You are on page 1of 6

Review TRENDS in Endocrinology and Metabolism Vol.16 No.

8 October 2005

Genetic complexity of FSH receptor


function
Jörg Gromoll and Manuela Simoni
Institute of Reproductive Medicine, Domagkstrasse 11, 48129 Münster, Germany

The interaction between follicle-stimulating hormone that receptor dimerization might participate in signal
(FSH) and the FSH receptor (FSHR) is essential for normal transduction.
oogenesis and spermatogenesis. Recently, single-nucleo- The description of naturally occurring mutations of
tide polymorphisms (SNPs) have been assigned to the the FSHR highlights the role of this gonadotropin in
FSHR gene. These give rise to different FSHR haplotypes male and female gonadal function (reviewed in [3,7–11])
that modify the action of FSH. In women, FSH sensi- and provides further evidence that spermatogenesis is
tivities during the menstrual cycle and different cycle impaired and follicular maturation is halted by disruption
lengths are observed, depending on the FSHR haplo- of FSH action. In addition, mutations that result in either
type. Thus, SNPs of the FSHR determine the ovarian constitutive activation [12] or loss of binding specificity for
response and should, therefore, be considered in con- FSH, which result in human chorionic gonadotropin
trolled ovarian hyperstimulation during assisted-repro- (hCG)-induced ovarian hyperstimulation syndrome
duction techniques in women with normal ovarian [13–15], have been described (Box 1). However, studies
function. In men, the impact of the FSHR SNPs is during the past decade demonstrate that mutations in
unclear. The genetic complexity of FSHR should be FSHR are rare. By contrast, mutation screening shows
considered when studying FSH action. These SNPs are that the FSHR gene contains several single-nucleotide
one of the first examples in which genetic changes polymorphisms (SNPs), some of which are common and
contribute to fine-tuning the endocrine regulation of might influence receptor activity. In this review we
reproduction. A rational pharmacogenetic approach summarize the current knowledge of the (patho)physio-
that combines FSH dose according to the FSHR haplo- logical importance of SNPs in the FSHR gene.
type is envisaged.
SNPs
From sequencing the human genome and searching for
Introduction mutations in several pathophysiological conditions, it is
Follicle-stimulating hormone (FSH) is a key player in evident that genetic variations caused by SNPs are
human reproduction [1,2]. This pituitary gonadotropin frequent and occur in the hormones and receptors of the
consists of two subunits, the a- and the b-subunits, and it hypothalamic–pituitary–gonadal axis [9,16]. In general,
exerts its throphic and stimulatory effects on gameto- the incidence of polymorphisms in a given gene ranges
genesis by binding to a specific receptor that is located
exclusively on the surface of Sertoli cells in the testis and Box 1. The FSH receptor
granulosa cells in the ovary (Box 1). The FSH receptor Gene: Chromosome 2 p21-p16; 10 exons spanning 54 kb (NCBI
(FSHR) is a G-protein-coupled receptor whose main database http://www.ncbi.nlm.nih.gov/entrez/; GeneID:2492; Locus
signal-transduction mechanism involves activation of tag:HGNC:3969).
Protein family: G-protein-coupled receptor.
protein kinase A (PKA); however, PKB and PKC are
Structure: 678 amino acids; seven transmembrane domains; the
also involved [3–5]. extracellular domain (359 amino acids) consists of 10 leucine-rich
Recently, the nature of the FSH–FSHR interaction has repeats that form a horseshoe-shaped pocket for hormone binding.
been resolved by crystallizing FSH in a complex with the Expression: Ovary, granulosa cells; testis, Sertoli cells.
extracellular domain of FSHR [6]. According to this, the Ligand: FSH.
Signal transduction pathway: PKA, PKB and PKC.
extracellular FSHR domain is shaped as a slightly curved Pathophysiology of FSHR mutations: OMIM 136435
tube. An a-subunit and a b-subunit of FSH bind to the Male:
inner, slightly concave face of the tube in a hand-clasp Activating mutation: sustained spermatogenesis without
fashion, and binding specificity is mediated by amino acids gonadotropins
in both subunits. Binding to the receptor involves con- Inactivating mutations: hypospermatogenesis
Female:
formational changes in the hormone, with the a-subunit- Activating mutation: no activating mutations have been identified
loops adjusting their shape to reach optimal interaction. Inactivating mutation: FSH-resistance syndrome, primary or second-
The extracellular FSHR domain that is occupied by the ary amenorrhoea with the presence of primordial follicles
hormone forms dimers in the crystal, which indicates Ligand-sensitive mutation: increased sensitivity to human
chorionic gonadotropin (hCG). Ovarian hyperstimulation syndrome
Corresponding author: Gromoll, J. (Joerg.Gromoll@ukmuenster.de). caused by hCG.
Available online 26 August 2005

