ArticlePDF Available

Abstract and Figures

Background: Increased secretion of growth hormone leads to gigantism in children and acromegaly in adults; the genetic causes of gigantism and acromegaly are poorly understood. Methods: We performed clinical and genetic studies of samples obtained from 43 patients with gigantism and then sequenced an implicated gene in samples from 248 patients with acromegaly. Results: We observed microduplication on chromosome Xq26.3 in samples from 13 patients with gigantism; of these samples, 4 were obtained from members of two unrelated kindreds, and 9 were from patients with sporadic cases. All the patients had disease onset during early childhood. Of the patients with gigantism who did not carry an Xq26.3 microduplication, none presented before the age of 5 years. Genomic characterization of the Xq26.3 region suggests that the microduplications are generated during chromosome replication and that they contain four protein-coding genes. Only one of these genes, GPR101, which encodes a G-protein-coupled receptor, was overexpressed in patients' pituitary lesions. We identified a recurrent GPR101 mutation (p.E308D) in 11 of 248 patients with acromegaly, with the mutation found mostly in tumors. When the mutation was transfected into rat GH3 cells, it led to increased release of growth hormone and proliferation of growth hormone-producing cells. Conclusions: We describe a pediatric disorder (which we have termed X-linked acrogigantism [X-LAG]) that is caused by an Xq26.3 genomic duplication and is characterized by early-onset gigantism resulting from an excess of growth hormone. Duplication of GPR101 probably causes X-LAG. We also found a recurrent mutation in GPR101 in some adults with acromegaly. (Funded by the Eunice Kennedy Shriver National Institute of Child Health and Human Development and others.).
Content may be subject to copyright.
The
new england journal
of
medicine
n engl j med nejm.org
1
original article
Gigantism and Acromegaly Due to Xq26
Microduplications and GPR101 Mutation
G. Trivellin, A.F. Daly, F.R. Faucz, B. Yuan, L. Rostomyan, D.O. Larco,
M.H. Schernthaner-Reiter, E. Szarek, L.F. Leal, J.-H. Caberg, E. Castermans,
C. Villa, A. Dimopoulos, P. Chittiboina, P. Xekouki, N. Shah, D. Metzger,
P.A. Lysy, E. Ferrante, N. Strebkova, N. Mazerkina, M.C. Zatelli, M. Lodish,
A. Horvath, R. Bertollo de Alexandre, A.D. Manning, I. Levy, M.F. Keil,
M. de la Luz Sierra, L. Palmeira, W. Coppieters, M. Georges, L.A. Naves,
M. Jamar, V. Bours, T.J. Wu, C.S. Choong, J. Bertherat, P. Chanson, P. Kamenický,
W.E. Farrell, A. Barlier, M. Quezado, I. Bjelobaba, S.S. Stojilkovic, J. Wess,
S. Costanzi, P. Liu, J.R. Lupski, A. Beckers, and C.A. Stratakis
The author s’ full names, academ ic degrees,
and affiliations are listed in the Appendix.
Address reprint requests to Dr. Stratakis at
10 Center Dr., Bldg. 10, National Institutes
of Health, Clinical Research Center, Rm.
1-3330, MSC1103, Bethesda, MD 20892-
1862, or at stratakc@mail.nih.gov; or to
Dr. Beckers at th e Department of Endocri-
nology, Centre Hospitalier Universitaire
de Liège (B35), Domain e Universitaire du
Sart-Tilman, 4000 Liege, Belgium, or at
albert.beckers@chu.ulg.ac.be.
Drs. Trivellin and Daly, and Drs. Beckers
and Stratakis, contributed equally to this
article.
This arti cle was published on De cember 3,
2014, at NEJM.org.
DOI: 10.1056/NEJMoa1408028
Copyright © 2014 Massachusetts Medical Society.
ABSTRACT
Background
Increased secretion of growth hormone leads to gigantism in children and acro-
megaly in adults; the genetic causes of gigantism and acromegaly are poorly under-
stood.
Methods
We performed clinical and genetic studies of samples obtained from 43 patients
with gigantism and then sequenced an implicated gene in samples from 248 pa-
tients with acromegaly.
Result s
We observed microduplication on chromosome Xq26.3 in samples from 13 patients
with gigantism; of these samples, 4 were obtained from members of two unrelated
kindreds, and 9 were from patients with sporadic cases. All the patients had disease
onset during early childhood. Of the patients with gigantism who did not carry an
Xq26.3 microduplication, none presented before the age of 5 years. Genomic char-
acterization of the Xq26.3 region suggests that the microduplications are generated
during chromosome replication and that they contain four protein-coding genes.
Only one of these genes, GPR101, which encodes a G-protein–coupled receptor, was
overexpressed in patients’ pituitary lesions. We identified a recurrent GPR101 mutation
(p.E308D) in 11 of 248 patients with acromegaly, with the mutation found mostly
in tumors. When the mutation was transfected into rat GH3 cells, it led to increased
release of growth hormone and proliferation of growth hormone–producing cells.
Conclusions
We describe a pediatric disorder (which we have termed X-linked acrogigantism
[X-LAG]) that is caused by an Xq26.3 genomic duplication and is characterized by
early-onset gigantism resulting from an excess of growth hormone. Duplication
of GPR101 probably causes X-LAG. We also found a recurrent mutation in GPR101 in
some adults with acromegaly. (Funded by the Eunice Kennedy Shriver National
Institute of Child Health and Human Development and others.)
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
The
new england journal
of
medicine
n engl j med nejm.org
2
S
omatic growth is orchestrated by a
complex hormonal crosstalk involving the
hypothalamus, pituitary, and peripheral tis-
sues.
1
Genetic disorders that affect this network
can lead to increased secretion of growth hor-
mone, which results in acromegaly. If the excess
in growth hormone occurs before epiphyseal fu-
sion, the result can be gigantism. Nonsyndromic
gigantism is most frequently caused by pituitary
adenomas occurring as familial isolated pituitary
adenomas or sporadically, usually as a result of
mutations in the gene encoding aryl hydrocarbon
receptor–interacting protein (AIP).
2-4
Other mono-
genic diseases can cause gigantism, but most of
these conditions develop in adulthood in associa-
tion with other tumors.
5
In young children, so-
matic overgrowth that is due to an excess of
growth hormone is rare, and the cause is un-
known. Other syndromic genetic overgrowth con-
ditions in children, such as the Sotos syndrome
and the Simpson–Golabi–Behmel syndrome, are
not associated with pituitary abnormalities.
6,7
We report a striking phenotype of gigantism
that has an onset in early childhood and that is
caused by an excess of growth hormone. The
disorder is associated with heritable microdupli-
cations on chromosome Xq26.3. There are four
genes in the duplicated stretch of DNA; one of
these, GPR101, encodes an orphan G-protein–
coupled receptor and is probably the gene that
drives the phenotype in young children and the
growth of sporadic growth hormone–producing
adenomas in some patients with acromegaly.
Methods
Patients
We analyzed samples obtained from 43 patients
with gigantism who had hypersecretion of
growth hormone, evidence of an anterior pitu-
itary lesion on magnetic resonance imaging, a
height on country-specific growth charts of ei-
ther more than the 97th percentile or more than
2 SD above the mean height for age, and negative
test results for mutations or deletions in genes
associated with pituitary adenomas (Table 1).
Details with respect to one family with this syn-
drome8,9 and two patients with sporadic dis-
ease10,11 have been described previously.
Genetic Analyses
We sequenced the four genes in the duplicated
region on chromosome Xq26.3 in 259 germline
and tumor DNA samples that were obtained
from 248 patients with sporadic acromegaly (Ta-
ble S1 in the Supplementary Appendix, available
with the full text of this article at NEJM.org). We
sequenced GPR101 and performed array compar-
ative genomic hybridization (aCGH) on germline
DNA in samples obtained from 13 families with
familial isolated pituitary adenomas without AIP
mutations. We used quantitative reverse-transcrip-
tase–polymerase-chain-reaction (qRT-PCR) assays
to measure the expression levels of duplicated
genes in both leukocytes and pituitary tumors.
We performed comparative protein-structure
modeling on GPR101 using Modeller software,
version 9.13.12 We determined the level of growth
hormone and cyclic AMP (cAMP) and the rate of
cellular proliferation after transient overexpres-
sion of each of the four implicated genes in GH3
cells obtained from rat pituitary tumors.
Study Oversight
The institutional review board at each of the par-
ticipating institutions approved our studies. We
studied the anonymized samples from interna-
tional acromegaly cohorts with approval from the
National Institutes of Health Intramural Office
for Human Research Protections. Written in-
formed consent was obtained from all adult pa-
tients and parents or guardians of children with
early-onset gigantism.
