Sunteți pe pagina 1din 11

Molecular and Cellular Endocrinology 383 (2014) 203213

Contents lists available at ScienceDirect

Molecular and Cellular Endocrinology


journal homepage: www.elsevier.com/locate/mce

Review

Luteinizing hormone and human chorionic gonadotropin: Origins


of difference
Janet Choi a,, Johan Smitz b,1
a
The Center for Womens Reproductive Care at Columbia University, New York, NY, United States
b
UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium

a r t i c l e i n f o a b s t r a c t

Article history: Luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are widely recognized for their roles
Received 25 September 2013 in ovulation and the support of early pregnancy. Aside from the timing of expression, however, the dif-
Received in revised form 6 December 2013 ferences between LH and hCG have largely been overlooked in the clinical realm because of their similar
Accepted 12 December 2013
molecular structures and shared receptor. With technologic advancements, including the development of
Available online 21 December 2013
highly puried and recombinant gonadotropins, researchers now appreciate that these hormones are not
as interchangeable as once believed. Although they bind to a common receptor, emerging evidence sug-
Keywords:
gests that LH and hCG have disparate effects on downstream signaling cascades. Increased understanding
Gonadotropin
Lutropin
of the inherent differences between LH and hCG will foster more effective diagnostic and prognostic
Luteinizing hormone assays for use in a variety of clinical contexts and support the individualization of treatment strategies
Human chorionic gonadotropin for conditions such as infertility.
Luteinizing hormone/choriogonadotropin 2013 The Authors. Published by Elsevier Ireland Ltd. Open access under CC BY-NC-SA license.
receptor
Luteinizing hormone receptor

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204
2. Materials and methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204
3. Molecular structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204
3.1. Gonadotropin variants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204
4. Circulating forms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
4.1. Luteinizing hormone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
4.2. Human chorionic gonadotropin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
5. Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
6. Receptor binding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
6.1. Luteinizing hormone/choriogonadotropin receptor. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
6.2. Signaling pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
6.3. Extragonadal LHCGR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
7. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
Disclosure statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211

Corresponding author. Address: The Center for Womens Reproductive Care at Columbia University, 1790 Broadway, 2nd Floor, New York, NY 10019, United States. Tel.:
+1 646 756 8282; fax: +1 646 756 8280.
E-mail addresses: jmc30@columbia.edu (J. Choi), johan.smitz@uzbrussel.be (J. Smitz).
1
Address: UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium. Tel.: +32 2 477 50 52; fax: +32 2 477 50 60.

0303-7207 2013 The Authors. Published by Elsevier Ireland Ltd. Open access under CC BY-NC-SA license.
http://dx.doi.org/10.1016/j.mce.2013.12.009
204 J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213

1. Introduction Table 1
Characteristics of hLHa and hCGb.

It is well established that luteinizing hormone (LH) and human hLH hCG
chorionic gonadotropin (hCG) play key roles in the reproductive Molecular weight (Da) 30,000 36,000c
cycle. Textbooks recognize the disparate endogenous functions of
a Subunit
these hormones, underscoring the role of LH in follicular matura- Gene location 6q12q21 6q12q21
tion and ovulation induction, while acknowledging the essential Size of mature subunit (no. of amino acids) 92 92
role of hCG in early pregnancy survival. During early pregnancy, b Subunit
hCG is vital to support secretion of progesterone by the corpus lut- Gene location 19q13.32 19q13.32
eum, without which a pregnancy cannot persist (Fritz and Speroff, Size of mature subunit (no. of amino acids) 121 145
2011; Mesiano, 2009). Nonetheless, LH and hCG are frequently de- hLH, human luteinizing hormone; hCG, human chorionic gonadotropin.
picted as interchangeable, with one contemporary text stating that a
Bulun (2011).
b
the b subunits [of human LH (hLH) and hCG] confer identical bio- Cole (2010).
c
Molecular weight of hyperglycosylated form is 40,00041,000 Da.
logic activities when associated with the a subunit (Bulun, 2011).
Although similar in structure and function, LH and hCG are dis-
tinct molecular entities with divergent patterns of expression and
physiologic functions. LH and hCG share a common receptor, yet
The high degree of sequence conservation among CGB genes,
each hormone triggers a unique cascade of events following recep-
combined with the fact that CG is detected only in equine and pri-
tor binding (Casarini et al., 2012; Gupta et al., 2012). Moreover,
mate species (whereas LH is found in all vertebrates) suggests that
within the same hormone family, individual isoforms exhibit un-
CG is a relatively recent evolutionary derivative of LH. The amino
ique characteristics regarding half-life and biologic functions.
acid sequences of human LH (hLH) and hCG are highly conserved,
Emerging data suggest that these differences between LH and
sharing 82% homology (Bulun, 2011). hCG retains the full
hCG have functional signicance. Identifying the unique roles of
145 amino-acid complement in its b-subunit. In contrast, the
LH and hCG is key to understanding both normal physiologic pro-
b-subunit of LH undergoes cleavage of its 24-amino acid leader se-
cesses (e.g., reproduction, placentation) and dysfunctional states
quence to generate its nal 121-amino acid sequence.
(e.g., infertility, gestational and non-gestational neoplasms). Ad-
As a result of structural differences and post-translational mod-
vances in gonadotropin purication and recombinant technology
ications, hCG is more stable and has a longer circulating half-life
have helped to differentiate the varied actions of LH and hCG. In
than LH. The half-lives of these molecules typically are expressed
turn, this has improved the diagnosis and treatment of human dis-
as a range (on the order of minutes for LH and hours for hCG),
ease, most signicantly in the context of infertility.
reecting the heterogeneity of circulating isoforms. The shorter
half-life of LH is physiologically important, as it allows for the pro-
duction of LH pulses. The longer half-life of hCG and its greater
2. Materials and methods
receptor binding afnity make it more biologically active than
hLH (Rahman and Rao, 2009; Rao, 1979).
A comprehensive literature review of LH and hCG was con-
ducted, focusing on differentiation in terms of hormone structure,
expression, modication, receptor activation, and clinical use. Sci- 3.1. Gonadotropin variants
entic literature was identied via interrogation of the MEDLINE
database using relevant search terms, including but not limited In the endogenous state, LH and hCG consists of isohormone
to: gonadotropin, human chorionic gonadotropin, luteinizing mixtures that result from (1) post-translational modication of
hormone, luteinizing hormone/choriogonadotropin receptor, the native proteins; (2) metabolism to form truncated or nicked
lutropin, and lutropin receptor. Results were limited to articles intermediates; and (3) natural sequence variants (Bergendah and
published in English. Additional resources were extracted from Veldhuis, 2001; Cole, 2009; Stanton et al., 1993). Post-translational
clinical texts and review articles. Article inclusion was predicated modication chiey consists of the addition of various carbohy-
on relevance to the topic without use of an objective set of criteria, drate side chains, including sialic and sulfonic acid moieties, thus
given that the review is descriptive rather than systematic in creating a variety of isoforms (upwards of 30 for LH and 15 for
nature. hCG) that differ with respect to half-life, bioactivity, and signaling
properties (Arey and Lopez, 2011; Bergendah and Veldhuis, 2001;
Cole, 2009; Stanton et al., 1993). Obviously, the number of isoform
3. Molecular structure classes for each hormone is determined by our capacity for analyt-
ical discrimination. Because glycosylation affects the size and con-
Both LH and chorionic gonadotropin (CG) are heterodimeric gly- formation of the molecule as well as access to binding sites,
coproteins comprised of a and b subunits. Along with thyroid- differences in interactions with their cognate receptor are to be ex-
stimulating hormone (TSH) and follicle-stimulating hormone pected (Arey and Lopez, 2011).
(FSH), they share a common 92-amino acid a subunit, whose gene Naturally occurring variants of both LH and hCG have been
resides on chromosome 6q12q21 in humans (Bulun, 2011; Naylor characterized. A variant of LH that has an additional glycosylation
et al., 1983). The unique functions and receptor-binding capacity of site compared with wild-type LH is relatively common within cer-
each of these hormones stem from differences among the b sub- tain populations (Lamminen and Huhtaniemi, 2001; Nilsson et al.,
units (Table 1). Transcription of the b-subunit is the rate-limiting 1997). Data have been collected showing that variant LH differs
step in LH and CG production (Nagirnaja et al., 2010). The genes from normal LH in its biological effects (Haavisto et al., 1995). This
for the LH and hCG b subunits are located within a cluster of seven LH variant has been associated with unexplained infertility and
similar sequences on human chromosome 19q13.32 (Boorstein subfertility due to ovulatory dysfunction in females, and with slo-
et al., 1982; Jameson and Hollenberg, 1993). One of those se- wed pubertal progression in males (Raivio et al., 1996; Takahashi
quences encodes the b-subunit of LH (LHB); four are hCG b-subunit et al., 1998, 1999). Preliminary data also suggest that the variant
coding genes (CGB, CGB5, CGB7, and CGB8), and the remaining two LH is more prevalent among women who demonstrate resistance
are pseudogenes (CGB1, CGB2). to ovarian stimulation with recombinant human FSH, compared
J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213 205