www.sciencedirect.com 1043-2760/$ - see front matter Q 2005 Elsevier Ltd. All rights reserved. doi:10.1016/j.tem.2005.05.011
Review TRENDS in Endocrinology and Metabolism Vol.16 No.8 October 2005 369

from 15 to 50% in the normal population. This indicates polymorphisms spread late in evolution and are most
that such genetic changes have no pronounced effects on prevalent in humans [17].
reproduction, otherwise evolution would have exerted
deleterious effects on them. Polymorphisms can affect The FSHR promoter
gene function by at least two mechanisms: by changing the The promoter of the FSHR gene in humans, rats, mice and
biochemical properties of the gene product directly; and sheep is in a class of promoters that lack conventional
by acting at the transcriptional level to modify the activity TATA and CCAAT boxes and have multiple transcription
of the promoter. SNPs generate genetic diversity and start sites. A core promoter region of human FSHR, which
complexity within species [17]. It is possible that genetic displays the highest transcriptional activity but no cell
changes, such as SNPs, either singly or in combination, specificity, is assigned to a region between K225 and K1
modify and fine-tune endocrine-feedback systems and relative to the translational start site [18–21]. To date, two
hormone action. This results in variable inter-individual response elements have been identified within this core
reproductive performance that ranges from fully func- promoter region. An E-box consensus sequence (CACATG),
tional gametogenesis to subfertility. which interacts with a family of basic helix–loop–helix
transcriptional factors such as USF, is required for full
SNPs in FSHR promoter function [19,21–24]. Recently, it has been shown
To date, the National Center for Biotechnology Infor- that the sequence region covering the E-box might also be
mation (NCBI) SNP database (http://www.ncbi.nlm.nih. the binding target for factors such as chicken ovalbumin
gov/SNP/) indicates that there are 731 SNPs in the FSHR upstream promoter-transcription factors (COUP-TF) that
gene. Only five of these are in the coding region (exons), repress FSHR expression. Thus, the net regulation of the
the rest are intronic. One SNP is located in the 5 0 untrans- FSHR gene might result from the relative availability of
lated region of FSHR mRNA at position –29 (ss2189241). repressors and activators in a given physiological state
The five SNPs in the coding region occur in exon 10, [22]. The second element is an initiator element (Inr),
at codons 307, 329, 524, 665 and 680, and four are which has been identified covering the region of the
non-synonymous (result in amino acid substitution). transcriptional start site, and is typical of housekeeping
Although the Ala307Thr (rs6165) and the Ser680Asn genes (Figure 1) [21].
(rs6166) polymorphisms are well characterized with
respect to frequency and ethnic distribution (see below), SNPs in the FSHR promoter
there is no information on the prevalence of the In accordance with the SNP database, a G/A SNP with a
Ala665Thr, Arg524Ser and the synonymous Thr329Thr heterozygosity rate of 0.6 occurs at position K29 within
polymorphisms. the core promoter region of the FSHR. The A nucleotide
There are differences in the number of SNPs in the is conserved in chimpanzees and sheep, whereas G is
FSHR genes of humans, mice and rats. Only 25 intronic present in rats (Figure 1). This SNP is embedded in the
SNPs are described in mice and only four SNPs in exon 10 consensus sequence GG/AAA, which represents a putative
of rat FSHR. Although SNPs in mice and other species binding site for a c-E-twenty-six specific (c-ETS) tran-
might have received less attention than in humans, scription factor. However, functional studies in COS-7
this marked difference supports the view that genetic cells, and in granulosa- and Sertoli-cell lines, which