Results
Clinical Presentation
The clinical and biochemical characteristics of
the 43 patients who had nonsyndromic gigan-
tism without abnormalities in genes associated
with pituitary tumors are presented in Table 1.
Genetic analyses delineated two phenotypes: an
early-childhood form of gigantism with a typical
onset in late infancy (Fig. 1) and a second form
with a typical onset in adolescence.
Identification of Xq26.3 Microduplication
We detected microduplications on chromosome
Xq26.3 in samples obtained from patients with
the early-childhood form of gigantism (Fig. 2,
and Fig. S1, S2, and S3 in the Supplementary Ap-
pendix). Nine of the 13 patients with an Xq26.3
microduplication and the 1 probable carrier (an
affected mother with gigantism) were female
and were of normal size at birth. All the patients
grew rapidly during infancy, attaining a median
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
Gigantism and Acromegaly Due to Xq26 abnormalities
n engl j med nejm.org
3
height score of +3.8 SD at diagnosis (median age,
36 months). At the time of diagnosis, they
showed marked overall somatic growth, with el-
evated weight and an enlarged head circumfer-
ence (median, 51.2 cm). The onset of accelerated
growth and the onset of accelerated weight gain
usually coincided but were not always synchro-
nous (Fig. 1, and Fig. S4 in the Supplementary
Appendix). As compared with patients who did
not have an Xq26.3 microduplication, those with
the microduplication had an earlier median age
at the onset of abnormal growth (12 months vs.
16 years), an increased acceleration in height,
and elevated levels of insulin-like growth factor 1
and prolactin (
Table 1
). We did not observe pre-
cocious puberty in the microduplication carriers.
Levels of peripheral growth hormone–releasing
hormone did not suggest ectopic secretion of this
hormone, and nuclear imaging scans were nega-
tive for other tumors.
Of the 13 patients who underwent surgery, 10
had pituitary macroadenomas alone (median
maximum diameter, 16 mm), and 3 patients had
pituitary hyperplasia, with or without an identi-
fied adenoma (Fig. 3H). In all the patients, hor-
monal control was not achieved with medical
therapy alone. Such control required either radi-
cal or repeated neurosurgery alone (in 4 pa-
tients) or in combination with the administra-
tion of the
growth hormone receptor antagonist
pegvisomant (in 3 patients) or radiotherapy (in
2 patients). Seven patients had permanent hypo-
pituitarism at the time of this study.
The common duplicated genomic segment
was approximately 500 kb in length, from posi-
tion 135,627,637 to 136,118,269 (GRCh37/hg19)
Table 1. Clinical Characteristics of 43 Patients with Gigantism with and without Xq26.3 Microduplications.
Characteristic Xq26.3 Microduplication
(N = 14) No Xq26.3 Microduplication
(N = 29) P
Value
Female sex — no. (%) 10 (71) 7 (24) 0.007
Median age at onset of rapid growth (range) — yr 1.0 (0.5 to 2.0) 16.0 (5.0 to 18.0) <0.001
Median age at diagnosis (range) — yr 3 (1 to 22) 21 (5 to 34) <0.001
Median height at diagnosis (range) — cm 116 (99 to 175) 187 (171 to 209) <0.001
Median standard-deviation score for height at
diagnosis (range) +3.8 (+1.9 to +7.1) +3.3 (+2.1 to +5.8) 0.45
Elevated levels of growth hormone and insulin-like
growth factor 1 at diagnosis — no. (%) 14 (100) 29 (100) 1.00
No suppression of growth hormone during oral
glucose-tolerance test — no. (%) 14 (100) 29 (100) 1.00
Median factor increase in insulin-like growth factor 1
at diagnosis (range) — multiple of ULN 4.4 (2.4 to 5.2) 2.1 (1.4 to 5.3) 0.005
Elevated prolactin level at diagnosis — no. (%) 13 (93) 6 (21) <0.001
Median maximum tumor diameter (range) — cm 16 (10 to 39) 20 (9 to 41) 0.16
Adenoma or hyperplasia — no. (%)†
Both adenoma and hyperplasia 2 (14) 0
Adenoma only 10 (71) 29 (100)
Hyperplasia only 1 (7) 0
Type of syndrome — no. (%)
Sporadic 9 (64) 29 (100)
Familial 5 (36)‡ 0
Siblings with normal growth — no./total no. (%) 9/11 (82) 29/29 (100)
* ULN denotes upper limit of the normal range.
The presence of hyperplasia or adenoma could not be determined in one patient who did not undergo surgery.
In one patient with the familial syndrome, pituitary gigantism was diagnosed in the mother and son at the same visit,
when the son was 8 years of age and the mother was 22 years of age. The mother had had tall stature and acromegalic
features since childhood for which she had not been referred for medical attention. The clinical data for the mother, for
whom DNA was not available, are included.
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
The
new england journal
of
medicine
n engl j med nejm.org
4
Birth to 24 months: Boys
Length-for-age and Weight-for-age percentiles
Published by the Centers for Disease Control and Prevention, November 1, 2009
SOURCE: WHO Child Growth Standards (http://www.who.int/childgrowth/en)
98
95
85
75
50
25
10
5
2
98
95
90
75
50
25
10
5
2
Birth to 24 months: Girls
Length-for-age and Weight-for-age percentiles
Published by the Centers for Disease Control and Prevention, November 1, 2009
SOURCE: WHO Child Growth Standards (http://www.who.int/childgrowth/en)
98
95
90
75
50
25
10
5
2
98
95
90
75
50
25
10
5
2
A
B C
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
Gigantism and Acromegaly Due to Xq26 abnormalities
n engl j med nejm.org
5
(Fig. 2). One patient had a complex genomic re-
arrangement, with two duplicated segments that
were separated by a short region of normal ge-
nomic sequence. No other patterns of duplica-
tion or deletion or homozygosity were shared
among the affected patients. One family with
familial isolated pituitary adenomas included an
affected mother and two affected sons (who
have been described previously
8
) with the same
Xq26.3 microduplication; the unaffected father
did not have the duplication. In another family
with this condition, the mother had childhood-
onset gigantism and a histologically confirmed
pituitary macroadenoma but had died of com-
plications of hypopituitarism. She had two
children: the son carried the Xq26.3 microdu-
plication and had childhood-onset gigantism
(Patient F2A), and the healthy daughter did not
have the duplication. The most parsimonious ex-
planation is that the son inherited the X-linked
disease from his carrier mother. Hence, Xq26.3
microduplications can be considered to be a
new pathogenic explanation in certain kindreds
with familial isolated pituitary adenomas that
have acrogigantism without AIP mutations.
Chromosomal position
Patient No.
S9
S8
S7
S6
S5
S4
S2
S1
F2A
F1A/F1B/F1C
135,500,000 135,700,000 135,900,000 136,100,000 136,300,000
Familial
Familial
Sporadic
Sporadic
Sporadic
Sporadic
Sporadic
Sporadic
Sporadic
Sporadic
SRO1 SRO2
BRS3
HTATSF1
VGLL1
CD40LG
ARHGEF6 RBMX GPR101
Figure 2. Summary of the Genomic Gains on Chromosome Xq26.3.
Shown are 10 different Xq26.3 microduplications, as seen on array comparative genomic hybridization, that were
found in 12 patients with familial or sporadic gigantism (with the inheritance pattern indicated at right). Duplicated
genomic segments (red) and nonduplicated segments (white) are shown. The genomic coordinates are provided at
base-pair resolution on the x axis. The two smallest regions of overlap (SRO), SRO1 and SRO2, are identified, show-
ing the genomic contents in the corresponding regions. The symbols next to the gene names represent the struc-
ture of the genes, with vertical lines representing exons and horizontal lines (with or without arrows) representing
introns. Adapted from the UCSC Genes track in the UCSC Genome Browser.
Figure 1 (facing page). Familial and Sporadic Cases
of Gigantism and Male and Female Growth Patterns
Due to the Xq26.3 Microduplications.
Panel A shows Patient F1C, who has familial gigantism,
at the age of 3 years. His growth chart up to 24 months
of age shows the rapid acceleration in weight, although
the acceleration in height did not begin until after his
second birthday (Fig. S4 in the Supplementary Appendix).
Panel B shows an unaffected mother and her daughter
(Patient S6), who has sporadic gigantism and whose
height was 120 cm at the age of 3 years. A growth chart
for Patient S4 (Panel C), another girl with sporadic gigan-
tism, illustrates the typical early increase in height and
weight seen in patients with Xq26.3 microduplications,
starting at the age of 6 months in this child.
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
The
new england journal
of
medicine
n engl j med nejm.org
6
ABCD
E G HF
20 µm 20 µm 20 µm 20 µm
Figure 3. Imaging and Histopathological Findings in Patients with Xq26.3 Microduplications.
