ArticlePDF Available

A Homozygous Mutation in the Luteinizing Hormone Receptor Causes Partial Leydig Cell Hypoplasia: Correlation between Receptor Activity and Phenotype

Authors:

Abstract and Figures

Leydig cell hypoplasia (LCH) is characterized by a decreased response of the Leydig cells to LH. As a result, patients with this syndrome display aberrant male development ranging from complete pseudohermaphroditism to males with micropenis but with otherwise normal sex characteristics. We have evaluated three brothers with a mild form of LCH. Analysis of their LH receptor (LHR) gene revealed a homozygous missense mutation resulting in a substitution of a lysine residue for a isoleucine residue at position 625 of the receptor. In vitro analysis of this mutant LHR, LHR(I625K), in HEK293 cells indicated that the signaling efficiency was significantly impaired, which explains the partial phenotype. We have compared this mutant LHR to two other mutant LHRs, LHR(A593P) and LHR(S616Y), identified in a complete and partial LCH patient, respectively. Although the ligand-binding affinity for all three mutant receptors was normal, the hormonal response of LHR(A593P) was completely absent and that of LHR(S616Y) and LHR(I625K) was severely impaired. Low cell surface expression explained the reduced response of LHR(S616Y), while for LHR(I625K) this diminished response was due to a combination of low cell surface expression and decreased coupling efficiency. For LHR(A593P), the absence of a reduced response resulted from both poor cell surface expression and a complete deficiency in coupling. Our experiments further show a clear correlation between the severity of the clinical phenotype of patients and overall receptor signal capacity, which is a combination of cell surface expression and coupling efficiency.
Content may be subject to copyright.
A Homozygous Mutation in the
Luteinizing Hormone Receptor
Causes Partial Leydig Cell
Hypoplasia: Correlation between
Receptor Activity and Phenotype
John W. M. Martens, Miriam Verhoef-Post, Neusa Abelin,
Marilza Ezabella, Sergio P. A. Toledo, Han G. Brunner, and
Axel P. N. Themmen
Department of Endocrinology and Reproduction (J.W.M.M., M.V.-P.,
A.P.N.T.)
Faculty of Medicine and Health Sciences
Erasmus University Rotterdam
3000 DR Rotterdam, The Netherlands
Endocrine Genetics Unit (N.A., M.E., S.P.A.T.)
Endocrine Division
Department of Medicine
Sa˜ o Paulo University School of Medicine
CEP 01246 Sa˜ o Paulo, Brazil
Department of Human Genetics (H.G.B.)
University Hospital
6500 HB Nijmegen, The Netherlands
Leydig cell hypoplasia (LCH) is characterized by a
decreased response of the Leydig cells to LH. As a
result, patients with this syndrome display aber-
rant male development ranging from complete
pseudohermaphroditism to males with micropenis
but with otherwise normal sex characteristics. We
have evaluated three brothers with a mild form of
LCH. Analysis of their LH receptor (LHR) gene re-
vealed a homozygous missense mutation resulting
in a substitution of a lysine residue for a isoleucine
residue at position 625 of the receptor. In vitro
analysis of this mutant LHR, LHR(I625K), in HEK293
cells indicated that the signaling efficiency was
significantly impaired, which explains the partial
phenotype. We have compared this mutant LHR to
two other mutant LHRs, LHR(A593P) and
LHR(S616Y), identified in a complete and partial
LCH patient, respectively. Although the ligand-
binding affinity for all three mutant receptors was
normal, the hormonal response of LHR(A593P) was
completely absent and that of LHR(S616Y) and
LHR(I625K) was severely impaired. Low cell sur-
face expression explained the reduced response of
LHR(S616Y), while for LHR(I625K) this diminished
response was due to a combination of low cell
surface expression and decreased coupling effi-
ciency. For LHR(A593P), the absence of a reduced
response resulted from both poor cell surface ex-
pression and a complete deficiency in coupling.
Our experiments further show a clear correlation
between the severity of the clinical phenotype of
patients and overall receptor signal capacity,
which is a combination of cell surface expression
and coupling efficiency. (Molecular Endocrinology
12: 775–784, 1998)
INTRODUCTION
Male sex differentiation begins when the sex-deter-
mining factor (SRY) is expressed from the Y chromo-
some, which causes the indifferent gonads to develop
into testes (1). In the testis, testicular cords are formed
containing Sertoli cells and germ cells, while mesen-
chymal cells migrate into the interstitial space between
the cords, giving rise to fetal Leydig cells (2). The
testicular Sertoli cells and Leydig cells produce, re-
spectively, anti-Mu¨ llerian hormone and testosterone,
two hormones that are essential for correct differenti-
ation of both primary and secondary sex characteris-
tics (for review see Refs. 3–5). Anti-Mu¨ llerian hormone
triggers the regression of the mu¨ llerian duct, the anla-
gen of the female urogenital tract, while testosterone,
0888-8809/98/$3.00/0
Molecular Endocrinology
Copyright © 1998 by The Endocrine Society
775
in some target tissues after its reduction to dihydrotes-
tosterone, stimulates differentiation and growth of the
epididymides, vasa deferentia, the prostate, seminal
vesicles, and other parts of the male urogenital tract
including the formation of the penis. Before birth, pro-
liferation and differentiation of Leydig cells and their
production of androgens are dependent on the pla-
cental hormone, human CG (hCG). Prenatal distur-
bance of male sex differentiation leads to a variety of
phenotypes ranging from males with micropenis, indi-
viduals with ambiguous genitalia and hypospadias, to
complete male pseudohermaphrodites (5). These phe-
notypes are either due to insensitivity of the target
cells to androgens (5, 6) or to impaired testicular an-
drogen production as a result of a steroidogenic en-
zyme defect [e.g.17
b
-hydroxysteroid dehydrogenase
type II (7)] or reduced sensitivity of the Leydig cels to
LH [Leydig cell hypoplasia (LCH) (8, 9)]. Two types of
LCH have been described (10). LCH type I is the
severe form of LCH identified in 46 XY individuals
displaying a predominantly female phenotype, which
is caused by mutations in the LH receptor (LHR) that
completely disrupt LH signaling (11–13). Milder forms
of LCH (type II), initially described in 1985 by Toledo et
al. (14), are also caused by a mutation in the LHR gene,
but this defect disrupts LHR signaling less severely,
and patients are characterized by hypospadias or mi-
cropenis (13, 15, 16).
In the present paper we report the identification and
characterization of a homozygous mutation in the LHR
in three brothers with LCH type II. This mutation par-
tially inactivates LH signaling, which explains the phe-
notype. In addition, we have compared this mutation
to other missense mutations previously identified in
patients with LCH. Our data show that a clear corre-
lation exists between receptor activity and the result-
ing phenotype.
RESULTS
Clinical Details
Three brothers with normal karyotype were referred at
the age of 28, 35, and 51 with micropenis, absence of
pubertal signs, and infertility. LH and FSH levels were
elevated but responded normally to GnRH. Basal tes-
tosterone and androstenedione levels, however, were
low and responded poorly to hCG (Table 1; data not
shown). Some variation in their basal and hCG-stim-
ulated testosterone levels was observed among the
brothers. However, this variation is probably the result
of interindividual variation that is also observed in the
normal population (Table 1) (17). Histological analysis
of a testis biopsy of one of the patients showed sem-
iniferous tubules with clearly thickened basal lamina
and an interstitium that lacked mature Leydig cells
(Fig. 1A). Spermatogenesis was present but did not
extend beyond the elongated spermatid stage (Fig.
1B). Taken together these observations indicate that
these patients have a mild form of LCH (LCH type II).
DNA Analysis of the Patients
Single-strand conformation polymorphism (SSCP)
analysis of the LHR gene in two of the affected broth-
ers was first performed on exon 11 because in the
transmembrane domain (TMD) of the receptor most of
the subtle mutations have been identified. An aberrant
migration pattern was found in one of the PCR frag-
ments from both affected brothers while normal
individuals showed only control bands (Fig. 2A). Se-
quencing of the PCR fragment revealed a T-to-A trans-
version at position 1874 of the cDNA (18), changing
codon 625 from ATA for isoleucine to AAA for lysine
(Fig. 2B). Isoleucine 625 is located in the seventh
transmembrane segment of the receptor near the cy-
toplasmic tail. Both tested brothers were homozygous
for the DNA change, and the mutation was not iden-
tified in any of the other samples that were analyzed
(n 525). Therefore, we conclude that the missense
mutation segregates with the disease, although
genomic DNA of the other family members could not
be analyzed.
