ArticlePDF Available

Somatic activating mutations in MAP2K1 cause melorheostosis

Authors:

Abstract and Figures

Melorheostosis is a sporadic disease of uncertain etiology characterized by asymmetric bone overgrowth and functional impairment. Using whole exome sequencing, we identify somatic mosaic MAP2K1 mutations in affected, but not unaffected, bone of eight unrelated patients with melorheostosis. The activating mutations (Q56P, K57E and K57N) cluster tightly in the MEK1 negative regulatory domain. Affected bone displays a mosaic pattern of increased p-ERK1/2 in osteoblast immunohistochemistry. Osteoblasts cultured from affected bone comprise two populations with distinct p-ERK1/2 levels by flow cytometry, enhanced ERK1/2 activation, and increased cell proliferation. However, these MAP2K1 mutations inhibit BMP2-mediated osteoblast mineralization and differentiation in vitro, underlying the markedly increased osteoid detected in affected bone histology. Mosaicism is also detected in the skin overlying bone lesions in four of five patients tested. Our data show that the MAP2K1 oncogene is important in human bone formation and implicate MEK1 inhibition as a potential treatment avenue for melorheostosis.
No caption available
… 
No caption available
… 
This content is subject to copyright. Terms and conditions apply.
ARTICLE
Somatic activating mutations in MAP2K1 cause
melorheostosis
Heeseog Kang1, Smita Jha2,3, Zuoming Deng4, Nadja Fratzl-Zelman5, Wayne A. Cabral1,10, Aleksandra Ivovic6,
Françoise Meylan 6, Eric P. Hanson7, Eileen Lange8, James Katz8, Paul Roschger5, Klaus Klaushofer5,
Edward W. Cowen9, Richard M. Siegel6, Joan C. Marini1& Timothy Bhattacharyya2
Melorheostosis is a sporadic disease of uncertain etiology characterized by asymmetric bone
overgrowth and functional impairment. Using whole exome sequencing, we identify somatic
mosaic MAP2K1 mutations in affected, but not unaffected, bone of eight unrelated patients
with melorheostosis. The activating mutations (Q56P, K57E and K57N) cluster tightly in the
MEK1 negative regulatory domain. Affected bone displays a mosaic pattern of increased
p-ERK1/2 in osteoblast immunohistochemistry. Osteoblasts cultured from affected bone
comprise two populations with distinct p-ERK1/2 levels by ow cytometry, enhanced ERK1/2
activation, and increased cell proliferation. However, these MAP2K1 mutations inhibit BMP2-
mediated osteoblast mineralization and differentiation in vitro, underlying the markedly
increased osteoid detected in affected bone histology. Mosaicism is also detected in the skin
overlying bone lesions in four of ve patients tested. Our data show that the MAP2K1
oncogene is important in human bone formation and implicate MEK1 inhibition as a potential
treatment avenue for melorheostosis.
DOI: 10.1038/s41467-018-03720-z OPEN
1Section on Heritable Disorders of Bone and Extracellular Matrix, National Institute of Child Health and Human Development, National Institutes of Health,
Bethesda, MD 20892, USA. 2Clinical and Investigative Orthopedics Surgery Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases,
National Institutes of Health, Bethesda, MD 20892, USA. 3Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of
Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA. 4Biodata Mining and Discovery Section, Ofce of Science
and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA. 5Ludwig
Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Center Meidling, 1st Medical Department Hanusch Hospital, UKH
Meidling, Kundratstr. 37, Vienna 1120, Austria. 6Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and
Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA. 7Immunodeciency and Inammation Unit, Autoimmunity Branch, National Institute
of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda 20892, USA. 8Ofce of the Clinical Director, National Institute of
Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA. 9Dermatology Branch, National Institute of
Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
10
Present address: Molecular Genetics Section,
Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
These authors contributed equally: Heeseog Kang, Smita Jha, Zuoming Deng. These authors jointly supervised this work: Richard M. Siegel, Joan C. Marini,
Timothy Bhattacharyya. Correspondence and requests for materials should be addressed to T.B. (email: timothy.bhattacharyya@nih.gov)
NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications 1
1234567890():,;
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Melorheostosis (OMIM%155950) is a rare dysostosis
characterized by excess bone formation in the classic
dripping candle waxpattern on the surface of bone,
revealed radiographically (Fig. 1a, b and Supplementary Fig. 1)1.
The disease occurs in males and females equally, without familial
clustering, and often affects multiple contiguous bones uni-
laterally in an asymmetric distribution2. Symptoms of melor-
heostosis may begin in childhood; the diagnosis is usually
apparent by the age of 20 years3. The bone overgrowth lesions are
associated with pain, functional impairment, joint contracture,
and deformity. There is a wide spectrum of clinical appearance2
with skin lesions overlying the affected bone noted in up to 30%
of patients. Bone lesions do not metastasize, but conversion to
osteosarcoma has rarely been reported4. Increased uptake on
bone scintigraphy suggests that melorheostotic bone is
metabolically active (Fig. 1ce). Patients are diagnosed by a
combination of clinical ndings, radiographs and bone scan.
There is no denitive diagnostic test or specic treatment for
melorheostosis2.
At present, two patterns of occurrence of melorheostosis have
been differentiated by genetics. Patients with the autosomal
dominant spotted bone diseaseosteopoikilosis and the related
condition BuschkeOllendorff syndrome (OMIM #166700) may
display features of melorheostosis, and have been shown to have
germline loss-of-function mutations in LEMD3, encoding the
inner nuclear membrane protein MAN15.LEMD3 mutations
have not been found in patients with the more common sporadic
melorheostosis without osteopoikilosis6; the genetic cause of their
condition is undened7. Recently, a patient with melorheostosis
and familial osteopoikilosis was found to have a germline LEMD3
mutation and a somatic KRAS mutation in an overlying area of
scleroderma-like skin8. Whether the KRAS mutation was causa-
tive for the bone ndings in this one patient remains unde-
termined. The lack of vertical genetic transmission and
asymmetric involvement suggests that sporadic melorheostosis
may be caused by somatic mutations in bone-forming cells9, but
bone tissue has not been previously investigated for somatic
mutations in bone forming cells.
We biopsied affected and unaffected bone of 15 patients with
melorheostosis and compared the tissue whole exome sequences
for each patient. We identied mutations in the MAP2K1 gene in
affected, but not unaffected, bone of eight patients. MAP2K1
encodes the protein MEK1, a kinase whose activity is modulated
by its negative regulatory domain. The MAP2K1 mutations
identied in melorheostosis patients cause substitutions in two
residues of the MEK1 negative regulatory domain. We present
genetic, functional and histological data supporting the
enhancement of MEK1 activity predicted by the location of the
mutations and the causative role of the MAP2K1 mutations in
melorheostosis. This study provides evidence of a bone disease
caused by mutations in MAP2K1.
Results
Identication of mosaicism for MAP2K1. Fifteen patients with
melorheostosis underwent biopsies of affected and unaffected
bone (Supplementary Table 1and 2). A consistent intraoperative
nding was extremely dense, rigid affected bone that often dulled
the osteotomes and drill bits. Genomic DNA was extracted from
both tissues and subjected to high-depth whole exome sequen-
cing. We identied 8365 sequence variants that were present in a
subjects affected bone but not unaffected bone. After restricting
these variants to those that were found in more than 1% of
sequences (but less than germline frequency), rare in the general
population, and predicted to alter protein coding or mRNA
splicing, we were left with 284 variants (Fig. 2a and Table 1). Only
ve genes contained putative somatic mutations in more than one
affected bone sample (MAP2K1,USH2A, CCDC13, SIRT5 and
RAB44). Among these ve genes, it was striking that sequences
from ve affected bone samples contained one of three tightly
clustered missense mutations in the coding region of MAP2K1,
c.167 A > C (p.Q56P), c.169 A > G (p.K57E), and c.171 G > T
(p.K57N), encoding substitutions in two adjacent amino acids of
the protein MEK1 (Fig. 2b and Supplementary Data 1). These
coding variants are not present in the exomes of 123,136 healthy
individuals searchable with the ExAC10. Both substituted residues
are located in a MEK1 α-helix that negatively regulates kinase
function (Fig. 2b and Supplementary Fig. 2)11. All three of these
mutations have been previously shown to lead to gain-of-function
through loss of the inhibitory role of the negative regulatory
domain and have been identied in malignancies, including
lung12, melanoma13 and hairy cell leukemia14 (Supplementary
Table 3). All other 281 variants were unique to the affected bone
of 1 of the 15 subjects (Supplementary Data 1). No germline or
somatic LEMD3,MAP2K2 or MAP2K3 variants were identied in
automated or manual re-examination of exome sequencing data
Fig. 1 Clinical ndings in melorheostosis. aClinical appearance of Melo-10 with melorheostosis of right lower extremity. Note irregular thickening of the
right leg. The affected bone was found to harbor a MAP2K1 mutation. bRadiograph of the right tibia/bula of Melo-10 with classic candle-wax appearance.
cMaximal intensity projection (MIP) 18F-NaF PET image of the lower extremities showing intensely increased 18F-NaF activity in the bones of the right leg,
primarily along the medial and distal femur, and along the tibia extending into the foot. d,eAxial CT and fused 18F-NaF PET/CT images at the level of
mid-calf demonstrating dense cortical bone formation in the anterior and posterior aspect of the right tibia (red arrows in dwith corresponding abnormally
increased 18F-NaF uptake (white arrows, in e)
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z
2NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications
Content courtesy of Springer Nature, terms of use apply. Rights reserved
123
P loop
Negative
regulatory
ERK
binding
4 5 6 7 8 910 11
Catalytic core
ATP binding
Catalytic loop
Mg
activation
segment
K57E
K57N
Q56P
Proline rich Catalytic core
10,000
8000
6000
4000
2000
0
HEX channel (mutant) [RFU]HEX channel (mutant) [RFU]
10,000
8000
6000
4000
2000
FAM channel (wildtype) [RFU] FAM channel (wildtype) [RFU]
39.2%
6.95%0%
0%
Unaffected
Bone biopsies from 15 subjects with
melorheostosis
Affected bone Contralateral
unaffected bone
Exome sequencing >100X depth
BWA genome alignment (hg19)
GATK/Picard:
duplicate removal, base quality score recalibration,
InDel realignment
MuTect and Strelka: somatic
variant calling in affected bone
Annovar: variant annotation
8365 sequence variants
Variant filtering
Called by both MuTect and Strelka (1073)
Population frequency <1% in EXAC and 1KGP
Alters protein coding sequence or splicing
Not in duplicate genomic region
Variants in same gene found in
multiple patients (frequency)
Mutations cluster in same domain
Shared mutations between patients
Mutation confirmed by ddPCR
Q56P (3)
K57E (1)
K57N (4)
MAP2K1 (8)
MAP2K1 (5)
CCDC13 (2)
USH2A (2)
SIRT5 (2)
RAB44 (2)
284 variants
Bone
Cultured
osteoblasts
Affected
1000 60002000 3000 4000 50000 1000 2000 3000 4000 5000 6000
a
c
b
Fig. 2 MAP2K1 somatic mutations in melorheostotic bone. aFlowchart of genetic analysis. bSchematic of the MAP2K1 exon structure encoding MEK1
protein domains. The three mutations from the eight patients are clustered in the negative regulatory domain. cQuantication of mutant allele abundance
by ddPCR in unaffected and affected bone biopsies (top row), as well as in cultured osteoblasts from the respective biopsies (bottom row), from patient
Melo-2 identied by WES to have a p.K57N mutation in affected bone. Each dot represents a droplet, with blue being mutant positive, green being wildtype
positive, and orange being positive for both. Color-matched numbers correspond to count of droplets per quadrant. The boxed number is the fractional
abundance of mutant allele in each sample
NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z ARTICLE
NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications 3
Content courtesy of Springer Nature, terms of use apply. Rights reserved
from any patient. A KRAS p.Q61R mutation was found in both
the affected and unaffected bone of one MAP2K1 mutation-
negative patient with skin lesions consistent with RASopathy,
who likely has melorheostosis as part of a more complex early
embryonic mosaic RASopathy, which will be reported separately.