Initiation complex
USF USF

mRNA

–99
Exon 1
OligoA E-box
–257 bp Inr

–157 to –141 –124 to –119 –100 to –94 –29


Human An, n=13–17 CACATG Consensus A/G
Chimp AACAACA7 G T/C T/C A N T/A T/C T/C A
Ovine GAA6GA8GGA3 G A
Rat TA7 G G
TRENDS in Endocrinology & Metabolism

Figure 1. Schematic representation of the core promoter of human FSHR and the localization of polymorphisms. Indicated are the regulatory elements, including the E-box,
initiatior element region (Inr), the K29 SNP (yellow) and the variable oligoA stretch (yellow), and the transcription initiation complex with the corresponding transcriptional
start site. A sequence comparison of the human, chimpanzee, sheep and rat FSHR promoter for the SNPs and the regulatory elements within the core promoter region is
given in the bottom part of the figure. The chimpanzee nucleotide sequences were compiled from the chimpanzee genome database at the NCBI using the Pan troglodytes
chromosome 2A genomic contig NW_103405. The other sequences were obtained from GenBank: human FSHR accession No. X91738, ovine FSHR accession No. AF090438
and rat FSHR accession No. S81117.

www.sciencedirect.com
370 Review TRENDS in Endocrinology and Metabolism Vol.16 No.8 October 2005