I625K Mutation Partially Inactivates Signal
Transduction
The I625K mutation was introduced in a wild-type
human LHR (hLHR) cDNA expression vector to pro-
duce pLHR(I625K). To determine the effect of the mu-
tation on LH signal transduction, HEK293 cells were
cotransfected with a luciferase reporter construct con-
taining a cAMP-responsive promoter (CRE
6
-Lux) (19)
and the wild-type expression vector (pLHR(WT)) or
pLHR(I625K). We used a CRE-driven luciferase re-
porter because this response is more sensitive and
more conveniently measured than the regularly used
cAMP response (not shown). Transiently transfected
cells expressing the wild-type LHR responded to hCG
with a 32-fold increase of luciferase activity whereas
cells transfected with LHR(I625K) showed only a 18-
fold increase (Fig. 3A). Furthermore, the EC
50
of this
response in the LHR(I625K) shifted more than a 1
order of magnitude to the right compared with the
EC
50
obtained with the wild-type LHR (Table 2). The
total number of binding sites and the affinity for hCG of
LHR(I625K) were not different from those of the wild-
type receptor (Table 2).
Table 1. Basal and hCG-Induced Testosterone Levels of
Three Brothers with Mild Leydig Cell Hypoplasia
hCG test (10000 IU im) Case I Case II Case III Normal
Testosterone (basal)
a
2.28 3.26 2.99 10–33
b
Testosterone (after hCG)
a
4.17 20.4 10.9 36–60
b
a
Testosterone in nanomoles per liter.
b
Basal and hCG-induced levels in normal men (17).
MOL ENDO · 1998 Vol 12 No. 6
776
Correlation of Receptor Activity and Phenotype
To date, three different missense mutations in the LHR
gene, A593P, S616Y, and, in the present report, I625K,
have been identified in patients with LCH (11, 13, 15).
The extent of the syndrome probably depends on the
residual level of androgen production by the Leydig
cells, which in turn might be a function of the severity
of the effect of the mutation on LH signaling. There-
fore, we compared the characteristics of the different
mutant LHRs in HEK293 cells (Fig. 3B and Table 2).
The three mutant receptors bound hCG with similar
affinity as the wild-type receptor (Table 2). The hCG-
induced cAMP regulatory element (CRE) activity of all
three mutant receptors was impaired (Fig. 3B). As
previously shown, LHR(A593P) did not respond to
hCG at all, while the maximal response of LHR(S616Y)
and LHR(I625K) was reduced to 50% and 65%, re-
spectively (Table 2). In addition, the EC
50
of CRE in-
duction by hCG of both mutant receptors shifted to the
right by a factor of 20. The results obtained with
LHR(S616Y) confirm those of Laue et al. (15); however,
they contradict the results of Latronico et al. (13), who
were unable to show signaling of LHR(S616Y). A pos-
sible explanation for this discrepancy may be the poor
sensitivity of the cAMP production assay. The CRE
luciferase assay used here is much more sensitive,
and in this case appears to be clearly superior to a
cAMP assay.
A reduced hormonal response of the mutant LHRs
could be caused by a reduced expression of the LHR.
Therefore, the amount of LHR receptor protein on a
Western blot (Fig. 4) and the total number of binding
sites were determined (Table 2). To visualize the LHR
molecules on Western blot, the LHR cDNAs were pro-
vided with an hemagglutinin (HA) epitope tag at the
39-end of the open reading frame and overexpressed
in COS-1 cells. The HA tag had no effect on the num-
ber of binding sites or on the hCG-induced CRE re-
sponse (data not shown). In COS-1 cells transfected
with the wild-type LHR cDNA, two major LHR-specific
bands of approximately 65 and 220 kDa were ob-
served (indicated with arrows). Furthermore a minor
band of approximately 300 kDa was present. The two
largest bands are probably multimers or aggregates
but they could also represent glycosylated forms of
the receptor. The two additional bands of approxi-
mately 55 and 70 kDa are the result of nonspecific
binding of the HA antibody, as they also occur in cells
transfected with the empty expression vector (Fig. 4,
right lane). In cells transfected with the different mu-
tant LHR cDNAs, a similar profile was observed but the
intensity of the LHR-specific bands was less, indicat-
ing that the amount of LHR protein is much less. The
total number of binding sites (B
max
) was also deter-
mined as a measure of the amount of receptor that has
been inserted properly into the plasma membrane.
The wild-type LHR and LHR(I625K) had the same
Fig. 1. Testis Histology of Testis of a Patient with Mild LCH
A, Testis biopsy taken from one of the patients. It shows seminiferous tubules with thickened basal lamina and spermato-
genesis arrested in elongated spermatid stage. In the interstitium, mature Leydig cells are absent. Magnification 1603. B, Higher
magnification (4003) of one tubule with elongated spermatids (see arrow). Sections were stained with hematoxylin/eosin.
Partial LHR Inactivation in LCH Type II 777
number of binding sites, but the B
max
values of
LHR(A593P) and LHR(S616Y) were reduced to 0.5%
and 14%, respectively (Table 2). These results indicate
that the processing and/or the transport of the latter
two mutant receptors to the cell surface was impaired.
In conclusion, the expression studies show mutant
LHR molecules do not behave as wild-type molecules
with respect to stability and/or trafficking to the cell
surface. We, therefore, decided to measure the recep-
tor activity of those receptor molecules that present at
the cell surface because these are responsible for
signal transduction. Thus, basal and hCG-induced
CRE activity per cell surface binding-site of the differ-
ent mutants was determined and was compared with
activity of the wild-type receptor expressed at different
levels. Thus, a reference curve for the wild type LHR
was constructed by transfecting different amounts
(0.1, 1, and 10
m
g) of expression vector in HEK293
cells. In these cells, basal and hCG-induced CRE lu-
ciferase activity as well as the number of cell surface
hCG-binding sites was determined (Fig. 5A). Subse-
quently, 10
m
g of the various mutant LHR expression
vectors were transfected into HEK293 cells, and the
same parameters were determined and compared
with the wild-type LHR reference curve (Fig. 5B). In
wild-type LHR-expressing cells, both basal and hCG-
Fig. 2. Homozygous Missense Mutation in LHR Gene
A, SSCP in the coding sequence in patient with mild LCH
(patient). For comparison, SSCP patterns of three normal
individuals (contol) are shown. Normal and aberrant SSCP
patterns are indicated by arrows and arrowheads, respec-
tively. B, Genomic sequence of the LHR gene (from position
1867 to 1882) of patient with mild LCH (left) and normal
individual (right). A homozygous T-to-A conversion was iden-
tified at position 1874 of the LHR gene in the patient. Fig. 3. hCG-Induced CRE Activation by Wild-Type and Mu-
tant LHR
HEK 293 cells were transfected with the expression vector
pSG5 containing the wild-type or indicated mutant hLHR
cDNA in combination with the cAMP responsive construct,
pCRE
6
Lux. The expression vector pRSV-lacZ was included in
the transfection assay as a control for transfection efficiency.
Basal and hCG-induced luciferase activity divided by
b
-ga-
lactosidase activity determined in the same cell lysate was
plotted as means 6SEM (n 54) against the dose of hCG. The
results of one representative experiment out of three is
shown.
MOL ENDO · 1998 Vol 12 No. 6
778
induced CRE activity increased proportionally with the
number of cell surface binding sites. Cell surface ex-
pression of all mutant receptor was reduced com-
pared with the wild-type receptor when 10
m
g expres-
sion vector DNA were also used in the transfection. In
addition, LHR(A593P) completely lacked hormone-de-
pendent signaling activity. In contrast, LHR(S616Y),
which is also poorly expressed at the cell surface,
displayed hardly reduced signaling activity compared
with the wild-type receptor at similar receptor densi-
ties. The expression of LHR(I625K) was also reduced
although to a lesser extent than LHR(S616Y). In addi-
tion, however, this mutant receptor displayed a re-
duced signaling capacity.
DISCUSSION
The characteristics of LCH II that are found in the
family investigated in the present paper are caused by
a homozygous T
1874
to A mutation in the LHR gene.