Panels A through D show progressive changes from normal pituitary tissue (Panel A) to adenoma (Panel D), as indicated by reticulin staining of the pituitary gland in Patient F1C.
In Panel A, normal pituitary gives way to the expanded hyperplastic acini (Panel B), and in Panel C, areas of transformation are evident (circled) with enlarged, hyperplastic, conflu-
ent acini that are caused by breakdown of reticulin fibers and that lead to adenoma (Panel D) with disruption of the reticulin fiber network. Increased GPR101 expression was ob-
served in five tested patients with Xq26 microduplications, whereas there is little if any expression in normal pituitary tissue or growth hormone–producing tumors without Xq26.3
microduplications or GPR101 defects (see also Fig. S7 in the Supplementary Appendix); an example is shown here (Panels E through G) from Patient S3. When the staining of growth
hormone (Panel E) and the staining of GPR101 (Panel F) are merged, GPR101 seems to be expressed in some of the growth hormone–secreting cells (Panel G, arrows) but not in all
such cells. Nuclei (blue) were stained with DAPI. Panel H shows a sagittal view of a macroadenoma on magnetic resonance imaging of Patient S5 with the Xq26.3 microduplication.
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
Gigantism and Acromegaly Due to Xq26 abnormalities
n engl j med nejm.org
7
Further Characterization of Xq26.3
Microduplication
Using high-definition analysis of the critical du-
plicated region, we analyzed 10 distinct genomic
duplications in 12 patients, including 4 patients
with the familial form of the disease and 8 pa-
tients with the sporadic form (Fig. S1 and S3 in
the Supplementary Appendix). On genomewide
aCGH, these mutations appeared to be simple
duplications. However, using high-resolution
aCGH, long-range PCR, and Sanger sequencing
of the breakpoints, we found various underlying
genomic complexities (Fig. S3 in the Supplemen-
tary Appendix).
All sporadic Xq26.3 duplications were nonre-
current; the boundaries of the duplicated seg-
ment were unique to each patient. On both
aCGH and breakpoint PCR assays, samples ob-
tained from unaffected parents and siblings of
patients with sporadic disease showed negative
results, documenting the microduplication as a
new mutation (Fig. S3A and S5A in the Supple-
mentary Appendix). The same duplication was
transmitted from an affected mother (Patient F1A)
to her affected offspring, Patients F1B and F1C
(Fig. 2, and Fig. S3 and S5B in the Supplemen-
tary Appendix).
The duplicated genomic regions that were
shared by all affected persons consisted of the
two smallest regions of overlap (SRO), which were
designated as SRO1 and SRO2 (Fig. 2). SRO1
(chromosomal posit ion, 135,627,637 to 135,986,830;
hg19) encompassed three genes in the Online
Mendelian Inheritance in Man (OMIM) data-
base: CD40LG (OMIM number, 300386), ARHGEF6
(OMIM number, 300267), and RBMX (OMIM
number, 300199), whereas SRO2 (chromosomal
position, 136,045,310 to 136,118,269; hg19) in-
cluded GPR101 (OMIM number, 300393) (Fig. 2).
Investigation of Candidate Genes
Sequencing of each of the four genes in the 43
patients with gigantism did not reveal any single-
nucleotide variants of likely pathogenicity. A
quantitative RT-PCR assay of pituitary tumor
RNA from 2 patients with Xq26.3 microduplica-
tions suggested that CD40LG was not expressed in
the pituitary tumors. Neither ARHGEF6 nor RBMX
showed up-regulated expression in the pituitary
tumors of 2 patients with the duplication (Fig. 4).
In contrast, the expression of GPR101 in the pitu-
itaries of the children carrying an Xq26.3 dupli-
cation was increased by a factor as high as 1000,
as compared with unaffected pituitary tissue and
pituitary tumors from persons who tested nega-
tive for microduplications (Fig. 4A). This result
was confirmed at the protein level by increased
immunostaining for GPR101 in pituitary tumors
from patients with Xq26.3 duplications (Fig. 3G,
and Fig. S7 in the Supplementary Appendix). Ex-
perimental overexpression of ARHGEF6, RBMX,
and GPR101 alone in the rat GH3 cell line did not
signif icantly increase either cell proliferation or
the secretion of growth hormone (Fig. 4D and
4E, and Fig. S8 in the Supplementary Appendix).
Nonmutated GPR101 in combination with ARH-
GEF6, RBMX, or both modestly increased cell pro-
liferation but not the secretion of growth hor-
mone (Fig. S8 in the Supplementary Appendix).
The X-chromosome–inactivation pattern was
random in the female patients with sporadic
disease and skewed in Patient F1A, who had fa-
milial disease; CpG islands were identified in
silico only in RBMX and GPR101 (Fig. S9 and S10
in the Supplementary Appendix).
Identification of p.E308D Mutation in GPR101
In a series of 248 patients with sporadic acro-
megaly, none carried a microduplication at
Xq26.3. However, 11 patients had a c.924G→C
substitution (p.E308D) in GPR101, which was not
found in 7600 control samples obtained from
public databases (Tables S1 and S2 in the Supple-
mentary Appendix). Of the 11 mutation carriers,
3 appeared to carry a constitutive mutation,
which was detected in DNA from peripheral-
blood mononuclear cells (PBMCs). We detected
the mutation in the tumor DNA in the remaining
8 patients (Fig. 5A). In one patient, we deter-
mined that the mutation was a de novo somatic
mutation — that is, the GPR101 mutation oc-
curred only in the tumor DNA sequence and not
in the PBMC sequence (Fig. 5B). None of the 13
families with familial isolated pituitary adeno-
mas carried the p.E308D mutation in GPR101.
GPR101 encodes an orphan
G-protein–coupled
receptor
that is highly expressed in rodent hypo-
thalamus (Fig. S11 and S12 in the Supplemen-
tary Appendix) and is predicted to couple to the
stimulatory G protein (G
s
), a potent activator of
adenylyl cyclase.
13,14
A model of human GPR101
in complex with a G
s
heterotrimer shows the phy-
sical relationship between the p.E308D amino
acid change and the activating p.A397K change,
a mutation that has been described previously.
15
The two amino acids, which are predicted to be
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
The
new england journal
of
medicine
n engl j med nejm.org
8
D
Cell Proliferation
1.6
1.4
1.2
1.0
0.8
0.6
0.4
0.2
0.0
Control GPR101-
WT GPR101-
E308D GPR101-
A397K
Relative Units
** *
Normalized mRNA Expression
0NP1 NP2 NP3 NP4 NP5 GH1 GH2 F1C S6
200
400
600
800
1000
1200
1400
A
GPR101
Growth Hormone
E
45
40
35
30
25
20
15
10
5
0
Control GPR101-
WT
GPR101-
E308D
GPR101-
A397K
Relative Units
Vehicle
***
***
***
Forskolin
***
**
***
CRE Activation
F
1.6
1.4
1.2
1.0
0.8
0.6
0.4
0.2
0.0
Control GPR101-
WT GPR101-
E308D GPR101-
A397K
Relative Units
***
**
**
*
0.0
Normalized mRNA Expression
0.5
1.0
1.5
2.0
2.5
NP1 NP2 GH1 GH2 F1C S6
B
ARHGEF6
Normalized mRNA Expression
4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0.0 NP1 NP2 GH1 GH2 F1C S6
C
RBMX
Figure 4. Expression of GPR101 in Pituitary Tissue from Children with Xq26.3 Microduplications.
The expression of GPR101 in pituitary tissue from children carrying Xq26.3 microduplications was increased by a factor as high as 1000, as compared with the expression in unaffected
pituitary tissue (in five samples [NP1 through NP5] obtained on autopsy) and in pituitary tumors from two patients with sporadic acromegaly (GH1 and GH2) who tested negative for
the microduplication (Panel A). These findings, which were obtained on quantitative reverse-transcriptase–polymerase-chain-reaction (qRT-PCR) assay and normalized by a housekeep-
ing gene, contrast with those for two other genes, ARHGEF6 (Panel B) and RBMX (Panel C), in the duplicated stretch of DNA; neither of these two genes showed up-regulated expres-
sion. Also shown are cell proliferation (Panel D), growth hormone secretion (Panel E), and activation of
DNA sequences called cyclic AMP response elements (CRE)
(Panel F) in rat
GH3 cells transfected with mutant (p.E308D and p.A397K) and nonmutant GPR101 constructs. Values for cells transfected with empty (control) vector were set at 1. Also shown are
values for untreated cells (vehicle) and forskolin (which increases CRE activation). Data are expressed as the mean results of three to five independent experiments, each of which was
performed in triplicate. The T bars indicate standard deviations. One asterisk denotes P<0.05, two asterisks P<0.01, and three asterisks P<0.001.