measure activity of a luciferase reporter gene driven by that the amount of FSH needed for COH to achieve similar
the core promoter region of FSHR (which contains this peak estradiol levels is significantly lower in women who
SNP), reveal no significant differences in promoter are homozygous for the Asn680 variant compared with
activity [25]. Therefore, in vitro data show no significant women who are either homozygous for Ser680 or are
effect of this SNP on gene transcription. heterozygous, indicating a lower ovarian sensitivity to
A detailed sequence comparison of the FSHR core FSH in vivo of the Ser680 variant [28]. Similar results
promoter region reveals another polymorphic region have been obtained by others [29–31].
covering an oligo A-stretch that is polymorphic in humans This observation has been corroborated recently in a
(A11–17) and varies between species (Figure 1) [25]. prospective interventional study in which a randomized,
However, neither the incidence of this polymorphism nor controlled trial design showed a differential response of
functional data are available. estradiol to FSH because of the SNP at codon 680 of
Recently, we have analyzed the effects of the SNP at FSHR. In this study, women undergoing COH were
position K29 in 186 normozoospermic and 345 azo- administered the same dose of FSH but this resulted in
ospermic men. The genotype distribution is similar in significantly lower serum levels of estradiol in Ser680/
both groups, with the A–29 allele less frequent (25%) than Ser680 homozygote women compared with women with
the G–29 allele (75%). In addition, this polymorphism does Asn680/Asn680 homozygote women variant. This lower
not correlate with serum levels of FSH [25]. Therefore, response is overcome by increasing the FSH dose [32].
these in vivo data confirm the in vitro observation that the Despite differences in estradiol levels, no significant
SNP at position K29 is unlikely to influence FSH activity differences were detected in either the number of follicles
directly when considered alone. or retrieved oocytes, fertilization rate, cumulative embryo
score and pregnancy rate. This indicates that, according to
SNPs in exon 10 of FSHR the current protocols, some women might receive too much
Exon 10 encodes the C-terminal end of the extracellular FSH, thus putting them at risk of ovarian hyperstimula-
domain, the entire transmembrane domain and the tion syndrome (OHSS), which depends indirectly on
intracellular domain of FSHR [3]. Although exon 10 is excessive stimulation by FSH. Indeed, a recent, retro-
fundamental for signal transduction, it is not necessary spective, association study has demonstrated that the
for ligand binding. The transmembrane domain, however, Ser680 variant occurs significantly more often in women
might contact hormone bound to the extracellular domain, that develop iatrogenic OHSS but that the Asn680 variant
in particular to loops L1 and L3 of the a-subunit [6]. is associated with the severity of OHSS [33]. Therefore,
The two most common SNPs in the coding region of women with the Asn680 genotype that are undergoing
FSHR occur at nucleotides 919 and 2039 in exon 10 COH might be at risk for excessive stimulation with FSH,
(where 1 is the A of the ATG translation start codon), which might increase the risk of severe iatrogenic OHSS.
which correspond to amino acid positions 307 and 680,
respectively, of the mature protein [26,27]. Residue 307 is Normal menstrual cycle
in the hinge region of the receptor. This region, which Generally, studies in women that are undergoing COH
is highly variable in the three glycoprotein hormone indicate that ovarian FSH threshold is influenced by SNPs
receptors (FSHR, luteinizing hormone receptor and in exon 10 of the FSHR. This concept is confirmed by a
thyroid-stimulating-hormone receptor), connects the recent study in which the menstrual cycle in women with
hormone-binding domain to the transmembrane domain. normal, mono-ovulatory cycles has been monitored.
The corresponding residue is not conserved in the During the luteo–follicular transition, serum levels of
glycoprotein hormone receptors. Residue 680 is in the estradiol, progesterone and inhibin A are significantly
intracellular, C-terminal domain in a region that also lower, and FSH starts to rise earlier, in women who are
varies between the three receptors. No roles have been homozygous for the allele that encodes Ser680 compared
identified for residues 307 and 680. with women who are homozygous for Asn680. In addition,
The two polymorphisms in exon 10 are in linkage dis- FSH levels are significantly higher during the follicular
equilibrium and result in two major, almost equally phase in this group (Figure 2), but no differences were
common, allelic variants in Caucasians: Thr307–Asn680 observed between groups in estradiol, inhibin B and growth
and Ala307–Ser680 [16,27], with the Ala307–Ser680 velocity of the dominant follicle, which shows that higher
variant occurring in w40% of the population world- concentrations of endogenous FSH are necessary to achieve
wide. The other two combinations (Thr307–Ser680 and physiological ovulation in women who are homozygous for
Ala307–Asn680) are less common and constitute !5% of Ser680. In addition, menstrual cycles are significantly
FSHR alleles. longer, with a difference of about three days in these
women. Thus, the Ser680/Ser680 genotype results in a
Effects of exon 10 SNPs on ovarian function higher ovarian threshold for FSH, decreased negative feed-
Controlled ovarian hyperstimulation back to the pituitary and a longer menstrual cycle [34].
Studies in women with normal ovarian function demon-
strate that SNPs in exon 10 modulate FSHR function and Pathological ovarian function
the ovarian response to FSH. This effect was observed first Although clear in women with normal ovarian function,
in a partly retrospective, non-randomized study involving the impact of this FSHR polymorphism in anovulation
women who were undergoing controlled ovarian hyper- and amenorrhea remains controversial. An association
stimulation (COH) for assisted reproduction. It was shown between the Ser680 genotype and amenorrhea/anovulation
www.sciencedirect.com
Review TRENDS in Endocrinology and Metabolism Vol.16 No.8 October 2005 371

higher in these women compared with those with the


15 Menstruation
Asn680 genotype [34]. These data indicate that inhibin A,
together with estradiol and progesterone in the luteal
phase, might be responsible for the difference in FSH
concentrations that correlates with the polymorphism in
10 exon 10 of the FSHR observed in women. These luteal
FSH (IU/L)

hormones are irrelevant for the feedback control of FSH


secretion in men and there are no differences in FSH
levels in men with different FSHR genotypes. Therefore,
5 although the SNPs in exon 10 might have physiological
effects in the testis, there is no good parameter to measure
this. Inhibin B is not a good marker because, unlike
estradiol in women, it does not respond directly to FSH
0 stimulation. In men, inhibin B responds indirectly to the
–25 –20 –15 –10 –5 0 effects of FSH on spermatogonial proliferation and, thus,
spermatogenesis. The total sperm count might be a better
Day relative to midcycle LH peak
parameter to discern functional differences that relate
to the SNPs in exon 10 in normal men, but this has not
Asn/Asn Ser/Ser
been investigated.
TRENDS in Endocrinology & Metabolism