This base change results in a substitution of a neutral
hydrophobic isoleucine residue by a positively
charged lysine residue at position 625 of the LHR. This
residue is located at the border between the seventh
transmembrane segment and the cytoplasmic tail of
the receptor. The substitution severely impairs hCG-
induced receptor activation, without interfering with
the total number of binding sites and the affinity for the
ligand. The reduced hCG response, however, does not
completely preclude all Leydig cell responses. An hCG
challenge elicited a slight plasma testosterone re-
sponse, which probably explains why spermatogene-
sis in these patients progressed up to late stages of
spermatid differentiation. Spermiation, the last com-
plicated step of sperm release involving major recon-
Table 2. Comparison of Different Mutant LHRs Identified in Patients with LCH
Mutation Wild Type I625K S616Y A593P
Binding
K
d
(nM) 2.4 2.5 1.4 0.6
B
max
[pmol/mg protein] 2.5 1.7 0.36 0.01
CRE response
EC
50
(ng/ml) 0.5 10 10 ND
b
Basal
1
1.0 60.1 1.0 60.1 1.3 60.2 1.0 60.1
Max.
1
30 61.4 18 61.6 14 60.6 1.5 60.2
a
Relative luciferase units (means 6SEM).
b
ND, not detectable.
Fig. 4. Western Blot of Mutant LHRs Expressed in COS-1
Cells
COS-1 cells were transfected with the empty expression
vector pSG5 or with pSG5 containing the indicated wild-type
or mutant HA-tagged hLHR cDNA. Three days after transfec-
tion, the cells were harvested and subjected to 10% SDS-
PAGE followed by Western blotting. Specific bands were
visualized using immunostaining with a HA tag-specific
monoclonal antiserum. The arrows indicated the two most
predominant bands that are specific for the wild-type LHR.
On the left the migration and the molecular weights of three
standard proteins are indicated.
Fig. 5. Comparison of LHR Signaling Capacity per Cell Sur-
face Binding Site
HEK 293 cells were cotransfected with the cAMP respon-
sive reporter gene, pCRE
6
Lux in combination with either dif-
ferent amounts (10, 1, 0.1
m
g) of wild-type LHR cDNA ex-
pression vector (A) or with equal amounts (10
m
g) of
expression vectors containing the wild-type hLHR cDNA or
mutant cDNA A593P, S616Y, or I625K (B). Basal and hCG-
induced luciferase activity (n 54) determined in cell lysate is
plotted as means 6SEM against the number of binding sites
(n 52). The results of one representative experiment out of
two is shown.
Partial LHR Inactivation in LCH Type II 779
struction of the spermatogenic epithelium, did not oc-
cur, which is in accordance with the dependence of
this step on sufficient androgens (20). In patients with
severe LCH who display very low levels of androgens,
spermatogenesis does not occur at all (11), indicating
that the first stages of spermatogenesis may also be
androgen-dependent, although an additional effect of
cryptorchidism in complete LCH patients cannot be
excluded.
The only other reported point mutation causing LCH
type II, S616Y, is located in the same receptor domain,
approximately two
a
-helical turns toward the extracel-
lular side in TMD 7 (Fig. 5) (13, 15). Two patients with
this mutation have been identified independently. One
compound heterozygote patient carrying the S616Y
mutation in combination with a completely inactive
LHR gene (deletion of exon 8) had a small penis and
severe hypospadias, while the other patient, homozy-
gous for S616Y, had a phenotype similar to the pa-
tients described in the present paper, micropenis but
no other indications of aberrant male sex differentia-
tion. Both patients with the S616Y mutations were too
young to be informative about the effect of their LHR
mutation on spermatogenesis.
The extent of the phenotype of patients correlates
well with the effect of the mutation on both receptor
expression and responsiveness to hCG. Homozygous
presence of two LHR(A593P) allelles, giving rise to a
receptor that is poorly expressed and deficient in sig-
naling, is associated with complete pseudohermaph-
roditism. Similar phenotypes are observed in patients
having a premature stop codon in both alleles; severe
truncation of the LHR also results in complete disrup-
tion of signal transduction (12, 13). Homozygocity for a
S616Y or a I625K mutation both cause a milder iden-
tical phenotype (micropenis). The overall receptor ac-
tivity of these two mutant receptors is equally reduced,
albeit this reduction results from different mecha-
nisms. LHR(S616Y) is poorly expressed but responds
normally to hCG while LHR(I625K) is both poorly ex-
pressed and impaired in responding to hCG. In addi-
tion, both mutant receptors require a higher dose of
hCG to respond compared with the wild-type recep-
tor. When the LHR(S616Y) allele is combined with a
completely inactive LHR allele (15), the phenotype of
the compound heterozygous patient is intermediate
between the mild and the complete form of LCH.
(micropenis with severe hypospadia). These correla-
tions of patient phenotype with receptor behavior in
vitro suggest that there may be no clear distinction
between complete and partial feminization of external
genitalia due to LH insensitivity (LCH type I and type II)
as proposed by Toledo et al. (14). Rather, a continuous
range of phenotypes from complete pseudohermaph-
rodism to patients that are only mildly affected is ob-
served depending on the different effects of mutations
on LHR signal transduction and on specific combina-
tions of abnormal alleles. During the preparation of this
manuscript, another mutation causing partial LCH was
described (16). This patient displayed sexual ambigu-
ity at birth due to a homozygous mutation (C131R) in
the extracellular domain of the receptor. The signaling
of the receptor was detectable but severely impaired,
which is in line with the observed phenotype.
Remarkably, LHR(A593P), LHR(S616Y) and
LHR(I625K) display normal binding affinity while their
EC
50
is severly affected. Thus, only the biological re-
sponse of these receptors is impaired. Similar discrep-
ancies have been observed in the TSH receptor (21),
which underscores the separate position of the glyco-
protein hormone receptors within the G protein-cou-
pled receptor (GPCR) superfamily and supports the
hypothesis that these receptors contain two indepen-
dent domains: a ligand-binding domain and a TMD
that transduces the signal into the cell. Only the func-
tion of this latter domain is affected in the three mutant
receptors.
The different characteristics of the tested mutants
also provide clues as to the importance of subdomains
of the receptor molecule. Both the S616Y and I625K
mutations are located on the cytoplasmic side of TMD
7, close to a conserved region in the superfamily of
GPCRs, the NPXXY motif (22). In vitro studies of other
GPCRs, including the glycoprotein hormone recep-
tors, have indicated that the NPXXY motif is important
for ligand-induced receptor activation (23–25) and re-
ceptor sequestration (26). Indeed, I625K and S616Y
may affect the function of the NPXXY motif and in this
way decrease hCG-induced receptor activation.
Based on homology studies of a number of GPCRs,
Baldwin (22, 27) has suggested a model of the most
probable orientation of the seven-transmembrane
a
-helices in the membrane (Fig. 6). In this model,
which appears to agree well with both our own pre-
liminary model (F. Fanelli, personal communication)
and a recently published LHR model (28), the hydro-
phobic isoleucine 625 points toward the hydrophobic
phospholipid membrane. Introduction of a positive
charge by the exchange of a lysine residue at this
position may cause the
a
-helix of TMD 7 to rotate and
move residue 625 toward the hydrophilic core of the
TMD (see arrow in Fig. 6). Alternatively, TMD 7 may be
shifted toward the cytoplasm. Movement of parts of
TMD 7 could be facilitated by the presence of two
helix-breaking proline residues. Furthermore, a turn of
TMD 6 has been observed in a constitutively active
mutant
b
2
-adrenergic receptor (29), indicating that
such a turn can affect signal transduction. A slight
rotation of TMD 7 changes the position of the con-
served NPXXY motif in relation to the rest of the re-
ceptor and in that manner may reduce the response to
hCG. However, the change must be small because the
mutation does not severely affect proper folding. In the
same model, serine 616 points toward TMD 1 and 2 in
the hydrophilic pocket between the transmembrane
helices of the receptor (F. Fanelli, personal communi-
cation). This serine is located exactly one helical turn
N-terminal of the asparagine in the NPXXY motif and is
very well conserved in the GPCR family, which indi-
cates that the size of this residue and its ability to form
MOL ENDO · 1998 Vol 12 No. 6
780
hydrogen bonds may be important. According to mo-
lecular modeling (F. Fanelli, personal communication)
a tyrosine residue at this position may fit in the recep-
tor pocket, although its side chain points toward TMD
3 instead of TMD 1 and 2. However, this fit must be
poor because receptor folding is disturbed as indi-
cated by the reduced cell surface expression. The
intrinsic receptor activity is, however, unaffected,
which indicates that the bulky tyrosine side chain does
not interfere with receptor activation, once it is in
place.
In conclusion, partial LCH in the present family is
due to a homozygous missense mutation (I625K) in
TMD 7 of the LHR. This mutation causes severe im-
pairment in hormone-dependent receptor signaling.