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
Gigantism and Acromegaly Due to Xq26 abnormalities
n engl j med nejm.org
9
C
D308
IL 3
A397
TM 6
TM 5
AB
G G GA A C
308 309
308 309 308 309 308 309
Germline
DNA
Normal
Tumor
DNA
Mutation
Male
(Hemizygote)
Female
(Heterozygote)
Normal
Tumor DNA
G G G G G G GA A A AAC C C CAS
Figure 5. Effect of the p.E308D Mutation in GPR101 in 11 Patients with Sporadic Acromegaly.
Panel A shows the sequence for GPR101 in growth hormone–producing pituitary tumors obtained from patients with
sporadic acromegaly, as compared with normal tissue. Panel B shows results for a patient with a somatic mutation,
which was determined by the presence of the mutation in the GPR101 sequence of DNA in the tumor sample but
not in the sequence in peripheral-blood mononuclear cells. None of the 13 families with familial isolated pituitary
adenomas carried the p.E308D mutation in GPR101. Panel C shows a structural model of GPR101
bearing the p.E308D
mutation. Residue A397 is located at the cytosolic end of transmembrane (TM) 6 of GPR101. The mutated D308 res-
idue and the nonmutated A397 residue are shown in space-filling representation and colored according to elements,
with carbon atoms in gray, oxygen atoms in red, and nitrogen atoms in blue. The backbone of the receptor and the
G protein heterotrimer is schematically represented as a ribbon, with the receptor shown with a spectrum of colors
that ranges from red at the N-terminal to purple at the C-terminal; the α, β, and γ subunits of the G protein are in gray,
blue, and pink, respectively. The cytosolic ends of TM 5 and TM 6 and intracellular loop (IL) 3, which connects them,
are indicated by labels. The blue arrows show directions of the β-sheet domains of the β subunit of the G protein.
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
The
new england journal
of
medicine
n engl j med nejm.org
10
affected by the mutations, are on the cytosolic
side of the receptor (Fig. 5C). The E308 residue
is located in the long intracellular loop 3, which
connects transmembrane domains 5 and 6.
Overexpression of the p.E308D and p.A397K
mutants, but not of nonmutant GPR101, signifi-
cantly increased cell proliferation and secretion
of growth hormone in rat GH3 cells (Fig. 4D and
4E). As in the construct containing the nonmu-
tant receptor, the two mutant constructs result-
ed in increased cAMP signaling in GH3 cells in
an in vitro reporter assay, both at baseline and
in the presence of 10 μM forskolin, a direct
stimulator of adenylyl cyclase (Fig. 4F).
Discussion
Several lines of evidence support the identifica-
tion of a new pituitary gigantism syndrome in
young children carrying microduplications on
chromosome Xq26.3, a disorder that is probably
caused by GPR101 overexpression. We propose that
this syndrome be called X-linked acrogigantism
(X-LAG). First, we did not find disruption of
Xq26.3 in patients with later-onset gigantism
(Table 1). Second, the finding that patients with
other conditions had different duplications with-
in the same region narrowed our focus to the
smallest region of overlap. A duplication encom-
passing CD40LG and ARHGEF6 but not RBMX and
GPR101 occurred in a family with low birth
weight, intellectual disability, and craniofacial
abnormalities,16 which suggests that duplica-
tions with the exclusion of RBMX and GPR101 do
not lead to gigantism. Third, short stature has
been reported in several patients with deletions
in this region, which suggests that the absence
of these genes may lead to the opposite pheno-
type (Table S4 in the Supplementary Appen-
dix).16-18 Other investigators have described at
least 15 additional patients with the same pheno-
type of early-onset growth who may be good
candidates for a diagnosis of X-LAG (Table S3 in
the Supplementary Appendix).
The breakpoint features of Xq26.3 duplica-
tions suggest that they were generated by means
of a replication-based mechanism that under-
lies the genesis of other copy-number variants
(CNVs) and the pathogenesis of other genomic
disorders.
19
The cytogenetic data narrowed the smallest
region of overlap to a segment spanning
CD40LG, ARHGEF6, RBMX, GPR101, one microRNA
(miR-934), and a small nucleolar RNA (SNORD61)
of unknown function. We did not detect CD40LG
expression in the pituitary tissues from our pa-
tients (Fig. 4). Messenger RNA for ARHGEF6 and
RBMX was expressed to a similar degree in af-
fected and unaffected tissues from duplication
carriers. Of all the genes and the noncoding
RNAs in the duplicated segment, only GPR101
had markedly increased expression in the pitu-
itary tumors from the duplication carriers (Fig. 4).
GPR101 is an orphan G-protein–coupled recep-
tor that is strongly expressed in the hypothalamus
in rodents (Fig. S11 and S12 in the Supplemen-
tary Appendix).
13,14,20
It was recently shown that
a fragment of the gonadotropin-releasing hor-
mone could be a ligand for this receptor.
21
The
GPR101 protein may also play a role in hypotha-
lamic control of energy homeostasis.
22
The ef-
fect of a mutation (p.A397K) that is predicted to
activate GPR101 when tested in vitro and in mice
supports such a role.
15
The pituitary-specific over-
expression of GPR101 may be due to a gene-dose
effect (as described in many genomic disorders
23
)
or to an unknown promoter sequence created by
the chromosomal rearrangement, although we did
not identify any putative new promoter, or to per-
turbed chromatin regulation due to the genomic
structural alteration from duplication CNVs.
24,25
On the basis of our data from transfection experi-
ments, we cannot rule out a modest contribution
of RBMX and ARHGEF6 coexpression to cell prolif-
eration. However, unlike GPR101, neither ARHGEF6
nor RBMX was overexpressed in the pituitary
tumors from children with microduplications.
Our studies of sporadic acromegaly provide
further support for a role of GPR101 in X-LAG.
We found a recurrent GPR101 mutation, p.E308D,
in 4.4% of DNA in tumor samples and in 1.9%
of DNA in PBMC samples obtained from patients
with isolated acromegaly. In at least one patient,
the mutation was present only in the tumor DNA.
We did not identify GPR101 mutations in families
with familial isolated pituitary adenomas. A model
of human GPR101 in complex with a G
s
hetero-
trimer showed that both the p.E308D mutation
and the previously described p.A397K mutation
15
are on the cytosolic side of the receptor that
interacts with heterotrimeric G proteins. Residue
E308 is located in a remarkably long intracellu-
lar loop, which connects two transmembrane
domains. But in the absence of a model template
for the GPR101 intracellular loop in which E308
resides, it is difficult to estimate the structural
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
Gigantism and Acromegaly Due to Xq26 abnormalities
n engl j med nejm.org
11
effect of the p.E308D substitution. However, trans-
fection of a construct expressing GPR101 contain-
ing the p.E308D mutation increased proliferation
and growth hormone secretion in a rat pituitary
cell line. Moreover, we showed that GPR101 can
strongly activate the cAMP pathway, for which the
mitogenic effects in pituitary somatotropes are
well established.
26
These data further support a
role for variant GPR101 in sporadic acromegaly.
The mechanism by which mutant GPR101
contributes to increased growth hormone secre-
tion is unclear. Some of the patients with early-
onset gigantism whom we evaluated had normal
or mildly elevated levels of circulating growth
hormone–releasing hormone (but below the
threshold required for ectopic tumoral secretion
of this hormone), as was previously noted in
Family F1.
8
The tumor tissue showed strong
expression of the growth hormone–releasing
hormone receptor, in contrast to its expression
of growth hormone–releasing hormone, which
was low or absent (Fig. S15 in the Supplemen-
tary Appendix).
In conclusion, our results suggest that Xq26.3
microduplication is associated with a clinical
syndrome of early-onset gigantism, which we
have termed X-LAG. An increased dose of GPR101
on chromosome Xq26.3 probably causes the dis-
ease, and its activation by mutation occurs in
patients with sporadic acromegaly. Xq26.3 micro-
duplications may explain other historical cases
of gigantism with features that closely resemble
those of X-LAG.
27,28
Our results offer an oppor-
tunity to study a new pathway involved in the
central regulation of human growth.
Supported by a grant from the Intramural Research Program
of the Eunice Kennedy Shriver National Institute of Child Health
and Human Development (NICHD) (Z01-HD008920, to Dr. Strata-
kis), by a grant from the National Institute of Neurological Dis-
orders and Stroke (NINDS) (RO1 NS058529), by a grant from the
National Human Genome Research Institute (U54HG006542, to
Dr. Lupski), by a grant from Fonds d’Investissement de Recherche
Scientifique of Centre Hospitalier Universitaire (CHU) de Liège
(to Dr. Beckers), by an educational grant from Pfizer Belgium (to
Dr. Beckers), and by the Jabbs Foundation (to Dr. Beckers). Com-
puting resources used for the molecular-modeling component
of this work were provided by the American University High
Performance Computing System, which is funded in part by a
grant from the National Science Foundation (BCS-1039497).
