switch in inhibin regulation
Figure 1 Measurement of serum inhibin B by a commercially
available ELISA. Results from the measurement of control serum
with normal inhibin B values in 111 consecutive assays are shown.
The interassay coef®cient of variation was 18.9%.
Inhibin B and male reproduction
563EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
occurring around puberty. While Sertoli cell prolifera-
tion and FSH govern inhibin activity in childhood,
germ cells are a major determinant of inhibin B
production in adulthood.
Inhibin B levels are low but detectable in cord blood
and increase into the adult range as early as in the ®rst
week after birth in male newborns (40), continue to
rise up to levels higher that those observed in adults by
3±6 months of age (30, 41, 42) and then decline
progressively to reach their nadir towards 3±6 years
(30, 42). This early postnatal increase is presumably
due to the activation of the hypothalamic±pituitary
testicular axis (41) and mirrors the proliferating
activity of Sertoli cells. Intriguingly, the early postnatal
rise of inhibin B is better correlated with LH and
testosterone than with FSH (42, 43), raising the
possibility that Sertoli cell proliferation in neonatal
life depends more on LH/testosterone than on FSH.
Inhibin B levels remain relatively high for several
months while FSH concentrations are low, suggesting
that the negative feedback control of FSH secretion
might already be established between the ®rst and
second years of age and that inhibin B production,
probably initially activated by FSH and/or by LH,
continues without gonadotrophin stimulation under
the in¯uence of yet unknown factors (41). It has been
suggested that inhibin B might be a marker of the
presence of the testis in case of sexual ambiguity with
cryptorchidism but no studies have been carried out yet
to verify this hypothesis. Inhibin B can be elevated by
recombinant FSH administration in prepubertal hypo-
gonadal boys (44).
After the initial postnatal rise, serum inhibin B levels
are low until puberty but still higher than in adults
with impaired spermatogenesis. This suggests that the
regulation of inhibin B secretion in childhood is
different from that in the adult. A critical change in
the regulation of inhibin B secretion occurs at puberty
(45) or just prior to mid puberty (44). Prepubertal boys
lacking spermatogenesis and the relevant germ cell
types secrete inhibin B in readily measureable amounts
(41, 46), whereas adult men with impaired spermato-
genesis (Sertoli-cell-only; SCO) have low to undetect-
able levels of serum inhibin B in the presence of
elevated FSH concentrations (11). Obviously, prior to
puberty, inhibin B levels are independent of the
presence of actively proliferating germ cells, while
after puberty serum inhibin B levels are closely related
to the spermatogenic status (41). During puberty, the
main control of inhibin B secretion switches from FSH
to spermatogenesis. Basal inhibin B increases under
FSH stimulation in the ®rst pubertal stages when the
last wave of Sertoli cell proliferation occurs and a
positive correlation between inhibin B and FSH is
observed. At Tanner stages G3 and G4, however, FSH
and inhibin B levels correlate negatively, suggesting
that the negative feedback regulation loop is fully
established at this stage of development (46±48).
Altogether, the existing data suggest that the stimula-
tory activity of FSH on inhibin B is the predominant
form of control as long as Sertoli cells proliferate (49±
52), although it is not known whether LH can also
in¯uence inhibin B production in childhood and
puberty. At puberty, when Sertoli cell proliferation
ceases and spermatogenesis starts, the basal, adult
inhibin B level is set and can be considered to be an
index of Sertoli cell density. Once full spermatogenesis is
ongoing, changes in inhibin B levels re¯ect mainly the
status of germ cell proliferation and development and
depend only secondarily on FSH. Therefore, the main
regulation of inhibin B production seems to change at
puberty. For instance, puberty could induce a switch in
the control of inhibin subunit expression, and it has
been postulated that testosterone might be responsible
for this maturational switch (53) in early pubertal boys
(47).
Adulthood
In the adult, FSH stimulates the production of inhibin B
in the testis and inhibin B inhibits the secretion of FSH.
Clinical ®ndings show a strong inverse correlation
between inhibin B and FSH in healthy men and in men
with testicular disorders (8, 11, 54±56). However,
unlike the LH±testosterone axis, the production of
inhibin B is not only dependent on FSH. After puberty,
in the presence of FSH, the prime regulator of inhibin B
levels is the spermatogenic status and inhibin B
production is directly proportional to the `amount' of
spermatogenesis, as shown by the direct correlation
between serum inhibin B and sperm count (57). When
spermatogenesis is damaged, as in SCO syndrome or
after testicular irradiation, inhibin B falls and FSH
increases. Under these circumstances, Sertoli cell
function might be altered as well, at least because, in
the absence of germ cells, Sertoli cells cannot ful®l their
primary function. In any case, although FSH stimulates
inhibin B secretion in normal men (11), inhibin B
production is resistant to the endogenous, elevated FSH
levels in infertile men. Therefore, FSH stimulation of
inhibin B secretion in the adult is mediated by and
necessitates the presence of germ cells. It is possible
that, in the adult, FSH stimulates the a subunit and
thereby the secretion of inhibin B, provided that
enough b
B
subunit is available: the availability of b
B
subunits for assembly into the dimeric, bioactive form
would be the limiting factor under the control of the
germ cells. In fact, the transcription of the a and b
B
subunit genes is regulated by different mechanisms
(58) and FSH stimulates the a subunit mRNA
expression but not that of the b
B
subunit (59).
Moreover, FSH stimulates pro-a-C secretion (11, 26)
and pro-a-C is elevated in infertile men while inhibin B
is low (6, 10). Expression of the gene encoding inhibin
a is upregulated through a cAMP mechansim (58, 60)
whereas the inhibin b
B
gene does not have a classical
564
S J Meachem and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
cAMP response element (61) and is not markedly
in¯uenced by hypophysectomy or FSH stimulation (62)
in the rat. Thus a decrease in the inhibin B feedback
signal results in a sustained increase in FSH secretion
which, in turn, directly stimulates inhibin a. This
hypothesis explains the lack of correlation between FSH
and inhibin originally described using the Monash
assay (6) which crossreacted with inhibin precursors
containing the a subunit.
Which germ cell type is responsible for the regulation
of inhibin B? Studies in the rat report that inhibin
secretion from Sertoli cells is regulated by interaction
with pachytene spermatocytes and early spermatids
(63, 64). However, in the primate, there is evidence to
suggest that spermatogonia may also modulate inhibin
B. Arrest of spermatogonia proliferation in irradiated
monkeys was associated with a rapid and marked
decrease of inhibin B in the presence of later germ cell
types (65) and normal inhibin B levels have been found
in a patient presenting with Reifenstein's syndrome in
whom spermatogonia were the only germ cell type
present (55). As a general rule, inhibin B concentra-
tions are progressively lower in groups of men with
increasing spermatogenic damage with undetectable
inhibin B in azoospermic men with SCO syndrome (8,
55, 66). However, in many individual cases it is not
possible to show any clear-cut relationship between
spermatogenic cell type, inhibin B and FSH (55, 66). It
might be that other, still unrecognised factor(s) take
part in the complex interplay between FSH, inhibin B
and spermatogenesis, which makes inhibin B a rather
unpredictable clinical marker for spermatogenesis in
individual patients (see below).
Clinical signi®cance of inhibin B
measurement
The demonstration that inhibin B is the relevant
inhibin form in man prompted a number of clinical
investigations aimed at the analysis of this `new'
endocrine parameter in several pathophysiological
settings. To what extent is inhibin B a reliable and
clinically useful biomarker of spermatogenesis? Does it
have any prognostic value?
Response to FSH stimulation
Is inhibin B a pharmacodynamic parameter of FSH
action, i.e. a marker of FSH-dependent Sertoli cell
function? If this is the case, it should be possible to
identify prospectively a subgroup of infertile patients
who might bene®t from FSH treatment. It has been
shown that FSH (3000 IU) induces a signi®cant
increase of serum FSH within 24 h from injection in
normal men (11). Such an increase is partially dose
dependent, and 1000 IU FSH are suf®cient to stimulate
inhibin B secretion signi®cantly (32). However, in
normal men, 225 IU recombinant FSH, i.e. a dose
capable of increasing inhibin B concentrations in
normal women (67), did not result in any appreciable
rise in serum inhibin B values (26). No acute FSH
stimulation test has been performed yet in men with
idiopathic infertility and normal or slightly elevated
FSH levels and the chronic treatment of infertile
patients yielded con¯icting results. In a placebo-
controlled study, FSH (150 IU daily for 12 weeks)
administration could not signi®cantly increase inhibin
B production in infertile men and no improvement in
semen parameters or pregnancy rate was observed
(68). On the other hand, when 11 patients with
oligozoospermia, moderate hypospermatogenesis and
normal FSH and inhibin levels were treated with FSH
(75 IU every second day for 3 months), inhibin B
increased signi®cantly in serum but sperm concentra-
tion improved only in six men (69). Therefore, in
infertile men as a group, serum inhibin B does not seem
to be of much predictive value.