Detailed analysis of three missense mutations that
result in LCH revealed a clear inverse relationship be-
tween residual receptor activity and severity of the
clinical phenotype.
MATERIALS AND METHODS
Patients
The patients studied here have a mild form of LCH that was
designated LCH type II (14). In short, three brothers, born to
consanguineous parents were referred at the ages of 28, 31,
and 51, because of infertility due to azoospermia. The pa-
tients, all with a 46 XY karyotype, had male external genitalia
with adult-sized testis, but an undervirilized penis (micrope-
nis). Baseline levels of testosterone were low and a single
hCG injection (10,000 IU Pregnyl; Organon International, Oss,
The Netherlands) elicited a slight but significant increase of
serum testosterone levels (Table 1). Levels of intermediates
of the testosterone biosynthetic route were not elevated,
indicating absence of enzyme defects (not shown). An acute
adrenal cortex stimulation using 250
m
g Cortrosyn (Organon
International) induced a normal elevation of corticosteroids,
showing that adrenal steroid production was normal (data not
shown). LH and FSH levels were elevated, but the pituitary
responded normally to GnRH (100
m
g iv; Ayerst Laboratories,
Rouse Point, IL) (data not shown). LH bioactivity of one of
these patients was tested and was found to be normal. The
two younger brothers were treated with testosterone enan-
thate (250 mg/3 weeks; Organon International). After 2 yr of
treatment, both patients showed sufficient virilization but pe-
nis size remained inadequate. In only one of the patients did
treatment result in a significant increase in sperm count (from
azoospermia to 3 310
6
/ml) and fertility. All procedures were
carried out in the course of normal patient care after appro-
priate informed consent had been obtained.
SSCP and Sequence Analysis
Genomic DNA was extracted from peripheral blood (30) of
two of the affected brothers. Six overlapping fragments of
exon 11 of the LHR gene were amplified by PCR and
analyzed by SSCP as described previously (Ref. 31 and H.
Kremer et al., submitted). For sequencing, PCR fragments
were treated with alkaline phosphatase and exonuclease I
and sequenced using the USB sequencing kit for PCR
fragments (US Biochemical Corporation, Cleveland, OH).
Fig. 6. Model of the TMD of the hLHR
Projection of the cytoplasmic part of the TMD of the LHR according to the model proposed by Baldwin (22, 27). The view is
from the intracellular side toward the outside. The size of the dots indicate the distance of the amino acid to the cytoplasm (a large
dot means that the amino acid is close to the cytoplasm). The amino acids of the NPXXY motif are indicated in squares; the two
amino acids mutated in patients with partial LCH are indicated in circles. The amino acid mutated in the patient with complete
LCH (A593P) is located at the border between TMD 6 and extracellular loop 3 and is therefore absent in the projection.
Partial LHR Inactivation in LCH Type II 781
Construction of Mutant hLHR Expression Vectors
Wild-type hLHR cDNA was introduced in the expression vec-
tor pSG5 (33), resulting in pLHR(WT) (34). Mutations were
introduced into this construct using standard PCR mutagen-
esis (34, 35) with the primers (Pharmacia, Uppsala, Sweden)
described below. The nucleotides that differ from the wild-
type hLHR cDNA are indicated in bold.
LHR1512FOR: 59-GTC GGT GTC AGC AAT TAC-39;
LHR2182REV: 59-GTT AAA ATT ACT GGT ACA GG-39;
LHR616SYFOR: 59-CCC ATC AAT TATTGCGCAAAT
CCA TTT-39;
LHR616SYREV: 59-AAA TGG ATT TGC GCA ATA ATT
GAT GGG-39;
LHR625IKFOR: 59-G TAT GCA AAA TTC ACT AAG-39;
LHR625IKREV: 59-CTT AGT GAA TTT TGC ATA C-39.
For constructing pLHR(I625K), primer sets LHR1512FOR/
LHR625IKREV and LHR625IKFOR/LHR2181REV were used
separately to perform the first PCR amplification. After mixing
of the fragments, the final mutant fragment was obtained by
PCR using the primer set LHR1512FOR and LHR2181REV.
To construct the mutant hLHR expression vector, a BstXI-
HpaI fragment of the reamplified fragment (669 bp) was used
to replace the wild-type sequence in pLHR(WT), resulting in
pLHR(I625K). For the construction of pLHR(S616Y), a similar
strategy was used using the LHR616 primer set. Both con-
structs were checked by DNA sequencing. The construct
pLHR(A593P) was described previously (11).
Transfection of COS-1 and HEK293 Cells
COS-1 and HEK293 cells were maintained in culture medium
(DMEM/Ham’s F12 (1:1 vol/vol) (GIBCO BRL, Gaithersburg,
MD), 2 310
5
IU/liter penicillin (Brocades Pharma, Leiderdorp,
The Netherlands) and 0.2 g/liter streptomycin (Radium
Farma, Milan, Italy) and 5% and 10% FCS (SEBAK, Aiden-
bach, Germany), respectively, and were incubated in a hu-
midified incubator at 37 C and 5% CO
2
. Before transfection
the cells were seeded at 15% confluence in 75-cm
2
flasks
(Nunc, Roskilde, Denmark) and transfected the next day with
1 ml precipitate containing 20
m
g DNA (36).
cAMP Reporter Activity Measurements
For measuring the hormonal response of the different mu-
tants, HEK293 cells were cotransfected with pCRE
6
Lux
(19), pRSVlacZ (37) and pSG5, pLHR(WT), pLHR(A593P),
pLHR(S616Y) or pLHR(I625K) (10
m
g expression construct, 1
m
g pRSVlacZ, 2
m
g pCRE
6
Lux, and 7
m
g carrier DNA per ml
precipitate). Three days after transfection the hCG-depen-
dent CRE response was determined in 24-well tissue culture
plates (Costar, Cambridge, MA) by incubating the cells for 4 h
in culture medium containing 0.1% BSA with increasing con-
centrations of hCG (0.001 to 1000 ng/ml; urinary hCG; Or-
ganon International). Subsequently, the cells were lysed and
luciferase activity was measured (38).
b
-Galactosidase activ-
ity of the lysates was determined to correct for transfection
efficiency (37).
Scatchard Analysis
To determine the binding affinity (K
d
) and total receptor num-
ber (B
max
), HEK293 cells were transfected with pSG5,
pLHR(WT), pLHR(A593P), pLHR(S616Y), or pLHR(I625K) (10
m
g expression construct and 10
m
g carrier DNA per ml pre-
cipitate). Three days after transfection, Scatchard analysis
using chloramine T
125
I-labeled hCG (39) was performed on
purified membranes according to Ketelslegers and Catt (40).
cAMP Reporter Activity per Cell Surface Receptor
Number
For determining the receptor activity per receptor number
expressed at the cell surface, HEK293 cells were trans-
fected with pCRE
6
Lux, pRSVlacZ and pSG5, pLHR(WT),
pLHR(A593P), pLHR(S616Y), or pLHR(I625K). Three days af-
ter transfection a part of the transfected cells was used to
measure the basal and maximal hCG (1000 ng/ml) CRE re-
sponse as described above while the rest of the cells were
used to measure LHR cell surface expression. Cell surface
expression was determined as described previously (41, 42).
Briefly, transfected cells were harvested and resuspended in
binding buffer (10 mMTris-HCl, pH 7.5, 5 mMMgCl
2
, 0.1%
BSA, 5 mMsodium azide, 200 mMsucrose) containing
125
I-
labeled hCG (1.5 310
6
cpm) in the presence or absence of
a 1,000 fold excess of unlabeled hCG in a volume of 0.2 ml.
The sodium azide was added to prevent internalization. After
incubation for1hat37C,thecells were washed twice with
excess of binding buffer, after which the binding as measured
by the amount of radioactive hCG bound to the cells was
counted in a
g
-counter.