Disclosure forms provided by the authors are available with
the full text of this article at NEJM.org.
We thank the patients and their families for their participa-
tion in t his study; the nursing and other support sta ff at the NIH
Clinical Research Center (CRC), in particular Dr. Edward Old-
field (now at the University of Virginia, Charlottesville) and Dr.
Russell Lonser (now at Ohio State University, Columbus) who
operated on most of the patients from the NIH who are de-
scribed in this report; Dr. Phillip Gorden of the National Insti-
tute of Diabetes and Digestive and Kidney Diseases for provid-
ing a list of patients with acromegaly who have been seen at the
NIH CRC during the past 30 years; Dr. Timothy Jones (Depart-
ment of Pediatric Endocrinology, Princess Margaret Hospital for
Children and School of Pediatrics and Child Health, University
of Western Australia); Vincent Schram at the Microscopy and
Imaging Core of the NICHD; Dr. Charalampos Lyssikatos and
Dr. Monalisa Azevedo (Section on Endocrinology and Genetics,
NICHD); Ms. Isabelle Besson and Dr. Michèle Bernier (Pathol-
ogy and Cytology Department, Hôpital Foch, Suresnes, France);
Drs. Antonella Forlino, Annalisa Vetro, and Orsetta Zuffardi
(Department of Molecular Medicine, University of Pavia, Pavia,
Italy); Dr. Anna Spada and Dr. Paolo Beck-Peccoz (Endocrinolo-
gy and Diabetology Unit, Fondazione Istituto de Ricovero e Cura
a Carattere Scientif ico Ca’ Granda Ospedale Maggiore Policlinico,
Department of Clinical Sciences, University of Milan); Ms. Carine
Mottard, Ms. Carine Deusings, Mr. Valery Leduc, and Ms. Nathalie
Sacre (Department of Clinical Genetics, CHU de Liege, Liege,
Belgium); Ms. Latifa Karim (GIGA-Genomics, Liège, Belgium);
Dr. Silvia Paoletta (Laboratory of Bioorganic Chemistry,
National
Institute of Diabetes and Digestive and Kidney Diseases
); Ms.
Silke Williams (Laboratory of Pathology, National Cancer Insti-
tute); Dr. Jack A. Yanovski (Section on Growth and Obesity,
NICHD); Dr. S.J. Levine (Cardiovascular and Pulmonary Branch,
National Heart, Lung, and Blood Institute); Dr. Say Viengchareun
(INSERM Unité 693 Le Kremlin-Bicêtre, France); Dr. Paul Hofman
(Liggins Institute, University of Auckland, Auckland, New Zea-
land); Dr. Stephen Butler (Taranaki District Health Board, New
Plymouth, New Zealand); Dr. Yvonne C. Anderson (Liggins In-
stitute, University of Auckland, Auckland, New Zealand, and
Taranaki Base Hospital, New Ply mouth, New Zealand); Dr. Ian
Holdaway (Auckland City Hospital and Greenlane Clinical
Centre, Auckland, New Zealand); and Dr. Karen Carpenter
(Department of Diagnostic Genomics, PathWest Laboratory
Medicine Western Australia, Perth, Australia).
Appendix
The authors are as follows: Giampaolo Trivellin, Ph.D., Adrian F. Daly, M.B., B.Ch., Ph.D., Fabio R. Faucz, Ph.D., Bo Yuan, B.S., Liliya
Rostomyan, M.D., Darwin O. Larco, Ph.D., Marie Helene Schernthaner-Reiter, M.D., Ph.D., Eva Szarek, Ph.D., Letícia F. Leal, Ph.D.,
Jean-Hubert Caberg, Ph.D., Emilie Castermans, Ph.D., Chiara Villa, M.D., Ph.D., Aggeliki Dimopoulos, M.D., Prashant Chittiboina,
M.D., Paraskevi Xekouki, M.D., D.Sc., Nalini Shah, M.D., D.M., Daniel Metzger, M.D., Philippe A. Lysy, M.D., Ph.D., Emanuele Fer-
rante, M.D., Ph.D., Natalia Strebkova, M.D., Ph.D., Nadia Mazerkina, M.D., Ph.D., Maria Chiara Zatelli, M.D., Ph.D., Maya Lodish,
M.D., Anelia Horvath, Ph.D., Rodrigo Bertollo de Alexandre, Ph.D., Allison D. Manning, M.Sc., Isaac Levy, M.D., Margaret F. Keil,
Ph.D., P.N.P., Maria de la Luz Sierra, M.S., Leonor Palmeira, Ph.D., Wouter Coppieters, Ph.D., Michel Georges, M.D., Ph.D., Luciana
A. Naves, M.D., Ph.D., Mauricette Jamar, M.D., Vincent Bours, M.D., Ph.D., T. John Wu, Ph.D., Catherine S. Choong, M.D., M.B., B.S.,
Jerome Bertherat, M.D., Ph.D., Philippe Chanson, M.D., Ph.D., Peter Kamenický, M.D., Ph.D., William E. Farrell, Ph.D., Anne Barlier,
M.D., Ph.D., Martha Quezado, M.D., Ivana Bjelobaba, Ph.D., Stanko S. Stojilkovic, Ph.D., Jurgen Wess, Ph.D., Stefano Costanzi, Ph.D.,
Pengfei Liu, Ph.D., James R. Lupski, M.D., Ph.D., D.Sc., Albert Beckers, M.D., Ph.D., and Constantine A. Stratakis, M.D., D.Sc.
The author’s affiliations are as follows: the Section on Endocrinology and Genetics, Program on Developmental Endocrinology and
Genetics and Pediatric Endocrinology Interinstitute Training Program (G.T., F.R.F., M.H.S.-R., E.S., L.F.L., A.D., P.X., M.L., A.H.,
R.B.A., A.D.M., I.L., M.F.K., M.L.S., C.A.S.), and the Section on Cellular Signaling, Program in Developmental Neuroscience (I.B.,
S.S.S.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, the Surgical Neurology Branch, National
Institute of Neurological Disorders and Stroke (P. Chittiboina), Laboratory of Pathology, National Cancer Institute (M.Q.), and the
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org
12
Gigantism and Acromegaly Due to X
q26 abnormalities
Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases
(J.W.) — all at the National Institutes of Health, and the Department of Obstetrics and Gynecology, Uniformed Services University of the
Health Sciences (D.O.L., T.J.W.) — both in Bethesda, MD; the Departments of Endocrinology (A.F.D., L.R., C.V., A. Beckers) and
Clinical Genetics (J.-H.C., E.C., M.J., V.B.), Centre Hospitalier Universitaire de Liège, University of Liège, Domaine Universitaire du
Sart-Tilman, and Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-Genomics, Domaine Universitaire Sart-Tilman
(L.P., W.C., M.G.), Liège, and the Pediatric Endocrinology Unit, Department of Pediatrics, Clinique Universitaire Saint-Luc, Université
Catholique de Louvain, Louvain (P.A.L.) — all in Belgium; School of Health and Biosciences, Pontifícia Universidade Católica do Paraná,
Curitiba (F.R.F., R.B.A.), and Department of Endocrinology, Faculty of Medicine, University of Brasilia, Brasilia (L.A.N.) — both in
Brazil; Departments of Molecular and Human Genetics (B.Y., P.L., J.R.L.) and Pediatrics (J.R.L.), Texas Children’s Hospital (J.R.L.),
Baylor College of Medicine, Houston; Department of Anatomical and Cytological Pathology, Hôpital Foch, Suresnes (C.V.), INSERM
Unité 1016, Institut Cochin, Université Paris Descartes, Hôpital Cochin, Service d’Endocrinologie, Paris (C.V., J.B.), Assistance Pu-
blique–Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Service d’Endo crinologie et des Maladies de la Reproduction et Centre
de Référence des Maladies Endocriniennes Rares de la Croissance, and Université Paris Sud 11, Faculté de Médecine, UMR-S693, Le
Kremlin-Bicêtre (P. Chanson, P.K.), and Aix-Marseille Université, Centre National de la Recherche Scientifique, UMR-7286, Centre de
Recherche en Neurobiologie et Neurophysiologie de Marseille, Marseille (A. Barlier) — all in France; Department of Endocrinology,
King Edward Memorial Hospital, Mumbai, India (N. Shah); Endocrinology and Diabetes Unit, BC Children’s Hospital, Vancouver, Brit-
ish Columbia (D.M.), and the Division of Endocrinology, Hospital for Sick Children, Toronto (I.L.) — both in Canada; Endocrinology
and Diabetology Unit, Fondazione Istituto de Ricovero e Cura a Carattere Scientifico Ca’ Granda Ospedale Maggiore Policlinico, Milan
(E.F.), and Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara (M.C.Z.) — both in Italy; Endocrino-
logical Research Center, Institute of Pediatric Endocrinology (N. Strebkova), and Burdenko Neurosurgery Institute (N.M.) — both in
Moscow; Department of Pharmacology and Physiology, George Washington University (A.H.), and Department of Chemistry and Center
for Behavioral Neuroscience, American University (S.C.) — both in Washington, DC; Department of Paediatric Endocrinology, Princess
Margaret Hospital for Children and School of Pediatrics and Child Health, University of Western Australia, Perth, Australia (C.S.C.); and
Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Stoke-on-Trent, United Kingdom (W.E.F.).