Thus 5 of 15 patients were identied to have MAP2K1 mutations
by whole exome sequencing.
The allele frequency of these somatic heterozygous
MAP2K1 mutations identied by automated somatic mutation
calling was relatively high (1025%). We performed amplicon-
based targeted sequencing to validate the exome sequencing
ndings and screen the remaining patients for MAP2K1
mutations at higher sensitivity. This approach revealed three
additional patients with MAP2K1 p.K57N mutations at lower
mutant allele frequencies (Table 1). Amplicon-seq polymerase
errors were less than 1% at the base positions in question
(Supplementary Tables 4,5and 6). Their mutations were missed
by WES due to low allele frequencies in patients Melo-16 and
Melo-6 (2.7% and 4.8%, respectively) or low coverage in patient
Melo-2 (58×). All eight MAP2K1 mutations were then conrmed
by droplet digital PCR (ddPCR) of DNA from affected bone
(Fig. 2c). The MAP2K1 mutant allele frequency in affected bone
by ddPCR ranged from 9 to 28% for p.Q56P, 334% for p.K57N,
and 18% for p.K57E and correlates well with amplicon and
WES data.
The ddPCR mutant allele frequency in osteoblasts cultured
from melorheostotic bone of seven subjects, varied from 0.05 to
47% and was similar in different samples from the same cell
culture, but often did not match the frequency found in bone
tissue, consistent with tissue mosaicism. For example, there was
a markedly higher frequency of the p.K57N mutation in
cultured osteoblasts than bone tissue from patient Melo-2
(4047% in cell passages 15 vs. 6.9% in bone) and a lower
frequency of p.Q56P in Melo-4 (0.050.2% in cell passages 15
vs. 10% in bone tissue).
Mosaicism for their respective MAP2K1 mutations
was also detected in skin tissue overlying affected bone
of patients Melo-2, Melo-4, Melo-6 and Melo-18, but not
detected in skin from patient Melo-16, who has a lower
disease burden. To look for germline mutations, we tested
blood samples available from seven patients. MAP2K1 mutations
were not identied in blood samples including those with
mutation-positive skin tissue, further conrming mosaicism
(Table 1).
Melorheostosis histology reveals intense remodeling. To gain
insights into the effects of MAP2K1 mutations on bone structure,
remodeling and mineralization, we compared histomorphometry
of tissue sections from affected and unaffected bone in six
patients. Melorheostotic bone was characterized in the outer
regions by distinctive parallel layers of primary lamellar bone, an
organization that underlies the surgical hardness of the bone
(Fig. 3a). This newly formed compact tissue was intensely
remodeled into a highly porous osteonal-like bone at greater
depth from the surface (Fig. 3b). Compared to unaffected bone,
affected bone showed increased active bone-resorbing osteoclasts,
elevated eroded surfaces and a higher number of bone-forming
osteoblasts (Fig. 3c). There was an approximately six-fold increase
in the thickness of osteoid (unmineralized bone matrix) and a
greater than 50-fold increase in osteoid surface/bone surface in
the affected bone samples compared to their respective unaffected
counterparts (Fig. 3c, d).
Table 1 Table of individual patients showing the mutation location, cDNA and protein consequences, and variant allele
frequencies (VAF)
Subject AA Changes VAF WES
affected
bone
VAF WES
unaffected
bone
VAF amplicon
affected bone
VAF ddPCR
affected bone
VAF ddPCR
unaffected
bone
VAF
ddPCR
skin
VAF ddPCR
unaffected
skin
VAF
ddPCR
blood
melo4 NM_002755:
exon2:c.A167C:
p.Q56P
10.1%a0.00% N.D. 9.4% 0.0% 12.5% 0.0% 0.0%
melo9 NM_002755:
exon2:c.A167C:
p.Q56P
17.19%a0.00% N.D. 20.0% 0.0% N.D. N.D. 0.0%
melo19 NM_002755:
exon2:c.A167C:
p.Q56P
30.86%a0.99% 27.9% 27.8% 0.0% N.D. N.D. N.D.
melo10 NM_002755:
exon2:c.
A169G:
p.K57E
12.07%a0.00% 16.7% 18.3% 0.0% N.D. N.D. 0.0%
melo2 NM_002755:
exon2:c.G171T:
p.K57N
6.09% 0.00% 7.2% 7.0% 0.0% 4.1% 0.0% 0.0%
melo6 NM_002755:
exon2:c.G171T:
p.K57N
3.12% 0.00% 4.8% 4.4% 0.0% 16.2% 0.0% 0.0%
melo16 NM_002755:
exon2:c.G171T:
p.K57N
1.04% 0.00% 2.7% 2.8% 0.0% 0.0% 0.0% 0.0%
melo18 NM_002755:
exon2:c.G171T:
p.K57N
25.21%a0.0% 34.4% 33.6% 0.0% 6.5% N.D. 0.0%
aCases where MAP2K1 mutations were identied through automated ltering of WES for somatic mutations. Others were found by manual inspection. Mutations were not identied in normal control
bone. Melo-19 had one mutant read in unaffected bone that was not seen in ddPCR, designating it as a sequencing error. The VAF results from ddPCR, amplicon and whole exome sequencing are highly
correlated (R=0.96 or greater for all comparisons, p< 0.003). MAP2K1 mutations are present in overlying skin in four of ve patients tested while contralateral skin is negative. N.D. not done
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z
4NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications
Content courtesy of Springer Nature, terms of use apply. Rights reserved
MEK1 signaling increased in melorheostosis. To determine how
the MAP2K1 mutations identied in our cohort affected the
MEK1 target kinases ERK1 and ERK2, we examined MEK1
activity by immunohistochemical analysis of phosphorylated
ERK1/2 (p-ERK1/2) in sections of affected bone tissue from two
patients with MAP2K1 mutations. Osteocytes and osteoblasts in
affected bone displayed stronger p-ERK1/2 signals than control
bone, but no p-ERK1/2 signal was detected in osteoclasts
(Fig. 4a). Consistent with somatic mosaicism, the p-ERK1/2
positive cells occurred in patches in affected bone tissue.
Furthermore, osteoblasts cultured from affected bone and having a
high frequency of mutant MAP2K1 cells (approximately 80%)
exhibited a bimodal ERK1/2 activation after serum-stimulation when
examined by ow cytometry for intracellular p-ERK1/2. Bimodal
ERK1/2 activation was not observed in cells from unaffected bone.
Both peaks of ERK1/2 activation in osteoblasts from affected bone,
Unaffected Affected Affected
MAP2K1 p.K57N (Melo-18)MAP2K1 p.K57N (Melo-2)
MAP2K1 p.Q56P (Melo-9)
Osteoid
thickness
Unaff Aff Unaff
Unaff
Unaffacted Affacted
Unaff
****
Aff
Aff Aff
Unaff Aff
20
15
10
5
0
4
3
2
1
0
10
8
6
4
2
0
80
60
40
20
0
Osteoid
surface
Eroded
surface
Osteoclast
surface
% Bone surface
% Bone surface
% Bone surface
% Bone surface
(μm)
Osteoblast
surface
*********
12
10
8
6
4
2
0
*
*
**
100 μm 100 μm
*
100 μm 100 μm
1000 μm
ab
c
d
Fig. 3 Intense bone remodeling and abnormal mineralization in melorheostosis. aLight microscopy images of cortical bone from patient Melo-2 showing
the orientation of the collagenous matrix. Goldners trichrome staining is viewed under polarized light. Affected bone tissue shows in the periosteal regions
appositional growth through deposition of layers of compact primary lamellar bone oriented parallel to the periosteal surface (open arrows). Subsequently
these regions become highly remodeled. The solid arrows point towards large canals traversing the parallel layers of primary bone. Note the thick osteoid
seams (red, unmineralized matrix) in affected bone tissue, whereas no osteoid is seen in unaffected bone (asterisks). bA backscattered electron image
(BEI) of affected bone tissue from patient Melo-18. Gray represents the mineralized bone tissue, black is non-mineralized tissue. Note the extreme porosity
of the bone tissue as a consequence of an intense bone remodeling activity (solid arrows). The open arrow is the same area labeled by the open arrow in a.
cBone histomorphometry. Comparison of indices of bone formation and bone resorption of affected vs. unaffected bone tissue from six patients with
MAP2K1 mutations. All indices of bone formation and bone resorption are signicantly increased in affected bone tissue compared to unaffected bone of
the same patient. *p< 0.05; **p< 0.01; ***p< 0.001; ****p< 0.0001 vs. values of unaffected bone tissue (paired t-test). Unaff: unaffected bone tissue, Aff:
affected bone tissue. dOverviews of unaffected and affected bone tissue obtained from patient Melo-9. Affected bone can be easily recognized by its
porosity and by the substantial portions of unmineralized matrix (solid arrows), while osteoid formation is not seen around osteonal canals in the
unaffected bone sample (asterisks). Goldners trichrome staining followed by light microscopy in the bright-eld mode. Green indicates mineralized bone
matrix and red indicates unmineralized matrix
NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z ARTICLE
NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications 5
Content courtesy of Springer Nature, terms of use apply. Rights reserved
likely representing wild-type and mutant cells, were reduced by the
MEK-specic inhibitor U0126. Baseline p-ERK1/2 was also higher in
a proportion of affected cells than in unaffected cells (Fig. 4b). These
ndingswouldbeexpectedfromamixedcellpopulationand
conrm MAP2K1 mosaicism at the cellular level.
We also investigated the MEK1-ERK1/2 pathway in cultured
osteoblasts harboring MAP2K1 mutations by western blot analysis
in seven patients (Fig. 4c). Osteoblasts from the same patient and
passage as those analyzed by ow cytometry showed increased p-
ERK1/2 compared to cells from unaffected bone, indicating MEK1
gain-of-function (Supplementary Fig. 4). FBS-induced ERK1/2
activation could more readily be seen in unaffected osteoblasts by
western blotting, likely due to the increased sensitivity in
comparison to ow cytometry. Pre-treatment of cells with
U0126 signicantly diminished serum-stimulated p-ERK1/2, sup-
porting enhanced MEK1 activity as mediating the increased ERK1/
2 activation. The level of ERK1/2 activation by MEK1 generally
correlated with mutant allele frequency. Activation was clearly seen
in Melo-2 and Melo-18, with the highest percentage of mutant cells.
In cells cultured from affected bone of Melo-10 with lower mutant
IgG control
p-ERK1/2
Colon cancer Normal bone Melorheostosis bone
MAP2K1 p.K57N (Melo-2)
p-ERK1/2
MAP2K1 p.K57N (Melo-2)
Melo-2
MFI
low peak
p-ERK1/2
MFI
high peak
p-ERK1/2
261
2301 0
345
Baseline
Baseline
Baseline
DMSO
U0126
U0126
Starv.
Starv.
Baseline
Affected
Affected
Affected
Unaffected
Unaffected
UnaffectedMelo-2
p-ERK1/2
Baseline Baseline
1.00 1.06 3.52 1.50 1.02 1.09 5.23 3.18
U0126U0126 DMSODMSO No. stim.No. stim.
FBS stim. FBS stim.
FBS stim.