SDS-PAGE
To determine LHR expression on Western blots, LHR cDNAs
were extended with an HA immuno tag (YPYDVPDYAS) at the
39-end. This HA tag did not affect the number of binding sites
or hormone-dependent signaling (not shown). COS-1 cells
were transfected with pSG5, pLHR(WT)HA, pLHR(A593P)HA,
pLHR(S616Y)HA, or pLHR(I625K)HA (10
m
g expression plas-
mid and 10
m
g carrier DNA per ml precipitate). Three days
after transfection, the cells were washed twice with PBS and
harvested in 1 ml PBS. After the protein concentration was
determined (Bradford), equal amounts of protein (3.5
m
g)
were separated on 10% SDS/PAGE (43) and subsequently
blotted onto nitrocellulose (Schleicher & Schuell, Dassel, Ger-
many) using the Mini-Protean II gel electrophoresis and elec-
tro-blotting apparatus (Bio-Rad, Hercules, CA). The tagged
LHR proteins were visualized using the Renaissance Western
blot chemiluminescence detection kit (DuPont/NEN, Du Pont
de Nemours GmbH, Dreieich, Germany) using as primary
antibody a 1:500 dilution of the HA-specific monoclonal an-
tiserum 12C5.
Acknowledgments
We would like to thank Dr. H. Kremer for her support during
the initial stages of this project, Marianna Timmerman for
technical assistence, Dr. A. Himmler for supplying the CRE
luciferase construct, Dr. E. Milgrom for supplying the expres-
sion vector containing the wild-type hLHR cDNA, Dr. F.
Fanelli for performing molecular modeling on the LHR, and
Dr. Th. van der Kwast for evaluating the testis pathology.
Received October 23, 1997. Revision received February
17, 1998. Accepted March 2, 1998.
Address requests for reprints to: J. W. M. Martens,
Department of Endocrinology and Reproduction, Erasmus
University Rotterdam, P.O. Box 1738, 3000 DR Rotterdam,
The Netherlands. E-mail: martens@endov.fgg.eur.nl.
The research of J. Martens is financed by the Netherlands
Organisation of Scientific Research and that of S. Toledo and
N. Abelin by the National Research Council of Brazil (CNPq),
the State Research Foundation (FAPESP), and the Medical
Research Laboratory (LIM).
MOL ENDO · 1998 Vol 12 No. 6
782
REFERENCES
1. Goodfellow PN, Lovell-Badge R 1993 SRY and sex de-
termination in mammals. Annu Rev Genet 27:71–92
2. Buehr M, Gu S, McLaren A 1993 Mesonephric contribu-
tion to testis differentiation in the fetal mouse. Develop-
ment 117:273–281
3. Huhtaniemi I, Pelliniemi LJ 1992 Fetal Leydig cells: cel-
lular origin, morphology, life span, and special functional
features. Proc Soc Exp Biol Med 201:125–140
4. Josso N, Cate RL, Picard JY, Vigier B, di Clemente N,
Wilson C, Imbeaud S, Pepinsky RB, Guerrier D, Boussin
L, et a 1993 Anti-mullerian hormone: the Jost factor.
Recent Prog Horm Res 48:1–59
5. Griffin JE, McPhaul MJ, Russel DW, Wilson JD 1995 The
androgen resistance syndromes: 5-alpha-reductase 2
deficiency, testicular feminization, and related disorders.
In: Scriver CR, Beaudet AL, Sly WS,Valle D (eds) The
Metabolic and Molecular Bases of Inherited Disease.
McGraw-Hill, Inc, New York, pp 2987–2998
6. Brinkmann AO, Kuiper GG, Ris-Stalpers C, van Rooij HC,
Romalo G, Trifiro M, Mulder E, Pinsky L, Schweikert HU,
Trapman J 1991 Androgen receptor abnormalities. J Ste-
roid Biochem Mol Biol 40:349–352
7. Andersson S, Geissler WM, Wu L, Davis DL, Grumbach
MM, New MI, Schwarz HP, Blethen SL, Mendonca BB,
Bloise W, Witchel SF, Cutler Jr GB, Griffin JE, Wilson JD,
Russel DW 1996 Molecular genetics and pathophysiol-
ogy of 17 beta-hydroxysteroid dehydrogenase 3 defi-
ciency. J Clin Endocrinol Metab 81:130–136
8. Lee PA, Rock JA, Brown TR, Fichman KM, Migeon CJ,
Jones Jr H 1982 Leydig cell hypofunction resulting in
male pseudohermaphroditism. Fertil Steril 37:675–679
9. El-Awady MK, Temtamy SA, Salam MA, Gad YZ 1987
Familial Leydig cell hypoplasia as a cause of male
pseudohermaphroditism. Hum Hered 37:36–40
10. Toledo SP 1992 Leydig cell hypoplasia leading to two
different phenotypes: male pseudohermaphroditism and
primary hypogonadism not associated with this. Clin En-
docrinol (Oxf) 36:521–522
11. Kremer H, Kraaij R, Toledo SPA, Post M, Fridman JB,
Hayashida CY, Van Reen M, Milgrom E, Ropers HH,
Mariman E, Themmen APN, Brunner HG 1995 Male
pseudohermaphroditism due to a homozygous missense
mutation of the luteinizing hormone receptor gene. Nat
Genet 9:160–164
12. Laue L, Wu S-M, Kudo M, Hsueh AJW, Cutler Jr GB,
Griffin JE, Wilson JD, Brain C, Berry AC, Grant DB, Chan
W-Y 1995 A nonsense mutation of the human luteinizing
hormone receptor gene in Leydig cell hypoplasia. Hum
Mol Genet 4:1429–1433
13. Latronico AC, Anasti J, Arnhold IJ, Rapaport R,
Mendonca BB, Bloise W, Castro M, Tsigos C, Chrousos
GP 1996 Brief report: testicular and ovarian resistance to
luteinizing hormone caused by inactivating mutations of
the luteinizing hormone-receptor gene. N Engl J Med
334:507–512
14. Toledo SP, Arnhold IJ, Luthold W, Russo EM, Saldanha
PH 1985 Leydig cell hypoplasia determining familial
hypergonadotropic hypogonadism. Prog Clin Biol Res
200:311–314
15. Laue LL, Wu S-M, Kudo M, Bourdony CJ, Cutler Jr GB,
Hsueh AJW, Chan W-Y 1996 Compound heterozygous
mutations of the luteinizing hormone receptor gene in
Leydig cell hypoplasia. Mol Endocrinol 10:987–997
16. Misrahi M, Meduri G, Pissard S, Bouvattier C, Beau I,
Loosfelt H, Jolivet A, Rappaport R, Milgrom E, Bougne-
res P 1997 Comparison of immunocytochemical and mo-
lecular features with the phenotype in a case of incom-
plete male pseudohermaphroditism associated with a
mutation of the luteinizing hormone receptor. J Clin En-
docrinol Metab 82:2159–2165
17. Mahoudeau JA, Valcke JC, Bricaire H 1975 Dissociated
response of plasma testosterone and estradiol to human
chorionic gonadotropin in adult men. J Clin Endocrinol
Metab 41:13–20
18. Minegishi T, Nakamura K, Takakura Y, Miyamoto K,
Hasegawa Y, Ibuki Y, Igarashi M 1990 Cloning and se-
quencing of human LH/hCG receptor cDNA. Biochem
Biophys Res Commun 172:1049–1054
19. Himmler A, Stratowa C, Czernilofsky AP 1993 Functional
testing of human dopamine D1 and D5 receptors ex-
pressed in stable cAMP-responsive luciferase reporter
cell lines. J Recept Res 13:79–94
20. Parvinen M 1993 Cyclic function of Sertoli cells. In: Rus-
sell LD, Griswold MD (eds) The Sertoli Cell. Cache River
Press, Clearwater, FL, pp 331–347
21. De Roux N, Misrahi M, Brauner R, Houang M, Carel JC,
Granier M, Le Bouc Y, Ghinea N, Boumedienne A,
Toublanc JE, Milgrom E 1996 Four families with loss of
function mutations of the thyrotropin receptor. J Clin
Endocrinol Metab 81:4229–4235
22. Baldwin JM 1993 The probable arrangement of the
helices in G protein-coupled receptors. EMBO J
12:1693–1703
23. Arora KK, Cheng Z, Catt KJ 1996 Dependence of agonist
activation on an aromatic moiety in the DPLIY motif of the
gonadotropin-releasing hormone receptor. Mol Endocri-
nol 10:979–986
24. Barak LS, Menard L, Ferguson SS, Colapietro AM, Caron
MG 1995 The conserved seven-transmembrane se-
quence NP(X)2,3Y of the G-protein-coupled receptor su-
perfamily regulates multiple properties of the beta 2-ad-
renergic receptor. Biochemistry 34:15407–15414
25. Puett D, Bhowmick N, Fernandez LM, Huang J, Wu C,
Narayan P 1996 hCG-receptor binding and transmem-
brane signaling. Mol Cell Endocrinol 125:55–64
26. Barak LS, Tiberi M, Freedman NJ, Kwatra MM, Lefkowitz
RJ, Caron MG 1994 A highly conserved tyrosine residue
in G protein-coupled receptors is required for agonist-
mediated beta 2-adrenergic receptor sequestration.