with those who exhibit moderate or strong responses (Alviggi pin-releasing hormone (GnRH), estradiol, and testosterone
et al., 2011). Men with one or two copies of the variant allele have (Bergendah and Veldhuis, 2001; Lambert et al., 1998; Wide and
lower circulating LH levels compared with homozygotes for the Bakos, 1993).
wild-type allele (Huhtaniemi et al., 2010). However, the ratio of Variations in LH isoform composition are also observed during
biological-to-immunological activity (a measure of relative LH the reproductive life cycle. In general, LH isoforms with shorter
biopotency) for variant LH is greater than that of wild-type LH half-lives but increased biopotency are present in younger post-
(Haavisto et al., 1995). pubertal women, whereas longer-lived LH species are the predom-
A less common LH variant resulting in a single amino acid sub- inant form detected in post-menopausal women (Reader et al.,
stitution in the b subunit (serine for glycine at amino acid 102) has 1983; Wide, 1985). LH in the latter group also has an increased ra-
been associated with menstrual disorders and male infertility tio of biological-to-immunological activity (Marrama et al., 1983).
(Hashad et al., 2012; Ramanujam et al., 1999, 2000). Little is known Interestingly, women with polycystic ovarian syndrome (PCOS)
about how this rare variant affects the functionality of LH. Based on an endocrine condition associated with reduced fertilityappear
in vivo experiments, Wide et. al. showed that an extra glycosylation to predominantly secrete LH isoforms that have a high ratio of bio-
site on variant LH compared to the wild type, allows for greater logical-to-immunological activity (Ding and Huhtaniemi, 1991;
sialylation and results in a longer half-life in serum. Granulosa Ropelato et al., 1999). Molecular analysis of LH in women with
cells are triggered typically by distinct pulses of LH; however, PCOS has revealed a substantial decrease in sulfonation at midcy-
because of its prolonged residence in serum, granulosa cells are cle, accompanied by a slight increase in sialylation compared with
exposed to decreased pulse amplitudes of variant LH. Thus, Wide, non-affected individuals (Wide et al., 2007). The net result is an
et. al. proposed that the longer half-life of variant LH may be a increase in basic LH isoforms relative to women without PCOS
contributing factor to the reproductive disorders observed (Wide (Ropelato et al., 1999; Wide et al., 2007). It is unclear what inu-
et al., 2010). ence, if any, these changes have on the half-life of LH. It is known
Although hCG polymorphisms with functional signicance are that that basic LH isoforms are, in general, cleared more rapidly
generally rare, variants in the b subunit of hCG with putative than acidic isoforms, yet based on ndings regarding the specic
protective or detrimental effects on recurrent miscarriage risk have effects of sialylation vs. sulfonation on LH half-life (Burgon et al.,
been identied (Nagirnaja et al., 2012; Rull et al., 2008). The se- 1996; Wide et al., 2009), the changes observed in women with
quence changes associated with increased risk of recurrent miscar- PCOS would be expected to increase LH residence time in circula-
riage occur in the most commonly transcribed hCG b-subunit tion. This question is made more complex by the apparent effect of
genes, CGB5 and CGB8, and result in conformational changes to body mass index (BMI) on the LH prole of women with PCOS
the proteins structure (Nagirnaja et al., 2012). One of the b-subunit (Srouji et al., 2007). Current dogma posits that increased basal
missense mutations in particular-hCG b p.Val56Leu adversely im- secretion of LH plays a greater role than altered post-translational
pacts its ability to bind efciently with its a-subunit with only modication in the elevated serum LH concentrations that have
10% forming the complete hCG heterodimer. However, additional been observed among women with PCOS.
studies show increased bioactivity of this resultant variant hCG In men, LH stimulates testosterone production by Leydig cells of
(possibly due to subtle conformational changes) when bound to the testis. The degree of LH sialylation and the ratio of biological-
LHCGR, increasing the impact of this more sparsely produced var- to-immunological activity of LH are generally greater in men than
iant hCG. The paucity of polymorphisms that have been identied in pre-menopausal women (Bergendah and Veldhuis, 2001; Wide
in hCG imply that only modest changes in CGB5 and CGB8 are et al., 2007). Unlike women, however, the biological-to-immuno-
compatible with a successful pregnancy outcome. logical activity ratio of LH decreases with age in men (Marrama
et al., 1984; Warner et al., 1985). A shift toward more LH species
that have a shorter serum half-life has been described in obese
4. Circulating forms men, which may be a source of the reduced testosterone levels
reported in those individuals (Castro-Fernandez et al., 2000).
4.1. Luteinizing hormone
4.2. Human chorionic gonadotropin
Gonadotrophic cells of the anterior pituitary produce LH, which
regulates ovulation. Over the course of a normal menstrual cycle, a At least four physiologically important isoforms of hCG have
surge in LH spurs ovulation from the dominant follicle. This pro- been described, including regular hCG, hyperglycosylated hCG (h-
cess initiates during the mid-follicular phase, when LH stimulates hCG), hyperglycosylated free hCG b subunit, and pituitary hCG. Dif-
androgen production in thecal cells. These androgens are aroma- ferent cell types produce these isoforms, which have unique,
tized in the granulosa cells to estrogen. Prolonged, sustained estro- although sometimes overlapping, functions. Each of the four vari-
gen levels generate positive feedback on the pituitary gland and ants shares a common protein sequence (at least for the b subunit),
the subsequent LH surge that results in ovulation (Liu and Yen, but each is modied differently, resulting in isoforms with dispa-
1983) Post-ovulation, LH promotes progesterone production, sup- rate half-lives and biologic functions. The distribution and quantity
porting development of the corpus luteum (Van de Wiele et al., of hCG isoforms varies over the course of pregnancy and in the con-
1970). text of aberrant development or disease. For example, in early
The composition of LH isoforms, their longevity, and their bio- pregnancy, an abnormally low proportion of h-hCG to total hCG
activity vary throughout the menstrual cycle. The ratio of biologi- (<50%) indicates a failing pregnancy, (Kovalevskaya et al., 2002;
cal-to-immunological activity is lowest during the luteal phase, Sasaki et al., 2008), while decreased rst trimester serum free b-
rising slowly during the follicular phase to a peak at mid-cycle hCG levels have been associated with ectopic pregnancies (Borrelli
(Anobile et al., 1998). This corresponds with the pinnacle of LH et al., 2003). Conversely, elevated h-hCG can indicate choriocarci-
sialylation and a marked trough in sulfonation (Wide and Eriksson, noma or Down syndrome pregnancy (Cole et al., 1999; Elliott
2013). Earlier studies had established that sialylation prolongs LH et al., 1997; Palomaki et al., 2007).
half-life, whereas sulfonation hastens protein elimination (Burgon Regular hCG is secreted by fused and differentiated syncytio-
et al., 1996; Wide et al., 2009). Changes in the extent of glycosyla- trophoblasts. It is the predominant hCG species measured in serum
tion during the menstrual cycle appear to be regulated by the during pregnancy, after the initial early surge of h-hCG (Cole,
pituitary gland, under the inuence of such factors as gonadotro- 2010). The chief role of hCG historically has been the promotion
206 J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213

Table 2
Production and biological functions of hCG isoforms.

Isoform (source) Function(s)


hCG (syncytiotrophoblast cells)  Promotes corpus luteum progesterone secretion
 Stimulates angiogenesis within pregnant uterus
 Differentiates cytotrophoblasts and may stimulate cytotrophoblast metalloproteinases, facilitating
pregnancy implantation
 Inhibits immune response and macrophage phagocytosis at utero-placental junction
 Relaxes uterine muscle contraction and permits uterine growth during pregnancy
 May promote differentiation and development of umbilical cord and fetal organs
 Blastocyst cross-talk with endometrium prior to implantation
 LHCG receptors found in sperm and fallopian tubes suggesting pre-pregnancy communication roles
 hCG receptors in hippocampus, hypothalamus and brain stem suggests its role in hyperemesis
gravidarum
Hyperglycosylated hCG (cytotrophoblast cells normal and  Promotes implantation via cytotrophoblast invasion; also inhibits apoptosis and stimulates invasion
malignant cells) by choriocarcinoma cells
Free b-hCG (non-trophoblastic malignancies)  Inhibits non-trophoblastic malignancies apoptosis; promotes cancer cell growth
Pituitary hCG (gonadotroph cells of the anterior pituitary)  Mimics LH functions during the normal menstrual cycle

hCG, human chorionic gonadotropin; LH, luteinizing hormone. Adapted from Cole (2010).