Hypogonadotropic hypogonadism
A few studies have been dedicated to the study of the
effects of gonadotrophin-releasing hormone (GnRH) or
gonadotrophin treatment on inhibin B secretion in
hypogonadotropic hypogonadism. Serum inhibin B
levels are in the prepubertal range in men with isolated
GnRH de®ciency and increase signi®cantly into the
normal adult range during pulsatile treatment (56,
70). Similar to that observed during physiological
puberty, the negative feedback relationship between
inhibin B and FSH is evident during treatment as
demonstrated by the establishment of a negative
correlation between these two parameters (56). Long-
term GnRH treatment does not further increase inhibin
B levels, again demonstrating that, beside FSH, local
testicular factors are involved in the regulation of
inhibin B secretion (70). A recent study demonstrated
that the gonadotrophin effect on inhibin B secretion is
entirely due to FSH and that Leydig cells do not
contribute to the pool of circulating hormone. Recom-
binant FSH administration (150 IU/day for 1 month)
to patients with acquired hypogonadotropic hypogo-
nadism was able to increase serum inhibin B levels
progressively, while recombinant LH had no effect (25).
Together with the data on FSH stimulation in normal
men (11, 32), these results show that FSH and not LH
stimulates inhibin B production. However, when
spermatogenesis is normal, large supraphysiological
doses of FSH are necessary in order to raise the inhibin
B values further (11, 26, 32).
Male infertility
To date the only predictors of the state of spermato-
genesis are semen analysis, the measurement of FSH in
blood and testicular histology. The diagnostic accuracy
of FSH is limited by the fact that spermatogenic arrest
Inhibin B and male reproduction 565EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
at late stages does not lead to changes in FSH secretion
and that FSH may be normal even in patients with focal
SCO or hypospermatogenesis (71). In fact, serum FSH is
not an absolute parameter for the selection of
azoospermic men as candidates for testicular sperm
extraction (TESE) in assisted reproduction (72, 73).
Testis biopsy is an invasive procedure associated with
potential complications (74). In addition, a biopsy may
not be representative for the whole testis (75). Multiple
biopsies which are performed for TESE often show a
large variation in the completeness of spermatogenesis.
This heterogeneity is even more conspicuous in
patients with impaired spermatogenesis, where sections
with complete spermatogenesis may be found along
with others with focal spermatogenesis (71). Given
these limitations, any demonstration that inhibin B can
discriminate between complete absence of germ cells in
the testis and less severe disturbances of sperm
production would be of considerable clinical value.
Indeed, it has been shown that inhibin B serum levels
represent a good marker of spermatogenesis which
correlates with FSH, testis volume, sperm counts and
the presence of SCO tubules (30, 41, 54±57, 69, 76±
78). In particular, when used in combination with FSH,
inhibin B is a more sensitive marker than FSH alone
(55, 77). However, inhibin B, alone or in combination
with FSH, cannot predict the presence of sperm in
individual testicular tissue samples (55). Moreover,
inhibin B cannot predict accurately the type of
spermatogenic damage. For instance, in cases of late
spermatogenic arrest, inhibin B concentrations may
remain normal, exactly like FSH (66). In addition, in a
considerable number of cases of non-obstructive
azoospermia with focal SCO, both inhibin B and FSH
are in the normal range (55, 66). Therefore, despite the
fact that the combination of inhibin B and FSH
represent a more sensitive predictor of the spermato-
genic state than either of them alone, inhibin B has not
dramatically improved the diagnostic and prognostic
value of FSH for individual patients.
This has important implications for TESE. We have
investigated the diagnostic sensitivity of inhibin B in
successful retrieval of sperm in 52 TESE patients.
Although the combination of serum FSH and inhibin B
measurements showed high diagnostic sensitivity
(75%) and speci®city (73%) in identifying patients in
whom sperm could be possibly retrieved, no sperm
could be retrieved in 25% of the cases with normal FSH
and normal inhibin B. On the other hand, sperm could
be extracted in 38% of the cases with inhibin B values
,20 pg/ml. Therefore, inhibin B alone or in combina-
tion with FSH cannot be used to decide if a patient
should undergo TESE (55). At odds with our observa-
tion, a recent study has suggested a cut-off of 40 pg/ml
as fully predictive of the presence of sperm upon TESE
(79). However, the low number of subjects analysed (17
vs 52 in our study) and the high variability of the
current inhibin B assay, especially at such low levels,
recommend caution. Many reports have suggested that
recovery of testicular spermatozoa may be possible in
.50% of cases of non-obstructive azoospermia regard-
less of clinical parameters such as testicular size or
serum FSH (73, 75, 79, 80) and that the success rate is
increased by collecting more tissue (81). Even if inhibin
B is useful, the clinical decision as to whether TESE
should be performed or not cannot be based on inhibin
B, either alone, or in combination with FSH and other
parameters.
Radiation and antineoplastic therapy
Elimination of the seminiferous epithelium through
radiation is followed by an increase in serum FSH
levels. An experimental study in cynomolgus monkeys
has recently shown that radiation is immediately
followed by a sharp drop in serum inhibin B values
which occurs well before the decrease in testis volume
and semen count and the increase in serum FSH (65).
Therefore, in this experimental model, inhibin B is an
early marker of testicular injury, most probably an
index of premeiotic germ cell proliferation. This
experimental observation in the monkey, however,
has not yet been followed by similar investigations in
the human. It is well known that even before
orchiectomy patients with testicular cancer have
impaired spermatogenesis (82) which deteriorates
even further after surgery (83). In this clinical setting,
inhibin B levels also correlate quite well with the
spermatogenic status and undetectable inhibin B levels
are associated with an absence of spermatogenic
activity (84). Similarly, treatment with radioiodine for
thyroid carcinoma (85) or chemotherapy (12, 86) are
followed by a decrease in spermatogenesis which is
re¯ected by decreased inhibin B and increased FSH
levels. Whether inhibin B is a more sensitive and
precocious parameter of the damage of the semini-
ferous epithelium than FSH has not been investigated.
However, on the basis of the results obtained in
monkeys after irradiation (65) or hemiorchiectomy
(87) it would seem that inhibin B could be useful as an
early marker of testicular toxicants.
Ageing
Few data are available on spermatogenesis in older
men, particulary in men over 60. Controlled studies
suggest that semen parameters do not necessarily
deteriorate as an effect of ageing in healthy men (88).
Recent studies have shown only a moderate decrease in
serum inhibin B in old men (30, 89) but, in the absence
of semen analysis, this decrease cannot be attributed to
the ageing process per se. Rather, similar to all other
clinical conditions examined until now, a decrease in
serum inhibin B could re¯ect a decrease in spermato-
genesis for whatever reason (90).
566
S J Meachem and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
Hormonal male contraception
Current regimens of hormonal male contraception are
based on administration of testosterone esters possibly
combined with a gestagen. This treatment leads to
rapid suppression of serum gonadotrophins and vari-
able suppression of spermatogenesis with azoospermia
being induced in about 60±90% of the Caucasian
population, mainly depending on the type of gestagen.
Some studies have investigated the concentrations of
inhibin B in serum (11, 91±94) and in seminal plasma
(94) in men treated with hormonal contraceptives.
However, the contradictory results of such studies are
puzzling. The administration of testosterone enanthate
(TE) for 65 weeks was followed by a rapid and
progressive decline in serum inhibin B concentrations
which continued until the end of the treatment (92). A
similar, signi®cant decline was already observed in
another study of 19±24 weeks of TE administration
(91). The treatment with TE plus levonorgestrel for 6
months was followed by a signi®cant decline of serum
inhibin B in one study (11), while, in our experience,
transdermal testosterone plus levonorgesterel for 22
weeks was not followed by a signi®cant decline in inhibin
B (93). Finally, treatment with testosterone pellets plus
different doses of desogestrel for 8 weeks did not result in
any signi®cant change of inhibin B in serum, while the
hormone was dose-dependently suppressed down to
undetectable levels in seminal plasma (94). Interestingly,
in the latter study, serum pro-a-C levels were signi®-
cantly decreased, again supporting the gonadotrophin
dependency of this parameter. In clinical trials of male
contraception based on testosterone undecanoate and
levonorgestrel or norethitestosterone enanthate (95,
96), we could not ®nd any signi®cant decrease in serum
inhibin B concentrations, despite a profound suppression
of gonadotrophins and spermatogenesis (Fig. 2). These
contradictory results are dif®cult to reconcile, but
differences in the duration of treatment, in the type of
gestagens and in their mode of administration might
play some role. Treatment with TE alone has been
reported to affect mainly type B spermatogonia which
stop proliferating as a result of gonadotrophin suppres-
sion (91). If spermatogonial proliferation is the main
determinant of inhibin b
B
production, a decline in
inhibin B has to be expected. The situation might be
different if a gestagen is added, which could have some
direct effect on inhibin b
B
transcription and/or might
lead to a spermatogenic arrest at later stages, not directly
in¯uencing inhibin B production. The effects of long-
term treatment (beyond 1 year) are not known. In any
case, the original hope of distinguishing between
responders and non-responders to hormonal contra-
ceptive regimens based on inhibin B determination
remains unful®lled and the dissociation between serum
inhibin B concentrations and sperm count in many
studies remains an enigma.