J Biol Chem 269:2790–2795
27. Baldwin JM 1994 Structure and function of receptors
coupled to G proteins. Curr Opin Cell Biol 6:180–190
28. Lin Z, Shenker A, Pearlstein R 1997 A model of the
lutropin/choriogonadotropin receptor: insights into the
structural and functional effects of constitutively activat-
ing mutations. Protein Eng 10:501–510
29. Javitch JA, Fu D, Liapakis G, Chen J 1997 Constitutive
activation of the
b
2 adrenergic receptor alters the orien-
tation of its sixth membrane-spanning segment. J Biol
Chem 272:18546–18549
30. Miller SA, Dykes DD, Polesky HF 1988 A simple salting
out procedure for extracting DNA from human nucleated
cells. Nucleic Acids Res 16:1215
31. Kremer H, Mariman E, Otten BJ, Moll Jr G, Stoelinga GB,
Wit JM, Jansen M, Drop SL, Faas B, Ropers HH, Brunner
HG 1993 Cosegregation of missense mutations of
the luteinizing hormone receptor gene with familial
male-limited precocious puberty. Hum Mol Genet
2:1779–1783
32. Deleted in proof
33. Green S, Issemann I, Sheer E 1988 A versatile in vivo and
in vitro eukaryotic expression vector for protein engineer-
ing. Nucleic Acids Res 16:369
34. Kraaij R, Post M, Kremer H, Milgrom E, Epping W,
Brunner HG, Grootegoed JA, Themmen APN 1995 A
missense mutation in the second transmembrane do-
main of the luteinizing hormone receptor causes familial
male-limited precocious puberty. J Clin Endocrinol
Metab 80:3168–3172
35. Higuchi R, Krummel B, Saiki RK 1988 A general method
of in vitro preparation and specific mutagenesis of DNA
fragments: study of protein and DNA interactions. Nu-
cleic Acids Res 16:7351–7367
Partial LHR Inactivation in LCH Type II 783
36. Chen C, Okayama H 1987 High-efficiency transformation
of mammalian cells by plasmid DNA. Mol Cell Biol
7:2745–2752
37. Hall CV, Jacob PE, Ringold GM, Lee F 1983 Expression
and regulation of Escherichia coli lacZ gene fusions in
mammalian cells. J Mol Appl Genet 2:101–109
38. Blok LJ, Themmen AP, Peters AH, Trapman J, Baarends
WM, Hoogerbrugge JW, Grootegoed JA 1992 Transcrip-
tional regulation of androgen receptor gene expression in
Sertoli cells and other cell types. Mol Cell Endocrinol
88:153–164
39. Dufau ML, Charreau EH, Catt KJ 1973 Characterisation
of a soluble gonadotropin receptor from the rat testis.
J Biol Chem 248:6973–6982
40. Ketelslegers JM, Catt KJ 1978 Follitropin receptors in rat
testis. Characterization with enzymatically 125I-labeled
human follitropin. Biochim Biophys Acta 541:360–371
41. Van Loenen HJ, Flinterman JF, Rommerts FFG 1994
Follicle stimulating hormone stimulates rat Sertoli cells
via relatively low affinity binding sites. Endocrine
2:1023–1029
42. Kawate N, Menon KM 1994 Palmitoylation of luteinizing
hormone/human choriogonadotropin receptors in trans-
fected cells. Abolition of palmitoylation by mutation of
Cys-621 and Cys-622 residues in the cytoplasmic tail
increases ligand-induced internalization of the receptor.
J Biol Chem 269:30651–30658
43. Leamli UK 1970 Cleavage of structural proteins during
assembly of the head bacteriophage T4. Nature
227:680–685
MOL ENDO · 1998 Vol 12 No. 6
784
... LCH type II is a rare subtype of LCH. Although it was first described in 1985 [30], few reports have described patients with partial (insufficient) LHCGR activation in 46,XY patients [1,8,9,11,12,34]. Most of the mutant proteins causing LCH type II produced partially active LHCGR, which retain some responsiveness to LH and hCG hormones, implying normal male development during fetal life. ...
... Most of the mutant proteins causing LCH type II produced partially active LHCGR, which retain some responsiveness to LH and hCG hormones, implying normal male development during fetal life. After birth, patients were presented with micropenis, cryptorchidism, and/or hypospadias and sometimes only infertility [8,12,16]. In contrast, LCH type I is the more severe form associated with complete inactivation of LHCGR, which causes complete unresponsiveness to LH and hCG hormones. ...
... Reported variants related to LCH type II, as shown in Supplementary Table 2, were located within the seventh TMD (p.Ser616Tyr and p.Ile625Lys) or the second TMD (p.Ile415Thr) [8,9,11,12,34]. Herein, the third TMD (440-462 aa) with the detected missense variant (p. Phe-444Cys) has broadened the genetic variation spectrum. ...
Article
Full-text available
Purpose Leydig cell hypoplasia (LCH) type II is a rare disease with only a few cases reported. Patients presented with hypospadias, micropenis, undescended testes, or infertility. In this study, we report a new patient with compound heterozygous variants in the LHCGR gene and LCH type II phenotype. Methods: Whole exome sequencing (WES) was performed followed by Sanger sequencing to confirm the detected variants in the patient and his parents. Results: A novel missense variant (p.Phe444Cys) was identified in a highly conserved site and is verified to be in trans with the signal peptide’s 33-bases insertion variant. Conclusion: Our research provides a more comprehensive clinical and genetic spectrum of Leydig cell hypoplasia type II. It highlighted the importance of WES in the diagnosis of this uncommon genetic disorder as well as the expansion of the genotype of LCH type II.
... hCG'ye yetersiz testosteron yanıtı vardır. Kısmi Leydig hücre hipoplazisinde klinik yetersiz virilize erkek (mikropenis ve/veya hipospadias) fenotipinden, erkek dış cinsel organlarının belirsizliği olmaksızın hipergonadotropik hipogonadizme kadar geniş bir yelpazeye sahiptir 44 . Olgularda hCG'ye kısmi testosteron yanıtı alınabilir ve pubertal dönemde artan androjenlere bağlı virilizasyon görülebilir. ...
Chapter
Full-text available
46,XY disorders of sex development (DSD) are characterized by variable development of the external genitalia, from an atypical appearance to a completely female external genitalia, as a result of intrauterine inadequate masculinization in the presence of a 46,XY karyotype. Müller structures may or may not be accompanied to varying degrees in some cases. Male gonads have been identified in the majority of 46, XY DSD patients, but some have no gonadal tissue.Cases with no masculinization have a completely female external genitalia and may present due to a lack of breast development and/or primary amenorrhea. 46,XY DSD may occur as a result of disorders at any stage of gonadal differentiation, disorders in androgen production and effect, and anti-müllerian hormone disorders. 46, XY DSD cases should be evaluated with a detailed history, a complete physical examination, and laboratory and imaging methods, and the management of the cases should be planned by the multidisciplinary team according to the underlying cause. (PDF) 46, XY cinsiyet gelişim bozuklukları. Available from: https://www.iuc-universitypress.org/books/disorders-of-gender-development https://www.researchgate.net/publication/378526679_46_XY_cinsiyet_gelisim_bozukluklari
... 32 However, a large proportion of LCF patients are not diagnosed until adulthood. 26,27 To our surprise, we herein found that adult mice responded to AAV-mediated gene therapy as effectively as pubertal mice, as demonstrated by similar increments of testosterone production and restart of sexual development. Notably, we observed massive spermatozoa in the testis and caudal epididymis of Lhcgr À/À mice in adult cohorts after AAV8-Lhcgr treatment. ...
Article
Full-text available
Leydig cell failure (LCF) caused by gene mutation results in testosterone deficiency and infertility. Serum testosterone levels can be recovered via testosterone replacement; however, established therapies have shown limited success in restoring fertility. Here, we use a luteinizing hormone/choriogonadotrophin receptor (Lhcgr)-deficient mouse model of LCF to investigate the feasibility of gene therapy for restoring testosterone production and fertility. We screen several adeno-associated virus (AAV) serotypes and identify AAV8 as an efficient vector to drive exogenous Lhcgr expression in progenitor Leydig cells through interstitial injection. We observe considerable testosterone recovery and Leydig cell maturation after AAV8-Lhcgr treatment in pubertal Lhcgr−/− mice. Of note, this gene therapy partially recovers sexual development, substantially restores spermatogenesis, and effectively produces fertile offspring. Furthermore, these favorable effects can be reproduced in adult Lhcgr−/− mice. Our proof-of-concept experiments in the mouse model demonstrate that AAV-mediated gene therapy may represent a promising therapeutic approach for patients with LCF.