DMSO
DMSO
DMSOU0126 U0126
409
2057 2725
447 713
5709 3784
637
FBS stimulation
Baseline DMSO U0126
LHLHLH
IgG control
Unaffected
Affected
–42/44 kDa
–42/44 kDa
–17 kDa
p-ERK1/2
Tot ERK1/2
COX IV
a
b
c
FBS stim.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z
6NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications
Content courtesy of Springer Nature, terms of use apply. Rights reserved
allele frequency of 10%, the activation of ERK1/2 by MEK1 was
equivocal (Supplementary Fig. 4).
MEK1 activation increases cell proliferation.MAP2K1 muta-
tions did not affect MAP2K1 transcripts (Fig. 5a) or MEK1
protein levels (Fig. 5b), supporting a gain-of-function mechanism
intrinsic to the protein itself. ERK1/2 activation promotes G1 to
S-phase cell cycle progression and accelerates cell proliferation.
The enhanced MEK1-ERK1/2 activation in affected osteoblasts
increased cell proliferation in vitro (Fig. 5c). For example, the
doubling time of osteoblasts from affected bone of patient Melo-2
(mutant allele frequency 45%) was 18 h, while osteoblasts from
this patients unaffected bone had a doubling time of 54 h.
Osteoblasts from affected bone did not lose contact inhibition.
Furthermore, affected osteoblasts showed increased levels of
cyclin D3 expression compared to unaffected cells, consistent
with accelerated G1-S cell cycle transition (Fig. 5d).
MEK1 activation decreases osteoblast differentiation in vitro.
To further investigate the functional consequences of the
MAP2K1 mutations causing melorheostosis, osteoblast differ-
entiation and mineralization were assessed in vitro. The presence
of the MAP2K1 mutation strikingly inhibited BMP2-stimulated
mineralization in affected compared to unaffected osteoblasts
(Fig. 5e). Expression of marker genes for osteoblast differentiation
was also reduced in cells with the MAP2K1 mutation during
BMP2-stimulated differentiation (Fig. 5f). Reduced expression of
RUNX2 (runt-related transcription factor 2), ALPL(alkaline
phosphatase) and COL1A1 (the α1 chain of type I collagen)
indicates the prevalence of immature bone forming cells. Osteo-
blasts early in differentiation secrete proteins to stimulate osteo-
clastogenesis15. In fact, we observed a markedly increased ratio of
RANKL/OPG transcripts in osteoblasts from affected bone, which
would generate a strongly positive context for osteoclastogenic
stimulus (Fig. 5f)16.
Together, these data support key histological ndings in
affected bone. Inhibition of mineralization by overactive MEK1
accounts for the massive accumulation of unmineralized osteoid
in affected bone tissue (Fig. 3d). Furthermore, the elevated
RANKL/OPG ratio stimulates the increased osteoclast number
and intense remodeling that occurs in melorheostotic bone
(Fig. 3c).
Discussion
Genetic osteosclerotic disorders are a rare but interesting subset
of bone diseases which can provide important insights into
bone biology. Identication of a germline mutation causing
sclerosteosis led to the development of a potential treatment for
osteoporosis1719. Investigations into brodysplasia ossicans
progressiva elucidated the role of inammation in heterotopic
ossication and exposed targets for treatment20. Here, our
investigation has revealed a role for MEK1 in a disorder char-
acterized by excessive bone formation.
We identied mutations in MAP2K1 in the affected, but not
unaffected, bone of 8 of 15 patients diagnosed clinically and
radiologically with melorheostosis. The mutations cluster at two
residues in the MEK1 negative regulatory domain, p.Q56 and p.
K57, where identical substitutions have been identied in multi-
ple malignancies, but not primary bone tumors. The occurrence
of the mutations in a MEK1 hot spot,and the evidence of
enhanced MEK1-ERK1/2 signaling in melorheostotic bone and
osteoblasts, strongly support MAP2K1 somatic mutations as
causing about half of cases of sporadic melorheostosis. In our
cohort, we found no occurrences of somatic or germline muta-
tions in LEMD3, which have previously been reported in patients
with familial osteopoikilosis and features of melorheostosis6,8.
Mosaicism for cells with increased ERK1/2 activation was
conrmed directly in melorheostotic bone by immunohis-
tochemistry. Furthermore, two populations of cells with distinct
levels of ERK1/2 activation were detected by ow cytometry.
Increased ERK1/2 activation was also detected in cultured
osteoblasts from affected, but not unaffected bone. In individual
patients, the proportion of cultured mutant osteoblasts often
differed from the proportion of mutant allele in genomic DNA
extracted from bone chips from the same lesion, consistent with
clustering of affected cells in mosaic conditions.
The three MAP2K1 mutations we identied in melorheostosis
have been shown to increase MEK1 activity in transfection stu-
dies21, and likely act by destabilizing the α-helix comprising the
MEK1 negative regulatory region, which normally keeps the ATP-
bindingsiteinaninactiveconformation
11.MEK1isadownstream
activator in the RAS pathway. Mutations in RAS pathway com-
ponents are associated with developmental defects falling under the
umbrella designation RASopathies22 and involve variable cardiac,
facial and neurodevelopmental defects. Interestingly, germline
mutations identied in leukocytes from patients with the RASo-
pathy cardio-facio-cutaneous syndrome23,24 include two MAP2K1
mutations (p.G128V/p.Y130C) in the catalytic core, and putatively
activating mutations in the negative regulatory region of MAP2K1
(p.F53S) or MAP2K2 (p.F57V, analogous to MAP2K1 p.F53). Bone
overgrowth is not reported in these RAS-MAPK syndromes,
implying that the somatic mutations we report in osteoblasts pro-
duce a different phenotype than the same mutation in the germline.
Mosaicism for mutations at the same residues (p.Q56P and
p.K57E) we identied in the MEK1 negative regulatory domain in
Fig. 4 Activation of the MEK1-ERK1/2 pathway by MAP2K1 mutations. aImmunohistochemical analysis of ERK1/2 activation in bone tissues from a
melorheostosis patient. Left column shows sections of colon cancer stained with p-ERK1/2-specic antibodies. Marked brown staining is visible. Middle
column shows a section of normal bone stained in a similar fashion. Right column shows representative section from melorheostotic bone (Melo-18,
MAP2K1 p.K57N, VAF 46%) stained with p-ERK1/2-specic antibodies. Osteocytes stain brown surrounded by woven bone. Cells positive for p-ERK1/2 are
also seen in the periosteum. Inset shows high-power view of positively staining cells. A multinucleated osteoclast which does not stain for p-ERK1/2 is
marked by the solid arrow. Cells negative for p-ERK1/2 are noted by the open arrows. See Supplementary Figure 3for staining of SW48 colon cancer cells,
which harbor the MAP2K1 p.K56P mutation. bp-ERK1/2-specicow cytometry analysis. Affected and unaffected osteoblasts from Melo-2 (MAP2K1 p.
K57N, VAF 45%) were stimulated with serum with or without MEK inhibitor U0126. Two peaks in the histogram indicates cell subpopulations of distinct
level of p-ERK1/2 upon serum-stimulation in osteoblasts from affected bone (red), cells from unaffected bone (blue) only showed a single peak. U0126
reduced p-ERK1/2 in cells from affected bone. Cells stained with matching rabbit IgG isotype control are also shown (gray). The geometrical mean channel
uorescence is shown below for the high and low peaks of p-ERK1/2 marked on the histograms. cWestern blot analysis of osteoblasts from affected and
unaffected bone of patient Melo-2 (MAP2K1 p.K57N, VAF 45%) shows increased ERK1/2 activation (p-ERK1/2) by MEK1 mutation in affected osteoblasts
(lane 7 of p-ERK1/2 blot), as compared to unaffected osteoblasts (lane 3), by serum-stimulation. Inhibition of MEK1 with U0126 signicantly diminished p-
ERK1/2 in both affected and unaffected, but the level of p-ERK1/2 was still higher in affected osteoblasts compared to unaffected (lanes 4 and 8).
Quantication data of band intensities are shown in a table. COX IV was used as control for equal amount protein loading
NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z ARTICLE
NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved
melorheostosis patients has recently been reported in endothelial,
but not skin or blood, cells of patients with extracranial arter-
iovenous malformations (AVM)25. Similar to melorheostotic bone
lesions, AVM lesions caused by MAP2K1 mutations expand but do
not metastasize.
While the MAPK pathway is known to be involved in skele-
togenesis and MEK1 is essential in embryogenesis, our study
documents the importance of the MEK1-ERK1/2 pathway in
human bone cell biology. While somatic mutations in MAP2K1
have also been reported in Langerhans cell histiocytosis and
Relative expression
1.5
1.0
0.5
0.0
Unaff Aff Unaff Aff Unaff Aff Unaff Aff Unaff Aff Unaff Aff Unaff Aff
Melo-18
Melo-06Melo-02
0 8 16 24 32 40 48 56 64 72 80 88 96 104 112 120
Time (h)
100
80
60
40
20
0
% confluence
Melo-10
K57E K57N
Q56P
MAP2K1
MAP2K1 p.K57N (Melo-18)
RANKL/OPGALPLRUNX2 COL1A1
Unaffected
Affected
Melo-04 Melo-09 Melo-19 Melo-10 Melo-02 Melo-06 Melo-18
Unaff
8
6
4
2
0
1.5
1.0
0.5
0.0
4
3
2
1
0
Relative expression
Relative expression
Relative expression
Relative expression
Aff
Unaff
Aff
Unaff
Aff
Unaff
Aff
50
40
30
20
10
0
–45 kDa
–17 kDa
MEK1
COXIV
Aff
Unaff
Aff
Unaff
Aff
Unaff
Aff
Unaff
Aff
Unaff
Aff
Unaff
Aff
Unaff
Melo-19Melo-09Melo-04
Affected
Unaffected D1 D3 D6
p-ERK1/2
Cyclin D3
GAPDH
–42/44 kDa
–31 kDa
–37 kDa
Aff
Unaff
Aff
Unaff
Aff
Unaff
MAP2K1 p.K57N (Melo-18)
K57NK57E
MEK1
Q56P
a
c
b
d
e
fg
Fig. 5 Increased cell proliferation and delayed osteoblast differentiation and mineralization by MAP2K1 mutations. aMAP2K1 variants found in seven
melorheostosis patients do not affect the levels of MAP2K1 transcripts. There was no statistically signicant difference in MAP2K1 transcript levels between
affected and unaffected osteoblasts (*p<0.05, paired t-test). Real-time qPCR was carried out in triplicate for each patient sample. bWestern blot analysis
displays comparable levels of MEK1 protein in affected and unaffected osteoblasts. cCell proliferation assay using live-cell imaging. Affected and
unaffected osteoblasts from melorheostosis patient, Melo-2 (MAP2K1 p.K57N, VAF 45%) were plated at various densities (1000~5000 cells/well) (n=
30). Percent cell conuence is shown at 2-h intervals with symbols indicating mean of replicates (error bars: SEM). Doubling time calculated from the
linear phase growth yielded a doubling time of 18 h for affected osteoblasts compared to 54 h for unaffected. Results are representative of two independent
experiments. (p< 0.0001). dWestern blot analysis shows that affected osteoblasts (lanes 2, 4, and 6) expressed higher level of cyclin D3 compared to
unaffected (lanes 1, 3, and 5), correlating with the increased p-ERK1/2 level in affected cells shown in Fig. 4c. Note that levels of cyclin D3 and p-ERK1/2
decreased on day 3 (D3) and day 6 (D6) compared to day 1 (D1), because culture media was not refreshed after day 1, similar to conditions used during the
live-cell imaging shown in Fig. 5c. eAlizarin Red S staining of mineralization in osteoblast cultures from patient Melo-18 (MAP2K1 p.K57N, VAF 46%). After
7 weeks of BMP2-stimulated mineralization in vitro, signicantly inhibited mineralization was observed in affected cells compared to unaffected. fReal-
time qPCR analysis of expression of osteogenic marker genes, RUNX2,COL1A1, and ALPL in osteoblasts from melorheostosis patient Melo-2 (MAP2K1 p.