Conclusion
The development of a speci®c and sensitive assay for the
measurement of inhibin B has led to signi®cant
breakthroughs in our understanding of inhibin B
biology and control of FSH secretion (97). Yet its
clinical usefulness is rather disappointing and the
measurement of this hormone in clinical samples does
not signi®cantly reinforce the diagnostic arsenal of the
clinical andrologist. Although FSH and inhibin B
together are more sensitive than either one alone in
Figure 2 Inhibin B levels in the serum of male volunteers undergoing
trials of hormonal male contraception. Upper panel: volunteers
n 6 were treated with the GnRH antagonist Cetrorelix (dose
10 mg/day for 5 days, followed by 2 mg/day up to week 12) for 12
weeks and 19-nortestosterone (400 mg in week 1, followed by
200 mg every 3 weeks) for 28 weeks (95). Lower panel: volunteers
received either testosterone undecanaote (1000 mg every 6 weeks)
and placebo (B) or testosterone undecanaote plus norethisterone
enanthate (200 mg every 6 weeks) (A) (96). The arrows indicate the
duration of the treatment. Results are expressed as means^
S.D. No
signi®cant change in serum inhibin B levels could be detected over
the treatment and observation periods.
Inhibin B and male reproduction
567EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
predicting the histological status of the testis and/or the
presence of sperm in bioptic tissue, this parameter gives
no absolute certainty. Concerning diagnostic and
therapeutic recommendations for the individual
patient, inhibin B measurement does not really add
much to FSH values, semen analysis and clinical
evaluation of the testis. On the contrary, inhibin B
values are sometimes at odds with the other parameters
and can generate interpretative uncertainty. Moreover,
it is still unclear whether inhibin B can be used as a
functional parameter of Sertoli cells. Finally, inhibin B
is not predictive of FSH responsiveness in idiopathic
infertility and cannot discriminate between responders
and non-responders in trials for hormonal male
contraception. Thus, inhibin B is not an indispensable
diagnostic tool in clinical practice.
On the other hand, the measurement of inhibin B is
very useful in experimental studies and can still add
important information to our knowledge of testicular
function and regulation of the pituitary±gonadal axis
in several pathophysiological conditions. For instance,
inhibin B seems to be a very fast marker of testicular
damage and could become important for rapid
identi®cation of spermatogenic disorders in populations
exposed to testicular toxicants. Moreover, many enig-
matic aspects of inhibin B physiology remain to be
clari®ed and deserve further investigation. Finally,
methodological aspects should not be disregarded. We
should keep in mind that the totality of the data on
which our current concept is based has been generated
using only one and the same assay kit. Several decades
of experience with other protein hormone assays
suggest that standardisation and availability and
marketing of new, competing assay kits could hold
some surprises in store.
Acknowledgements
S J M was a recipient of a Wellcome Trust Fellowship. Our
own work reported here was in part supported by the
Deutsche Forschungsgemeinschaft Confocal Research
Group: `The Male Gamete: Production, Maturation,
Function' (Ni 130/15). The language editing of the
manuscript by S Nieschlag is gratefully acknowledged.
References
1 McCullagh DR. Dual endocrine activity of the testes. Science 1932
76 19±20.
2 Robertson DM, Foulds LM, Leversha L, Morgan FJ, Hearn MT,
Burger HG et al. Isolation of inhibin from bovine follicular ¯uid.
Biochemical and Biophysical Research Communications 1985 126
220±226.
3 Ling N, Ying SY, Ueno N, Esch F, Denoroy L & Guillermin R.
Isolation and partial characterisation of a Mr 32 000 protein
with inhibin activity from porcine follicular ¯uid. PNAS 1985 82
7217±7221.
4 Burger HG & Igarashi M. Inhibin: de®nition and nomenclature,
including related substances. Endocrinology 1988 122 1701±
1702.
5 Burger HG. Clinical review 46: Clinical utility of inhibin
measurements. Journal of Clinical Endocrinology and Metabolism
1993 76 1391±1396.
6 de Kretser DM, McLachlan RI, Robertson DM & Burger HG.
Serum inhibin levels in normal men and men with testicular
disorders. Journal of Endocrinology 1989 120 517±523.
7 de Kretser DM & McFarlane JR. Inhibin in the male. Journal of
Andrology 1996 17 179±182.
8 Illingworth PJ, Groome NP, Byrd W, Rainey WE, McNeilly AS,
Mather JP et al. Inhibin-B: a likely candidate for the physiologi-
cally important form of inhibin in men. Journal of Clinical
Endocrinology and Metabolism 1996 81 1321±1325.
9 Bhasin S & de Kretser DM. Measurement of circulating inhibin
levels: revisiting the inhibin hypothesis. Journal of Clinical
Endocrinology and Metabolism 1996 81 1318±1320.
10 Burger HG & Robertson DM. Editorial: inhibin in the male ±
progress at last. Endocrinology 1997 138 1361±1362.
11 Anawalt BD, Bebb RA, Matsumoto AM, Groome NP,
Illingworth PJ, McNeilly AS et al. Serum inhibin B levels re¯ect
Sertoli cell function in normal men and men with testicular
dysfunction. Journal of Clinical Endocrinology and Metabolism
1996 81 3341±3345.
12 Wallace EM, Groome NP, Riley SC, Parker AC & Wu FC. Effects of
chemotherapy-induced testicular damage on inhibin, gonado-
tropin, and testosterone secretion: a prospective longitudinal
study. Journal of Clinical Endocrinology and Metabolism 1997 82
3111±3115.
13 Anderson RA, Irvine DS, Balfour C, Groome NP & Riley SC.
Inhibin B in seminal plasma: testicular origin and relationship to
spermatogenesis. Human Reproduction 1998 13 920±926.
14 Plant TM, Padmanabhan V, Ramaswamy S, McConnell DS,
Winters SJ, Groome N et al. Circulating concentrations of dimeric
inhibin A and B in the male rhesus monkey (Macaca mulatta).
Journal of Clinical Endocrinology and Metabolism 1997 82 2617±
2621.
15 Woodruff TK, Besecke LM, Groome N, Draper LB, Schwartz NB &
Weiss J. Inhibin A and inhibin B are inversely correlated to
follicle-stimulating hormone, yet are discordant during the
follicular phase of the rat estrous cycle, and inhibin A is
expressed in a sexually dimorphic manner. Endocrinology 1996
137 5463±5467.
16 Grootenhuis AJ, Timmerman MA, Hordijk PL & de Jong FH.
Inhibin in immature rat Sertoli cell conditioned medium: a
32 kDa alpha beta-B dimer. Molecular and Cellular Endocrinology
1990 70 109±116.
17 Handelsman DJ, Spaliviero JA & Phippard AF. Highly vectorial
secretion of inhibin by primate Sertoli cells in vitro. Journal of
Clinical Endocrinology and Metabolism 1990 71 1235±1238.
18 Risbridger GP, Clements J, Robertson DM, Drummond AE, Muir J,
Burger HG et al. Immuno- and bioactive inhibin and inhibin
alpha-subunit expression in rat Leydig cell cultures. Molecular
and Cellular Endocrinology 1989 66 119±122.
19 Andersson AM, Muller J & Skakkebaek NE. Different roles of
prepubertal and postpubertal germ cells and Sertoli cells in the
regulation of serum inhibin B levels. Journal of Clinical
Endocrinology and Metabolism 1998 83 4451±4458.
20 Majdic G, McNeilly AS, Sharpe RM, Evans LR, Groome NP &
Saunders PT. Testicular expression of inhibin and activin
subunits and follistatin in the rat and human fetus and neonate
and during postnatal development in the rat. Endocrinology 1997
138 2136±2147.
21 Berensztein E, Saraco N, Belgorosky A & Rivarola MA. Secretion
of inhibin B by human prepubertal testicular cells in culture.
European Journal of Endocrinology 2000 142 481±485.
22 Vliegen MK, Schlatt S, Weinbauer GF, Bergmann M, Groome NP
& Nieschlag E. Localization of inhibin/activin subunits in the
testis of adult nonhuman primates and men. Cell and Tissue
Research 1993 273 261±268.
23 Forti G, Vannelli GB, Barni T, Balboni GC, Orlando C &
Serio M. Sertoli-germ cells interactions in the human testis.
568 S J Meachem and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
Journal of Steroid Biochemistry and Molecular Biology 1992 43
419±422.