Figure 2. Menstrual-cycle-dependent serum levels of FSH referenced to the day of


Functional effects of FSHR SNPs in vitro
the luteinizing hormone (LH) surge (0) in women with the Asn680/Asn680 (nZ12) The molecular mechanism of the ‘resistance’ of Ser680-
and the Ser680/Ser680 (nZ9) genotype from the luteo–follicular transition phase FSHR to FSH is unclear. Studies in vitro following
(day K25) until ovulation (day 0). Results are meanGSEM. There is a statistically
significant difference between the two groups (Mann-Whitney-U test P!0.05) on
transient transfection of COS7 and HEK cells show that
most days from day K18 until ovulation. (Reproduced from [34] with permission). SNPs do not alter FSH binding and signal transduction
measured by cAMP and inositol (1,4,5)-triphosphate 3
is described occasionally [29,35], whereas other studies [Ins(1,4,5)P3] stimulation [26,29]. FSH-dependent events
find no association between FSHR polymorphisms and in granulosa cells, such as progesterone and estradiol
premature ovarian failure [36,37]. The Asn/Ser allele production, might be differentially regulated by poorly
variant is more common in Japanese women with poly- understood mechanism(s) that either do not involve or
cystic ovary syndrome than in normal controls [29]. No only partly involve cAMP production and PKA stimu-
difference was found in women with polycystic ovary lation. However, nuclear transcription factors such as
syndrome in the UK [36] and Singapore [38]. Similarly, no liver receptor homolog-1, and, possibly, steroidogenic
association has been identified between FSHR polymorph- factor-1 and dosage-sensitive sex reversal-adrenal hypo-
isms and twinning [27,39]. In a non-randomized trial, plasia congenita critical region on the X-chromosome 1
normogonadotropic anovulatory women who failed to (DAX-1) might be involved in the differential control of
either ovulate or conceive after administration of clomi- estrogen and progesterone production by granulosa cells
phene citrate have been treated with low-dose FSH to [41]. No differences are seen when cAMP (the earliest
induce ovulation [35]. No significant difference in the dose measurable effect of FSH stimulation in the signal-
of FSH needed for mono-follicular development was transduction cascade) is measured after stimulation of
detected between women with different allele variants of the two FSHR variants. Future studies, possibly in human
the FSHR. Therefore, it appears that the effect of the granulosa cells, are needed to ascertain if the two variants
FSHR allele variants on the estradiol response is evident activate different signal-transduction pathways down-
only in women with normal ovarian function. However, stream of cAMP/ Ins(1,4,5)P3 and/or whether quantitative
more extensive, prospective trials are necessary to inves- differences are observed. For example, the Ser680 (i.e. a
tigate the precise physiological role of the allele variants potential site of phosphorylation) is located in a region
in women with pathologic ovarian function. that is important for receptor recycling [42]. Interestingly,
the C-terminal truncation of the human FSHR involving
Effects of exon 10 SNPs on testicular function the amino acid at position 680 results in rerouting of
In contrast to observations in women with normal ovarian the internalized receptor to lysosomes and increases
function, SNPs in exon 10 of the FSHR have no effect on FSH-induced downregulation without affecting the pro-
serum levels of FSH and other clinical parameters in men duction of either cAMP or Ins(1,4,5)P3 [42]. It should be
with either normal or impaired spermatogenesis [26,40]. considered that, in vivo, receptors are usually activated at
This sex-related difference is striking and might be caused a low level of occupancy. If there is less Ser680 FSHR at
by the still unsolved role of FSH in spermatogenesis. the cell surface, both promoter activity and intracellular
Studies in women indicate that inhibin A, which is not fate of the receptor protein might play a role in deter-
present in men, is a major factor in the feedback control mining a quantitatively lower stimulation in response to
of serum FSH, whereas inhibin B might function as a FSH, but this is difficult to address in vitro.
marker of the number of developing follicles: higher Alternatively, factors that are not related directly to
concentrations of inhibin B in women with the Ser680 the FSHR polymorphism should also be considered. For
variant of the FSHR do not suppress serum FSH, which is example, it is established that the development of ovarian
www.sciencedirect.com
372 Review TRENDS in Endocrinology and Metabolism Vol.16 No.8 October 2005