K57N, VAF 45%) after two weeks of osteogenic stimulation. Expression level of RUNX2, COL1A1, and ALPL was signicantly lower in affected osteoblasts
compared to unaffected. gThe RANKL/OPG transcript ratio, an index of osteoclastogenic stimulus, was assessed by real-time qPCR with Melo-2 patient
osteoblasts as in Fig. 5f. Signicantly higher ratio of RANKL/OPG in affected osteoblasts indicates increased osteoclastogenesis compared to unaffected
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z
8NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications
Content courtesy of Springer Nature, terms of use apply. Rights reserved
ErdheimChester disease (ECD)26, the mutated cells in ECD are
derived from monocytes rather than osteoblasts. Bone lesions in
ECD are typically bilateral and symmetrical and occur in trabe-
cular bone of the epiphysis27. In contrast, activation of ERK1/2
MAP kinases in melorheostosis occurs in osteoblast lineage and
the lesions are primarily cortical.
Transgenic murine models, in which the entire MEK1 negative
regulatory domain was deleted, share the elevated ERK1/2 sig-
naling seen in melorheostosis28. However, the mice displayed
accelerated bone remodeling rather than bone overgrowth. The
mouse osteoblasts also showed increased differentiation and
mineralization, but unchanged proliferation in vitro.
In melorheostosis, activation of the MEK1-ERK1/2 pathway in
mature bone results in an increase in osteoblast surface and
increased production of unmineralized bone matrix (osteoid).
This is consistent with the faster proliferation of osteoblasts
cultured from melorheostotic bone and with the less differ-
entiated osteoblast phenotype revealed in vitro in BMP2-
stimulated cells, in which transcripts of osteogenic genes
RUNX2,COL1A1, and ALPL were decreased. Histologically, we
observed a signicant increase in osteoid formation and osteoclast
numbers in affected bone from the MAP2K1 variant cohort.
Interestingly, these features were previously noted in a single case
report29 with unknown genetics. The increased osteoid seen
in vivo is consistent with the decreased mineralization deposited
by affected osteoblasts in vitro with BMP2 stimulation. Increased
osteoclast numbers and bone turnover in affected bone is con-
sistent with the increased RANKL secretion by cultured cells,
which would stimulate osteoclast development and intense bone
remodeling. Furthermore, it is known that chronic activation of
ERK1/2 strongly inhibits BMP2-mediated mineralization, as
previously shown in a murine model for neurobromatosis30, and
provides an underlying mechanism for the increased unminer-
alized osteoid found in melorheostotic bone. The phenotypic
distinction between murine models and melorheostosis patients
may be ascribed to overexpression of the MEK1 transgene in all
murine osteoblasts as opposed to the mosaic mutations in
melorheostosis, or greater disruption of function by deletion of
the full negative regulatory domain28,31. Furthermore, studies in
fruit ies and zebrash indicate that activating MAP2K1 muta-
tions affect ERK1/2 activation differently depending on pathways
intrinsic to the target tissue and developmental state22.
Identication of a somatic mosaic mutation in osteoblasts
accounts for the unique clinical features of melorheostosis. The
dripping candle waxpattern observed on radiographs is due to
marked appositional bone growth followed by aggressive remo-
deling. The radiographic pattern of multiple contiguous bones
affected while others are spared may be due to longitudinal
expansion of mutant osteoblasts precursors along the axis of
development (Supplementary Fig. 5)32. The marked variability in
the pattern of bone lesions between patients is likely related to
mutations arising during different points during development,
with earlier mutations resulting in an increased burden of disease.
Further studies will delineate the roles of specicMAP2K1
mutations in different tissues at various developmental stages.
Melorheostosis can now be viewed as a genetically hetero-
geneous group of dysostoses. The melorheostosis seen in patients
who have spotted boneosteopoikilosis is associated with a
mutation in germline LEMD36. Whyte et al. have shown that
additional somatic mutations in KRAS (and possibly other genes)
may play a role in the development of dripping candle wax
bone lesions on radiographs in some patients in an osteopoiki-
losis kindred8. Patients who have sporadic melorheostosis are a
distinct group and do not have germline6,33, or as we now show,
somatic mutations in LEMD3. It remains to be seen if the
radiographic similarities in melorheostosis caused by LEMD3
mutations and MAP2K1 mutations are due to involvement of a
common pathway with activation of SMADs, or are simply a
similar clinical outcome.
Our nding that three of four patients demonstrated mutations
in the overlying skin raises the possibility that testing of skin for
MAP2K1 mutations may be a diagnostic test for melorheostosis.
However, if the skin sample is negative, bone biopsy may still be
necessary to detect the mutation, especially in patients with a low
mutant allele frequency. Although the causative gene is an
oncogene, the clinical data from many years is reassuring to
patients that malignant degeneration is rare2,4.
Development of a diagnostic test is especially valuable because
identication of somatic MAP2K1 mutations in melorheostosis
raises the possibility of inhibiting MEK1 to treat melorheostotic
bone lesions. Several MEK1 inhibitors have already been devel-
oped for malignancies with MAP2K1 mutations identical to those
we identied in melorheostosis34. Because melorheostosis is one
of few osteosclerotic bone-forming diseases, study of the MEK1-
ERK1/2 pathway may lead to insights relevant to diseases where
bone formation is impaired, such as delayed fracture healing and
osteoporosis.
Methods
Patient cohort and bone sampling. We recruited 20 unrelated adults with a
diagnosis of melorheostosis to a NIAMS Institutional Review Board-approved
protocol (NCT02504879). Diagnoses were conrmed by radiographs and increased
uptake on 18F-NaF bone scan (Fig. 1). Patients were excluded if pregnant or
lactating, if under the age of 18, if actively infected or if unable to provide informed
consent. Study subjects were not compensated for their participation. Fifteen
patients consented to bone biopsies of affected and contralateral unaffected bone
(see Supplementary Table 1for clinical characteristics). Patients consented to
publication of photographs.
Patients underwent open surgical biopsy of the affected bone under uoroscopic
guidance. The site of the biopsy of affected bone was chosen to minimize risk. Thus
the most supercial area of melorheostotic bone was chosen. Under general
anesthesia, the limb was exsanguinated and a tourniquet elevated (in all but one
patient with proximal femur lesion). The bone was exposed and any thickened
periosteum divided. Fluoroscopy was used to conrm the site of affected bone.
Four drill holes were used to create a rectangle, and an osteotome used to connect
the drill holes. The rectangular piece of cortical bone was then removed and
sectioned in the operating room. The sections were placed immediately in cell
culture media (for DNA extraction and osteoblast culture) or 70% ethanol for
histology. Hemostasis was obtained with electrocautery and the wound closed in
layers. A contralateral control sample of bone with no radiographic evidence of
melorheostosis was also obtained.
For some patients, such as Melo-2 and Melo-18, abundant melorheostotic bone
was available for evaluation. For other patients, only a small amount of affected
bone could be safely sampled; priority was given to bone for DNA extraction
followed by osteoblast culture over other studies. In all patients, the smallest
informative sample of normal bone was taken to minimize morbidity. Thus,
normal bone was often a limiting reagent. See Supplementary Table 2for details on
which patient samples underwent particular experiments.
Additional tissue sampling. In our initial study design, we were concerned that
surgical biopsy of melorheostosis (which had never been done before) could lead to
worsening of the disease, infection, or other complication. Therefore, only the
smallest amount of usable bone was collected. After a shared mutation was iden-
tied and the absence of surgical complications assured, four patients returning for
a follow-up visit underwent 6 mm punch biopsy of skin overlying an area of
affected bone, and a control biopsy of unaffected skin from the contralateral side.
Seven of the eight patients with MAP2K1 mutations agreed to have blood tested for
mutations. DNA from peripheral blood leukocytes was tested by ddPCR for pre-
sence of the mutation.
Genomic DNA extraction and mutation detection in bone tissue. Soft tissues
and cartilage were manually dissected from affected and unaffected bone of each
patient. Following repeated washes in phosphate-buffered saline (PBS) containing
no Ca2+or Mg2+to remove marrow, mineralized tissues were minced using a
rongeur and spring scissors in a sterile conical bottom glass tube (Roboz, Gai-
thersburg, MD).
Genomic DNA was isolated from minced bone chips following the
manufacturers instructions (Gentra Puregene, Qiagen). In brief, minced bone
samples were resuspended in cell lysis solution and digested with 0.2 mg/ml
proteinase K at 55 °C for 12 h. Digested proteins were precipitated and removed by
centrifugation at 14,000×gfor 3 min. The cleared supernatant was transferred to a
NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z ARTICLE
NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/s41467-018-03720-z |www.nature.com/naturecommunications 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved
new tube and genomic DNA was precipitated with isopropanol and collected by
centrifugation at 14,000×gfor 5 min. The DNA pellet was washed once with 70%
ethanol and air-dried at room temperature for 5 min. DNA was resuspended in
DNA hydration buffer (10 mM Tris, 1 mM EDTA, pH 8.0) and its concentration
was measured using a NanoDrop spectrophotometer (ThermoFischer Scientic).
Genomic DNA samples underwent deep Whole Exome Sequencing (Otogenetics),
with 107270× coverage for each sample. Most exome targets (82%) had at least
50× coverage in each sample. Agilent SureSelect Human 51 Mbp All Exon kit was
used for exome capture. Paired-end sequencing was performed on Illumina HiSeq
sequencers. Sequencing reads were returned in FASTQ format and were aligned to
Human Reference Genome Build 37 using BurrowsWheeler alignment Tool. The
standard PICARD-GATK pipeline was used to remove duplicate reads, rene
alignment around indels, and recalibrate base quality scores. The resulting BAM
les (one per sample) served as inputs to somatic mutation callers (Fig. 2a).
Two somatic mutation callers (muTect v1.1.7 and Strelka v1.0.1435) were used,
with default parameters for WES data. Somatic variants called by either method
were annotated with functional impact and population frequency using
ANNOVAR. We ltered somatic variants for those that were not present in the
unaffected bone, caused protein or ncRNA sequence changes or splicing changes,
had less than 1% frequency in ExAC and 1000 Genomes Project databases, were
not in a duplicated genomic region, and were called by both muTect and Strelka.
Using this method, 284 putative somatic variants were identied in 15 affected
bone samples (Supplementary Data 1and Supplementary Fig. 2).
Exome sequencing was complemented by high-coverage amplicon sequencing
with an average sequencing coverage of more than 10,000 × per sample. A targeted
NGS amplicon library prep kit from Swift Biosciences (56 G Oncology) was used to
screen patients for MAP2K1 somatic mutations. The panel targets cancer-
associated mutations in 56 genes including 5 mutation hotspot regions in MAP2K1
(exons 2, 3, 6, 7, 11), as well as the RAS codon 12, 13 and 61 hotspot mutations.
The amplicon libraries were sequenced on Illumina MiSeq and variants were called
with LoFreq 2.1.2.