References
1. Veldhuis JD, Iranmanesh A, Erickson
E, Roelfsema F, Bowers CY. Lifetime reg-
ulation of growth hormone (GH) secre-
tion. In: Fink G, Pfaff DW, Levine JW, eds.
Handbook of neuroendocrinology. New
York: Academic Press, 2012:237-55.
2. Beckers A, Aaltonen LA, Daly AF, Ka rhu
A. Familial isolated pituitary adenomas
(FIPA) and the pituitary adenoma predis-
position due to mutations in the aryl hy-
drocarbon receptor interacting protein
(AIP) gene. Endocr Rev 2013;34:239-77.
3. Strat akis CA, Tichom irowa MA, Boiko s
S, et al. The role of germline AIP, MEN1,
PRKAR1A, CDKN1B and CDKN2C muta-
tions in causing pituitary adenomas in a
large cohort of children, adolescents, and
patients with genetic syndromes. Clin
Genet 2010;78:457-63.
4. Daly AF, Jaffrain-Rea ML, Ciccarelli A,
et al. Clinical characterization of familial
isolated pituitar y adenomas. J Clin Endo-
crinol Metab 2006;91:3316-23.
5. Xekouki P, Azevedo M, Stratakis CA.
Anterior pituitary adenomas: inherited
syndromes, novel genes and molecular
pathways. Expert Rev Endocrinol Metab
2010;5:697-709.
6. Tatton-Brow n K, Rahman N. Sot os syn-
drome. Eur J Hum Genet 2007;15:264-71.
7. Gurrieri F, Pomponi MG, Pietrobono
R, et al. The Simpson-Golabi-Behmel syn-
drome: a clinical case and a detective
story. Am J Med Genet A 2011;155A:145-8.
8. Gläsker S, Vortmeyer AO, Laffert y AR,
et al. Hereditary pituitary hyperplasia
with infantile gigantism. J Clin Endocri-
nol Metab 2011;96(12):E2078-E2087.
9. Espiner EA, Carter TA, Abbott GD,
Wrightson P. Pituitary gigantism in a 31
month old girl: endocrine studies and
successful response to hypophysectomy.
J Endocrinol Invest 1981;4:445-50.
10. Bergamaschi S, Ronchi CL, Giavoli C,
et al. Eight-year follow-up of a child with
a GH/prolactin-secreting adenoma: effi-
cacy of pegvisomant therapy. Horm Res
Paediatr 2010;73:74-9.
11. Leontiou CA, Gueorguiev M, van der
Spuy J, et al. The role of the ar yl hydrocar-
bon receptor-interacting protein gene in
familial and sporadic pituitary adenomas.
J Clin Endocrinol Metab 2008;93:2390-401.
12. Sali A, Overington JP. Derivation of
rules for comparative protein modeling
from a database of protein structure
alignments. Protein Sci 1994;3:1582-96.
13. Bates B, Zhang L, Nawoschik S, et al.
Characterization of Gpr101 expression
and G-protein coupling selectivity. Brain
Re s 20 06;1087:1-14.
14. Lee DK, Nguyen T, Lynch KR, et al.
Discovery and mapping of ten novel
G protein-coupled receptor genes. Gene
2001;275:83-91.
15. Lowell BB, Dhillon H. Patents: novel
gpr101 transgenic mice and methods of
use thereof. 2011 (http://www.google.com/
patents/US20110173706).
16. Madrigal I, Fernández-Burriel M, Ro-
driguez-Revenga L, et al. Xq26.2-q26.3
microduplication in two brothers with
intellectual disabilities: clinical and mo-
lecular characterization. J Hum Genet
2010;55:822-6.
17. Møller RS, Jensen LR, Maas SM, et al.
X-linked congenital ptosis and associated
intellectu al disability, short st ature, micro-
cephaly, cleft palate, digital and genital
abnormalities define novel Xq25q26 dupli-
cation syndrome. Hum Genet 2014;133:
625-38.
18. Stankiewicz P, Thiele H, Schlicker M,
et al. Duplication of Xq26.2-q27.1, includ-
ing SOX3, in a mother and daughter with
short stature and dyslalia. Am J Med Genet
A 2005;138:11-7.
19. Zhang F, Khajavi M, Connolly AM,
Towne CF, Batish SD, Lupski JR. The DNA
replication FoSTeS/MMBIR mechanism
can generate genomic, genic and exonic
complex rearrangements in humans. Nat
Genet 2009;41:849-53.
20. Regard JB, Sato IT, Coughlin SR. Ana-
tomical profiling of G protein-coupled
receptor expression. Cell 2008;135:561-71.
21. Cho-Clark M, Larco DO, Semsa rzadeh
NN, Vasta F, Mani SK, Wu TJ. GnRH-(1-5)
transactivates EGFR in Ishikawa human
endometrial cells via an orphan G protein-
coupled receptor. Mol Endocrinol 2014;28:
80-98.
22. Nilaweera KN, Ozanne D, Wilson D,
Mercer JG, Morga n PJ, Barrett P. G protein-
coupled receptor 101 mRNA expression in
the mouse brain: a ltered expression in the
posterior hypothalamus and amygdala by
energetic challenges. J Neuroendocrinol
20 07;19:34- 45.
23. Vissers LE, Pawel S. Microdeletion and
microdupl ication syn dromes. In: Feuk L, e d.
Genomic structural variants: methods and
protocols. Hatfield, Hertfordshire, United
Kingdom: Humana Press, 2012:29-76.
24. Kurth I, Klopocki E, Stricker S, et al.
Duplications of noncoding elements 5′ of
SOX9 are associated with brachydactyly-
anonychia. Nat Genet 2009;41:862-3.
25. Nord KH, Lilljebjörn H, Vezzi F, et al.
GRM1 is upregulated through gene fu-
sion and promoter swapping in chondro-
myxoid f ibroma. Nat Genet 2014;46:474-7.
26. Peverelli E, Mantovani G, Lania AG,
Spada A. cAMP in the pituitary: an old
messenger for multiple signals. J Mol En-
docrinol 2014;52:R67-R77.
27. de Herder WW. Acromegaly and g igan-
tism in the medical literature: case de-
scriptions in the era before and the early
years after the initial publication of Pierre
Marie (1886). Pituitary 2009;12:236-44.
28. Behrens LH, Barr DP. Hyperpituita-
rism beginning in infancy: the Alton giant.
Endocrinology 1932;16:120-8.
Copyright © 2014 Massachusetts Medical Society.
The New England Journal of Medicine
Downloaded from nejm.org on December 7, 2014. For personal use only. No other uses without permission.
Copyright © 2014 Massachusetts Medical Society. All rights reserved.

Supplementary resource (1)

... The next most common genetic cause of pituitary gigantism is X-linked acrogigantism (X-LAG) (2). This very rare disease has a distinctive presentation beginning in the first 3 years of life (5). Most cases present with mixed GH and prolactin-positive macroadenomas, but individuals with hyperplasia alone have been described (5)(6)(7). ...
... This very rare disease has a distinctive presentation beginning in the first 3 years of life (5). Most cases present with mixed GH and prolactin-positive macroadenomas, but individuals with hyperplasia alone have been described (5)(6)(7). In X-LAG, the young patients can have markedly increased height and weight at diagnosis compared with their age-peers (8). ...
... In X-LAG, the young patients can have markedly increased height and weight at diagnosis compared with their age-peers (8). X-LAG is caused by duplications on chromosome Xq26.3 that include the gene GPR101, which encodes an orphan G protein-coupled receptor (GPCR) (5). In patients with X-LAG, there is a very marked over-expression of GPR101 in pituitary adenomatous/hyperplastic tissue (9). ...