24 Mason AJ, Farnworth PG & Sullivan J. Characterization and
determination of the biological activities of noncleavable high
molecular weight forms of inhibin A and activin A. Molecular
Endocrinology 1996 10 1055±1065.
25 Young J, Couzinet B, Chanson P, Brailly S, Loumaye E &
Schaison G. Effects of human recombinant luteinizing hormone
and follicle-stimulating hormone in patients with acquired
hypogonadotropic hypogonadism: study of Sertoli and Leydig
cell secretions and interactions. Journal of Clinical Endocrinology
and Metabolism 2000 85 3239±3244.
26 Kinniburgh D & Andersen RA. Differential patterns of inhibin
secretion in response to gonadotrophins stimulation in normal
men. International Journal of Andrology 2001 24 95±101.
27 Maddocks S & Sharpe RM. The route of secretion of inhibin from
the rat testis. Journal of Endocrinology 1989 120 R5±R8.
28 Scott RS & Burger HG. An inverse relationship exists between
seminal plasma inhibin and serum follicle-stimulating hormone
in man. Journal of Clinical Endocrinology and Metabolism 1981 52
796±803.
29 Franchimont P, Millet D, Vendrely E, Letawe J, Legros JJ &
Netter A. Relationship between spermatogenesis and serum
gonadotropin levels in azoospermia and oligospermia. Journal of
Clinical Endocrinology and Metabolism 1972 34 1003±1008.
30 Byrd W, Bennett MJ, Carr BR, Dong Y, Wians F & Rainey W.
Regulation of biologically active dimeric inhibin A and B from
infancy to adulthood in the male. Journal of Clinical Endocrinology
and Metabolism 1998 83 2849±2854.
31 Carlsen E, Olsson C, Petersen JH, Andersson AM & Skakkebaek NE.
Diurnal rhythm in serum levels of inhibin B in normal men:
relation to testicular steroids and gonadotropins. Journal of Clinical
Endocrinology and Metabolism 1999 84 1664±1669.
32 Kamischke A, Simoni M, Schrameyer K & Nieschlag E. Is inhibin
B pharmacodynamic parameter for FSH in normal men?
European Journal of Endocrinology 2001 144 629±637.
33 de Kretser DM, Meinhardt A, Meehan T, Phillips DJ, O'Bryan MK
& Loveland KA. The roles of inhibin and related peptides in
gonadal function. Molecular and Cellular Endocrinology 2000 161
43±46.
34 Matzuk MM, Finegold MJ, Mishina Y, Bradley A & Behringer RR.
Synergistic effects of inhibins and Mu
È
llerian-inhibiting substance
on testicular tumorigenesis. Molecular Endocrinology 1995 9
1337±1345.
35 Robertson DM, Hertan R & Farnworth PG. Is the action of
inhibin mediated via a unique receptor? Reviews in Reproduction
2000 5 131±135.
36 Lewis KA, Gray PC, Blount AL, MacConnel LA, Wiater E,
Bilezikjian LM et al. Bataglycan binds inhibin and can mediate
functional antagonism of activin signalling. Nature 2000 404
411±414.
37 Chong H, Pangas SA, Bernard DJ, Wang E, Gitch J, Chen W et al.
Structure and expression of a membrane component of the
inhibin receptor system. Endocrinology 2000 141 2600±2607.
38 Chapman SC & Woodruff TK. Modulation of activin signal
transduction by inhibin B and inhibin-binding protein (InhBP).
Molecular Endocrinology 2001 15 666±679.
39 Groome NP, Illingworth PJ, O'Brien M, Pai R, Rodger FE,
Mather JP et al. Measurement of dimeric inhibin B throughout
the human menstrual cycle. Journal of Clinical Endocrinology and
Metabolism 1996 81 1401±1405.
40 De Scheppers J, Verlinde F, Cortvrindt R, Callewaert M & Smitz J.
Serum inhibin B in normal term-born male and female neonates
during the ®rst week of life. European Journal of Pediatrics 2000
159 465±469.
41 Andersson AM, Toppari J, Haavisto AM, Petersen JH, Simell T,
Simell O et al. Longitudinal reproductive hormone pro®les in
infants: peak of inhibin B levels in infant boys exceeds levels in
adult men. Journal of Clinical Endocrinology and Metabolism 1998
83 675±681.
42 Bergada
Â
I, Rojas G, Ropelato G, Ayuso S, Bergada
Â
C & Campo S.
Sexual dimorphism in circulating monomeric and dimeric
inhibins in normal boys and girls from birth to puberty. Clinical
Endocrinology 1999 51 455±460.
43 Borgato S, Giacchero R, Morpurgo P, Persani L & Beck-Peccoz P.
Physiological secretion of gonadotropins and inhibin B during
the ®rs year of postnatal life in both sexes. 5th European Congress
of Endocrinology, Turin, Italy, 2001, Abstract O-079.
44 Raivio T, Toppari J, Perheentupa A, McNeilly AS & Dunkel L.
Treatment of prepubertal gonadotrophin-de®cient boys with
recombinant human follicle-stimulating hormone. Lancet 1997
350 263±264.
45 Crofton PM, Illingworth PJ, Groome NP, Stirling HF, Swanston I,
Gow S et al. Changes in dimeric inhibin A and B during normal
early puberty in boys and girls. Clinical Endocrinology 1997 46
109±114.
46 Andersson AM, Juul A, Petersen JH, Muller J, Groome NP &
Skakkebaek NE. Serum inhibin B in healthy pubertal and
adolescent boys: relation to age, stage of puberty, and follicle-
stimulating hormone, luteinizing hormone, testosterone, and
estradiol levels. Journal of Clinical Endocrinology and Metabolism
1997 82 3976±3981.
47 Raivio T & Dunkel L. Inverse relationship between serum inhibin
B and FSH levels in prepubertal boys with cryptorchidism.
Pediatric Research 1999 46 496±500.
48 Raivio T, Saukkonen S, Jaaskelainen J, Komulainen J & Dunkel L.
Signaling between the pituitary gland and the testes: inverse
relationship between serum FSH and inhibin B concentrations in
boys in early puberty. European Journal of Endocrinology 2000
142 150±156.
49 Cortes D, Muller J & Skakkebaek NE. Proliferation of Sertoli
cells during development of the human testis assessed by
stereological methods. International Journal of Andrology 1987
10 589±596.
50 Marshall GR & Plant TM. Puberty occurring either sponta-
neously or induced precociously in rhesus monkey (Macaca
mulatta) is associated with a marked proliferation of Sertoli cells.
Biology of Reproduction 1996 54 1192±1199.
51 Sharpe RM, Turner KJ, McKinnell C, Groome NP, Atanassova N,
Millar MR et al. Inhibin B levels in plasma of the male rat from
birth to adulthood: effect of experimental manipulation of Sertoli
cell number. Journal of Andrology 1999 20 94±101.
52 Ramaswamy S, Marshall GR, McNeilly AS & Plant TM. Evidence
that in a physiological setting Sertoli cell number is the major
determinant of circulating concentrations of inhibin B in the
adult male rhesus monkey (Macaca mulatta). Journal of Andrology
1999 20 430±434.
53 Gromoll J, Eiholzer U, Nieschlag E & Simoni M. Male hypogonad-
ism caused by homozygous deletion of exon 10 of the luteinizing
hormone (LH) receptor: differential action of human chorionic
gonadotropin and LH. Journal of Clinical Endocrinology and
Metabolism 2000 85 2281±2286.
54 Leifke E, Simoni M, Kamischke A, Gromoll J, Bergmann M &
Nieschlag E. Does the gonadotrophic axis play a role in the
pathogenesis of Sertoli-cell-only syndrome? International Journal
of Andrology 1997 20 29±36.
55 von Eckardstein S, Simoni M, Bergmann M, Weinbauer GF,
Gassner P, Schepers AG et al. Serum inhibin B in combination
with serum follicle-stimulating hormone (FSH) is a more
sensitive marker than serum FSH alone for impaired spermato-
genesis in men, but cannot predict the presence of sperm in
testicular tissue samples. Journal of Clinical Endocrinology and
Metabolism 1999 84 2496±2501.
56 Seminara SB, Boepple PA, Nachtigall LB, Pralong FP, Khoury RH,
Sluss PM et al. Inhibin B in males with gonadotropin-releasing
hormone (GnRH) de®ciency: changes in serum concentration
after short term physiologic GnRH replacement. A clinical
research center study. Journal of Clinical Endocrinology and
Metabolism 1996 81 3692±3696.
Inhibin B and male reproduction
569EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
57 Pierik FH, Vreeburg JT, Stijnen T, De Jong FH & Weber RF. Serum
inhibin B as a marker of spermatogenesis. Journal of Clinical
Endocrinology and Metabolism 1998 83 3110±3114.