follicles depends on members of the transforming growth 4 Ohtani, K. et al. (2001) Follicle-stimulating hormone promotes the
factor b superfamily [43], and that FSH and Sma and Mad growth of human epithelial ovarian cancer cells through the protein
kinase C-mediated system. Cancer Lett. 166, 207–213
homolog 3 (Smad-3) interact in signaling downstream of
5 Zeleznik, A.J. (2003) Protein kinase B is obligatory for follicle-
the FSHR in the mouse ovary [44]. In this respect, the stimulating hormone-induced granulosa cell differentiation. Endo-
polymorphism at residue 680 of the FSHR might be a crinology 144, 3985–3994
marker of another, unrecognized factor in linkage dysequi- 6 Fan, Q.R. and Hendrickson, W.A. (2005) Structure of human follicle-
librium that is responsible for the effect observed in vivo. stimulating hormone in complex with its receptor. Nature 433,
269–277
7 Gromoll, J. et al. (1996c) Functional and clinical consequences of
FSHR haplotypes mutations in the FSH receptor. Mol. Cell. Endocrinol. 125, 177–182
There are four common allelic combinations of 8 Gromoll, J. et al. (2000) Detection of mutations in glycoprotein
FSHR: A-29-A919-A2039 (A-Thr-Asn), G-29-A919-A2039 hormone receptors. Methods 21, 83–97
(G-Thr-Asn) A-29G919-G2039 (A-Ala-Ser) and G-29G919- 9 Huhtaniemi, I. (2003) Mutations affecting gonadotropin secretion and
action. Horm. Res. 60 (Suppl. 3), 21–30
G2039 (G-Ala-Ser). Based on O600 subjects of both sexes 10 Themmen, A.P.N. and Huhtaniemi, I.T. (2000) Mutations of gonado-
that we have screened, these four haplotypes account for tropins and gonadotropin receptors: elucidating the physiology and
O99% of FSHR alleles in the Caucasian population. pathophysiology of pituitary-gonadal function. Endocr. Rev. 21,
Expression studies using the K29 SNP and a reporter 551–583
gene in vitro, identified no major changes in transcrip- 11 Vassart, G. et al. (2004) A molecular dissection of the glycoprotein
hormone receptors. Trends Biochem. Sci. 29, 119–126
tional activity induced by this polymorphism [23]. How-
12 Gromoll, J. et al. (1996b) An activating mutation of the follicle-
ever, this does not exclude the possibility that subtle stimulating hormone receptor autonomously sustains spermato-
functional effects that are not evident in this system might genesis in a hypophysectomized man. J. Clin. Endocrinol. Metab.
become relevant when considered in combination with 81, 1367–1370
SNPs in exon 10 and/or following stimulation with FSH. 13 Vasseur, C. et al. (2003) A chorionic gonadotropin-sensitive mutation
Interestingly, the two allelic variants A-Ala-Ser and in the follicle-stimulating hormone receptor as a cause of familial
gestational spontaneous ovarian hyperstimulation syndrome.
G-Thr-Asn showed a statistically significant different dis- N. Engl. J. Med. 349, 753–759
tribution between controls and men with non-obstructive 14 Smits, G. et al. (2003) Ovarian hyperstimulation syndrome due to
azoospermia [45]. Although these data await confirma- a mutation in the follicle-stimulating hormone receptor. N. Engl.
tion, they indicate that the FSHR haplotype might be one J. Med. 349, 760–766
of many genetic factors that contributes to spermato- 15 Montanelli, L. et al. (2004) A Mutation in the follicle-stimulating
hormone receptor as a cause of familial spontaneous ovarian hyper-
genetic failure. The role of FSHR haplotypes in ovarian
stimulation syndrome. J. Clin. Endocrinol. Metab. 89, 1255–1258
function has not been investigated. 16 Simoni, M. et al. (2002) Isoforms and single nucleotide polymorphisms