of progesterone secretion by the corpus luteum in early pregnancy. Hyperglycosylated free hCG b subunits are found at low levels
However, as noted by Cole, hCG levels continue to increase after during normal pregnancy and are also produced by non-tropho-
hCG is no longer needed for progesterone production, suggesting blastic malignancies (Butler et al., 2000; Cole, 2009; Cole et al.,
other functions for this hormone during pregnancy (Cole, 2009). 1988; Iles et al., 1990). Akin to h-hCG, the hyperglycosylated free
It is now believed that hCG is involved in placentation through b subunit is characterized by N- and O-linked oligosaccharides
activities such as maintaining angiogenesis of the uterine vascula- containing a large number of sugar residues. Free b subunits are
ture and promoting differentiation of cytotrophoblasts into syncy- not only a biomarker of malignancy but are also believed to play
tiotrophoblasts (Rao and Alsip, 2001; Shi et al., 1993; Zygmunt an active role in cancer development by preventing apoptosis
et al., 2002). Emerging evidence also suggests roles for regular and promoting carcinogenesis (Butler et al., 2000; Cole and Butler,
hCG in fostering implantation, preventing fetal rejection, coordi- 2008). Indeed, elevated levels of free b-hCG in urine samples of pa-
nating uterine and fetal growth, and, potentially, growth and tients with malignancies increase the likelihood for metastasis and
development of fetal organs (Cole, 2010). The varieties of estab- mortality (Butler et al., 2000; Iles et al., 1996). The anti-apoptotic
lished and putative functions for regular hCG are listed in Table 2. effects of free b-hCG may result from antagonistic inhibition of
Secreted by root or extravillous cytotrophoblasts, h-hCG con- other growth factor receptors, rather than interaction with the
centrations peak early in the rst trimester. The percentage of canonical receptor (Butler and Iles, 2004). Because it lacks the a
hCG in the form of h-hCG subsequently declines, becoming a minor subunit, free b-hCG cannot bind to the lutropin receptor (Cole,
component of total hCG measurement during the last two trimes- 2009).
ters. The key differentiator between regular hCG and h-hCG is the Pituitary hCG is secreted by gonadotrophic cells of the anterior
complexity of the oligosaccharide side chains: h-hCG tends toward pituitary. The higher number of sulfonated side chains of pituitary
oligosaccharides with a greater number of sugar residues. Conse- hCG gives it a shorter half-life than its placental counterpart (Bir-
quently, the molecular weight of h-hCG is in the range of ken et al., 1996). Pituitary hCG is present in all adults, including
40,00041,000 Da, whereas that of regular hCG is approximately men and pre-/post-menopausal women, and is suppressed with
36,000 Da. The additional sugar moieties prevent normal protein GnRH agonist treatment (Odell and Grifn, 1987, 1989). During a
folding, thus exposing alternative receptor binding sites and alter- regular menstrual cycle, pituitary hCG secretion parallels that of
ing the hormones functionality (Cole, 2010). Indeed, results of re- LH, although the half-life of pituitary hCG is shorter than that of
cent experiments by Berndt and colleagues indicate that h-hCG- LH. As much as one-third of follicular-stage LH activity may derive
mediated angiogenesis may occur independent of activation of its from hCG during a natural menstrual cycle (Cole, 2010). The func-
cognate receptor (Berndt et al., 2013). tion of pituitary hCG is generally not known, but its concomitant
The association between pregnancy loss and low h-hCG levels temporal appearance with LH during the menstrual cycle and com-
supports a key role for this variant in implantation (Kovalevskaya mon receptor suggest that its functionality may mimic LHstimu-
et al., 2002; Sasaki et al., 2008). The importance of h-hCG in lating follicular maturation, inducing ovulation, and supporting
implantation/invasion is also evident in the context of choriocarci- progesterone production during the luteal phase (Cole, 2010). In
noma, as a pathologic process (Cole, 2010). Current data suggest contrast, expression of the LHB gene is down-regulated in preg-
that h-hCG and hCG act in conjunction to forewarn the endome- nancy (Rull and Laan, 2005) suggesting that while hCG might have
trium of impending implantation, to foster growth and differentia- a functional role during the normal menstrual cycle, LH cannot
tion of trophoblast cells, and to establish placental villous substitute for hCG in supporting pregnancy.
structures.
During pregnancy, the amount of total and h-hCG in serum and
urine demonstrates a high degree of inter-individual variability 5. Metabolism
(Cole, 2010). This endogenous variation may be normalized at
the receptor level via downregulation in the presence of high Gonadotropins undergo a series of metabolic conversions be-
hCG concentrations, or through the spare receptor concept, which fore ultimately being excreted in urine (Braunstein, 2011; Cole,
generates the same response regardless of the proportion of recep- 2009; OConnor et al., 1998). This process has been well character-
tors activated. The end result is that, unless the hCG level is insuf- ized for hCG. One of the initial steps in hCG degradation is proteo-
cient to support its basic functionality, variation is compatible lytic cleavage of the b subunit (possibly by human leukocyte
with normal pregnancy. elastase), which generates a nicked form of the protein. Nicked
J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213 207

hCG rapidly dissociates into its component a and b subunits. that occur within certain clinical contexts may generate erroneous
(Alternately, hCG that has not been nicked may slowly dissociate results. It is important, therefore, to be aware of the limitations for
prior to enzymatic cleavage.) Therefore, h-hCG dissociates more a particular assay when interpreting laboratory ndings.
readily than regular hCG in this context. The nicked b subunit is
further degraded, predominantly in the kidneys, resulting in a b-
core fragment. A comparable LH b-core fragment has been isolated 6. Receptor binding
from human pituitary and urine samples (OConnor et al., 1998).
Given the array of glycosylated hCG forms and the potential for 6.1. Luteinizing hormone/choriogonadotropin receptor
nicked and dissociated varieties therein, a multitude of hCG degra-
dation products are detected in the urine of pregnant women, of LH and hCG bind to and activate a common receptor, the LH/
which the b-core fragment is most prevalent from 7 weeks gesta- choriogonadotropin receptor (LHCGR), also known as the LH or
tion onward (Cole, 2009). In total, more than 30 hLH isoforms and lutropin receptor (LHR) (Ascoli et al., 2002; Puett et al., 2007). This
15 variants of hCG have been identied from serum and urine sam- 675-amino-acid, G-protein-coupled receptor is a member of the
ples (Cole, 2009; Stanton et al., 1993). rhodopsin subfamily of glycoprotein hormone receptors, as are
The extent of gonadotropin glycosylation dictates molecular the FSH and TSH receptors. Like all G-protein coupled receptors,
charge, which, in turn, governs clearance rate, such that more acidic LHCGR is anchored within the cell membrane by seven transmem-
isoforms have a longer half-life in vivo (Lambert et al., 1998). Inves- brane domains. The unusually large extracellular domain of LHCGR
tigators have also noted that the sialic acid content of LH positively is characterized by leucine-rich repeats and multiple sites for gly-
correlates with metabolic clearance rate (Burgon et al., 1996). In cosylation (Fig. 1). The human LHCGR gene is located on chromo-
other words, a greater degree of sialylation increases LH retention some 2p21, in the same general region as the FSH receptor
in circulation. This observation holds true in the context of GnRH (Rousseau-Merck et al., 1993, 1990).
receptor blockade as well. In response to GnRH receptor blockade, Consistent with its role in female reproduction, LHCGR is ex-
LH levels decrease and the overall glycosylation prole shifts: the pressed by multiple cell types in the ovary, including thecal, luteal,
mean number of sialic acid residues per molecule increases and interstitial, and differentiated granulosa cells (Ascoli et al., 2002).
the number of sulfonated residues decreases (Wide et al., 2009). Expression of LHCGR in these cell types during the ovarian cycle
The more sialylated and less sulfonated LH isoforms have longer is dynamic rather than constitutive, reecting changes in the hor-
circulating half-lives. A similar effect occurs naturally via an LH monal milieu (Menon and Menon, 2012; Menon et al., 2010).
polymorphism that results in an extra site for glycosylation. This LHCGR is downregulated transiently after the pre-ovulatory LH
LH variant has an increased number of sialic acid residues per mol- surge, reaches a maximum at mid-luteal phase, and decreases with
ecule, resulting in a circulating half-life approximately 40% longer corpus luteum regression. This pattern is the inverse of what has
than wild-type LH in humans (Wide et al., 2010). been observed for bioactive LH isoforms, wherein peak biologic-
hCG glycosylation also affects its longevity in circulation. Meta- to-immunologic activity occurs at midcycle and reaches its lowest
bolic clearance rates of deglycosylated, b-core fragment, and desi- point during the luteal phase (Anobile et al., 1998). The mecha-
alylated hCG are all faster relative to hCG, with the greatest degree nisms that govern temporal changes in LHCGR expression are not
of acceleration observed for desialylated hCG (Liu et al., 1989; Rosa yet fully elucidated but are generally believed to involve transcrip-
et al., 1984). The effects of naturally occurring hCG polymorphisms tional as well as post-transcriptional regulation (Ascoli et al., 2002;
on metabolic clearance rate or protein half-life have yet to be Menon and Menon, 2012). Induction of LHCGR expression in differ-
described. entiating granulosa cells in response to FSH is an example of pre-
As a valuable early marker of pregnancy as well as various dis- dominantly transcriptional control, whereas desensitization of
ease states, the ability to accurately measure hCG in its myriad LHCGR in response to LH or hCG exposure includes an element of
forms is particularly important. Beyond pregnancy detection, post-transcriptional regulation. Agonist-mediated desensitization
hCG measurement is useful for identifying gestational trophoblas- of LHCGR involves a reduction in cell surface receptor concentra-
tic disease, risk of failing or abnormal pregnancy, and non-gesta- tion, as well as diminished mRNA abundance. The decrease in
tional malignancies. The predominant hCG species characteristic mRNA has been attributed to increased transcript degradation
of each of these conditions differs. In early pregnancy and gesta- rather than suppressed transcription (Lu et al., 1993).
tional trophoblastic disease, h-hCG is the dominant form, whereas Through a series of studies involving rat and human ovarian
elevated free b-subunit production is characteristic of certain models, Menon and colleagues identied a binding protein, meva-
cancers (Cole et al., 1988; Elliott et al., 1997; Hoermann et al., lonate kinase, which appears to regulate LHCGR expression by has-
1992; Iles et al., 1990; Mock et al., 2000). Abnormalities in h- tening degradation of LHCGR mRNA (Menon et al., 2010). An
hCG are also used to assess pregnancy-related riskselevated inverse correlation was observed between LHCGR mRNA and mev-
maternal blood levels during the rst and second trimester are alonate kinase activity: when one was elevated, the other was de-
associated with fetal aneuploidy risk such as Down syndrome creased (Nair et al., 2002). Moreover, inhibition of mevalonate
while low h-hCG in the latter stages of pregnancy is emerging kinase prevented downregulation of LHCGR in response to hCG,
as a putative marker of preeclampsia risk (Bahado-Singh et al., whereas overexpression of mevalonate kinase ameliorated induc-
2002; Cole, 2009). tion of LHCGR in the presence of FSH and estradiol (Ikeda et al.,
In the case of both LH and hCG, the multiple circulating forms 2008; Wang et al., 2007).
complicate their measurement in blood and urine (Cole, 2009; Alternative splicing of LHCGR mRNA has been implicated in reg-
OConnor et al., 1998). For example, total hCG assays, which are ulation of the ovarian cycle and luteolysis (Dickinson et al., 2009;
the preferred measurement tool used by clinical laboratories, are Minegishi et al., 2007). Splice variants differ in exon content and,
effective for quantifying regular hCG, but are generally suboptimal consequently, in their ability to bind or respond to LH or hCG.
for detecting h-hCG (Cole, 2009). Moreover, in addition to differ- Using human corpora lutea obtained at various stages of the ovar-
ences in glycosylation and degradation rates, genetic variants have ian cycle, investigators have demonstrated quantitative, but not
been found to impede detection of LH by immunoassay (OConnor qualitative, differences in the prole of LHCGR splice variants, indi-
et al., 1998; Pettersson and Soderholm, 1991). Detection limits must cating that the relative abundance of mRNA species changes
also be taken into account when assessing gonadotropin presence or throughout the ovarian cycle but their identity did not (Dickinson
absence in clinical testing. Extremes in gonadotropin concentrations et al., 2009; Madhra et al., 2004).
208 J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213