58 Feng ZM, Li YP & Chen CL. Analysis of the 5
0
-¯anking regions of
rat inhibin alpha- and beta-B-subunit genes suggests two
different regulatory mechanisms. Molecular Endocrinology 1989
3 1914±1925.
59 Namiki M, Kondoh N, Matsumiya K, Sakoda S, Nishimune Y &
Okuyama A. Hormonal regulation of human testicular inhibin
alpha and beta B subunit messenger RNAS. Journal of Urology
1993 150 1007±1009.
60 Eramaa M, Heikinheimo K & Voutilainen R. Developmental and
cyclic adenosine 3
0
,5
0
monophosphate-dependent regulation of
inhibin subunit messenger ribonucleic acids in human fetal
testes. Journal of Clinical Endocrinology and Metabolism 1992 75
806±811.
61 Dykema JC & Mayo KE. Two messenger ribonucleic acids
encoding the common beta B-chain of inhibin and activin have
distinct 5
0
-initiation sites and are differentially regulated in rat
granulosa cells. Endocrinology 1994 135 702±711.
62 Krummen LA, Toppari J, Kim WH, Morelos BS, Ahmad N,
Swerdloff RS et al. Regulation of testicular inhibin subunit
messenger ribonucleic acid levels in vivo: effects of hypophysect-
omy and selective follicle-stimulating hormone replacement.
Endocrinology 1989 125 1630±1637.
63 Pineau C, Sharpe RM, Saunders PT, Gerard N & Jegou B.
Regulation of Sertoli cell inhibin production and of inhibin
alpha-subunit mRNA levels by speci®c germ cell types. Molecular
and Cellular Endocrinology 1990 72 13±22.
64 Allenby G, Foster PM & Sharpe RM. Evidence that secretion of
immunoactive inhibin by seminiferous tubules from the adult rat
testis is regulated by speci®c germ cell types: correlation between
in vivo and in vitro studies. Endocrinology 1991 128 467±476.
65 Foppiani L, Schlatt S, Simoni M, Weinbauer GF, Hacker-Klom U &
Nieschlag E. Inhibin B is a more sensitive marker of spermato-
genetic damage than FSH in the irradiated non-human primate
model. Journal of Endocrinology 1999 162 393±400.
66 Foresta C, Bettella A, Petraglia F, Pistorello M, Luisi S &
Rossato M. Inhibin B levels in azoospermic subjects with
cytologically characterized testicular pathology. Clinical Endo-
crinology 1999 50 695±701.
67 Burger HG, Groome NP & Roberston DM. Both inhibin A and B
respond to exogenous follicle-stimulating hormone in the
follicular phase of the human menstrual cycle. Journal of Clinical
Endocrinology and Metabolism 1998 83 4167±4169.
68 Kamischke A, Behre HM, Bergmann M, Simoni M, SchaÈfer T &
Nieschlag E. Recombinant human FSH for treatment of male
idiopathic infertility: a randomized, double-blind, placebo-con-
trolled, clinical trial. Human Reproduction 1998 13 596±603.
69 Foresta C, Bettella A, Rossato M, La Sala G, De Paoli M &
Plebani M. Inhibin B plasma concentrations in oligozoospermic
subjects before and after therapy with follicle stimulating
hormone. Human Reproduction 1999 14 906±912.
70 Nachtigall LB, Boepple PA, Seminara SB, Khoury RH, Sluss PM,
Lecain AE et al. Inhibin B secretion in males with gonadotropin-
releasing hormone (GnRH) de®ciency before and during long-
term GnRH replacement: relationship to spontaneous puberty,
testicular volume, and prior treatment ± a clinical research
center study. Journal of Clinical Endocrinology and Metabolism
1996 81 3520±3525.
71 Bergmann M, Behre HM & Nieschlag E. Serum FSH and
testicular morphology in male infertility. Clinical Endocrinology
1994 40 133±136.
72 Chen CS, Chu SH, Lai YM, Wang ML & Chan PR. Reconsideration of
testicular biopsy and follicle-stimulating hormone measurement in
the era of intracytoplasmic sperm injection for non-obstructive
azoospermia? Human Reproduction 1996 11 2176±2179.
73 Ezeh UI, Moore HD & Cooke ID. Correlation of testicular sperm
extraction with morphological, biophysical and endocrine
pro®les in men with azoospermia due to primary gonadal failure.
Human Reproduction 1998 13 3066±3074.
74 Schlegel PN & Su LM. Physiological consequences of
testicular sperm extraction. Human Reproduction 1997 12
1688±1692.
75 Tournaye H, Verheyen G, Nagy P, Ubaldi F, Goossens A, Silber S
et al. Are there any predictive factors for successful testicular
sperm recovery in azoospermic patients? Human Reproduction
1997 12 80±86.
76 Mahmoud AM, Comhaire FH & Depuydt CE. The clinical and
biologic signi®cance of serum inhibins in subfertile men.
Reproduction and Toxicology 1998 12 591±599.
77 Bohring C & Krause W. Serum levels of inhibin B in men with
different causes of spermatogenic failure. Andrologia 1999 31
137±141.
78 Jensen TK, Andersson AM, Hjollund NH, Scheike T, Kolstad H,
Giwercman A et al. Inhibin B as a serum marker of
spermatogenesis: correlation to differences in sperm concentra-
tion and follicle-stimulating hormone levels. A study of 349
Danish men. Journal of Clinical Endocrinology and Metabolism
1997 82 4059±4063.
79 Ballesca
Á
JL, Balasch J, Calafell JM, Alvarez R, Fabregues F, de
Osaba MJ et al. Serum inhibin B determination is predictive of
successful testicular sperm extraction in men with non-
obstructive azoospermia. Human Reproduction 2000 15 1734±
1738.
80 Cha KY, Oum KB & Kim HJ. Approaches for obtaining sperm in
patients with male factor infertility. Fertility and Sterility 1997 67
985±995.
81 Ezeh UI, Moore HD & Cooke ID. A prospective study of multiple
needle biopsies versus a single open biopsy for testicular sperm
extraction in men with non-obstructive azoospermia. Human
Reproduction 1998 13 3075±3080.
82 Petersen PM, Skakkebaek NE, Vistisen K, Rorth M &
Giwercman A. Semen quality and reproductive hormones before
orchiectomy in men with testicular cancer. Journal of Clinical
Oncology 1999 17 941±947.
83 Petersen PM, Skakkebaek NE, Rorth M & Giwercman A. Semen
quality and reproductive hormones before and after orchiectomy
in men with testicular cancer. Journal of Urology 1999 161 822±
826.
84 Petersen PM, Andersson AM, Rorth M, Daugaard G &
Skakkebaek NE. Undetectable inhibin B serum levels in men
after testicular irradiation. Journal of Clinical Endocrinology and
Metabolism 1999 84 213±215.
85 Wichers M, Benz E, Palmedo H, Biersack HJ, Grunwald F &
Klingmu
È
ller D. Testicular function after radioiodine therapy for
thyroid carcinoma. European Journal of Nuclear Medicine 2000 27
503±507.
86 Grigg AP, McLachlan R, Zaja J & Szer J. Reproductive status in
long-term bone marrow transplant survivors receiving busulfan-
cyclophosphamide (120 mg/kg). Bone Marrow Transplantation
2000 26 1089±1095.
87 Ramaswamy S, Marshall GR, McNeilly AS & Plant TM. Dynamics
of the follicle-stimulating hormone (FSH)±inhibin B feedback
loop and its role in regulating spermatogenesis in the adult male
rhesus monkey (Macaca mulatta) as revealed by unilateral
orchidectomy. Endocrinology 2000 141 18±27.
88 Nieschlag E, Lammers U, Freischem CW, Langer K & Wickings EJ.
Reproductive functions in young fathers and grandfathers.
Journal of Clinical Endocrinology and Metabolism 1982 55 676±
681.
89 Mahmoud AM, Goemaere S, De Bacquer D, Comhaire FH
& Kaufman JM. Serum inhibin B levels in community-
dwelling elderly men. Clinical Endocrinology 2000 53 141±
147.
90 Andersen AG, Jensen TK, Carlsen E, Jorgensen N, Andersson AM,
Krarup T et al. High frequency of sub-optimal semen quality in
an unselected population of young men. Human Reproduction
2000 15 366±372.
570 S J Meachem and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
91 Zhengwei Y, Wreford NG, Royce P, de Kretser DM &
McLachlan RI. Stereological evaluation of human spermato-
genesis after suppression by testosterone treatment: hetero-
geneous pattern of spermatogenic impairment. Journal of Clinical
Endocrinology and Metabolism 1998 83 1284±1291.
92 Anderson RA, Wallace EM, Groome NP, Bellis AJ & Wu FC.
Physiological relationships between inhibin B, follicle stimulating
hormone secretion and spermatogenesis in normal men and
response to gonadotrophin suppression by exogenous testoster-
one. Human Reproduction 1997 12 746±751.
