ArticlePDF Available

Type V OI Primary Osteoblasts Display Increased Mineralization Despite Decreased COL1A1 Expression

Authors:

Abstract and Figures

Context: Patients with type V osteogenesis imperfecta (OI) are heterozygous for a dominant IFITM5 c.-14C>T mutation, which adds five residues to the N terminus of bone-restricted interferon-induced transmembrane-like protein (BRIL), a transmembrane protein expressed in osteoblasts. Type V OI skeletal findings include hyperplastic callus formation, ossification of the forearm interosseous membrane, and dense metaphyseal bands. Objective: The objective of this study was to examine the role of osteoblasts in the active mineralization traits of type V OI and the effect of the IFITM5 mutation on type I collagen. Methods: We identified eight patients with the IFITM5 c.-14C>T mutation. Cultured osteoblasts from type V OI patients were used to study osteoblast differentiation and mineralization. Results: We verified the expression and stability of mutant IFITM5 transcripts. In differentiated type V OI primary osteoblasts in culture, the IFITM5 expression and BRIL level is comparable with control. Both early and late markers of osteoblast differentiation are increased in type V OI osteoblasts. Mineralization, assayed by alizarin red staining, was increased in type V OI osteoblasts compared with control. However, type V OI osteoblasts have significantly decreased COL1A1 transcripts in mid- to late differentiation. Type I collagen protein is concomitantly decreased, with decreased cross-linked collagen in matrix and altered appearance of fibrils deposited in culture. Conclusions: This study demonstrates that type V OI mineralization has a gain-of-function mechanism at the osteoblast level, which likely underlies the overactive tissue mineralization seen in patients. Decreased type I collagen expression, secretion, and matrix incorporation establish type V OI as a collagen-related defect.
Content may be subject to copyright.
Type V OI primary osteoblasts display increased
mineralization despite decreased COL1A1 expression
Adi Reich
1
, Alison S. Bae
1
, Aileen M. Barnes
1
, Wayne A. Cabral
1
,
Aleksander Hinek
2
, Jennifer Stimec
3
, Suvimol C. Hill
4
, David Chitayat
5,6
, and
Joan C. Marini
1
1Bone and Extracellular Matrix Branch, NICHD, NIH, Bethesda, MD, USA;
2
Physiology and Experimental
Medicine Program, Heart Center, Hospital for Sick Children, University of Toronto, ON, Canada;
3
Division of Diagnostic Imaging, Department of Pediatrics, Hospital for Sick Children, University of
Toronto, Toronto, ON, Canada;
4
Diagnostic Radiology Department, NIH Clinical Center, NIH, Bethesda,
MD, USA;
5
The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and
Gynecology, Mt Sinai Hospital, University of Toronto, Toronto, ON, Canada;
6
Division of Clinical and
Metabolic Genetics, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto,
ON, Canada
Context: Patients with type V osteogenesis imperfecta (OI) are heterozygous for a dominant IFITM5
c.-14C!T mutation, which adds 5 residues to the N-terminus of BRIL, a transmembrane protein
expressed in osteoblasts. Type V OI skeletal findings include hyperplastic callus formation, ossifi-
cation of the forearm interosseous membrane and dense metaphyseal bands.
Objective: The objective of this study was to examine the role of osteoblasts in the active miner-
alization traits of type V OI, and the effect of the IFITM5 mutation on type I collagen.
Methods: We identified 8 patients with the IFITM5 c.-14C!T mutation. Cultured osteoblasts from
type V OI patients were used to study osteoblast differentiation and mineralization.
Results: We verified expression and stability of mutant IFITM5 transcripts. In differentiated type V
OI primary osteoblasts in culture, IFITM5 expression and BRIL protein level is comparable to control,
Both early and late markers of osteoblast differentiation are increased in type V OI osteoblasts.
Mineralization, assayed by alizarin red staining, was increased in type V OI osteoblasts compared
to control. However, type V OI osteoblasts have significantly decreased COL1A1 transcripts in mid
to late differentiation. Type I collagen protein is concomitantly decreased, with decreased cross-
linked collagen in matrix, and altered appearance of fibrils deposited in culture.
Conclusions: This study demonstrates that type V OI mineralization has a gain-of-function mech-
anism at the osteoblast level, which likely underlies the overactive tissue mineralization seen in
patients. Decreased type I collagen expression, secretion and matrix incorporation establish type