To validate and quantitate the allele frequency of somatic mutations in other
tissues and cells cultured ex vivo, we utilized ddPCR (Fig. 2c). The approach
determines the abundance of each allele using allele-specic hydrolysis probes
(Supplementary Table 7) complementary to mutant allele (FAM-labeled) and wild-
type allele (HEX-labeled) and is based on partitioning of each sample into >10,000
nanoliter-sized droplets, in which PCR is carried out. gBlock oligos of 200 bp
bearing the mutation of interest were designed by Integrated DNA Technologies,
Inc. (Coralville, IA) and used as mutation-positive controls. Reactions were
performed on the QX200 Droplet Digital PCR System (Bio-Rad), using standard
PCR cycling conditions based on manufacturers instructions (10-min enzyme
activation at 95 °C, followed by 40 cycles of 30-s 94 °C denaturation and 1-min 55 °
C annealing/extension, nishing with a 10-min enzyme deactivation at 98 °C).
QuantaSoft software was used to quantitate the concentration of each allele
as the number of copies per microliter, using a Poisson distribution model to
account for the number of target copies per individual droplet. Fractional
abundance of mutant allele was calculated as the concentration (copies per
microliter) obtained in FAM channel divided by sum of concentrations of FAM
and HEX channels.
Bone histology and histomorphometry. Bone samples from a subset of 6 patients
with MAP2K1 mutations were embedded in polymethylmethacrylate and
prepared for histomorphometric analyses using standard procedures36. For
histological examinations, thin sections (4 µm) were cut from the tissue blocks
with a hard tissue microtome (Leica SM2500, Leica Microsystems Nußloch
GmbH), deplasticized with 2-methoxyethyl-acetate and stained with
modied Goldners Trichrome36 (Fig. 3d). A light microscope equipped with a
video camera (Zeiss Axiophot, Zeiss AxioCam, Oberkochen, Germany) was used to
obtain digital images of the sections that were analyzed using NIH ImageJ software
(version 1.63)37. Bone histomorphometric analyses were performed on 4 randomly
chosen areas throughout each bone section (Fig. 3c). Because bone samples
from the various skeletal sites contained predominantly compact/osteonal bone,
only indices of bone formation and resorption were evaluated: osteoid thickness
(O.Th), osteoid surface per bone surface (OS/BS), osteoblast surface per bone
surface (Ob.S/BS), osteoclast surface per bone surface (Oc.S/BS) and eroded
surface per bone surface (ES/BS). Bone lamellar organization was observed
under polarized light.
Subsequently, the residual blocks were prepared for backscattered electron
imaging by grinding and polishing to obtain plane parallel surfaces, then carbon
coated (Fig. 3a, b). The entire cross-sectioned bone sample area was imaged with a
spatial resolution of 1.8 µm per pixel using a eld emission scanning electron
microscope (FESEM) (Zeiss Supra 40, Oberkochen, Germany) equipped with a
four-quadrant semiconductor backscatter electron detector. The FESEM was
operated with electron energy of 20 keV. The gray levels reecting the mineral/
calcium content were calibrated by the material contrast of pure carbon and
aluminum.
Statistical evaluation was performed with GraphPad Prism 6.0 (GraphPad
Software, Inc., La Jolla, CA, USA). Comparison of histomorphometric indices were
based on paired t-test comparing affected vs. unaffected tissue in each patient.
Statistical signicance was considered as p< 0.05.
Cell culture. Primary osteoblasts were grown from freshly minced bone chips
according to the method of Robey and Termine38. Bone chips (~ 1 mm) were
incubated in alpha-minimal essential medium (α-MEM, Life Technologies) con-
taining type II collagenase (200 U/ml, Worthington Biochemical Corp., Lakewood,
NJ) for 2 h at 37 °C with gentle rocking. Bone chips were allowed to settle briey,
then collagenase solution was removed and washed twice with PBS. Bone chips
were placed in T75 tissue culture asks with α-MEM supplemented with 10% fetal
bovine serum (Gemini Bio-Products, West Sacramento, CA) and antibiotics
(penicillin (100 U/ml) and streptomy cin (100 µg/ml), Thermo Fisher Scientic
Inc.). Bone chips were incubated for 23 weeks at 37 °C/8% CO
2
, refreshing culture
media every 3 days. Osteoblasts of passages 14 were used in this study.
Genomic DNA was extracted from cultured osteoblasts using the Gentra
Puregene kit (Qiagen), as described above. The proportion of mutant MAP2K1
allele in DNA from multiple cell passages and revivals was determined by ddPCR
as above.
Cell proliferation assay by live-cell imaging. Osteoblasts were plated at the
indicated densities in 96-well tissue culture dish and imaged at 2 h intervals for 120
h using the IncuCyte ZOOM Kinetic Imaging System (Essen Bioscience) at 10×
magnication (Fig. 5c). During the live-cell imaging, cells were incubated in 37 °C/
8% CO
2
. Percent conuence was calculated from the percentage of the well area
occupied by cells. Doubling time was calculated using GraphPad Prism 6.0 using
non-linear regression. To analyze comparable populations of cells between groups,
only wells with conuency at the rst time-point of image acquisition within one
half standard deviation of the mean were included in the analysis. Statistical
comparison was done by two-way ANOVA.
Gene expression in osteoblasts by real-time quantitative PCR. Total RNA was
extracted from osteoblasts using the RNeasy mini kit (Qiagen) following the
manufacturers instruction. RNA concentration was measured by the NanoDrop
spectrophotometer. Synthesis of cDNA was performed using 1 µg of total RNA and
the HighCapacity cDNA Reverse Transcription Kit (Thermo Fisher Scientic),
following the manufacturers instructions. Comparative real-time PCR was per-
formed in triplicate with an Applied Biosystems Prism 7500 Fast Sequence
Detection System using TaqMan universal PCR master mix, according to the
manufactures protocol (Applied Biosystems Inc., Foster City, CA) (Fig. 5a, f, g).
The TaqMan probes and primers were purchased from Applied Biosystems:
Human RUNX2 (Hs01047973_m1), COL1A1 (Hs00164004_m1), ALPL
(Hs01029144_m1), RANKL (TNFSF11, Hs00243522_m1), OPG (TNFRSF11B,
Hs00900358_m1). Human GAPDH (Hs02786624_g1) and TBP (Hs00427620_m1)
were used as endogenous controls for normalization. Levels of MAP2K1,RUNX2,
ALPL and COL1A1, RANKL (TNFSF11), and OPG (TNFRSF11B) transcripts
were determined using the 7500 Fast System SDS software version 1.3.1 (Applied
Biosystems). Relative expression was calculated using the comparative ΔΔCt
method.
Flow cytometric analysis for intracellular p-ERK1/2. Osteoblasts cultured from
affected and unaffected bone of melorheostosis patients were serum-starved for 1 h
and stimulated with 20% FBS with or without MEK inhibitor U0126 (10 μM) for
30 min. Osteoblasts were then harvested for intracellular staining with antibodies
for p-ERK1/2. After xation with 4% paraformaldehyde and permeabilization with
ice cold methanol, cells were washed in PBS containing 1% BSA. Cells were stained
for 60 min in the dark at room temperature with the following antibodies (Cell
Signaling Technology, Cambridge, MA): Phospho-p44/42 MAPK (ERK1/2)
(Thr202/Tyr204), (197G2), (clone E10) rabbit monoclonal antibody (Alexa Fluor
647®Conjugate) (#13148) (1:100 dilution) or rabbit IgG isotype control (Alexa
Fluor®647 Conjugate) #3452). Cells were acquired on a FACSVerseow cyt-
ometer (Becton Dickinson) and analyses for p-ERK1/2 levels were performed by
using FlowJo®software (Tree Star) on live gated cells (Fig. 4b). The gating strategy
is shown in Supplementary Figure 7.
Western blot analysis of MEK1 and ERK1/2 activation. Osteoblast lysates were
analyzed by western blotting using standard methods and indicated antibodies
(Fig. 4c, b, d). Briey, osteoblasts from affected and unaffected bone of melor-
heostosis patients were grown in α-MEM supplemented with 10% FBS (Gemini
Bio-Products, West Sacramento, CA) and antibiotics (penicillin (100 U/ml) and
streptomycin (100 µg/ml), Thermo Fi sher Scientic Inc.). Cells were seeded at
75,000 cells/well in 12-well tissue culture vessel containing 1 ml/well of culture
media. On the next day, cells were pre-treated with U0126 (1, 4-diamino-2, 3-
dicyano-1,4-bis[2-aminophenylthio] butadiene, a MEK1/2 inhib itor, for 1 h.
Inhibitor concentration in culture was 10 µM (#9903, Cell Signaling Technology)
or DMSO (vehicle control, Sigma) in serum-free α-MEM. Cells were then stimu-
lated with FBS (20% in media) for 30 min before being lysed in cell lysis buffer (50
mM Tris (pH 8.0), 150 mM NaCl, 1% IGEPAL, 0.5% sodium deoxycholate, 0.1%
sodium disodium sulfate, 1× protease inhibitor cocktail, and 1× phosphatase
inhibitor cocktail). Cell lysates were cleared by centrifugation at 20,000×gfor 20
min at 4 °C. Soluble cell lysates were mixed with SDS-PAGE sample buffer (50 mM
Tris, pH 6.8, 2% SDS, 10% glycerol, 0.1% bromophenol blue, and 100 mM
dithiothreitol) and denatured at 80 °C for 10 min. Proteins were resolved in 415%
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z
10 NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/ s41467-018-03720-z |www.nature.com/naturecommunications
Content courtesy of Springer Nature, terms of use apply. Rights reserved
mini-PROTEAN precast gels (Bio-Rad, Hercules, CA) at 100 V for 90 min. The
resolved proteins were transferred to nitrocellulose membranes (Bio-Rad) at 100 V
for 90 min at 4 °C. The membranes were immersed in 5% bovine serum albumin in
Tris-buffered saline containing 0.5% Tween-20 (TBS-T) for 1 h at room tem-
perature to block non-specic binding of primary antibodies. Subsequently, the
membranes were probed with the following antibodies (Cell Signaling Technology,
Cambridge, MA): phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) (clone
#D13.14.4E) rabbit monoclonal antibody (#4370, 1:2000), p44/42 MAPK (Erk1/2)
(clone # L34F12) mouse monoclonal antibody (#4696, 1:2000), MEK1/2 rabbit
antibody (#9122, 1:1000), cyclin D3 (clone #DCS22) mouse monoclonal antibody
(#2936, 1:2000), GAPDH (clone #D16H11) rabbit monoclonal antibody (#5174,
1:1000), and COX IV (clone # 3E11) rabbit monoclonal antibody (#4850, 1:1000).
After incubating membranes with primary antibodies for 16 h at 4 °C, membranes
were washed in TBS-T three times for 5 min each at room temperature. Protein
bands were visualized by incubating membranes for 1 h at room temperature with
the following secondary antibodies (LI-COR Biosciences, Lincoln, NE): IRDye®
680RD anti-rabbit IgG (#926-68071, 0.1 ug/ml) or IRDye®800CW anti-mouse IgG
(#926-32210, 0.1 ug/ml). The membranes were washed in TBS-T three times for 5
min each at room temperature before scanning with Odyssey®Infrared Imaging
Systems (LI-COR Biosciences, Lincoln, NE). Uncropped western blots are available
in Supplementary Figure 6.
Immunohistochemistry for p-ERK1/2. Fresh bone tissue was xed in 10%
neutral buffered formalin, decalcied by formic acid, and embedded in parafn.