93 Bu
È
chter D, von Eckardstein S, von Eckardstein A, Kamischke A,
Simoni M, Behre HM et al. Clinical trial of transdermal
testosterone and oral levonorgestrel for male contraception.
Journal of Clinical Endocrinology and Metabolism 1999 84 1244±
1249.
94 Martin CW, Riley SC, Everington D, Groome NP, Riemersma RA,
Baird DT et al. Dose-®nding study of oral desogestrel with
testosterone pellets for suppression of the pituitary±testicular
axis in normal men. Human Reproduction 2000 15 1515±1524.
95 Behre HM, Kliesch S, Lemke B, von Eckardstein S & Nieschlag E.
Suppression of spermatogenesis to azoospermia by combined
administration of GnRH antagonist and 19-nortestosterone
cannot be maintained by this non-aromatizable androgen
alone. Human Reproduction 2001 (In Press).
96 Kamischke A, Venherm S, PloÈger D, von Eckardstein S &
Nieschlag E. Intramuscular testosterone undecanoate and
noresthisterone enanthate in a clinical trial for male contra-
ception. Journal of Clinical Endocrinology and Metabolism 2001 86
303±309.
97 Anderson RA & Sharpe RM. Regulation of inhibin production in
the human male and its clinical applications. International Journal
of Andrology 2000 23 136±144.
Received 27 March 2001
Accepted 19 July 2001
Inhibin B and male reproduction
571EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 145
www.eje.org
... Recent advancements in specific inhibin assays have unveiled that circulating inhibin B can have a role over MFI diagnostic work-up. Inhibin B is exclusively produced by the testis, primarily by Sertoli cells in prepubertal testes, while its production in adults remains controversial [86]. Inhibin B regulates FSH secretion through negative feedback [86]. ...
... Inhibin B is exclusively produced by the testis, primarily by Sertoli cells in prepubertal testes, while its production in adults remains controversial [86]. Inhibin B regulates FSH secretion through negative feedback [86]. During life, inhibin B production changes, with peaks after birth and during puberty, influenced by FSH and spermatogenic status. ...
... During life, inhibin B production changes, with peaks after birth and during puberty, influenced by FSH and spermatogenic status. In general, serum inhibin B levels are positively correlated with TV and sperm counts, and they decrease in infertile patients [86][87][88][89]. Inhibin B levels per se are not a reliable predictor of sperm presence in testicular biopsy samples [89,90]. ...
... Similarly, serum inhibin B levels were significantly lower in patients with TS and those with MPHD than in patients with GHD and SGA, each independently, whereas serum inhibin B levels were slightly higher in patients with MPHD than in TS (p < 0.05; Figure 3A). Notably, no significant differences in inhibin B serum concentrations were observed between female patients with congenital MPHD (n = 2) and acquired MPHD (n = 5), although the sample size was limited (median 15.5 [10][11][12][13][14][15][16][17][18][19][20][21] vs. 13 ; not significant). Additionally, six girls with TS mosaicism who experienced spontaneous menarche displayed higher serum AMH and inhibin B levels than in other girls with TS (data not shown). ...
... In male patients, slightly lower inhibin B levels were observed in those with congenital MPHD and HH than in acquired MPHD, likely due to fewer Sertoli cells and reduced germ cell count resulting in smaller testes [21,22]. Serum inhibin B levels demonstrated a positive correlation with testicular volumes [21], indicating its utility as a marker for discriminating HH from constitutional delay of puberty [23]. ...
... In male patients, slightly lower inhibin B levels were observed in those with congenital MPHD and HH than in acquired MPHD, likely due to fewer Sertoli cells and reduced germ cell count resulting in smaller testes [21,22]. Serum inhibin B levels demonstrated a positive correlation with testicular volumes [21], indicating its utility as a marker for discriminating HH from constitutional delay of puberty [23]. Additionally, males with KS exhibited lower inhibin B levels but higher AMH levels, reflecting Sertoli cell dysfunction with impaired spermatogenesis. ...
Article
Full-text available
Objective During the process of transition from paediatric to adult health care, counselling concerning fertility is an important issue and is based mainly on serum markers of gonadal function. Here, we analysed these markers in adolescents with various underlying endocrine diseases at the time of transition. Methods After reaching near adult height and late puberty (girls: bone age [BA] ≥14 years, and boys: BA ≥16 years), we assessed stages of puberty according to Tanner and measured testes or ovarian volumes and serum markers of gonadal function (anti‐Mullerian hormone [AMH], inhibin B, 17β‐estradiol, testosterone). Results One hundred and ten patients (56 females and 54 males) were included from May 2010 to March 2016 with multiple pituitary hormone deficiency (MPHD; n = 17), growth hormone deficiency (GHD; n = 35), Turner syndrome (TS; n = 27), short stature after being born small for gestational age (SGA; n = 20) and Klinefelter syndrome (KS; n = 11). Female and male adolescents exhibited mature secondary sexual characteristics. The levels of serum inhibin B and AMH were lower in TS and female MPHD than in GHD and SGA, each independently (p < 0.05). The levels of serum AMH were higher whereas serum inhibin B were lower in male MPHD and KS (p < 0.05). Ovary volumes were significantly smaller in patients with TS, and testicular volumes were smaller in patients with KS. Conclusions After current established treatments with sex steroids, the development of secondary sexual characteristics was mature. However, impaired markers of fertility have been identified in patients with TS, KS and MPHD, reflecting gonadal dysgenesis in TS and KS, but gonadal immaturity in MPHD as gonadal gonadotropin stimulation is lacking throughout development. Consequently, in patients with MPHD, these markers cannot reliably predict individual fertility, which warrants consideration and incorporation in future treatment concepts.
... Another aspect that should be considered is the hormone profile linked to male infertility: in particular, inhibin B is a glycoprotein hormone produced by Sertoli testicular cells, that regulates the Follicle-Stimulating Hormone (FSH) level throughout a negative feedback mechanism [24]. Its serum levels are strongly positively correlated with testicular volume and sperm counts. ...
... Its serum levels are strongly positively correlated with testicular volume and sperm counts. Generally, in infertile patients, inhibin B decreases and FSH increases [24]. ...
Article
Full-text available
Purposeof Review Male fertility is an emergent issue that should be considered in clinical practice, when dealing with chronic inflammatory diseases in young men. As it is known, the chronic inflammation is the main pathophysiologic mechanism in some rheumatological conditions such as spondyloarthritis (SpA), Ankylosing Spondylitis (AS) and Psoriatic Arthritis (PsA). Therefore, it is paramount to be aware if these diseases could impair male fertility, both due to the inflammation or to the treatments needed: we reviewed the literature on the most relevant and recent evidence on male fertility in patients affected by SpA, AS and PsA. Recent Findings Rheumatological inflammatory diseases (included SpA, AS and PsA) could impair the family planning in man life, especially when diagnosed at young age. Moreover, focusing on sperm quality, it seems that a link between sperm quality impairment and a higher disease activity exist. Focusing on therapies, Tumor Necrosis Factor inhibitors showed a safety profile on human male fertility in clinical studies. Recently, a prospective study and two double-blind placebo-controlled trials assessed the impact of methotrexate and Filgotinib on semen parameters, respectively, showing a safety profile of these drugs on human semen quality. However, there are no clinical data on the impact of Interleukin (IL)17 inhibitors(i), IL12-23i and IL23i. Summary Concerning male fertility in SpA, AS and PsA, an unmet clinical need is still present and new studies are needed to understand the association between these diseases and male fertility, and the implication of the therapies used for these diseases. This narrative review provides an overview of the available data on male fertility in patients affected by SpA, AS and PsA.
... Inhibin (INH) plays a vital role in the hypothalamus-pituitary gonadal (HPG) axis, which is secreted by Sertoli cells of testis in males and granulosa cells of ovarian follicles in females (Ying 1988), (Rehman et al. 2021). As a member of the transforming growth factor (TGF-β) superfamily, INH down-regulates the production of follicle-stimulating hormone (FSH) by the gonadotropic cells of the anterior pituitary in males (Meachem et al. 2001). INH has been proposed as an autocrine/paracrine factor that regulates follicular atresia, growth, steroidogenesis and gonadotropin responsiveness (Knight 1996;Akhtar et al. 2021). ...