Article
Full-text available
X-linked acrogigantism (X-LAG) is a rare form of pituitary gigantism that is associated with growth hormone (GH) and prolactin-secreting pituitary adenomas/pituitary neuroendocrine tumors (PitNETs) that develop in infancy. It is caused by a duplication on chromosome Xq26.3 that leads to the misexpression of the gene GPR101, a constitutively active stimulator of pituitary GH and prolactin secretion. GPR101 normally exists within its own topologically associating domain (TAD) and is insulated from surrounding regulatory elements. X-LAG is a TADopathy in which the duplication disrupts a conserved TAD border, leading to a neo-TAD in which ectopic enhancers drive GPR101 over-expression, thus causing gigantism. Here we trace the full diagnostic and therapeutic pathway of a female patient with X-LAG from 4C-seq studies demonstrating the neo-TAD through medical and surgical interventions and detailed tumor histopathology. The complex nature of treating young children with X-LAG is illustrated, including the achievement of hormonal control using a combination of neurosurgery and adult doses of first-generation somatostatin analogs.
... heterogeneous when PA is of different secretory subtypes [5,7]. Overall, about 15% to 25% of nonsyndromic FIPA occur in patients with germline mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene [6,8,9] or in patients with X-linked acrogigantism (XLAG) due to microduplication of GPR101 [10]. About 50% of homogenous somatotropinoma FIPA kindreds have AIP mutations or XLAG duplications [8,11]. ...
Article
Full-text available
Context Data on germline genetics of pituitary adenomas (PA) using whole exome sequencing (WES) are limited. This study investigated the germline genetic variants in patients with PA using WES. Patients and Methods We studied 134 consecutive functioning (80.6%) and non-functioning (19.4%) PA in 61 females (45.5%) and 73 males (54.5%). Their median age was 34 years (range 11-85) and 31 had microadenomas (23.0%) and 103 macroadenomas (77%). None of these patients had family history of PA or a known PA-associated syndrome. Peripheral blood DNA was isolated and whole exome sequenced. We used ACMG criteria and a number of in silico analysis tools to characterize genetic variant pathogenicity levels and focused on previously reported PA-associated genes. Results We identified 35 variants of unknown significance (VUS) in 17 PA-associated genes occurring in 40 patients (29.8%). Although designated VUS by the strict ACGM criteria, they are predicted to be pathogenic by in silico analyses and their extremely low frequencies in 1000 genome, gnomAD, and the Saudi Genome Project databases. Further analysis of these variants by the Alpha Missense analysis tool yielded 8 likely pathogenic variants in 9 patients in the following genes: AIP:c.767C > T (p.S256F), CDH23:c.906G > C (p.E302D), CDH23:c.1096G > A (p.A366T), DICER1: c.620C > T (p.A207 V), MLH1:c.955G > A (p.E319 K), MSH2:c.148G > A (p.A50T), and SDHA: c.869T > C (p.L290P) and USP48 (2 Patients): c.2233G > A (p.V745 M). Conclusions This study suggests that about 6.7% of patients with apparently sporadic PA carry likely pathogenic variants in PA-associated genes. These findings need further studies to confirm them.
... Several genes have been identified in association with pituitary adenomas in CYP occurring as isolated pituitary adenomas (familial isolated pituitary adenoma) or as part of a syndromic disease [67][68][69][70][71] . Although these conditions are rare, the number of identified germline (and somatic) genetic alterations is expected to increase in the future with the implementation of wider genetic testing. ...
Article
Full-text available
Tumours of the anterior part of the pituitary gland represent just 1% of all childhood (aged <15 years) intracranial neoplasms, yet they can confer high morbidity and little evidence and guidance is in place for their management. Between 2014 and 2022, a multidisciplinary expert group systematically developed the first comprehensive clinical practice consensus guideline for children and young people under the age 19 years (hereafter referred to as CYP) presenting with a suspected pituitary adenoma to inform specialist care and improve health outcomes. Through robust literature searches and a Delphi consensus exercise with an international Delphi consensus panel of experts, the available scientific evidence and expert opinions were consolidated into 74 recommendations. Part 1 of this consensus guideline includes 17 pragmatic management recommendations related to clinical care, neuroimaging, visual assessment, histopathology, genetics, pituitary surgery and radiotherapy. While in many aspects the care for CYP is similar to that of adults, key differences exist, particularly in aetiology and presentation. CYP with suspected pituitary adenomas require careful clinical examination, appropriate hormonal work-up, dedicated pituitary imaging and visual assessment. Consideration should be given to the potential for syndromic disease and genetic assessment. Multidisciplinary discussion at both the local and national levels can be key for management. Surgery should be performed in specialist centres. The collection of outcome data on novel modalities of medical treatment, surgical intervention and radiotherapy is essential for optimal future treatment.
Article
Pituitary gigantism is a rare manifestation of chronic growth hormone (GH) excess that begins before closure of the growth plates. Nearly half of patients with pituitary gigantism have an identifiable genetic cause. X-linked acrogigantism (X-LAG; 10% of pituitary gigantism) typically begins during infancy and can lead to the tallest individuals described. In the 10 years since its discovery, about 40 patients have been identified. Patients with X-LAG usually develop mixed GH and prolactin macroadenomas with occasional hyperplasia that secrete copious amounts of GH, and frequently prolactin. Circulating GH-releasing hormone is also elevated in a proportion of patients. X-LAG is caused by constitutive or sporadic mosaic duplications at chromosome Xq26.3 that disrupt the normal chromatin architecture of a topologically associating domain (TAD) around the orphan G-protein–coupled receptor, GPR101. This leads to the formation of a neo-TAD in which GPR101 overexpression is driven by ectopic enhancers (“TADopathy”). X-LAG has been seen in 3 families due to transmission of the duplication from affected mothers to sons. GPR101 is a constitutively active receptor with an unknown natural ligand that signals via multiple G proteins and protein kinases A and C to promote GH/prolactin hypersecretion. Treatment of X-LAG is challenging due to the young patient population and resistance to somatostatin analogs; the GH receptor antagonist pegvisomant is often an effective option. GH, insulin-like growth factor 1, and prolactin hypersecretion and physical overgrowth can be controlled before definitive adult gigantism occurs, often at the cost of permanent hypopituitarism.
Article
Full-text available
G protein-coupled receptors (GPCRs) make up the largest receptor superfamily, accounting for 4% of protein-coding genes. Despite the prevalence of such transmembrane receptors, a significant number remain orphans, lacking identified endogenous ligands. Since their conception, the reverse pharmacology approach has been used to characterize such receptors. However, the multifaceted and nuanced nature of GPCR signaling poses a great challenge to their pharmacological elucidation. Considering their therapeutic relevance, the search for native orphan GPCR ligands continues. Despite limited structural input in terms of 3D crystallized structures, with advances in machine-learning approaches, there has been great progress with respect to accurate ligand prediction. Though such an approach proves valuable given that ligand scarcity is the greatest hurdle to orphan GPCR deorphanization, the future pairings of the remaining orphan GPCRs may not necessarily take a one-size-fits-all approach but should be more comprehensive in accounting for numerous nuanced possibilities to cover the full spectrum of GPCR signaling.
Article
Full-text available
Pituitary adenomas are rare in children and young people under the age of 19 (hereafter referred to as CYP) but they pose some different diagnostic and management challenges in this age group than in adults. These rare neoplasms can disrupt maturational, visual, intellectual and developmental processes and, in CYP, they tend to have more occult presentation, aggressive behaviour and are more likely to have a genetic basis than in adults. Through standardized AGREE II methodology, literature review and Delphi consensus, a multidisciplinary expert group developed 74 pragmatic management recommendations aimed at optimizing care for CYP in the first-ever comprehensive consensus guideline to cover the care of CYP with pituitary adenoma. Part 2 of this consensus guideline details 57 recommendations for paediatric patients with prolactinomas, Cushing disease, growth hormone excess causing gigantism and acromegaly, clinically non-functioning adenomas, and the rare TSHomas. Compared with adult patients with pituitary adenomas, we highlight that, in the CYP group, there is a greater proportion of functioning tumours, including macroprolactinomas, greater likelihood of underlying genetic disease, more corticotrophinomas in boys aged under 10 years than in girls and difficulty of peri-pubertal diagnosis of growth hormone excess. Collaboration with pituitary specialists caring for adult patients, as part of commissioned and centralized multidisciplinary teams, is key for optimizing management, transition and lifelong care and facilitates the collection of health-related quality of survival outcomes of novel medical, surgical and radiotherapeutic treatments, which are currently largely missing.
Article
Full-text available
Somatostatin (SST), a growth hormone inhibitory peptide, is expressed in endocrine and non-endocrine tissues, immune cells and the central nervous system (CNS). Post-release from secretory or immune cells, the first most appreciated role that SST exhibits is the antiproliferative effect in target tissue that served as a potential therapeutic intervention in various tumours of different origins. The SST-mediated in vivo and/or in vitro antiproliferative effect in the tumour is considered direct via activation of five different somatostatin receptor subtypes (SSTR1-5), which are well expressed in most tumours and often more than one receptor in a single cell. Second, the indirect effect is associated with the regulation of growth factors. SSTR subtypes are crucial in tumour diagnosis and prognosis. In this review, with the recent development of new SST analogues and receptor-specific agonists with emerging functional consequences of signaling pathways are promising therapeutic avenues in tumours of different origins that are discussed.