Thin sections (5 µm) were mounted onto tape adhesive slides. Following depar-
afnization and rehydration with ethanol, endogenous peroxidase was blocked
using 3% hydrogen peroxide/distilled water. Antigen retrieval was performed
using a citrate buffer (BioGenex) for 10 s at 100 °C, then slid es were cooled and
rinsed in distilled water and Tris-buffered saline/Tween-20 (TBS-T). After blocking
in 5% normal goat serum (Vector Labs), sections were incubated with anti-phos-
pho-p44/42 MAPK ERK1/2 rabbit monoclonal antibody (Thr202/Tyr204; diluted
1:200, Cell Signaling Technology, USA #4376) overnight at 4 °C or monoclonal
rabbit IgG isotype control. Subsequently, biotinylated goat anti-rabbit IgG (1:100,
Vector Labs) was applied for 30 min, before slides were incubated with ABC
Elite Standard (Vector Labs) for 30 min. p-ERK1/2 was visualized by a 5-min
incubation in DAB Reagent (Sigma) and subsequent hematoxylin counterstain.
Slides were dehydrated, cleared in xylene and mounted with coverslips with Per-
mount. Parafn embedded colon cancer cells (SW48, carrying a p.Q56P mutation
in MAP2K1) was used as positive control13. Normal bone from patients under-
going surgical resection for unrelated indications served as negative control
(Fig. 4a).
Osteoblast mineralization and differentiation in vitro. Osteoblasts from affected
and unaffected bone of patient Melo-18 were plated at passage 2 with 50,000 cells/
well in 12-well plates and cultured to conuency in α-MEM supplemented with
10% FBS and antibiotics at 37 °C/8% CO
2
. Following conuence, osteoblasts were
grown in osteoblast differentiation medium (50 µg/mL L-ascorbic acid, 10 nM
dexamethasone, and 2.5 mM β-glycerop hosphate), with or without 100 ng/mL
recombinant BMP2 (#355-BM, R&D Systems, Minneapolis, MN), refreshing
osteogenic media every 3 days for 7 weeks.
To visualize mineralization in osteoblast culture, cells were washed once with
PBS and xed with 4% paraformaldehyde for 30 min at room temperature. Fixed
cells were washed three times with PBS and stained with 2% Alizarin Red S
solution (pH 4.2) for 30 min at room temperature. Excess Alizarin Red S stain was
removed with three distilled water washes. Cells were air-dried in the dark before
imaging (Fig. 5e). During osteoblast mineralization in osteogenic medium, total
RNA was collected at 2 weekspost-induction and used to examine gene expression
by real-time quantitative PCR analysis.
Data availabilty. The data that support the ndings of this study are available from
the corresponding author upon reasonable request.
Received: 7 November 2017 Accepted: 7 March 2018
References
1. Freyschmidt, J. Melorheostosis: a review of 23 cases. Eur. Radiol. 11, 474479
(2001).
2. Kotwal, A. & Clarke, B. L. Melorheostosis: a rare sclerosing bone dysplasia.
Curr. Osteoporos. Rep. 15, 335342 (2017).
3. Younge, D., Drummond, D., Herring, J. & Cruess, R. L. Melorheostosis in
children. Clinical features and natural history. J. Bone Jt. Surg. Br. 61-B, 4158
(1979).
4. Bostman, O. M., Holmstrom, T. & Riska, E. B. Osteosarcoma arising in
a melorheostotic femur. A case report. J. Bone Jt. Surg. Am. 69, 12327
(1987).
5. Hellemans, J. et al. Loss-of-function mutations in LEMD3 result in
osteopoikilosis, Buschke-Ollendorff syndrome and melorheostosis. Nat. Genet
36, 12138 (2004).
6. Mumm, S. et al. Deactivating germline mutations in LEMD3 cause
osteopoikilosis and Buschke-Ollendorff syndrome, but not sporadic
melorheostosis. J. Bone Miner. Res. 22, 24350 (2007).
7. Bonafe, L. et al. Nosology and classication of genetic skeletal disorders: 2015
revision. Am. J. Med Genet A 167A, 286992 (2015).
8. Whyte, M. P. et al. Melorheostosis: exome sequencing of an associated
dermatosis implicates postzygotic mosaicism of mutated KRAS. Bone 101,
145155 (2017).
9. Fryns, J. P. Melorheostosis and somatic mosaicism. Am. J. Med. Genet. 1995,
199 (1995).
10. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans.
Nature 536, 28591 (2016).
11. Fischmann, T. O. et al. Crystal structures of MEK1 binary and ternary
complexes with nucleotides and inhibitors. Biochemistry 48, 26612674
(2009).
12. Marks, J. L. et al. Novel MEK1 mutation identied by mutational analysis of
epidermal growth factor receptor signaling pathway genes in lung
adenocarcinoma. Cancer Res. 68, 55248 (2008).
13. Murugan, A. K., Dong, J., Xie, J. & Xing, M. MEK1 mutations, but not ERK2
mutations, occur in melanomas and colon carcinomas, but none in thyroid
carcinomas. Cell Cycle 8, 21224 (2009).
14. Waterfall, J. J. et al. High prevalence of MAP2K1 mutations in variant and
IGHV4-34-expressing hairy-cell leukemias. Nat. Genet. 46,810 (2014).
15. Atkins, G. J. et al. RANKL expression is related to the differentiation state of
human osteoblasts. J. Bone Miner. Res. 18, 108898 (2003).
16. Boyce, B. F. & Xing, L. Biology of RANK, RANKL, and osteoprotegerin.
Arthritis Res. Ther. 9(Suppl 1), S1 (2007).
17. Cosman, F. et al. Romosozumab treatment in postmenopausal women with
osteoporosis. N. Engl. J. Med. 375, 15321543 (2016).
18. Balemans, W. et al. Increased bone density in sclerosteosis is due to the
deciency of a novel secreted protein (SOST). Hum. Mol. Genet. 10, 53743
(2001).
19. Brunkow, M. E. et al. Bone dysplasia sclerosteosis results from loss of the
SOST gene product, a novel cystine knot-containing protein. Am. J. Hum.
Genet 68, 57789 (2001).
20. Shore, E. M. & Kaplan, F. S. Insights from a rare genetic disorder of extra-
skeletal bone formation, brodysplasia ossicans progressiva (FOP). Bone 43,
42733 (2008).
21. Arcila, M. E. et al. MAP2K1 (MEK1) mutations dene a distinct subset of lung
adenocarcinoma associated with smoking. Clin. Cancer Res. 21, 193543
(2015).
22. Goyal, Y. et al. Divergent effects of intrinsically active MEK variants on
developmental Ras signaling. Nat. Genet. 49, 465469 (2017).
23. Rodriguez-Viciana, P. et al. Germline mutations in genes within the MAPK
pathway cause cardio-facio-cutaneous syndrome. Science 311, 12871290
(2006).
24. Schulz, A. L. et al. Mutation and phenotypic spectrum in patients with
cardio-facio-cutaneous and Costello syndrome. Clin. Genet. 73,6270
(2008).
25. Couto, J. A. et al. Somatic MAP2K1 mutations are associated with extracranial
arteriovenous malformation. Am. J. Hum. Genet. 100, 546554 (2017).
26. Chakraborty, R. et al. Mutually exclusive recurrent somatic mutations in
MAP2K1 and BRAF support a central role for ERK activation in LCH
pathogenesis. Blood 124, 300715 (2014).
27. Estrada-Veras, J. I. et al. The clinical spectrum of Erdheim-Chester disease: an
observational cohort study. Blood Adv. 1, 357366 (2017).
28. Ge, C., Xiao, G., Jiang, D. & Franceschi, R. T. Critical role of the extracellular
signal-regulated kinase-MAPK pathway in osteoblast differentiation and
skeletal development. J. Cell Biol. 176, 70918 (2007).
29. Hoshi, K. et al. Histopathological characterization of melorheostosis.
Orthopedics 24, 2737 (2001).
30. de la Croix Ndong, J. et al. Combined MEK inhibition and BMP2 treatment
promotes osteoblast differentiation and bone healing in Nf1Osx -/- mice. J.
Bone Miner. Res. 30,5563 (2015).
31. Matsushita, T. et al. Extracellular signal-regulated kinase 1 (ERK1) and ERK2
play essential roles in osteoblast differentiation and in supporting
osteoclastogenesis. Mol. Cell Biol. 29, 584357 (2009).
32. Arques, C. G., Doohan, R., Sharpe, J. & Torres, M. Cell tracing reveals a
dorsoventral lineage restriction plane in the mouse limb bud mesenchyme.
Development 134, 371322 (2007).
33. Hellemans, J. et al. Germline LEMD3 mutations are rare in sporadic patients
with isolated melorheostosis. Hum. Mutat. 27, 290 (2006).
34. Caunt, C. J., Sale, M. J., Smith, P. D. & Cook, S. J. MEK1 and MEK2 inhibitors
and cancer therapy: the long and winding road. Nat. Rev. Cancer 15, 57792
(2015).
NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z ARTICLE
NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/ s41467-018-03720-z |www.nature.com/naturecommunications 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved
35. Saunders, C. T. et al. Strelka: accurate somatic small-variant calling from
sequenced tumor-normal sample pairs. Bioinformatics 28, 18117 (2012).
36. Glorieux, F. H. et al. Normative data for iliac bone histomorphometry in
growing children. Bone 26, 1039 (2000).
37. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25
years of image analysis. Nat. Methods 9, 6715 (2012).
38. Robey, P. G. & Termine, J. D. Human-bone cells-invitro. Calcif. Tissue Int. 37,
453460 (1985).
Acknowledgements
We thank Dr. William Gahl for early support of the study, Donna Butcher for assistance
with immunohistochemistry, Daniela Gabriel, Petra Keplinger, Sonja Lueger and Phae-
dra Messmer for careful preparation of bone samples for histomorphometry. We are
especially grateful to Kathleen Harper, the Melorheostosis Association, and the patients
for their support of this study.
Author contributions
T.B. conceived the overall project. S.J. and T.B. designed and wrote the clinical protocol,
provided clinical care for patients and analyzed data. J.K., E.W.C. and E.L. supported
clinical care of patients. With assistance from E.P.H., W.A.C. and F.M. H.K.
designed, conducted experiments with cultured osteoblasts and analyzed data under
supervision of J.C.M. Z.D., A.I., and R.S. analyzed the data from whole exome sequen-
cing. N.F.Z., P.R. and K.K. performed histological evaluation of bone specimens. E.W.C.
and S.J. obtained and analyzed skin samples. J.C.M., T.B., R.S., H.K., Z.D., and S.J. wrote
the manuscript.
Additional information
Supplementary Information accompanies this paper at https://doi.org/10.1038/s41467-
018-03720-z.
Competing interests: The authors declare no competing interests.
Reprints and permission information is available online at http://npg.nature.com/
reprintsandpermissions/
Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.
© The Author(s) 2018
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03720-z
12 NATURE COMMUNICATIONS | (2018) 9:1390 |DOI: 10.1038/ s41467-018-03720-z |www.nature.com/naturecommunications
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com

Supplementary resources (3)

... [2][3][4] In classical melorheostosis, bone lesions are metabolically active on 18 F-NaF scans and have a characteristic "dripping candlewax" appearance on radiographs. [5,6] In approximately 30% of patients, skin overlying affected bone is thickened and erythematous. [7] Histology of melorheostosis reveals dense, lamellar bone overgrowth along the periosteal surface that subsequently undergoes intense remodeling, whereas affected skin is characterized by increased blood vessel density and thickness. ...
... [7] Histology of melorheostosis reveals dense, lamellar bone overgrowth along the periosteal surface that subsequently undergoes intense remodeling, whereas affected skin is characterized by increased blood vessel density and thickness. [5,7] Although melorheostotic lesions do not metastasize, bone overgrowth continues throughout the patient's life. There is no known treatment. ...