Article
Full-text available
The objective of the present study was to investigate the potential role of immunization against INH on testicular development, serum reproductive hormone (FSH, LH and T) concentrations, testicular biochemical indexes (GGT, ALP, ACP, LDH, α-neutral glucosidase and carnitine) and testicular cell apoptosis in rats. Forty healthy male rats were randomly divided into four groups (n = 10 per group), and respectively immunized with 0.5 mL of C500/pVAX-asd-IS-L (10⁸ CFU/ mL), C500/pVAX-asd-IS-M (10⁹ CFU/ mL), C500/pVAX-asd-IS-H (10¹⁰ CFU/ mL) or C500 (10¹⁰ CFU/ mL) (thrice with an interval of 2 weeks). P/N values increased after vaccination and differed (p < 0.05) at 1 week and sharply increased at 2 weeks following the booster vaccination (p < 0.01); P/N values indicated that the immune response was induced and production of anti-INH antibodies in rats; Immunization of INH vaccines enhanced serum concentrations of T and FSH, while reduced LH concentrations (p < 0.05); The testis weight, volume, testis coefficient, epididymal weight, sperm count in the epididymis, and the sperm deformity results indicated that INH gene vaccine immunization could promote testicular development and enhance sperm production rate in rats. After immunization of INH gene vaccines, GGT activity was markedly increased, but ALP, LDH and α-neutral glucosidase activities were observably reduced (p < 0.05). Furthermore, flow cytometry results demonstrated that the apoptosis rate of the testicular cells was extremely increased in the C500/pVAX-asd-IS-M group (p < 0.01). These findings illustrate a functional cross-link between FSH, T, LH and spermatogenic markers via inhibin to maintain spermatogenesis. INH immunization may have a positive effect on spermatogenesis and testicular development in male rats.
... The role of AMH in adult males is not completely understood, but it is believed to correlate with spermatogenesis. As for InhB, AMH may be considered as a marker of Sertoli cell maturation, whereby more immature cells produce more AMH than less immature cells, possibly reflecting the degree of spermatogenesis [21,[33][34][35][36]. This led to the hypothesis that AMH levels could predict SR at mTESE. ...
Article
Full-text available
Background and objective: No clear-cut markers for predicting positive sperm retrieval (+SR) at microdissection testicular sperm extraction (mTESE) have been identified thus far. Our aim was to conduct a systematic review and meta-analysis to evaluate the ability of follicle-stimulating hormone (FSH), inhibin B (InhB), and anti-Müllerian hormone (AMH) to predict +SR in men with nonobstruc-tive azoospermia (NOA) undergoing mTESE. Methods: We performed a search in the PubMed, EMBASE, Web of Science, and Scopus databases according to the Preferred Reporting Items for Systematic Review and Meta-analysis statement. Thirty-four publications were selected for inclusion in the analysis. Key findings and limitations: Overall, the mean +SR rate was 45%. Pooled standardized mean difference (SMD) values revealed significant hormonal differences between the +SR and ÀSR groups, with lower FSH (SMD À0.30), higher InhB (SMD 0.54), and lower AMH (SMD À0.56) levels in the +SR group. Pooled odds ratios (Ors) revealed no significant prediction of +SR by either FSH (OR 1.03, 95% confidence interval [CI] 1.00-1.06) or InhB (OR 1.01, 95% CI 1.00-1.02), despite variations in baseline levels and study heterogeneity. Conversely, AMH had significant predictive value (OR 0.82, 95% CI 0.73-0.92), with lower baseline levels in the +SR group. InhB and FSH levels were higher in the +SR group, while InhB exhibited the opposite trend. Conclusions and clinical implications: Despite study heterogeneity, our meta-analysis findings support the ability of AMH to predict +SR for men with NOA undergoing mTESE. Patient summary: We conducted a review and analysis of results from previous studies. Our findings show that for men with an infertility condition called nonob-structive azoospermia, blood levels of anti-Müllerian hormone can predict successful extraction of sperm using a microsurgical technique. Levels of two other hormones did not predict successful sperm extraction.
... It acts as a negative feedback mechanism for the FSH secretion process, and it appears to be an essential marker of the seminiferous tubules' ability to perform their role (Pierik, 1998). there is a positive correlation between testicular volume and serum inhibin B levels (Meachem, 2001). the combination of inhibitin B and follicle-stimulating hormone (FSH) appears to be a more sensitive and specific marker of spermatogenesis than either one by itself (Kong, 2021 ). ...
Thesis
Full-text available
I Summary The reproductive toxicity due to exposure to aflatoxins was investigated in adult male rats. The present study aims to document the harmful effects of exposure to aflatoxin-B1 in the pre-pubertal stage on the reproductive efficiency of male rats after puberty. The field experiment was extended from 1 November, 2022 to 30 January, 2023. A total of 80 male rats (35 days old and 75-80 g weight) and 40 mature female rats (75 days old and 155-165 g weight) were included in the current study. Male rats were divided equally to control and treatment groups (AFB1 group). The males were daily administered with distilled water and aflatoxin-B1 (0.3mg/kg/day) per os, respectively. After 15, 25, and 35 days of treatment (pre-puberty, puberty, and post-puberty periods, respectively), ten males from each group were weighted, anesthetized, and sacrificed. Testes, epididymis, seminal vesicles, and prostates were dissected and weighted. Blood samples were collected for assessment the serum concentrations of reproductive hormones (gonadotrophin releasing hormone; GnRH, follicle stimulating hormone; FSH, luteinizing hormone; LH, and testosterone; T). Pituitary and testicular tissue samples were obtained to analyze the expression levels of pituitary GnRHR, FSHβ and LHβ genes and testicular FSHR, LHR, ABP, 3β-HSD, and 17β-HSD genes. Testes and epididymis were obtained for histopathological examination. At puberty (25 days of treatment) and post-puberty (35 days of treatment), tail of epididymis was dissected for semen analysis, including sperm motility, sperm count, sperm viability, and sperm abnormality. The remaining 10 males from each group were matted with experienced females (1 male: 2 females) to find out the fertility index, included pregnancy rate (%), offspring number/dam, duration of pregnancy (day), and weight at birth (g). Following 15, 25, and 35 days of exposure, the AFB1 treated group revealed decline in the relative weight of testes, epididymis, seminal vesicle, and prostate than control, early at pre-pubertal stage, which continued at pubertal and post-pubertal stages. Compared to control, treated males showed a decrease in serum concentration of GnRH, FSH, LH, and testosterone, and the expression level of pituitary GNRHR, FSHβ, and LHβ genes and testicular LHR, FSHR, ABP, 3β-HSD, and 17β-HSD genes, in all experimental periods. Histological sections of the testicles of treated males showed atrophy of some seminiferous tubules, empty lumen, and massive vacuolization and exfoliation of the germ cells. The histological results of the II epididymis showed obliteration of the lumen, necrosis of the epithelial layer, deformed cavities, a climbing epithelial layer, and epithelium hyperplasia compared to the control group. Sperm motility, sperm count, and sperm viability were significantly decreased, while sperm abnormality was significantly increased in the AFB1 group, at puberty and post-puberty. Females matted with AFB1 treated males revealed significant decrease of pregnancy rate, number of offspring, and litter weight at birth in comparison with those matted with control males. In conclusion, altogether, the current results showed that exposure to aflatoxin-B1 at the pre-pubertal stage have adverse effects on reproduction represented by reduced reproductive efficiency and performance with impaired spermatogenesis after puberty.
... In particular, in adult men FSH stimulates the production of InhB in the testis, a glycoprotein member of the transforming growth factor-β (TGF-β) superfamily, which in turn activates a negative feedback on the pituitary secretion of FSH itself. Moreover, the release of InhB is regulated by other factors over FSH such as the amount for sperm count, and its levels could reflect the functional status of seminiferous epithelium [2]. ...
Article
Full-text available
Purpose: Purpose: We aimed to investigate the relationship between follicle stimulating hormone (FSH) and inhibin B (InhB). Materials and Methods: Materials and Methods: Data from 1,230 consecutive men presenting for primary couple's infertility were analyzed. Health-significant comorbidities were scored with Charlson comorbidity index. Quartiles of FSH and InhB were considered to determine threshold values. Descriptive statistics and logistic regression models tested association between FSH and InhB values. Results: Results: Overall, 1,080 (87.8%) men had concordant FSH and InhB values. Conversely, 150 patients (12.2%) had discrepancies in FSH and InhB, with 78 (6.3%) and 72 (5.9%) men reporting both low and high FSH and InhB values, respectively. Infertile men with discordant values were younger (median [interquartile range] 38.0 years [34-41 years] vs. 36.0 years [31-40 years]); had smaller testicular volume (TV) (12 mL [10-15 mL] vs. 15 mL [12-20 mL]); and, had more frequently a sperm DNA fragmentation test >30% (179 [59.1%] vs. 40 [78.4%]) than those with concordant values (all p<0.05). Moreover, a higher frequency of previous cryptorchidism (27.3% vs. 11.9%), lower sperm concentration (3.0 million/mL [0.9-11.0 mil-lion/mL] vs. 13.8 million/mL [3.1-36.0 million/mL]), lower progressive sperm motility rates (12.0% [5.0%-25.3%] vs. 20.0% [7.0%-36.0%]), and greater rates of non-obstructive azoospermia (36.4% vs. 23.9%) were found in men with discordant FSH and InhB values (all p≤0.005). At multivariable logistic regression analysis, higher body mass index (odds ratio [OR], 1.08; p=0.001), smaller TV (OR, 0.91; p<0.001), and a history of cryptorchidism (OR, 2.49; p<0.001) were associated with discordant FSH and InhB values. Conclusions: Conclusions: More than one out of ten infertile men had discordant FSH and InhB values in the real-life setting showing worse clinical profiles than those with concordant levels. Smaller TV and history of cryptorchidism could be used as clinical markers to better tailor the need to test InhB.