of the FSH receptor gene: implications for human reproduction. Hum.
Perspectives Reprod. Update 8, 413–421
17 Shastry, B.S. (2002) SNP alleles in human disease and evolution.
The Ala307Thr and Asn680Ser polymorphisms in the
J. Hum. Genet. 47, 561–566
FSHR represent the first example of a genetic marker that 18 Gromoll, J. et al. (1994a) Characterization of the 5 0 flanking region of
might have important therapeutic implications for fertil- the human follicle-stimulating hormone receptor gene. Mol. Cell.
ity treatment. Clinical studies show that SNPs in exon 10 Endocrinol. 102, 93–102
of the FSHR determine the ovarian FSH threshold. 19 Heckert, L.L. et al. (2000) The USF proteins regulate transcription of
Therefore, these SNPs need to be considered in COH for the follicle-stimulating hormone receptor but are insufficient for cell
specific expression. Mol. Endocrinol. 14, 1836–1848
assisted reproduction techniques in women with normal
20 Nordhoff, V. et al. (2003) Targeted expression of human FSH receptor
ovarian function. Future interventional, prospective Asp567Gly mutant mRNA in testis of transgenic mice: role of human
studies should be based on a pharmacogenetic approach FSH receptor promoter. Asian J. Androl. 5, 267–275
to ovarian stimulation that assesses the efficacy and safety 21 Putowski, L.T. (2004) Human follicle-stimulating hormone receptor
of patient-tailored stimulation protocols based on the promoter/enhancer activity is inhibited by transcriptional factors,
from the upstream stimulating factors family, via E-box and newly
FSHR genotype. Finally, elucidation of the mechanism by
identified initiator element (Inr) in FSHR non-expressing cells.
which different FSHR isoforms respond to different Gynecol. Endocrinol. 19, 9–17
isoforms of FSH might provide new therapeutic concepts 22 Xing, W. and Sairam, M.R. (2001) Characterization of regulatory
in reproductive endocrinology based on the genetic com- elements of ovine follicle-stimulating hormone (FSH) receptor gene:
plexity of FSHR function. The role of E-box in the regulation of ovine FSH receptor expression.
Biol. Reprod. 64, 579–589
23 Heckert, L.L. and Griswold, M.D. (2002) The expression of the follicle-
Acknowledgements stimulating hormone receptor in spermatogenesis. Recent Prog.
The authors work was supported by the German Research Foundation Horm. Res. 57, 129–148
(DFG-526/1–1). The language editing of S. Nieschlag, MA is gratefully 24 Griswold, M.D. and Kim, J.S. (2001) Site-specific methylation of the
acknowledged.
promoter alters DNA-protein interactions and prevents follicle-
stimulating hormone receptor gene transcription. Biol. Reprod. 64,
References 602–610
1 Simoni, M. and Nieschlag, E. (1995) FSH in therapy: Physiological 25 Wunsch, A. et al. Single-nucleotide polymorphisms in the promoter
basis, new preparations and clinical use. Reprod. Med. Rev. 4, 163–177 region influence the expression of the human follicle-stimulating
2 Nieschlag, E. et al. (1999) Role of FSH in the regulation of hormone receptor. Fertil. Steril. (in press)
spermatogenesis: clinical aspects. Clin. Endocrinol. (Oxf.) 51, 139–146 26 Simoni, M. et al. (1999) Mutational analysis of the follicle-stimulating
3 Simoni, M. et al. (1997) The follicle-stimulating hormone receptor: hormone (FSH) receptor in normal and infertile men: identification
biochemistry, molecular biology and pathophysiology. Endocr. Rev. 18, and characterization of two discrete FSH receptor isoforms. J. Clin.
739–773 Endocrinol. Metab. 84, 751–755
www.sciencedirect.com
Review TRENDS in Endocrinology and Metabolism Vol.16 No.8 October 2005 373