Fig. 1. Glycoprotein hormone receptor structure. (A) Illustration of the short intracellular, 7-pass transmembrane, and large extracellular domains characteristic of
glycoprotein hormone receptors (e.g., the luteinizing hormone/choriogonadotropin receptor). (B) Three-dimensional schematic of the leucine-rich repeats (LRRs) within the
extracellular domain. (C) Structure of an individual LRR, wherein X represents any amino acid. Reproduced with permission from Vassart et al. (2004).

In vitro evidence suggests that gonadotropin signaling through with infertility, including PCOS. A genome-wide association study
LHCGR inuences the expression of individual splice variants that detected a link between a polymorphic marker in the region
(Dickinson et al., 2009). Previous experiments had shown that of LHCGR and PCOS in Han Chinese women (which was conrmed
one of the identied LHCGR splice variants forms complexes with in a subsequent case-control cohort) spurred several similar investi-
LHCGR or the FSH receptor, inuencing receptor expression and gations in other ethnic groups (Chen et al., 2011; Shi et al., 2012). The
preventing the latter from reaching the cell surface (Nakamura specic polymorphic marker associated with PCOS in the Han Chi-
et al., 2004; Yamashita et al., 2005). More recently, a truncated nese population failed to correlate with PCOS in European popula-
form of LHCGR lacking transmembrane or carboxyterminal do- tions (Eriksen et al., 2012; Mutharasan et al., 2013); however,
mains negatively regulated the expression and activity of the linkage within the genomic region of LHCGR was maintained upon
full-length protein (Dickinson et al., 2009). Further study is needed ne mapping in one study (Mutharasan et al., 2013).
to elucidate the physiologic signicance of LHCGR splice variants. Utilizing an actual LHCGR sequence variant rather than a neigh-
Frank mutations in LHCGR are associated with developmental boring polymorphic marker, Capalbo and colleagues reported a cor-
and reproductive abnormalities, including psuedohermaphrodism, relation between the 312N LHCGR allele and increased risk for PCOS
micropenis, hypospadias, and infertility (Ascoli et al., 2002; Menon in women of Sardinian origin (Capalbo et al., 2012). A dose-depen-
and Menon, 2012). Emerging data also suggest that polymorphisms dent effect was observed for this polymorphism, such that homozy-
in the LHCGR sequence contribute to risk for conditions associated gosity of the 312N allele was associated with a further elevated risk

Table 3
Half-life in serum and differential downstream effects of r-hLH and r-hCG binding to LHCGR.

Cell type r-hLH r-hCG


Half-life
t1/2a, range of mean, hrs 0.61.3a,b 3.95.5c
t1/2b, range of mean, hrs 912a,b 2331c
Response
ED50 (pM) COS-7/LHCGRe 530.0 51.2 107.1 14.3
Time to maximal cAMP accumulationd COS-7/LHCGRe 10 min 1h
ERK1/2 activationf hGLC Strong Weak
AKT activationf hGLC Strong Minimal
Neuregulin 1 inhibition in presence of ERK1/2 and AKT pathway blockade hGLC Abrogated Unaffected
CYP19A1 expression in presence of ERK1/2 pathway blockade hGLC Increased Unaffected

ERK1/2, extracellular signal-regulated protein kinases 1 and 2; r-hLH, recombinant human luteinizing hormone; r-hCG, recombinant human chorionic gonadotropin; LHCGR,
luteinizing hormone/choriogonadotropin receptor; hGLC, human granulosa cells; t1/2a = distribution (initial) half-life; t1/2b = elimination (terminal) half-life
a
Le Cotonnec et al. (1998a)
b
Le Cotonnec et al. (1998b)
c
Trinchard-Lugan et al. (2002)
d
Doses used were consistent with ED50 for r-hLH and for r-hCG in COS-7/LHCGR cells.
e
COS-7/LHCGR cells constitutively express LHCGR. LHCGR expression in hGLC cells is under physiologic regulation.
f
100 pM dose used for both r-hLH and r-hCG (Casarini et al., 2012).
J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213 209

Fig. 2. Response of human granulosa cells (hGLC) to long-term stimulation with recombinant human luteinizing hormone (r-hLH) or recombinant human chorionic
gonadotropin (r-hCG). Intracellular cyclic adenosine monophosphate (cAMP) (bars) and extracellular progesterone (lines) concentrations during chronic exposure to 500 pM
r-hLH or 100 pM r-hCG in the presence of a non-selective phosphodiesterase inhibitor (IBMX 500 lM); control cells were not exposed to gonadotropins. Panels illustrate 3
time ranges: (A) 012 h; (B) 1324 h; and (C) 2536 h. Extracellular accumulation of cAMP continued to increase during the 36-h evaluation period (D). Reproduced with
permission from Casarini et al. (2012).
210 J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213