Article
Full-text available
The decapeptide GnRH is known for its central role in the regulation of the hypothalamo-pituitary-gonadal (HPG) axis. In addition, it is also known to have local effects within peripheral tissues. The zinc metalloendopeptidase, EC 3.4.24.15 (EP24.15), can cleave GnRH at the Tyr(5)-Gly(6) bond to form the pentapeptide, GnRH-(1-5). The central and peripheral effect of GnRH-(1-5) is different from its parent peptide, GnRH. In the current study, we examined the effect of GnRH-(1-5) on EGFR phosphorylation and cellular migration. Using the Ishikawa cell line as a model of endometrial cancer, we demonstrate that GnRH-(1-5) stimulates EGF release, increases the phosphorylation of EGFR (p<0.05) at three tyrosine sites (992, 1045, 1068), and promotes cellular migration. In addition, we also demonstrate that these actions of GnRH-(1-5) are mediated by the orphan G-protein coupled receptor 101 (GPR101). Down-regulation of GPR101 expression blocked the GnRH-(1-5)-mediated release of EGF and the subsequent phosphorylation of EGFR and cellular migration. These results suggest that GPR101 is a critical requirement for GnRH-(1-5) transactivation of EGFR in Ishikawa cells.
Article
Full-text available
Glutamate receptors are well-known actors in the central and peripheral nervous systems, and altered glutamate signaling is implicated in several neurological and psychiatric disorders. It is increasingly recognized that such receptors may also have a role in tumor growth. Here we provide direct evidence of aberrant glutamate signaling in the development of a locally aggressive bone tumor, chondromyxoid fibroma (CMF). We subjected a series of CMFs to whole-genome mate-pair sequencing and RNA sequencing and found that the glutamate receptor gene GRM1 recombines with several partner genes through promoter swapping and gene fusion events. The GRM1 coding region remains intact, and 18 of 20 CMFs (90%) showed a more than 100-fold and up to 1,400-fold increase in GRM1 expression levels compared to control tissues. Our findings unequivocally demonstrate that direct targeting of GRM1 is a necessary and highly specific driver event for CMF development.
Article
Full-text available
The cyclic nucleotide cyclic adenosine 3',5'-monophosphate (cAMP) is a universal regulator of a variety of cell functions in response to activated G protein-coupled receptors (GPCRs). In particular, cAMP exerts positive or negative effects on cell proliferation in different cell types. As demonstrated by several in vitro studies, in somatotrophs and in other endocrine cells cAMP is a mitogenic factor. In agreement with this notion, it has been found that mutations of genes coding for proteins that contribute to increase cAMP signalling cascade may cause endocrine tumor development. This review discusses the central role of cAMP signalling in the pituitary, focusing on the cAMP pathway alterations involved in pituitary tumorigenesis, as well as the poorly investigated aspects of the cAMP cascade, such as crosstalk with the ERK signalling pathway and new cAMP effectors.
Article
Full-text available
The most frequent conditions that are associated with inherited/familial pituitary adenomas are familial isolated pituitary adenoma (FIPA) and multiple endocrine neoplasia type 1 (MEN1), which together account for up to 5% of pituitary adenomas. One important genetic cause of FIPA are inactivating mutations or deletions in the aryl hydrocarbon receptor interacting protein (AIP) gene. FIPA is the most frequent clinical presentation of AIP mutations. This article traces the current state of knowledge regarding the clinical features of FIPA and the particular genetic, pathologic, and clinical characteristics of pituitary adenomas due to AIP mutations. Copyright © 2015 Elsevier Inc. All rights reserved.
Chapter
Dynamic characteristics of the growth hormone (GH) axis are determined in part by developmental age, including fetal, neonatal, childhood, pubertal, young-adult and older-adult stages of life. The growth-promoting pituitary-dependent GH axis mediates soft-tissue and skeletal growth in puberty, and maintains body composition within relatively narrow bounds in puberty and adulthood. The hypothalamo-pituitary unit comprises a unique neuroendocrine interface, in which intermittent brain signals are delivered via a finite portal microvasculature to the multicellular anterior pituitary gland. The GHRH receptor is a GTP-dependent adenylyl cyclase-activating protein, with sequence homology to secretin and vasoactive intestinal polypeptide (VIP) receptors. The prototypical endogenous GHS is ghrelin, a 28-amino acid acylated peptide, which requires an octanoyl group esterified to serine in N-terminal position 3 for maximal receptor activation. Other fatty acyl groups can also confer bioactivity. Low pulsatile GH secretion in aged individuals may be reversed acutely by simultaneous administration of GHRH, GHS and L-arginine.
Article
Submicroscopic duplications along the long arm of the X-chromosome with known phenotypic consequences are relatively rare events. The clinical features resulting from such duplications are various, though they often include intellectual disability, microcephaly, short stature, hypotonia, hypogonadism and feeding difficulties. Female carriers are often phenotypically normal or show a similar but milder phenotype, as in most cases the X-chromosome harbouring the duplication is subject to inactivation. Xq28, which includes MECP2 is the major locus for submicroscopic X-chromosome duplications, whereas duplications in Xq25 and Xq26 have been reported in only a few cases. Using genome-wide array platforms we identified overlapping interstitial Xq25q26 duplications ranging from 0.2 to 4.76 Mb in eight unrelated families with in total five affected males and seven affected females. All affected males shared a common phenotype with intrauterine- and postnatal growth retardation and feeding difficulties in childhood. Three had microcephaly and two out of five suffered from epilepsy. In addition, three males had a distinct facial appearance with congenital bilateral ptosis and large protruding ears and two of them showed a cleft palate. The affected females had various clinical symptoms similar to that of the males with congenital bilateral ptosis in three families as most remarkable feature. Comparison of the gene content of the individual duplications with the respective phenotypes suggested three critical regions with candidate genes (AIFM1, RAB33A, GPC3 and IGSF1) for the common phenotypes, including candidate loci for congenital bilateral ptosis, small head circumference, short stature, genital and digital defects.
Article
Human adiposity has long been associated with insulin resistance and increased cardiovascular risk, and abdominal adiposity is considered particularly adverse. Intra-abdominal fat is associated with insulin resistance, possibly mediated by greater lipolytic activity, lower adiponectin levels, resistance to leptin, and increased inflammatory cytokines, although the latter contribution is less clear. Liver lipid is also closely associated with, and likely to be an important contributor to, insulin resistance, but it may also be in part the consequence of the lipogenic pathway of insulin action being up-regulated by hyperinsulinemia and unimpaired signaling. Again, intramyocellular triglyceride is associated with muscle insulin resistance, but anomalies include higher intramyocellular triglyceride in insulin-sensitive athletes and women (vs men). Such issues could be explained if the “culprits” were active lipid moieties such as diacylglycerol and ceramide species, dependent more on lipid metabolism and partitioning than triglyceride amount. Subcutaneous fat, especially gluteofemoral, appears metabolically protective, illustrated by insulin resistance and dyslipidemia in patients with lipodystrophy. However, some studies suggest that deep sc abdominal fat may have adverse properties. Pericardial and perivascular fat relate to atheromatous disease, but not clearly to insulin resistance. There has been recent interest in recognizable brown adipose tissue in adult humans and its possible augmentation by a hormone, irisin, from exercising muscle. Brown adipose tissue is metabolically active, oxidizes fatty acids, and generates heat but, because of its small and variable quantities, its metabolic importance in humans under usual living conditions is still unclear. Further understanding of specific roles of different lipid depots may help new approaches to control obesity and its metabolic sequelae.
Article
During the past decade, widespread use of microarray-based technologies, including oligonucleotide array comparative genomic hybridization (aCGH) and single nucleotide polymorphism (SNP) genotyping arrays have dramatically changed our perspective on genome-wide structural variation. Submicroscopic genomic rearrangements or copy-number variation (CNV) have proven to be an important factor responsible for primate evolution, phenotypic differences between individuals and populations, and susceptibility to many diseases. The number of diseases caused by chromosomal microdeletions and microduplications, also referred to as genomic disorders, has been increasing at a rapid pace. Microdeletions and microduplications are found in patients with a wide variety of phenotypes, including Mendelian diseases as well as common complex traits, such as developmental delay/intellectual disability, autism, schizophrenia, obesity, and epilepsy. This chapter provides an overview of common microdeletion and microduplication syndromes and their clinical phenotypes, and discusses the genomic structures and molecular mechanisms of formation. In addition, an explanation for how these genomic rearrangements convey abnormal phenotypes is provided.