27 Gromoll, J. and Simoni, M. (2001) Follicle-stimulating hormone of the follicle stimulating hormone receptor gene in women with
receptor and twinning. Lancet 357, 230 premature ovarian failure, resistant ovary syndrome and polycystic
28 Perez Mayorga, M. et al. (2000) Ovarian response to FSH stimulation ovary syndrome. Clin. Endocrinol. (Oxf.) 51, 97–99
depends on the FSH receptor genotype. J. Clin. Endocrinol. Metab. 85, 37 Sundblad, V. et al. (2004) Screening of FSH receptor gene in Argentine
3365–3369 women with premature ovarian failure (POF). Mol. Cell. Endocrinol.
29 Sudo, S. et al. (2002) Genetic and functional analyses of polymorph- 222, 53–59
isms in the human follicle-stimulating hormone receptor gene. Mol. 38 Tong, Y. et al. (2001) Absence of mutations in the coding regions
Hum. Reprod. 8, 893–899 of follicle-stimulating hormone receptor gene in Singapore Chinese
30 de Castro, F. et al. (2003) Role of follicle-stimulating hormone receptor women with premature ovarian failure and polycystic ovary syn-
Ser680Asn polymorphism in the efficacy of follicle-stimulating hor- drome. Horm. Metab. Res. 33, 221–226
mone. Fertil. Steril. 80, 571–576 39 Hasbargen, U. et al. (2001) S680N substitution of the follicle-
31 de Castro, F. et al. (2004) Human controlled ovarian hyperstimulation stimulating hormone receptor is a common polymorphism not
outcome is a polygenic trait. Pharmacogenetics 14, 285–293 associated with spontaneous human twinning. Eur. J. Med. Res. 6,
32 Behre, H.M. et al. Significance of a common single nucleotide 315–316
polymorphism in exon 10 of the follicle-stimulating hormone (FSH) 40 Asatiani, K. et al. (2002) Distribution and function of FSH receptor
receptor gene for the ovarian response to FSH: a pharmacogenetic genetic variants in normal men. Andrologia 34, 172–176
approach to controlled ovarian hyperstimulation. Pharmacogenetics 41 Saxena, D. et al. (2004) Liver receptor homolog-1 stimulates the
15, 451–456 progesterone biosynthetic pathway during follicle-stimulating
33 Daelemans, C. et al. (2004) Prediction of severity of symptoms in hormone-induced granulosa cell differentiation. Endocrinology 145,
iatrogenic ovarian hyperstimulation syndrome by follicle-stimulating 3821–3829
hormone receptor Ser680Asn polymorphism. J. Clin. Endocrinol. 42 Krishnamurthy, H. et al. (2003) Postendocytotic trafficking of the
Metab. 89, 6310–6315 follicle-stimulating hormone (FSH)-FSH receptor complex. Mol.
34 Greb, R. et al. A common single nucleotide polymorphism in exon 10 Endocrinol. 17, 2162–2176
of the human follicle stimulation hormone receptor is a major deter- 43 Findlay, J.K. et al. (2002) Recruitment and development of the follicle;
minant of length and hormonal dynamics of the menstrual cycle. the roles of the transforming growth factor-beta superfamily. Mol.
J. Clin. Endocrinol. Metab.: in press Cell. Endocrinol. 191, 35–43
35 Laven, J.S. et al. (2003) Follicle-stimulating hormone receptor 44 Tomic, D. et al. (2004) Ovarian follicle development requires Smad3.
polymorphisms in women with normogonadotropic anovulatory Mol. Endocrinol. 18, 2224–2240
infertility. Fertil. Steril. 80, 986–992 45 Ahda, Y. et al. FSH receptor gene haplotype distribution in normo-
36 Conway, G.S. et al. (1999) Mutation screening and isoform prevalence zoospermic and azoospermic men. J. Androl. 26, 494–499

Disease models of autoimmunity


Look out for the November Special Issue of Trends in Immunology!

This issue will include clinical and scientific perspectives on whether, and how, disease models of autoimmunity have increased
our understanding of human autoimmune diseases. The authors are all experts in their field and selected articles within the issue
include:
Virtues and pitfalls of EAE for development of therapies for multiple sclerosis
Lawrence Steinman and Scott S. Zamvil
SLE: translating lessons from model systems to human disease
Ram Raj Singh
Of mice and men: use of animal models to identify possible interventions for the prevention of
autoimmune type 1 diabetes in humans
David V. Serreze and Yi-Guang Chen
NK cells: elusive players in autoimmunity
Sofia Johansson, Louise Berg, Håkan Hall and Petter Höglund

MHC II molecules in inflammatory diseases: interplay of qualities and quantities


Manuel A. Friese, E. Yvonne Jones and Lars Fugger

www.sciencedirect.com

You might also like