... In patients with type 2 LCH, partial receptor inactivation is present. In these patients, testosterone is partly synthesized and they manifest a wide clinical spectrum ranging from micropenis to hypospadias, depending on the testosterone level [20][21][22]. Type 1 LCH was considered in all of the patients in our study. Because although the karyotypes of all of the patients were 46,XY, their external genitalia were female. ...
Article
Introduction The luteinizing hormone/choriogonadotropin receptor (LHCGR) plays a critical role in sexual differentiation and reproductive functions in men and women. Inactivating mutations in this gene lead to Leydig cell hypoplasia (LCH), and cause disorders of sex development (DSD) in patients with 46,XY. In this study, it was aimed to discuss the clinical, laboratory and molecular genetic analysis results of nine patients with 46,XY karyotype who had mutations in the LHCGR gene. Materials and methods The ages, complaints, anthropometric measurements and hormonal results (follicle stimulating hormone (FSH), luteinizing hormone (LH), testosterone) of the patients at the time of admission were recorded retrospectively from their medical records. The mutations in the LHCGR gene were investigated using the Sanger sequencing method. Findings In this study, LHCGR gene mutations were detected in a total of nine patients as a result of the analysis of the index patients presenting with primary amenorrhea from four different families and the examination of the families. In the first three families with no consanguinity between, the same mutation was detected in seven patients in total (Homozygous c.161 + 4A > G). A different mutation was detected in the fourth family (Homozygous p.A483D c.1448C > A). Conclusion In this study, nine patients with karyotype 46,XY, most of whom presented with the complaint of delayed puberty/primary amenorrhea, were diagnosed with LCH. Especially in patients, in whom the elevation of LH is pronounced and there is no testosterone synthesis, LCH should be considered.
... In the androgen signalling pathway, androgen signals are passed by the nuclear Androgen Receptor (Lee et al., 2016) Luteinizing hormone/chorionic gonadotropin receptor (LHCGR) mutations lead to Leydig cell hypoplasia and reduction of testosterone during the prenatal stages (Richter-Unruh et al., 2002;Wu and Chan, 1999). The external genitalia of XY patients can be complete female or undervirilized (Martens et al., 2014). Additionally, persistent Müllerian duct syndrome can be cause by the mutations in AMH and its receptor AMHR2, which is responsible for the regression of Müllerian ducts during fetal development. ...
Thesis
Sex determination is a highly sophisticated and ordered process where both male and female gonads develop from a common bipotential gonad depending on different activated signaling pathways. XY gonads develop into a testis promoted by the SRY/SOX9 pathway, whereas XX gonads develop into an ovary through the action of the RSPO1/WNT4/ß-Catenin pathway. R-spondin (Rspo) genes encode one kind of secreted proteins that activate the canonical WNT/β-Catenin pathway by inhibiting the degradation of WNT receptors. After binding to its receptors LGR4/5, RSPO1-LGR4/5 recruit E3 Ubiquitin-Protein Ligases ZNRF3 and RNF43 to release WNT receptors from being degraded by ubiquitination process, therefore WNT/β-Catenin signaling can be activated continuously. Rspo1 is a major regulator of ovary development across species. In the developing mouse gonad, Rspo1 is mainly expressed in the female supporting cells, and Rspo1 XX mutant gonads undergo female-to-male sex reversal by developing into ovotestis, gonads with both male and female characteristics. The molecular and cellular mechanisms behind this partial sex reversal remain unclear. In this work, we have developed a new mouse model allowing a conditional mutation of Rspo1. We have established that the critical window for Rspo1 requirement in the developing XX mouse gonad is around E11.5, and that Rspo1 function is dispensable for ovarian differentiation after this time point. We have shown that ectopic steroidogenesis is an early event in the phenotypic changes of XX Rspo1 mutant gonads. Through pharmacological inhibition of the androgen receptor we have identified androgen signaling pathway as a major player in the female-to-male sex reversal of XX Rspo1 mutant gonads. In the second part of this work we have studied the phenotype of Sox9cKOWnt4KO double mutants where both the male and female pathway are impaired. We have found that XX Sox9cKOWnt4KO gonads develop as ovotestis indicating that the additional deletion of Sox9 did not rescue the female-to-male sex reversal caused by the Wnt4 mutation. We have also shown that XY Sox9cKOWnt4KO double knockout mutants undergo a transient female-like developmental phase before the gonads develop into ovotestis. This result demonstrates that Wnt4 deletion cannot rescue the initial steps of the male-to-female sex reversal caused by the Sox9 mutation. Together, these results reveal the timing of requirement of Rspo1 in ovarian development and highlight the pro-male role of androgen signaling in the XX female-to-male sex reversal process. It also rises new thoughts on the interactions between male and female pathway in mouse sex determination.
Article
G protein‐coupled receptors (GPCRs) are central to the functioning of the hypothalamic–pituitary–gonadal axis (HPG axis) and include the rhodopsin‐like GPCR family members, neurokinin 3 receptor, kappa‐opioid receptor, kisspeptin 1 receptor, gonadotropin‐releasing hormone receptor, and the gonadotropin receptors, luteinizing hormone/choriogonadotropin receptor and follicle‐stimulating hormone receptor. Unsurprisingly, inactivating variants of these receptors have been implicated in a spectrum of reproductive phenotypes, including failure to undergo puberty, and infertility. Clinical induction of puberty in patients harbouring such variants is possible, but restoration of fertility is not always a realisable outcome, particularly for those patients suffering from primary hypogonadism. Thus, novel pharmaceuticals and/or a fundamental change in approach to treating these patients are required. The increasing wealth of data describing the effects of coding‐region genetic variants on GPCR function has highlighted that the majority appear to be dysfunctional as a result of misfolding of the encoded receptor protein, which, in turn, results in impaired receptor trafficking through the secretory pathway to the cell surface. As such, these intracellularly retained receptors may be amenable to ‘rescue’ using a pharmacological chaperone (PC)‐based approach. PCs are small, cell permeant molecules hypothesised to interact with misfolded intracellularly retained proteins, stabilising their folding and promoting their trafficking through the secretory pathway. In support of the use of this approach as a viable therapeutic option, it has been observed that many rescued variant GPCRs retain at least a degree of functionality when ‘rescued’ to the cell surface. In this review, we examine the GPCR PC research landscape, focussing on the rescue of inactivating variant GPCRs with important roles in the HPG axis, and describe what is known regarding the mechanisms by which PCs restore trafficking and function. We also discuss some of the merits and obstacles associated with taking this approach forward into a clinical setting.
Article
Spermatogenesis is a complex process of germ cell division and differentiation that involves extensive cross-talk between the developing germ cells and the somatic testicular cells. Defective endocrine signaling and/or intrinsic defects within the testes can adversely affect spermatogenic progression, leading to subfertility/infertility. In recent years, male infertility has been recognized as a global public health concern, and research over the last few decades has elucidated the complex etiology of male infertility. Congenital reproductive abnormalities, genetic mutations, and endocrine/metabolic dysfunction have been demonstrated to be involved in infertility/subfertility in males. Furthermore, acquired factors like exposure to environmental toxicants and lifestyle-related disorders such as illicit use of psychoactive drugs have been shown to adversely affect spermatogenesis. Despite the large body of available scientific literature on the etiology of male infertility, a substantial proportion of infertility cases are idiopathic in nature, with no known cause. The inability to treat such idiopathic cases stems from poor knowledge about the complex regulation of spermatogenesis. Emerging scientific evidence indicates that defective functioning of testicular Sertoli cells (Sc) may be an underlying cause of infertility/subfertility in males. Sc plays an indispensable role in regulating spermatogenesis, and impaired functional maturation of Sc has been shown to affect fertility in animal models as well as humans, suggesting abnormal Sc as a potential underlying cause of reproductive insufficiency/failure in such cases of unexplained infertility. This review summarizes the major causes of infertility/subfertility in males, with an emphasis on infertility due to dysregulated Sc function.