The effects of gonadotropin binding appear to be regulated by


both temporal and spatial control of cAMP-mediated signaling
(Weedon-Fekjaer and Tasken, 2012). This cascade begins when
LHCGR binds LH or hCG. This induces a conformational change in
LHCGR, leading to the activation of the stimulatory G-protein Gs
(Puett et al., 2007). Gs, in turn, activates adenylate cyclase, which
increases the intracellular pool of cAMP.
Acting through an independent mechanism, LHCGR stimulation
also activates the phospholipase C/inositol phosphate (PLC/IP) sig-
naling pathway (Gilchrist et al., 1996). Given the high levels of LH
or hCG required to activate PLC via LHCGR, it has been suggested
that PLC-based signaling only occurs during the pre-ovulatory LH
surge and during pregnancy (Gudermann et al., 1992). Consistent
with this role, investigators have recently identied PLC as a medi-
ator of nal granulosa cell differentiation in response to LH (Dona-
deu et al., 2011).
Other signaling molecules/pathways including extracellular sig-
nal-regulated protein kinases 1 and 2 (ERK1/2) and AKT have been
identied as major downstream players in LHCGR-mediated sig-
naling, with putative roles in cell proliferation, differentiation,
and survival (Ben-Ami et al., 2009; Noma et al., 2011; Palaniappan
and Menon, 2010; Shiraishi and Ascoli, 2007). Together with cAMP/
PKA, the ERK1/2, and AKT pathways contribute to the regulation of
oocyte and follicle maturation (Brown et al., 2010; Reizel et al.,
2010).
Although both hLH and hCG bind to LHCGR, emerging evidence
suggests that the downstream effects differ. Much like LH and hCG
were considered to be functionally equivalent, the result of recep-
tor binding was traditionally presumed to be alike. New data, how-
ever, suggest that this is not true. Advances in gonadotropin
purication and recombinant technology have allowed investiga-
tors to explore hormone-specic signaling cascades, with
thought-provoking results.
Casarini and colleagues performed a series of experiments
examining changes in gene and protein expression/activation in
response to recombinant hLH (r-hLH) or recombinant hCG
(r-hCG) exposure in COS-7 cells constitutively expressing LHCGR
Fig. 3. Theoretical pathways for the divergence in receptor-mediated signaling (COS-7/LHCGR) and in human granulosa cells (hGLC) (Casarini
between luteinizing hormone (LH) and human chorionic gonadotropin (hCG).
Although acting on the same receptor, accumulating evidence suggests that LH
et al., 2012). Recombinant hCG was found to be ve times more
binding (A) has a greater impact (thicker arrow) on AKT and extracellular signal- potent than hLH with respect to cAMP production, with a com-
regulated protein kinase (ERK1/2) phosphorylation than hCG does, whereas binding mensurate lower ED50 (Table 3). cAMP concentration reached its
of hCG (B) generates a greater intracellular accumulation (thicker arrow) of cAMP maximum more quickly with r-hLH than with r-hCG in COS-7/
than does LH (Casarini et al., 2012; Gupta et al., 2012). Pathway components
LHCGR cells (10 minutes and 1 hour, respectively). During ex-
derived from Arey and Lopez (2011). AC, adenylate cyclase; ATP, adenosine
triphosphate; cAMP, cyclic adenosine monophosphate; Gs/Gq, G proteins; IP3, tended gonadotropin exposure in hGLC cells, in which LHCGR
inositol triphosphate; LHCGR, luteinizing hormone/choriogonadotropin receptor; expression was physiologically regulated, cAMP levels cycled, with
PKA, protein kinase A; PLC, phospholipase C; PKC, protein kinase C. a period of approximately 45 h (Fig. 2). Interestingly, extracellu-
lar cAMP levels were not cyclical. Peak intracellular cAMP levels
of PCOS beyond that of a single copy. Interestingly, the 312N allele were generally higher with r-hCG than with r-hLH. Nonetheless,
has been found to be less common among men with maldescended progesterone production was similar, with supernatant concentra-
testes compared with unaffected controls (Simoni et al., 2008). tions continuing to increase throughout the 36-hour evaluation
period. In addition, recombinant hLH stimulation of hGLC cells
strongly activated ERK1/2 and AKT, but hCG only weakly affected
6.2. Signaling pathways these pathways (Table 3). Blockage of the ERK1/2 and/or AKT
pathways inuenced downstream gene expression in hGLC ex-
LHCGR is capable of binding LH, hCG, and h-hCG, but not hCG posed to r-hLH but not those stimulated with r-hCG. To exclude
free b-subunit (Cole, 2009, 2010). Nicked hCG binds LHCGR, but the possibility that these ndings were specic to recombinant
with a substantially lower afnity (Cole et al., 1991). Experiments gonadotropins that do not undergo post-translational modication
using hCG and ovine LH indicate that binding of these hormones to of native proteins, the authors repeated the dose-response exper-
LHCGR differentially inuences receptor interaction with plasma iments using extractive hLH and hCG, with results comparable to
membrane proteins (Roess et al., 1997), which may contribute to that of r-hLH and r-hCG.
the subsequent divergence in signaling pathway activation. The Disparate effects on signaling between LH and hCG also have
intricacies of LHCGR signaling are a subject of active investigation, been described in goat ovarian granulosa cells (Gupta et al.,
with many components and interfaces not yet dened. It is gener- 2012). In this model, sustained exposure in cell culture led to sig-
ally understood that the cyclic adenosine monophosphate/protein nicantly greater cAMP accumulation with hCG vs. ovine LH
kinase A (cAMP/PKA) pathway is a signicant effector of down- (p < 0.05). Long-term culture in the presence of LH supported
stream events that lead to ovulation and steroid biosynthesis. growth and proliferation of granulosa cells; hCG, on the other
J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213 211

hand, had the opposite effect, slowing growth and increasing cell therapeutic tools to help manage clinical conditions such as
doubling time. Consistent with the previously described experi- infertility.
ments using human granulosa cells, phospho-ERK1/2 levels were
greater after long-term exposure to LH compared with hCG.
Using the aforementioned data in combination with known ele- Disclosure statement
ments of LHCGR signaling, we have constructed a schematic illus-
trating potential differences between LH- and hCG-mediated J.C. has nothing to disclose, J.S. has nothing to disclose.
signaling pathways (Fig. 3). This diagram is an acknowledged sim-
plication of the complex interactions between diverse cascades Acknowledgements
that coordinately regulate gonadotropin response. Further explora-
tion in different cell types and physiologic conditions is needed to Financial support for manuscript development was provided by
fully characterize the unique aspects of signaling via LH or hCG. Ferring Pharmaceuticals, Inc.
The authors thank Crystal Murcia, PhD, and Jill McCollam,
6.3. Extragonadal LHCGR PharmD, of The JB Ashtin Group, Inc., for assistance in preparing
this manuscript for publication based on the authors initial draft