Article
Full-text available
The testes serve as the primary source of androgens and the site of spermatogenesis, with their development and function governed by hormonal actions via endocrine and paracrine pathways. Male fertility hinges on the availability of testosterone, a cornerstone of spermatogenesis, while follicle-stimulating hormone (FSH) signaling is indispensable for the proliferation, differentiation, and proper functioning of Sertoli and germ cells. This review covers the research on how androgens, FSH, and other hormones support processes crucial for male fertility in the testis and reproductive tract. These hormones are regulated by the hypothalamic–pituitary–gonad (HPG) axis, which is either quiescent or activated at different stages of the life course, and the regulation of the axis is crucial for the development and normal function of the male reproductive system. Hormonal imbalances, whether due to genetic predispositions or environmental influences, leading to hypogonadism or hypergonadism, can precipitate reproductive disorders. Investigating the regulatory network and molecular mechanisms involved in testicular development and spermatogenesis is instrumental in developing new therapeutic methods, drugs, and male hormonal contraceptives.
Article
The endocrine disruptor hexavalent chromium [Cr(VI)] is a proven reproductive toxicant. We recently demonstrated that prenatal Cr(VI) exposure causes testicular resistance to gonadotropins, resulting in hypergonadotropic hypoandrogenism in F 1 rats. However, the mechanism driving hypergonadotropism in F 1 rats exposed to Cr(VI) prenatally remains an enigma. Therefore, we hypothesized that ‘Prenatal Cr(VI) exposure may disrupt steroid hormones‐mediated negative feedback regulation of the hypothalamic GnRH, and its receptor in the pituitary of F1 rats, leading to hypergonadotropism.’ We administered potassium dichromate (50, 100, or 200 mg/L) to pregnant rats through drinking water between days 9 and 14, and their male F1 offspring were euthanized at 60 days of age. Prenatal Cr(VI) exposure in F 1 rats resulted in the accumulation of Cr in the hypothalamus and pituitary. Western blot detected decreased hypothalamic GnRH, Kisspeptin1, and its receptor GPR54, along with diminished ERα, AR, aromatase, and 5α reductase, and GnRH regulatory transcription factors Pit‐1 and GATA‐4 proteins. Immunohistochemical studies revealed increased immunopositivity of GnRH receptor, AR, 5α reductase, ERα, ERβ, and aromatase proteins in the pituitary, whereas decreased Kisspeptin1, GPR54, and inhibin β. Our findings imply that Cr(VI) exposure during the prenatal period disrupts the hypothalamic Kisspeptin‐GPR54‐Pit‐1/GATA4‐GnRH network, boosting the pituitary GnRH receptor. We conclude that prenatal exposure to Cr(VI) alters GnRH expression in the hypothalamus and its receptor in the pituitary of F1 progeny through interfering with the negative feedback effect of androgens and estrogens.
Article
PURPOSE: To obtain information about preorchiectomy gonadal function in patients with testicular germ cell cancer to improve the clinical management of fertility and other andrologic aspects in these men. PATIENTS AND METHODS: In group 1, a group of 83 consecutive patients with testicular germ cell cancer (TGCC) investigated before orchiectomy, semen analysis was carried out in 63 patients and hormonal investigations, including measurement of follicle-stimulating hormone, luteinizing hormone (LH), testosterone, estradiol, sex hormone–binding globulin (SHBG), inhibin B, and human chorionic gonadotropin (hCG), in 71 patients. Hormone levels in patients with elevated hCG (n = 41) were analyzed separately. To discriminate between general cancer effects and specific effects associated with TGCC, the same analyses were carried out in a group of 45 consecutive male patients with malignant lymphoma (group 2). Group 3 comprised 141 men employed in a Danish company who served as controls in the comparison of semen parameters. As a control group in hormone investigations, 193 men were selected randomly from the Danish National Personal Register to make up group 4. RESULTS: We found significantly lower sperm concentration (median, 15 × 10 ⁶ /mL; range, 0 to 128 × 10 ⁶ /mL) and total sperm count (median, 29 × 10 ⁶ /mL; range, 0 to 589 × 10 ⁶ ) in patients with testicular cancer than in patients with malignant lymphomas (sperm concentration: median, 48 × 10 ⁶ /mL; range, 0.04 to 250 × 10 ⁶ /mL; sperm count: median, 146 × 10 ⁶ ; range, 0.05 to 418 × 10 ⁶ ) (P < .001 and P < .001) and healthy men (sperm concentration: median, 48 × 10 ⁶ /mL; range, 0 to 402 × 10 ⁶ /mL; sperm count: median, 162 × 10 ⁶ ; range, 0 to 1253 × 10 ⁶ ) (P < .001 and P < .001). FSH levels were increased in men with testicular cancer (median, 5.7 IU/L; range, 2.0 to 27 IU/L) compared with both men with malignant lymphomas (median, 3.3 IU/L; range, 1.01 to 12.0 IU/L) and healthy controls (median, 4.1 IU/L; range, 1.04 to 21 IU/L) (P = .001 and P = .007, respectively). Surprisingly, we found significantly lower LH in the group of men with TGCC (median, 3.6 IU/L; range, 1.12 to 11.9 IU/L) than in healthy men (median, 4.7 IU/L; range, 1.3 to 11.9 IU/L) (P = .01). We could not detect any differences between men with testicular cancer and men with malignant lymphomas and healthy men with regard to serum levels of testosterone, SHBG, and estradiol. Men with testicular cancer who had increased hCG levels had significantly lower LH and significantly higher testosterone and estradiol than those without detectable hCG levels. CONCLUSION: Spermatogenesis is already impaired in men with testicular cancer before orchiectomy. Neither local suppression of spermatogenesis by tumor pressure nor a general cancer effect seems to fully explain this impairment. The most likely explanation is pre-existing impairment of spermatogenesis in the contralateral testis in men with testicular cancer. The question of whether also a pre-existing Leydig cell dysfunction is present in men with testicular cancer could not be answered in this study because the tumor seems to have a direct effect on the Leydig cells. Men with testicular cancer had low LH values as compared with controls. We speculate that increased intratesticular level of hCG also in men without measurable serum hCG may play a role by exerting LH-like effects on the Leydig cells, causing increased testosterone and estrogen levels and low LH values in the blood.
Article
To elucidate the role of germ cells in the regulation of inhibin B secretion, serum inhibin B levels in prepubertal boys and adult men whom had a concurrent testicular biopsy showing either normal or impaired testicular function were compared. In addition, by immunohistochemistry the cellular localization of the two subunits of inhibin B (α and βB) were examined in adult testicular tissue with normal spermatogenesis, spermatogenic arrest, or Sertoli cell only tubules (SCO) as well as in normal testicular tissue from an infant and a prepubertal boy. Adult men with testicular biopsy showing normal spermatogenesis (n = 8) or spermatogenic arrest (n = 5) had median inhibin B levels of 148 pg/mL (range, 37–463 pg/mL) and 68 pg/mL (range, 29–186 pg/mL), respectively, corresponding to normal or near-normal levels of our reference population (165 and 31–443 pg/mL; n = 358). Men with SCO (n = 9) had undetectable or barely detectable (n = 1) serum levels of inhibin B. In contrast to adults, prepubertal boys with SC...
Article
The purification and cloning of a membrane-anchored proteoglycan with affinity for inhibin A are described. Bovine pituitary membranes were isolated, and membrane-anchored proteins were solubilized and used as an enriched source of inhibin binding protein. The extract was passed over an inhibin A affinity column, and a protein, designated p120, was identified as an inhibin-binding moiety. A partial amino acid sequence was determined for the protein, which matched two human complementary DNAs (cDNAs) in the database. The full-length cDNA predicts a 1336-amino acid glycoprotein. Full-length p120-encoding cDNAs were isolated from human testis RNA and cloned into expression vectors. Two p120 messenger RNA transcripts of 4.6 kb and 2 kb are detected in rat pituitary by RNA blot analysis. Similar analysis of rat testis RNA revealed transcripts of identical molecular mass, albeit at lower abundance. To determine the cellular localization of p120 in pituitary and testis, an antibody directed against the predicted extracellular domain of the protein was generated and used in an immunohistochemical analysis of thin tissue sections. p120 immunostaining is coincident with FSHβ immunopositive gonadotrope cells in rat pituitary. p120 staining is intense in the testicular Leydig cells, which bind iodinated inhibin but not iodinated activin. In summary, an inhibin-binding protein has been isolated that is produced in tissues that are targets of inhibin action.