Article
Objectives We present the first cases of two male brothers with Leydig cell hypoplasia secondary to a novel mutation in the LHCGR gene that has never been described before. Case presentation We report the case of two brothers with Leydig cell hypoplasia (LCH) type II caused by novel homozygous inactivating mutation of the LHCGR gene, located in exon 10 in c 947 position. The two patients presented at 11 years 7 months and 1 year 6 months, respectively, with abnormal sexual development, micropenis and cryptorchidism. Genetic analysis revealed a homozygous deletion of approximately 4 bp encompassing exon 10 of the LHR gene in the two brothers indicating autosomal recessive inheritance. An hCG stimulation test induced testosterone secretion within the normal range. Subsequently, a treatment with enanthate of testosterone was started, with an increase in the length of the penis. Conclusions Leydig cell hypoplasia is a rare form of disorder of sex development. We report the occurrence of a new mutation of the LHCGR gene in two Moroccan brothers in whom the clinical features and the molecular diagnosis were correlated.
Chapter
The androgen receptor (AR) mediates the endocrine functions of endogenous androgens like testosterone and exerts pleiotropic androgenic (male phenotype development, sexual function) and anabolic (growth and maintenance of musculoskeletal system) effects across most tissues. The AR‐axis, i.e. AR and its downstream effects, is extensively targeted to stimulate anabolism or treat prostatic diseases. Tissue‐selective, direct‐acting synthetic AR agonists are limited in scope by their inadequate separation of anabolic from untoward androgenic or virilizing effects and include FDA approved steroidal androgens (anabolic‐androgenic steroids) and investigational nonsteroidal selective AR modulators (SARMs). Gaining in popularity is testosterone replacement therapy in hypogonadal men. Antagonism of the AR‐axis is common in aging males with androgen‐dependent prostate hyperproliferation [prostate cancer or benign prostatic hyperplasia (BPH)]. Androgen ablation (indirect antagonism) and direct AR antagonism have been the mainstays of prostate cancer therapies for many decades including recent approvals. Indirect antagonists of the AR‐axis include gonadotropin‐releasing hormone agonists or antagonists (androgen‐deprivation therapy), which achieve systemic androgen and estrogen ablation for prostate or breast cancers as a monotherapy or sometimes in combination with steroidogenic enzyme (lyase) inhibitors. DHT‐dependent diseases are treated with 5α‐reductase to block intracrine production of DHT for male pattern baldness and BPH; whereas aromatase inhibitors block androgen metabolism to estrogen in breast cancer. Direct AR antagonists (antiandrogens) with improved potency and anti‐androgenic scope continue to be approved for prostate cancer. Research into innovative ways to directly or indirectly modulate the AR‐axis remains robust, suggesting new therapies will likely be introduced regularly for the foreseeable future.
Article
Full-text available
Leydig cell hypoplasia is a rare autosomal recessive condition that interferes with normal development of male external genitalia in 46,XY individuals. We have studied two Leydig cell hypoplasia patients (siblings born to consanguineous parents), and found them to be homozygous for a missense mutation (Ala593Pro) in the sixth transmembrane domain of the luteinizing hormone (LH) receptor gene. In vitro expression studies showed that this mutated receptor binds human choriogonadotropin with a normal KD, but the ligand binding does not result in increased production of cAMP. We conclude that a homozygous LH receptor gene mutation underlies the syndrome of autosomal recessive congenital Leydig cell hypoplasia in this family. These results have implications for the understanding of the development of the male genitalia.
Article
We describe a simple calcium phosphate transfection protocol and neo marker vectors that achieve highly efficient transformation of mammalian cells. In this protocol, the calcium phosphate-DNA complex is formed gradually in the medium during incubation with cells and precipitates on the cells. The crucial factors for obtaining efficient transformation are the pH (6.95) of the buffer used for the calcium phosphate precipitation, the CO2 level (3%) during the incubation of the DNA with the cells, and the amount (20 to 30 micrograms) and the form (circular) of DNA. In sharp contrast to the results with circular DNA, linear DNA is almost inactive. Under these conditions, 50% of mouse L(A9) cells can be stably transformed with pcDneo, a simian virus 40-based neo (neomycin resistance) marker vector. The NIH3T3, C127, CV1, BHK, CHO, and HeLa cell lines were transformed at efficiencies of 10 to 50% with this vector and the neo marker-incorporated pcD vectors that were used for the construction and transduction of cDNA expression libraries as well as for the expression of cloned cDNA in mammalian cells.
Article
We investigate the contribution of the Iberian bat fauna to the cryptic diversity in Europe using mitochondrial (cytb and ND1) and nuclear (RAG2) DNA sequences. For each of the 28 bat species known for Iberia, samples covering a wide geographic range within Spain were compared to samples from the rest of Europe. In this general screening, almost 20% of the Iberian species showed important mitochondrial discontinuities (K2P distance values > 5%) either within the Iberian or between Iberian and other European samples. Within Eptesicus serotinus and Myotis nattereri, levels of genetic divergence between lineages exceeded 16%, indicating that these taxa represent a complex of several biological species. Other well-differentiated lineages (K2P distances between 5–10%) appeared within Hypsugo savii, Pipistrellus kuhlii and Plecotus auritus, suggesting the existence of further cryptic diversity. Most unsuspected lineages seem restricted to Iberia, although two have crossed the Pyrenees to reach, at leas...
Article
The interaction between enzymatically radioiodinated human follitropin and the follitropin receptors in testis homogenate was investigated in immature and adult rats. The 125I-labeled human follitropin exhibited high binding activity with specific binding of up to 17% in the presence of an excess of testis homogenate. Approx. 50% of the bound hormone could be eluted at pH 5, and the receptor purified tracer exhibited a 3.6-fold increase in binding activity when compared with the original tracer preparation. Quantitative analysis of equilibrium binding data was performed with corrections for the measured specific activity and maximum binding activity of the tracer hormone. The equilibrium association constants (Ka) determined 24 degrees C were not significantly different in immature and adult rat testis, and the mean value for Ka was 3.9 . 10(9) M-1. At 37 degrees C, the Ka value obtained using immature rat testis was 1.3 . 10(10) M-1. The association of 125I-labeled human follitropin with immature rat testis homogenate was time and temperature dependent. In the presence of an excess of unlabeled hormone, 30--60% of the preformed hormone . receptor complex was dissociated after 24 h incubation. A specific and sensitive radioligand-receptor assay for follitropin was developed using immature rat testis homogenate. The minimum detectable dose of purified human follitropin was 0.6 ng, and human urinary and pituitary follitropin, ovine follitropin and pregnant mare serum gonadotropin reacted in the assay with equivalent slopes. The potencies of highly purified pregnent mare serum gonadotropin and highly purified human follitropin were similar in the radioligand-receptor assay, consistent with the follitropin bioactivity of the equine gonadotropin.
Article
Plasma testosterone (T), dihydrotestosterone (DHT) and estradiol (E2) were determined by radioimmunoassay in 10 normal males receiving hCG im 5000 IU on days 1, 2 and 3. The mean increase of plasma steroid on days 2, 3 and 1, respectively, was: 1.42, 1.79 and 1.87 times for T; 1.17, 1.56 and 1.49 times for DHT; 4.04, 3.29 and 2.33 times for E2. While T was still significantly increasing from day 2 to day 4, E2 significantly decreased. A positive correlation (P less than 0.01) was found between the basal T and the E2 peak after hCG, suggesting a release of E2 from a storage compartment in the testis. No significant change of either steroid was detected 4 h after the first hCG injection. In 6 cases of primary male hypogonadism, the mean basal values of T to hCG was defective, despite considerable individual variations. In 14 males with gonadotropin deficiency, basal values of T and E2 were very low; the T response to hCG ranged from undetectable to dramatic, and was correlated with the degree and duration of previous exposure to gonadotropin; and impaired response of E2 in all cases porvides a better estimate of the actual gonadotropin deficiency.
Article
Regulation of androgen receptor (AR) mRNA expression was studied in Sertoli cells and peritubular myoid cells isolated from immature rat testis, and in the lymph node carcinoma cell line derived from a human prostate (LNCaP). Addition of dibutyryl-cyclic AMP (dbcAMP) to Sertoli cell cultures resulted in a rapid transient decrease in AR mRNA expression (5 h), which was followed by a gradual increase in AR mRNA expression (24–72 h). This effect of dbcAMP mimicked follicle-stimulating hormone (FSH) action. In peritubular myoid cells, there was only a moderate but prolonged decrease during incubation in the presence of dbcAMP, and in LNCaP cells no effect of dbcAMP on AR mRNA expression was observed.