Detection of LHCGR in regions other than ovarian cell types and and direction.
Leydig cells of the testes, including the decidua, uterine vascula-
ture, umbilical cord, fetal organs, cytotrophoblast cells, and adrenal References
cortex, has spurred ongoing debate regarding the potential role for
gonadotropins in extragonadal locations (Cole, 2010; Pakarainen Alviggi, C., Clarizia, R., Pettersson, K., Mollo, A., Humaidan, P., Strina, I., et al., 2011.
Suboptimal response to GnRHa long protocol is associated with a common LH
et al., 2007; Rao, 2010). It has been suggested that the absence of
polymorphism. Reprod. Biomed. Online 22 (Suppl. 1), S67S72.
overt non-gonadal phenotypes in individuals with naturally occur- Anobile, C.J., Talbot, J.A., McCann, S.J., Padmanabhan, V., Robertson, W.R., 1998.
ring LHCGR mutations indicates that LH and hCG have only modest Glycoform composition of serum gonadotrophins through the normal
menstrual cycle and in the post-menopausal state. Mol. Hum. Reprod. 4, 631
effects on the normal physiologic activity of these tissues (Ascoli
639.
et al., 2002). Moreover, the extragonadal phenotypes in LHCGR- Arey, B.J., Lopez, F.J., 2011. Are circulating gonadotropin isoforms naturally
knockout and hCG overexpressing mice have been attributed occurring biased agonists? Basic and therapeutic implications. Rev. Endocr.
chiey to changes in gonadal function rather than a physiologic Metab. Disord. 12, 275288.
Ascoli, M., Fanelli, F., Segaloff, D.L., 2002. The lutropin/choriogonadotropin receptor,
role for LHCGR in non-gonadal tissue (Pakarainen et al., 2007; a 2002 perspective. Endocr. Rev. 23, 141174.
Rahman and Rao, 2009). It should be noted, however, that CG is Bahado-Singh, R.O., Oz, A.U., Kingston, J.M., Shahabi, S., Hsu, C.D., Cole, L., 2002. The
not a native constituent of the mouse genome; yet it is hCG for role of hyperglycosylated hCG in trophoblast invasion and the prediction of
subsequent pre-eclampsia. Prenat. Diagn. 22, 478481.
which the strongest evidence of extragonadal activity has been Banerjee, P., Fazleabas, A.T., 2011. Extragonadal actions of chorionic gonadotropin.
unearthed. Rev. Endocr. Metab. Disord. 12, 323332.
As previously discussed, the enduring presence of hCG beyond Ben-Ami, I., Armon, L., Freimann, S., Strassburger, D., Ron-El, R., Amsterdam, A.,
2009. EGF-like growth factors as LH mediators in the human corpus luteum.
the time period associated with luteal support implies additional Hum. Reprod. 24, 176184.
functionality. hCG is implicated in multiple modes of supporting Bergendah, M., Veldhuis, J.D., 2001. Is there a physiological role for gonadotrophin
pregnancy in addition to promoting progesterone production. oligosaccharide heterogeneity in humans? III. Luteinizing hormone
heterogeneity: a medical physiologists perspective. Hum. Reprod. 16, 1058
Implantation, trophoblast invasion, placentation, uterine growth
1064.
control, growth and differentiation of fetal organs, as well as a host Berndt, S., Blacher, S., Munaut, C., Detilleux, J., Perrier dHauterive, S., Huhtaniemi, I.,
of other putative or known functions of hCG that are listed in et al., 2013. Hyperglycosylated human chorionic gonadotropin stimulates
angiogenesis through TGF-beta receptor activation. FASEB J. 27, 13091321.
Table 2 (Banerjee and Fazleabas, 2011; Cole, 2010). While it seems
Birken, S., Maydelman, Y., Gawinowicz, M.A., Pound, A., Liu, Y., Hartree, A.S., 1996.
clear that extragonadal LHCGR has functional signicance, further Isolation and characterization of human pituitary chorionic gonadotropin.
study is needed to clarify its role in normal and aberrant cellular Endocrinology 137, 14021411.
processes and to characterize the individual effects of various Boorstein, W.R., Vamvakopoulos, N.C., Fiddes, J.C., 1982. Human chorionic
gonadotropin beta-subunit is encoded by at least eight genes arranged in
isoforms. tandem and inverted pairs. Nature 300, 419422.
Borrelli, P.T.A., Butler, S.A., Docherty, S.M., Staite, E.M., Borrelli, A.L., ILes, R.K., 2003.
Human chorionic gonadotropin isoforms in the diagnosis of ectopic pregnancy.
7. Conclusions Clin. Chem. 49, 20452049.
Braunstein, G.D., 2011. Endocrine changes in pregnancy. In: Melmed, S., Polonsky,
K.S., Larsen, P.R., Kronenberg, H.M. (Eds.), Williams Textbook of Endocrinology.
The signicance of differences between LH and hCG in terms of Elsevier Saunders, Philadelphia, PA, pp. 581660.
structure, expression, regulation, and function is just beginning to Brown, C., LaRocca, J., Pietruska, J., Ota, M., Anderson, L., Smith, S.D., et al., 2010.
be fully realized. It has long been known that LH and hCG funda- Subfertility caused by altered follicular development and oocyte growth in
female mice lacking PKB alpha/Akt1. Biol. Reprod. 82, 246256.
mentally differ in their expression patterns; however, we now Bulun, S.E., 2011. Physiology and pathology of the female reproductive axis. In:
appreciate that categorization based on this attribute alone fails Melmed, S., Polonsky, K.S., Larsen, P.R., Kronenberg, H.M. (Eds.), Williams
to adequately describe the complexity and unique aspects of these Textbook of Endocrinology, 12th ed. Elsevier Saunders, Philadelphia, PA,
pp. 581660.
hormones. Moreover, an array of isoforms exists within a single Burgon, P.G., Stanton, P.G., Robertson, D.M., 1996. In vivo bioactivities and clearance
hormone family, the composition of which uctuates during the patterns of highly puried human luteinizing hormone isoforms. Endocrinology
course of the ovarian cycle and pregnancy as well as throughout 137, 48274836.
Butler, S.A., Iles, R.K., 2004. The free monomeric beta subunit of human chorionic
the lifespan in both men and women. Emerging evidence suggests gonadotrophin (hCG beta) and the recently identied homodimeric beta-beta
that these diverse isoforms have distinct, though often overlap- subunit (hCG beta beta) both have autocrine growth effects. Tumour Biol. 25,
ping, functions that are reected by their relative abundance in 1823.
Butler, S.A., Ikram, M.S., Mathieu, S., Iles, R.K., 2000. The increase in bladder
normal and aberrant physiologic processes. The importance of
carcinoma cell population induced by the free beta subunit of human chorionic
quantitative and qualitative distinctions in signaling cascades acti- gonadotrophin is a result of an anti-apoptosis effect and not cell proliferation.
vated by LH and hCG and the clinical relevance of extragonadal Br. J. Cancer 82, 15531556.
activity are nascent areas of study that merit further exploration. Capalbo, A., Sagnella, F., Apa, R., Fulghesu, A.M., Lanzone, A., Morciano, A., et al.,
2012. The 312N variant of the luteinizing hormone/choriogonadotropin
All of these advances in knowledge broaden the ability to utilize receptor gene (LHCGR) confers up to 2.7-fold increased risk of polycystic
LH and hCG as diagnostic and prognostic biomarkers, as well as ovary syndrome in a Sardinian population. Clin. Endocrinol. (Oxf.) 77, 113119.
212 J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213