... Most cases of classical melorheostosis are caused by somatic, mosaic mutations (Q56P, K57N, or K57E) clustered in the negative regulatory domain of MAP2K1. [5] These mutations result in a loss of the negative regulatory domain's inhibitory role in the encoded MEK1 protein. The resulting MEK1 protein variant exhibits kinase activity on its downstream targets, extracellular signal-regulated kinase 1 (ERK1) and ERK2, in the absence of upstream RAF proto-oncogene, serine/threonine-protein kinase (RAF1) signaling. ...
Article
Full-text available
Patients with classical melorheostosis exhibit exuberant bone overgrowth in the appendicular skeleton, resulting in pain and deformity with no known treatment. Most patients have somatic, mosaic mutations in MAP2K1 (encoding the MEK1 protein) in osteoblasts and overlying skin. As with most rare bone diseases, lack of affected tissue has limited the opportunity to understand how the mutation results in excess bone formation. The aim of this study was to create a cellular model to study melorheostosis. We obtained patient skin cells bearing the MAP2K1 mutation (affected cells), and along with isogenic control normal fibroblasts reprogrammed them using the Sendai virus method into induced pluripotent stem cells (iPSCs). Pluripotency was validated by marker staining and embryoid body formation. iPSCs were then differentiated to mesenchymal stem cells (iMSCs) and validated by flow cytometry. We confirmed retention of the MAP2K1 mutation in iMSCs with PCR and confirmed elevated MEK1 activity by immunofluorescence staining. Mutation‐bearing iMSCs showed significantly elevated VEGF secretion, proliferation and collagen I and IV secretion. iMSCs were then differentiated into osteoblasts, which showed increased mineralization at 21 days and increased VEGF secretion at 14 and 21 days of differentiation. Administration of VEGF to unaffected iMSCs during osteogenic differentiation was sufficient to increase mineralization. Blockade of VEGF by bevacizumab reduced mineralization in iMSC‐derived affected osteoblasts and in affected primary patient‐derived osteoblasts. These data indicate that patient‐derived induced pluripotent stem cells recreate the elevated MEK1 activity, increased mineralization, and increased proliferation seen in melorheostosis patients. The increased bone formation is driven, in part, by abundant VEGF secretion. Modifying the activity of VEGF (a known stimulator of osteoblastogenesis) represents a promising treatment pathway to explore. iPSCs may have wide applications to other rare bone diseases. This article is protected by copyright. All rights reserved.
... j pLM of the periosteal part of a melorheostotic lesion from an adult patient with somatic MAP2K1 mutation. This outer shell of the lesion consists of compact multi-layered lamellae oriented parallel to the periosteum and covers the zone with intensively remodeled osteonal-like structure and prominent osteoid accumulation due to the activating MAP2K1 mutation (shown and discussed in Kang et al. [207] and Fratzl-Zelman et al. [211]). Similar to i, the primary bone was formed through periosteal apposition leading here to focal osteosclerosis (in the case of LRP5-HBM to generalized osteosclerosis). ...
... Recent studies have identified somatic gain-of-function mutations in MAP2K1 and SMAD3 as the cause of this very rare sclerosing disorder (prevalence 1:100,000). Characteristics are the eccentric hyperostosis or focal cortical thickening of the periosteal and endosteal surfaces of the cortex of a single bone or several adjacent bones resulting in local pain, joint stiffness, and limb swelling [206][207][208][209][210]. Biopsy samples of the lesions have revealed that the hyperostotic tissue consists of two morphologically distinctive regions: a shell formed by periosteal bone apposition, consisting of dense multilayered lamellae oriented parallel to the bone surface, which covers a distinct zone of either highly remodeled osteonallike bone that is very porous and hypomineralized in the case of somatic MAP2K1 mutations or poorly remodeled, less porous, and higher mineralized in the case of SMAD3 [210,211]. ...
Article
Full-text available
Metabolic and genetic bone disorders affect not only bone mass but often also the bone material, including degree of mineralization, matrix organization, and lacunar porosity. The quality of juvenile bone is moreover highly influenced by skeletal growth. This review aims to provide a compact summary of the present knowledge on the complex interplay between bone modeling and remodeling during skeletal growth and to alert the reader to the complexity of bone tissue characteristics in children with bone disorders. We describe cellular events together with the characteristics of the different tissues and organic matrix organization (cartilage, woven and lamellar bone) occurring during linear growth. Subsequently, we present typical alterations thereof in disorders leading to over-mineralized bone matrix compared to those associated with low or normal mineral content based on bone biopsy studies. Growth spurts or growth retardation might amplify or mask disease-related alterations in bone material, which makes the interpretation of bone tissue findings in children complex and challenging.
... There have been several proposed mechanisms of melorheostosis that focus on somatic genetic defects of growth factors in bone formation and remodeling dysplasias [5][6][7][8]. The unpredictable rate and occurrence of somatic mutation also helps explain the sporadic incidence of the disease. ...
Article
Full-text available
Melorheostosis is a rare benign bone pathology involving bone dysplasia and hyperostosis. The disease can be recognized with a characteristic radiographic feature of radiopaque lesions dripping along a long bone’s diaphysis. The aberrant bone formation and development manifests mainly as pain, edema, and paresthesia of the affected limb. Severe cases may report limb deformity as well as limited range of motion. Until now, there have been approximately 300 cases reported about melorheostosis worldwide and its diverse clinical picture and age distribution. In Vietnam, there is only one known case of melorheostosis discovered incidentally via radiography. The scarcity of cases presents a challenge within the medical community in recognizing and diagnosing the condition, and a delayed diagnosis can lead to severe contracture and compromised limb motility. In this article, we reported an 82-year-old case of polyostotic melorheostosis with late onset and predominant edema, affecting the sternum, the ribs, and multiple bones of the right extremities and presented our clinical approach for a geriatric patient with chronic limb edema. Our case is distinctive in terms of anatomical location as well as the predominant 20-year non-pitting edema. A prompt diagnosis was made upon the classic dripping candle wax radiographic features emphasizing the role of plain X-ray in establishing the diagnosis without extraneous utilization of other modalities and invasive procedures. Exclusion of other causes of chronic edema such as lymphadenopathy, malignancy as well as parasitic infection is of clinical importance.
Article
O diagnóstico de displasias esqueléticas requer avaliação de aspectos clínicos, radiológicos, metabólicos/bioquímicos, funcionais e moleculares, além de correlação genótipo-fenótipo. Apesar de se tratar de doenças raras, são de extrema importância para os reumatologistas que devem estar atentos aos detalhes para a correta identificação e adequado manejo destes pacientes. Descrevemos, nesta breve revisão, características epidemiológicas, fisiopatológicas, clínicas, radiográficas e terapêuticas de melorreostose, paquidermoperiostose, osteocondrite vertebral (histiocitose de células de Langerhans) e fibrodisplasia ossificante progressiva. Unitermos: displasia esquelética; melorreostose; osteocondrite vertebral; histiocitose de células de Langerhans; paquidermoperiostose; fibrodisplasia ossificante progressiva.
Article
Full-text available
Transforming growth factor-βs (TGF-βs) and bone morphometric proteins (BMPs) belong to the TGF-β superfamily and perform essential functions during osteoblast and chondrocyte lineage commitment and differentiation, skeletal development, and homeostasis. TGF-βs and BMPs transduce signals through SMAD-dependent and -independent pathways; specifically, they recruit different receptor heterotetramers and R-Smad complexes, resulting in unique biological readouts. BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner. BMPs and TGF-β have opposite functions in articular cartilage homeostasis. Moreover, TGF-β has a specific role in maintaining the osteocyte network. The precise activation of BMP and TGF-β signaling requires regulatory machinery at multiple levels, including latency control in the matrix, extracellular antagonists, ubiquitination and phosphorylation in the cytoplasm, nucleus-cytoplasm transportation, and transcriptional co-regulation in the nuclei. This review weaves the background information with the latest advances in the signaling facilitated by TGF-βs and BMPs, and the advanced understanding of their diverse physiological functions and regulations. This review also summarizes the human diseases and mouse models associated with disordered TGF-β and BMP signaling. A more precise understanding of the BMP and TGF-β signaling could facilitate the development of bona fide clinical applications in treating bone and cartilage disorders.
Article
Aneurysmal bone cyst (ABC) is a rare and usually painful condition, representing about 1% of all bone tumors. A geographical lytic, expansile, and septated radiological pattern, with fluid–fluid levels on MRI, is classically displayed. ABC can be a primary bone lesion (70% of patients) or can arise in an underlying condition and is subsequently named “ABC-like changes” (30%). ABC-like changes are more frequently encountered in skeletal segments affected by chondroblastoma, fibrous dysplasia, giant cell tumor, osteoblastoma, non-ossifying fibroma, and osteosarcoma. In this article, we describe the first case of ABC-like changes developed in association with an ultra-rare sclerosing bone disease: melorheostosis. Melorheostosis is characterized by recognizable patterns on radiological studies with a pathological increased bone density and a cortical thickening within the periosteal or endosteal space, usually with a “dripping candle wax” appearance. More rarely, other different radiological patterns can be observed, such as “osteopatia striata-like,” “osteoma-like,” “myositis ossificans-like,” and mixed patterns. Pain and limb hypotrophy are the most common clinical manifestations. We report the case of a Caucasian male with a clinic-radiological diagnosis of melorheostosis (with epiphyseal osteopoikilosis) since the age of twelve. At the age of nineteen, he suffered from increased pain in the proximal right thigh, and the radiological control revealed an expansive septated lesion at the right proximal femoral bone. The diagnosis of ABC-like changes developed in melorheostosis was obtained after CT-guided bone biopsy and confirmed by open-incisional biopsy.
Article
The periosteum is a membrane that covers almost all bones in the body. It is a living structure but attracts little attention unless it reacts excessively. We highlight the important points in the anatomy, histology, and physiology of the periosteum, the stimuli and various aspects of periosteal reaction, and the main conditions underlying periosteal reaction.
Article
Case: A 22-year-old female patient was referred to the orthopaedic department for further examination after a radiopaque area was observed in the T6 vertebra in her chest radiograph. Computed Tomography (CT) showed a sclerotic mass with smooth borders, involving the entire body of the T6 vertebra, left posterior elements, posterior of the rib past the left zygapophyseal joint, and a "flowing candle wax" image toward the T7 vertebra. Spinal melorheostosis was considered radiologically in the patient, but malignancy could not be completely excluded. Thereupon, open biopsy was performed under general anesthesia. Conclusion: Spinal melorheostosis is a rare condition. Histological examination should be considered in cases where the diagnosis remains suspicious after clinical and radiological evaluations.
Article
Full-text available
Purpose of review: Melorheostosis is a rare sclerosing bone dysplasia that affects both cortical bone and adjacent soft tissue structures in a sclerotomal distribution. In this review, we describe the natural history, radiological features, proposed pathogenesis, and management options for this debilitating condition. Recent findings: Since its first description in 1922, about 400 cases of melorheostosis have been reported, either as single reports or in small case series. Melorheostosis affects the appendicular skeleton more commonly than the axial skeleton and usually presents with lower limb deformity. Diagnosis is based on a combination of clinical and radiological features that help differentiate this condition from other sclerosing bone dysplasias. LEM domain-containing protein 3 (LEMD3) gene mutations have been demonstrated in several familial cases, but these have been more strongly correlated with other hereditary dysplasias, such as osteopoikilosis, and are not thought to be the causative gene for melorheostosis. The exact etiology of classic sporadically occurring melorheostosis remains unknown, with possible causes being somatic LEMD3 mutations, somatic mutations in the bone morphogenetic protein/transforming growth factor-beta pathway, mutations in multiple genes, or other non-genetic causes. Management in recent years has involved nitrogen-containing bisphosphonates in addition to traditional orthopedic surgical approaches and physical therapy. Melorheostosis may present as mixed or atypical osseous involvement in addition to the classically described "dripping candle wax" appearance of hyperostosis. Some patients may have overlap with osteopoikilosis or Buschke-Ollendorff syndrome. In the future, better characterization of genetic and developmental factors predisposing to melorheostosis may lead to the development of targeted therapy for this condition, as well as for more commonly encountered skeletal abnormalities.