Casarini, L., Lispi, M., Longobardi, S., Milosa, F., La Marca, A., Tagliasacchi, D., et al., Kovalevskaya, G., Birken, S., Kakuma, T., Ozaki, N., Sauer, M., Lindheim, S., et al.,
2012. LH and hCG action on the same receptor results in quantitatively and 2002. Differential expression of human chorionic gonadotropin (hCG)
qualitatively different intracellular signalling. PLoS One 7, e46682. glycosylation isoforms in failing and continuing pregnancies: preliminary
Castro-Fernandez, C., Olivares, A., Soderlund, D., Lopez-Alvarenga, J.C., Zambrano, E., characterization of the hyperglycosylated hCG epitope. J. Endocrinol. 172,
Veldhuis, J.D., et al., 2000. A preponderance of circulating basic isoforms is 497506.
associated with decreased plasma half-life and biological to immunological Lambert, A., Talbot, J.A., Anobile, C.J., Robertson, W.R., 1998. Gonadotrophin
ratio of gonadotropin-releasing hormone-releasable luteinizing hormone in heterogeneity and biopotency: implications for assisted reproduction. Mol.
obese men. J. Clin. Endocrinol. Metab. 85, 46034610. Hum. Reprod. 4, 619629.
Chen, Z.J., Zhao, H., He, L., Shi, Y., Qin, Y., Li, Z., et al., 2011. Genome-wide association Lamminen, T., Huhtaniemi, I., 2001. A common genetic variant of luteinizing
study identies susceptibility loci for polycystic ovary syndrome on hormone; relation to normal and aberrant pituitary-gonadal function. Eur. J.
chromosome 2p16.3, 2p21 and 9q33.3. Nat. Genet. 43, 5559. Pharmacol. 414, 17.
Cole, L.A., 2009. New discoveries on the biology and detection of human chorionic Le Cotonnec, J.Y., Porchet, H.C., Beltrami, V., Munafo, A., 1998a. Clinical
gonadotropin. Reprod. Biol. Endocrinol. 7, 8. pharmacology of recombinant human luteinizing hormone: Part I.
Cole, L.A., 2010. Biological functions of hCG and hCG-related molecules. Reprod. Pharmacokinetics after intravenous administration to healthy female
Biol. Endocrinol. 8, 102. volunteers and comparison with urinary human luteinizing hormone. Fertil.
Cole, L.A., Butler, S.A., 2008. Hyperglycosylated human chorionic gonadotropin and Steril. 69, 189194.
human chorionic gonadotropin free beta-subunit: tumor markers and tumor Le Cotonnec, J.Y., Porchet, H.C., Beltrami, V., Munafo, A., 1998b. Clinical
promoters. J. Reprod. Med. 53, 499512. pharmacology of recombinant human luteinizing hormone: Part II.
Cole, L.A., Wang, Y.X., Elliott, M., Latif, M., Chambers, J.T., Chambers, S.K., et al., 1988. Bioavailability of recombinant human luteinizing hormone assessed with an
Urinary human chorionic gonadotropin free beta-subunit and beta-core immunoassay and an in vitro bioassay. Fertil. Steril. 69, 195200.
fragment: a new marker of gynecological cancers. Cancer Res. 48, 13561360. Liu, J.H., Yen, S.S., 1983. Induction of midcycle gonadotropin surge by ovarian
Cole, L.A., Kardana, A., Andrade-Gordon, P., Gawinowicz, M.A., Morris, J.C., Bergert, steroids in women: a critical evaluation. J. Clin. Endocrinol. Metab. 57, 797802.
E.R., et al., 1991. The heterogeneity of human chorionic gonadotropin (hCG). III. Liu, L., Southers, J.L., Cassels Jr., J.W., Banks, S.M., Wehmann, R.E., Blithe, D.L., et al.,
The occurrence and biological and immunological activities of nicked hCG. 1989. Structurekinetic relationships of choriogonadotropin and related
Endocrinology 129, 15591567. molecules. Am. J. Physiol. 256, E721E724.
Cole, L.A., Shahabi, S., Oz, U.A., Bahado-Singh, R.O., Mahoney, M.J., 1999. Lu, D.L., Peegel, H., Mosier, S.M., Menon, K.M., 1993. Loss of lutropin/human
Hyperglycosylated human chorionic gonadotropin (invasive trophoblast choriogonadotropin receptor messenger ribonucleic acid during ligand-induced
antigen) immunoassay: a new basis for gestational Down syndrome down-regulation occurs post transcriptionally. Endocrinology 132, 235240.
screening. Clin. Chem. 45, 21092119. Madhra, M., Gay, E., Fraser, H.M., Duncan, W.C., 2004. Alternative splicing of the
Dickinson, R.E., Stewart, A.J., Myers, M., Millar, R.P., Duncan, W.C., 2009. Differential human luteal LH receptor during luteolysis and maternal recognition of
expression and functional characterization of luteinizing hormone receptor pregnancy. Mol. Hum. Reprod. 10, 599603.
splice variants in human luteal cells: implications for luteolysis. Endocrinology Marrama, P., Zaidi, A.A., Montanini, V., Celani, M.F., Cioni, K., Carani, C., et al., 1983.
150, 28732881. Age and sex related variations in biologically active and immunoreactive serum
Ding, Y.Q., Huhtaniemi, I., 1991. Preponderance of basic isoforms of serum luteinizing hormone. J. Endocrinol. Invest. 6, 427433.
luteinizing hormone (LH) is associated with the high bio/immune ratio of LH Marrama, P., Montanini, V., Celani, M.F., Carani, C., Cioni, K., Bazzani, M., et al., 1984.
in healthy women and in women with polycystic ovarian disease. Hum. Reprod. Decrease in luteinizing hormone biological activity/immunoreactivity ratio in
6, 346350. elderly men. Maturitas 5, 223231.
Donadeu, F.X., Esteves, C.L., Doyle, L.K., Walker, C.A., Schauer, S.N., Diaz, C.A., 2011. Menon, K.M., Menon, B., 2012. Structure, function and regulation of gonadotropin
Phospholipase Cbeta3 mediates LH-induced granulosa cell differentiation. receptors a perspective. Mol. Cell. Endocrinol. 356, 8897.
Endocrinology 152, 28572869. Menon, K.M., Menon, B., Wang, L., Gulappa, T., Harada, M., 2010. Molecular
Elliott, M.M., Kardana, A., Lustbader, J.W., Cole, L.A., 1997. Carbohydrate and peptide regulation of gonadotropin receptor expression: relationship to sterol
structure of the alpha- and beta-subunits of human chorionic gonadotropin metabolism. Mol. Cell. Endocrinol. 329, 2632.
from normal and aberrant pregnancy and choriocarcinoma. Endocrine 7, Mesiano, S., 2009. The endocrinology of human pregnancy and fetoplacental
1532. neuroendocrine development. In: Strauss, J.E., III, Barbieri, R.L. (Eds.), Yen &
Eriksen, M.B., Brusgaard, K., Andersen, M., Tan, Q., Altinok, M.L., Gaster, M., et al., Jaffes Reproductive Endocrinology: Physiology, Pathophysiology, and Clinical
2012. Association of polycystic ovary syndrome susceptibility single nucleotide Management, sixth ed. Saunders Elsevier, Philadelphia, PA, p. 254.
polymorphism rs2479106 and PCOS in Caucasian patients with PCOS or Minegishi, T., Nakamura, K., Yamashita, S., Omori, Y., 2007. The effect of splice
hirsutism as referral diagnosis. Eur. J. Obstet. Gynecol. Reprod. Biol. 163, 3942. variant of the human luteinizing hormone (LH) receptor on the expression of
Fritz, M.A., Speroff, L., 2011. Clinical Gynecologic Endocrinology and Infertility, gonadotropin receptor. Mol. Cell. Endocrinol. 260262, 117125.
eighth ed. Lippincott Williams & Wilkins, Philadelphia, PA. Mock, P., Kovalevskaya, G., OConnor, J.F., Campana, A., 2000. Choriocarcinoma-like
Gilchrist, R.L., Ryu, K.S., Ji, I., Ji, T.H., 1996. The luteinizing hormone/chorionic human chorionic gonadotrophin (HCG) and HCG bioactivity during the rst
gonadotropin receptor has distinct transmembrane conductors for cAMP and trimester of pregnancy. Hum. Reprod. 15, 22092214.
inositol phosphate signals. J. Biol. Chem. 271, 1928319287. Mutharasan, P., Galdones, E., Penalver Bernabe, B., Garcia, O.A., Jafari, N., Shea, L.D.,
Gudermann, T., Birnbaumer, M., Birnbaumer, L., 1992. Evidence for dual coupling of et al., 2013. Evidence for chromosome 2p16.3 polycystic ovary syndrome
the murine luteinizing hormone receptor to adenylyl cyclase and susceptibility locus in affected women of European ancestry. J. Clin. Endocrinol.
phosphoinositide breakdown and Ca2+ mobilization. Studies with the cloned Metab. 98, E185E190.
murine luteinizing hormone receptor expressed in L cells. J. Biol. Chem. 267, Nagirnaja, L., Rull, K., Uuskula, L., Hallast, P., Grigorova, M., Laan, M., 2010. Genomics
44794488. and genetics of gonadotropin beta-subunit genes: unique FSHB and duplicated
Gupta, C., Chapekar, T., Chhabra, Y., Singh, P., Sinha, S., Luthra, K., 2012. Differential LHB/CGB loci. Mol. Cell. Endocrinol. 329, 416.
response to sustained stimulation by hCG & LH on goat ovarian granulosa cells. Nagirnaja, L., Venclovas, C., Rull, K., Jonas, K.C., Peltoketo, H., Christiansen, O.B., et al.,
Ind. J. Med. Res. 135, 331340. 2012. Structural and functional analysis of rare missense mutations in human
Haavisto, A.M., Pettersson, K., Bergendahl, M., Virkamaki, A., Huhtaniemi, I., 1995. chorionic gonadotrophin beta-subunit. Mol. Hum. Reprod. 18, 379390.
Occurrence and biological properties of a common genetic variant of luteinizing Nair, A.K., Kash, J.C., Peegel, H., Menon, K.M., 2002. Post-transcriptional regulation of
hormone. J. Clin. Endocrinol. Metab. 80, 12571263. luteinizing hormone receptor mRNA in the ovary by a novel mRNA-binding
Hashad, D., Mohamed, N., Hashad, M.M., 2012. Luteinising hormone beta-subunit protein. J. Biol. Chem. 277, 2146821473.
gene Gly102Ser variant and oxidative stress biomarkers in Egyptian infertile Nakamura, K., Yamashita, S., Omori, Y., Minegishi, T., 2004. A splice variant of the
males. Andrologia 44 (Suppl. 1), 484489. human luteinizing hormone (LH) receptor modulates the expression of wild-
Hoermann, R., Gerbes, A.L., Spoettl, G., Jungst, D., Mann, K., 1992. Immunoreactive type human LH receptor. Mol. Endocrinol. 18, 14611470.
human chorionic gonadotropin and its free beta subunit in serum and ascites of Naylor, S.L., Chin, W.W., Goodman, H.M., Lalley, P.A., Grzeschik, K.H., Sakaguchi, A.Y.,
patients with malignant tumors. Cancer Res. 52, 15201524. 1983. Chromosome assignment of genes encoding the alpha and beta subunits
Huhtaniemi, I.T., Pye, S.R., Holliday, K.L., Thomson, W., ONeill, T.W., Platt, H., et al., of glycoprotein hormones in man and mouse. Somatic Cell Genet. 9, 757770.
2010. Effect of polymorphisms in selected genes involved in pituitary-testicular Nilsson, C., Pettersson, K., Millar, R.P., Coerver, K.A., Matzuk, M.M., Huhtaniemi, I.T.,
function on reproductive hormones and phenotype in aging men. J. Clin. 1997. Worldwide frequency of a common genetic variant of luteinizing
Endocrinol. Metab. 95, 18981908. hormone: an international collaborative research. Int. Collab. Res. Group
Ikeda, S., Nakamura, K., Kogure, K., Omori, Y., Yamashita, S., Kubota, K., et al., 2008. Fertil. Steril. 67, 9981004.
Effect of estrogen on the expression of luteinizing hormone-human chorionic Noma, N., Kawashima, I., Fan, H.Y., Fujita, Y., Kawai, T., Tomoda, Y., et al., 2011. LH-
gonadotropin receptor messenger ribonucleic acid in cultured rat granulosa induced neuregulin 1 (NRG1) type III transcripts control granulosa cell
cells. Endocrinology 149, 15241533. differentiation and oocyte maturation. Mol. Endocrinol. 25, 104116.
Iles, R.K., Purkis, P.E., Whitehead, P.C., Oliver, R.T., Leigh, I., Chard, T., 1990. OConnor, J.F., Kovalevskaya, G., Birken, S., Schlatterer, J.P., Schechter, D., McMahon,
Expression of beta human chorionic gonadotrophin by non-trophoblastic non- D.J., et al., 1998. The expression of the urinary forms of human luteinizing
endocrine normal and malignant epithelial cells. Br. J. Cancer 61, 663666. hormone beta fragment in various populations as assessed by a specic
Iles, R.K., Persad, R., Trivedi, M., Sharma, K.B., Dickinson, A., Smith, P., et al., 1996. immunoradiometric assay. Hum. Reprod. 13, 826835.
Urinary concentration of human chorionic gonadotrophin and its fragments as a Odell, W.D., Grifn, J., 1987. Pulsatile secretion of human chorionic gonadotropin in
prognostic marker in bladder cancer. Br. J. Urol. 77, 6169. normal adults. N. Engl. J. Med. 317, 16881691.
Jameson, J.L., Hollenberg, A.N., 1993. Regulation of chorionic gonadotropin gene Odell, W.D., Grifn, J., 1989. Pulsatile secretion of chorionic gonadotropin during the
expression. Endocr. Rev. 14, 203221. normal menstrual cycle. J. Clin. Endocrinol. Metab. 69, 528532.
J. Choi, J. Smitz / Molecular and Cellular Endocrinology 383 (2014) 203213 213

Pakarainen, T., Ahtiainen, P., Zhang, F.P., Rulli, S., Poutanen, M., Huhtaniemi, I., 2007. Shi, Y., Zhao, H., Cao, Y., Yang, D., Li, Z., Zhang, B., et al., 2012. Genome-wide
Extragonadal LH/hCG action not yet time to rewrite textbooks. Mol. Cell. association study identies eight new risk loci for polycystic ovary syndrome.
Endocrinol. 269, 916. Nat. Genet. 44, 10201025.
Palaniappan, M., Menon, K.M., 2010. Human chorionic gonadotropin stimulates Shiraishi, K., Ascoli, M., 2007. Lutropin/choriogonadotropin stimulate the
theca-interstitial cell proliferation and cell cycle regulatory proteins by a cAMP- proliferation of primary cultures of rat Leydig cells through a pathway that
dependent activation of AKT/mTORC1 signaling pathway. Mol. Endocrinol. 24, involves activation of the extracellularly regulated kinase 1/2 cascade.
17821793. Endocrinology 148, 32143225.
Palomaki, G.E., Neveux, L.M., Haddow, J.E., Wyatt, P., 2007. Hyperglycosylated-hCG Simoni, M., Tuttelmann, F., Michel, C., Bockenfeld, Y., Nieschlag, E., Gromoll, J., 2008.
(h-hCG) and Down syndrome screening in the rst and second trimesters of Polymorphisms of the luteinizing hormone/chorionic gonadotropin receptor
pregnancy. Prenat. Diagn. 27, 808813. gene: association with maldescended testes and male infertility.
Pettersson, K.S., Soderholm, J.R., 1991. Individual differences in lutropin Pharmacogenet. Genomics 18, 193200.
immunoreactivity revealed by monoclonal antibodies. Clin. Chem. 37, 333340. Srouji, S.S., Pagan, Y.L., DAmato, F., Dabela, A., Jimenez, Y., Supko, J.G., et al., 2007.
Puett, D., Li, Y., DeMars, G., Angelova, K., Fanelli, F., 2007. A functional Pharmacokinetic factors contribute to the inverse relationship between
transmembrane complex: the luteinizing hormone receptor with bound luteinizing hormone and body mass index in polycystic ovarian syndrome. J.
ligand and G protein. Mol. Cell. Endocrinol. 260262, 126136. Clin. Endocrinol. Metab. 92, 13471352.
Rahman, N.A., Rao, C.V., 2009. Recent progress in luteinizing hormone/human Stanton, P.G., Pozvek, G., Burgon, P.G., Robertson, D.M., Hearn, M.T., 1993. Isolation
chorionic gonadotrophin hormone research. Mol. Hum. Reprod. 15, 703711. and characterization of human LH isoforms. J. Endocrinol. 138, 529543.
Raivio, T., Huhtaniemi, I., Anttila, R., Siimes, M.A., Hagenas, L., Nilsson, C., et al., 1996. Takahashi, K., Kurioka, H., Ozaki, T., Kanasaki, H., Kohsaka, M., Miyazaki, K., et al.,
The role of luteinizing hormone-beta gene polymorphism in the onset and 1998. Increased prevalence of luteinizing hormone beta-subunit variant in
progression of puberty in healthy boys. J. Clin. Endocrinol. Metab. 81, 32783282. Japanese infertility patients. Hum. Reprod. 13, 33383344.
Ramanujam, L.N., Liao, W.X., Roy, A.C., Loganath, A., Goh, H.H., Ng, S.C., 1999. Takahashi, K., Ozaki, T., Okada, M., Kurioka, H., Kanasaki, H., Miyazaki, K., 1999.
Association of molecular variants of luteinizing hormone with menstrual Increased prevalence of luteinizing hormone beta-subunit variant in patients
disorders. Clin. Endocrinol. (Oxf.) 51, 243246. with premature ovarian failure. Fertil. Steril. 71, 96101.
Ramanujam, L.N., Liao, W.X., Roy, A.C., Ng, S.C., 2000. Association of molecular Trinchard-Lugan, I., Khan, A., Porchet, H.C., Munafo, A., 2002. Pharmacokinetics and
variants of luteinizing hormone with male infertility. Hum. Reprod. 15, 925928. pharmacodynamics of recombinant human chorionic gonadotrophin in healthy
Rao, C.V., 1979. Differential properties of human chorionic gonadotrophin and male and female volunteers. Reprod. Biomed. Online 4, 106115.
human luteinizing hormone binding to plasma membranes of bovine corpora Van de Wiele, R.L., Bogumil, J., Dyrenfurth, I., Ferin, M., Jewelewicz, R., Warren, M.,
lutea. Acta Endocrinol. (Copenh.) 90, 696710. et al., 1970. Mechanisms regulating the menstrual cycle in women. Recent Prog.
Rao, C.V., 2010. Human adrenal LH/hCG receptors and what they could mean for Horm. Res. 26, 63103.
adrenal physiology and pathology. Mol. Cell. Endocrinol. 329, 3336. Vassart, G., Pardo, L., Costagliola, S., 2004. A molecular dissection of the glycoprotein
Rao, C.V., Alsip, N.L., 2001. Use of the rat model to study hCG/LH effects on uterine hormone receptors. Trends Biochem. Sci. 29, 119126.
blood ow. Semin. Reprod. Med. 19, 7585. Wang, L., Nair, A.K., Menon, K.M., 2007. Ribonucleic acid binding protein-mediated
Reader, S.C., Robertson, W.R., Diczfalusy, E., 1983. Microheterogeneity of luteinizing regulation of luteinizing hormone receptor expression in granulosa cells:
hormone in pituitary glands from women of pre- and postmenopausal age. Clin. relationship to sterol metabolism. Mol. Endocrinol. 21, 22332241.
Endocrinol. (Oxf.) 19, 355363. Warner, B.A., Dufau, M.L., Santen, R.J., 1985. Effects of aging and illness on the
Reizel, Y., Elbaz, J., Dekel, N., 2010. Sustained activity of the EGF receptor is an pituitary testicular axis in men: qualitative as well as quantitative changes in
absolute requisite for LH-induced oocyte maturation and cumulus expansion. luteinizing hormone. J. Clin. Endocrinol. Metab. 60, 263268.
Mol. Endocrinol. 24, 402411. Weedon-Fekjaer, M.S., Tasken, K., 2012. Review: spatiotemporal dynamics of hCG/
Roess, D.A., Jewell, M.A., Philpott, C.J., Barisas, B.G., 1997. The rotational diffusion of cAMP signaling and regulation of placental function. Placenta 33 (Suppl.), S87
LH receptors differs when receptors are occupied by hCG versus LH and is S91.
increased by cytochalasin D. Biochim. Biophys. Acta 1357, 98106. Wide, L., 1985. Median charge and charge heterogeneity of human pituitary FSH, LH
Ropelato, M.G., Garcia-Rudaz, M.C., Castro-Fernandez, C., Ulloa-Aguirre, A., Escobar, and TSH. II. Relationship to sex and age. Acta Endocrinol. (Copenh.) 109, 190
M.E., Barontini, M., et al., 1999. A preponderance of basic luteinizing hormone 197.
(LH) isoforms accompanies inappropriate hypersecretion of both basal and Wide, L., Bakos, O., 1993. More basic forms of both human follicle-stimulating
pulsatile LH in adolescents with polycystic ovarian syndrome. J. Clin. hormone and luteinizing hormone in serum at midcycle compared with the
Endocrinol. Metab. 84, 46294636. follicular or luteal phase. J. Clin. Endocrinol. Metab. 76, 885889.
Rosa, C., Amr, S., Birken, S., Wehmann, R., Nisula, B., 1984. Effect of desialylation of Wide, L., Eriksson, K., 2013. Dynamic changes in glycosylation and glycan
human chorionic gonadotropin on its metabolic clearance rate in humans. J. composition of serum FSH and LH during natural ovarian stimulation. Ups. J.
Clin. Endocrinol. Metab. 59, 12151219. Med. Sci. 118, 153164.
Rousseau-Merck, M.F., Misrahi, M., Atger, M., Loosfelt, H., Milgrom, E., Berger, R., Wide, L., Naessen, T., Sundstrom-Poromaa, I., Eriksson, K., 2007. Sulfonation and
1990. Localization of the human luteinizing hormone/choriogonadotropin sialylation of gonadotropins in women during the menstrual cycle, after
receptor gene (LHCGR) to chromosome 2p21. Cytogenet. Cell Genet. 54, 7779. menopause, and with polycystic ovarian syndrome and in men. J. Clin.
Rousseau-Merck, M.F., Atger, M., Loosfelt, H., Milgrom, E., Berger, R., 1993. The Endocrinol. Metab. 92, 44104417.
chromosomal localization of the human follicle-stimulating hormone receptor Wide, L., Eriksson, K., Sluss, P.M., Hall, J.E., 2009. Serum half-life of pituitary
gene (FSHR) on 2p21-p16 is similar to that of the luteinizing hormone receptor gonadotropins is decreased by sulfonation and increased by sialylation in
gene. Genomics 15, 222224. women. J. Clin. Endocrinol. Metab. 94, 958964.
Rull, K., Laan, M., 2005. Expression of b-subunit of HCG genes during normal and Wide, L., Eriksson, K., Sluss, P.M., Hall, J.E., 2010. The common genetic variant of
failed pregnancy. Hum. Reprod. 20, 33603368. luteinizing hormone has a longer serum half-life than the wild type in
Rull, K., Nagirnaja, L., Ulander, V.M., Kelgo, P., Margus, T., Kaare, M., et al., 2008. heterozygous women. J. Clin. Endocrinol. Metab. 95, 383389.
Chorionic gonadotropin beta-gene variants are associated with recurrent Yamashita, S., Nakamura, K., Omori, Y., Tsunekawa, K., Murakami, M., Minegishi, T.,
miscarriage in two European populations. J. Clin. Endocrinol. Metab. 93, 2005. Association of human follitropin (FSH) receptor with splicing variant of
46974706. human lutropin/choriogonadotropin receptor negatively controls the
Sasaki, Y., Ladner, D.G., Cole, L.A., 2008. Hyperglycosylated human chorionic expression of human FSH receptor. Mol. Endocrinol. 19, 20992111.
gonadotropin and the source of pregnancy failures. Fertil. Steril. 89, 17811786. Zygmunt, M., Herr, F., Keller-Schoenwetter, S., Kunzi-Rapp, K., Munstedt, K., Rao,
Shi, Q.J., Lei, Z.M., Rao, C.V., Lin, J., 1993. Novel role of human chorionic C.V., et al., 2002. Characterization of human chorionic gonadotropin as a novel
gonadotropin in differentiation of human cytotrophoblasts. Endocrinology angiogenic factor. J. Clin. Endocrinol. Metab. 87, 52905296.
132, 13871395.

S-ar putea să vă placă și