Article
Full-text available
Erdheim-Chester Disease (ECD) is a rare, potentially fatal, multi-organ myeloid neoplasm occurring mainly in adults. The diagnosis is established by clinical, radiologic, and histologic findings; ECD tumors contain foamy macrophages that are CD68+, CD163+, CD1a-, and frequently S100-. The purpose of this report is to describe the clinical and molecular variability of ECD. Sixty consecutive ECD patients (45 males, 15 females) were prospectively evaluated at the NIH Clinical Center between 2011 and 2015. Comprehensive imaging and laboratory studies were performed, and tissues were examined for BRAF V600E and MAPK pathway mutations. Mean age at first manifestations of ECD was 46 years; a diagnosis was established, on average, 4.2 years after initial presentation. Bone was the most common tissue affected, with osteosclerosis in 95% of patients. Other manifestations observed in one-third to two-thirds of patients include cardiac mass and periaortic involvement, diabetes insipidus, retro-orbital infiltration, retroperitoneal, lung, CNS, skin and xanthelasma, usually in combination. Methods of detection included imaging studies of various modalities. Mutation in BRAF V600E was detected in 51% of 57 biopsies. One patient had an ARAF D228V mutation, and one had an activating ALK fusion. Treatments included interferon alpha, imatinib, anakinra, cladribine, vemurafenib and dabrafenib with trametinib; eleven patients received no therapy. The diagnosis of ECD is elusive because of the rarity and varied presentations of the disorder. Identification of BRAF and other MAPK pathway mutations in biopsies improves ECD diagnosis, allows for development of targeted treatments, and demonstrates that ECD is a neoplastic disorder.
Article
Full-text available
Arteriovenous malformation (AVM) is a fast-flow, congenital vascular anomaly that may arise anywhere in the body. AVMs typically progress, causing destruction of surrounding tissue and, sometimes, cardiac overload. AVMs are difficult to control; they often re-expand after embolization or resection, and pharmacologic therapy is unavailable. We studied extracranial AVMs in order to identify their biological basis. We performed whole-exome sequencing (WES) and whole-genome sequencing (WGS) on AVM tissue from affected individuals. Endothelial cells were separated from non-endothelial cells by immune-affinity purification. We used droplet digital PCR (ddPCR) to confirm mutations found by WES and WGS, to determine whether mutant alleles were enriched in endothelial or non-endothelial cells, and to screen additional AVM specimens. In seven of ten specimens, WES and WGS detected and ddPCR confirmed somatic mutations in mitogen activated protein kinase kinase 1 (MAP2K1), the gene that encodes MAP-extracellular signal-regulated kinase 1 (MEK1). Mutant alleles were enriched in endothelial cells and were not present in blood or saliva. 9 of 15 additional AVM specimens contained mutant MAP2K1 alleles. Mutations were missense or small in-frame deletions that affect amino acid residues within or adjacent to the protein's negative regulatory domain. Several of these mutations have been found in cancers and shown to increase MEK1 activity. In summary, somatic mutations in MAP2K1 are a common cause of extracranial AVM. The likely mechanism is endothelial cell dysfunction due to increased MEK1 activity. MEK1 inhibitors, which are approved to treat several forms of cancer, are potential therapeutic agents for individuals with extracranial AVM.
Article
Full-text available
Germline mutations in Ras pathway components are associated with a large class of human developmental abnormalities, known as RASopathies, that are characterized by a range of structural and functional phenotypes, including cardiac defects and neurocognitive delays. Although it is generally believed that RASopathies are caused by altered levels of pathway activation, the signaling changes in developing tissues remain largely unknown. We used assays with spatiotemporal resolution in Drosophila melanogaster (fruit fly) and Danio rerio (zebrafish) to quantify signaling changes caused by mutations in MAP2K1 (encoding MEK), a core component of the Ras pathway that is mutated in both RASopathies and cancers in humans. Surprisingly, we discovered that intrinsically active MEK variants can both increase and reduce the levels of pathway activation in vivo. The sign of the effect depends on cellular context, implying that some of the emerging phenotypes in RASopathies may be caused by increased, as well as attenuated, levels of Ras signaling.
Article
Full-text available
Background Romosozumab, a monoclonal antibody that binds sclerostin, increases bone formation and decreases bone resorption. Methods We enrolled 7180 postmenopausal women who had a T score of –2.5 to –3.5 at the total hip or femoral neck. Patients were randomly assigned to receive subcutaneous injections of romosozumab (at a dose of 210 mg) or placebo monthly for 12 months; thereafter, patients in each group received denosumab for 12 months, at a dose of 60 mg, administered subcutaneously every 6 months. The coprimary end points were the cumulative incidences of new vertebral fractures at 12 months and 24 months. Secondary end points included clinical (a composite of nonvertebral and symptomatic vertebral) and nonvertebral fractures. Results At 12 months, new vertebral fractures had occurred in 16 of 3321 patients (0.5%) in the romosozumab group, as compared with 59 of 3322 (1.8%) in the placebo group (representing a 73% lower risk with romosozumab; P<0.001). Clinical fractures had occurred in 58 of 3589 patients (1.6%) in the romosozumab group, as compared with 90 of 3591 (2.5%) in the placebo group (a 36% lower risk with romosozumab; P=0.008). Nonvertebral fractures had occurred in 56 of 3589 patients (1.6%) in the romosozumab group and in 75 of 3591 (2.1%) in the placebo group (P=0.10). At 24 months, the rates of vertebral fractures were significantly lower in the romosozumab group than in the placebo group after each group made the transition to denosumab (0.6% [21 of 3325 patients] in the romosozumab group vs. 2.5% [84 of 3327] in the placebo group, a 75% lower risk with romosozumab; P<0.001). Adverse events, including instances of hyperostosis, cardiovascular events, osteoarthritis, and cancer, appeared to be balanced between the groups. One atypical femoral fracture and two cases of osteonecrosis of the jaw were observed in the romosozumab group. Conclusions In postmenopausal women with osteoporosis, romosozumab was associated with a lower risk of vertebral fracture than placebo at 12 months and, after the transition to denosumab, at 24 months. The lower risk of clinical fracture that was seen with romosozumab was evident at 1 year. (Funded by Amgen and UCB Pharma; FRAME ClinicalTrials.gov number, NCT01575834.)
Article
Full-text available
Large-scale reference data sets of human genetic variation are critical for the medical and functional interpretation of DNA sequence changes. Here we describe the aggregation and analysis of high-quality exome (protein-coding region) DNA sequence data for 60,706 individuals of diverse ancestries generated as part of the Exome Aggregation Consortium (ExAC). This catalogue of human genetic diversity contains an average of one variant every eight bases of the exome, and provides direct evidence for the presence of widespread mutational recurrence. We have used this catalogue to calculate objective metrics of pathogenicity for sequence variants, and to identify genes subject to strong selection against various classes of mutation; identifying 3,230 genes with near-complete depletion of predicted protein-truncating variants, with 72% of these genes having no currently established human disease phenotype. Finally, we demonstrate that these data can be used for the efficient filtering of candidate disease-causing variants, and for the discovery of human 'knockout' variants in protein-coding genes.
Article
Full-text available
The role of the ERK signalling pathway in cancer is thought to be most prominent in tumours in which mutations in the receptor tyrosine kinases RAS, BRAF, CRAF, MEK1 or MEK2 drive growth factor-independent ERK1 and ERK2 activation and thence inappropriate cell proliferation and survival. New drugs that inhibit RAF or MEK1 and MEK2 have recently been approved or are currently undergoing late-stage clinical evaluation. In this Review, we consider the ERK pathway, focusing particularly on the role of MEK1 and MEK2, the 'gatekeepers' of ERK1/2 activity. We discuss their validation as drug targets, the merits of targeting MEK1 and MEK2 versus BRAF and the mechanisms of action of different inhibitors of MEK1 and MEK2. We also consider how some of the systems-level properties (intrapathway regulatory loops and wider signalling network connections) of the ERK pathway present a challenge for the success of MEK1 and MEK2 inhibitors, discuss mechanisms of resistance to these inhibitors, and review their clinical progress.
Article
Melorheostosis (MEL) is the rare sporadic dysostosis characterized by monostotic or polyostotic osteosclerosis and hyperostosis often distributed in a sclerotomal pattern. The prevailing hypothesis for MEL invokes postzygotic mosaicism. Sometimes scleroderma-like skin changes, considered a representation of the pathogenetic process of MEL, overlie the bony changes, and sometimes MEL becomes malignant. Osteopoikilosis (OPK) is the autosomal dominant skeletal dysplasia that features symmetrically distributed punctate osteosclerosis due to heterozygous loss-of-function mutation within LEMD3. Rarely, radiographic findings of MEL occur in OPK. However, germline mutation of LEMD3 does not explain sporadic MEL. To explore if mosaicism underlies MEL, we studied a boy with polyostotic MEL and characteristic overlying scleroderma-like skin, a few bony lesions consistent with OPK, and a large epidermal nevus known to usually harbor a HRAS, FGFR3, or PIK3CA gene mutation. Exome sequencing was performed to ~ 100 × average read depth for his two dermatoses, two areas of normal skin, and peripheral blood leukocytes. As expected for non-malignant tissues, the patient's mutation burden in his normal skin and leukocytes was low. He, his mother, and his maternal grandfather carried a heterozygous, germline, in-frame, 24-base-pair deletion in LEMD3. Radiographs of the patient and his mother revealed bony foci consistent with OPK, but she showed no MEL. For the patient, somatic variant analysis, using four algorithms to compare all 20 possible pairwise combinations of his five DNA samples, identified only one high-confidence mutation, heterozygous KRAS Q61H (NM_033360.3:c.183A > C, NP_203524.1:p.Gln61His), in both his dermatoses but absent in his normal skin and blood. Thus, sparing our patient biopsy of his MEL bone, we identified a heterozygous somatic KRAS mutation in his scleroderma-like dermatosis considered a surrogate for MEL. This implicates postzygotic mosaicism of mutated KRAS, perhaps facilitated by germline LEMD3 haploinsufficiency, causing his MEL.
Article
The purpose of the nosology is to serve as a “master” list of the genetic disorders of the skeleton to facilitate diagnosis and to help delineate variant or newly recognized conditions. This is the 9th edition of the nosology and in comparison with its predecessor there are fewer conditions but many new genes. In previous editions, diagnoses that were phenotypically indistinguishable but genetically heterogenous were listed separately but we felt this was an unnecessary distinction. Thus the overall number of disorders has decreased from 456 to 436 but the number of groups has increased to 42 and the number of genes to 364. The nosology may become increasingly important today and tomorrow in the era of big data when the question for the geneticist is often whether a mutation identified by next generation sequencing technology in a particular gene can explain the clinical and radiological phenotype of their patient. This can be particularly difficult to answer conclusively in the prenatal setting. Personalized medicine emphasizes the importance of tailoring diagnosis and therapy to the individual but for our patients with rare skeletal disorders, the importance of tapping into a resource where genetic data can be centralized and made available should not be forgotten or underestimated. The nosology can also serve as a reference for the creation of locus-specific databases that are expected to help in delineating genotype– phenotype correlations and to harbor the information that will be gained by combining clinical observations and next generation sequencing results.