V OI as a collagen-related defect.
Osteogenesis imperfecta (OI) is a genetically heteroge-
neous heritable connective tissue disorder charac-
terized by intrinsic bone fragility, resulting in frequent
fractures and deformities of the long bones and spine (1).
The phenotypic severity of OI ranges from perinatal lethal
to subtle fracture susceptibility and generalized osteope-
nia (2). Most cases of osteogenesis imperfecta are caused
by autosomal dominant mutations in the genes encoding
type I collagen, COL1A1 or COL1A2 (3). Autosomal re-
cessive osteogenesis imperfecta, with lethal to moderate
phenotypes, is caused by defects in genes whose products
interact with type I collagen for folding, post-translation
modification or processing (4). Most recessive cases have
null mutations in genes whose proteins are involved in
ISSN Print 0021-972X ISSN Online 1945-7197
Printed in U.S.A.
Copyright © 2014 by the Endocrine Society
Received July 31, 2014. Accepted October 31, 2014.
Abbreviations:
ORIGINAL ARTICLE
Endocrine Research
doi: 10.1210/jc.2014-3082 J Clin Endocrinol Metab jcem.endojournals.org 1
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
collagen prolyl 3-hydroxylation (CRTAP,LEPRE1 and
PPIB) (5–7) or proper helical folding (FKBP10 and SER-
PINH1) (8–11). Additional genes causing recessive OI
include SERPINF1, which encodes pigment epithelium-
derived factor (PEDF) (12, 13), collagen C-propeptide
cleavage enzyme (BMP1) (14), and WNT1 (15, 16).
Type V OI is the only dominantly inherited form of OI
not caused by mutations in COL1A1 or COL1A2 (4). It
has distinctive clinical and radiographic features, includ-
ing variable occurrence of hyperplastic callus, calcifica-
tion of the forearm interosseous membrane, radial-head
dislocation, and a subphyseal metaphyseal radiodense
band (17, 18). The histomorphometry of type V OI is also
distinct, with a mesh-like lamellation pattern (17). Recent
reports have shown that type V OI is caused by a single
recurrent heterozygous mutation in Interferon-Induced
Transmembrane Protein 5 (IFITM5), which encodes
Bone-restricted (IFITM)-like protein (BRIL), a protein in-
volved in mineralization and expressed in the skeleton (19,
20). The type V OI mutation is a C!T transition at posi-
tion –14 of the 5"untranslated region (UTR) of IFITM5
(c.-14C!T) (21, 22). This mutation generates a new start
codon, adding five amino acids to the N-terminus of the
protein. However, the mutant protein maintains the same
intramembrane topology and palmitoylation as normal
BRIL (23), and has been speculated to have a gain-of-
function mechanism. The understanding that the same
mutation is responsible for all cases of type V OI then led
to recognition of phenotypic variability (24). We describe
here 8 patients with type V OI from 5 different families,
who display phenotypic variability.
The mechanism of type V OI is not understood at the
cellular level, in this case, the bone forming osteoblasts.
Using primary cultured osteoblasts from type V OI pa-
tients, we verified the expression of mutant IFITM5 tran-
script and protein. We utilized these primary mutant os-
teoblasts to demonstrate that the presence of the IFITM5
mutation increases multiple markers of osteoblast differ-
entiation. Type V OI osteoblasts also display increased
mineral deposition in culture, demonstrating that a gain-
of-function mechanism at the cellular level underlies the
active mineralization traits in type V OI patients. Despite
increased osteoblast developmental markers, COL1A1
expression, secretion and deposition in matrix by type V
OI osteoblasts is significantly decreased, which establishes
type V OI as a collagen-related dysplasia.
Materials and Methods
Patients and Cells
Patients 2, 3, 5, and 8 are patients at the NIH Clinical Center,
whose samples were collected under an IRB approved protocol.
Osteoblasts were outgrowths from bone chips collected as sur-
gical discard during medically indicated orthopedic procedures.
patient 7 is followed at the Genetic Clinic at Mt Sinai Hospital,
Toronto, Ontario, Canada; samples were collected with parental
consent. Detailed case reports on the patients are presented in
Supplemental Material. Control fibroblasts were ATCC line
2127, which is well-validated for collagen biochemistry, while
control osteoblasts were obtained with parental consent from
surgical discard bone chips of an unaffected child during an elec-
tive orthopedic procedure. These control cells have been used
extensively in our lab and validated for experiments involving
osteoblast development and collagen biochemistry.
Mutation identification and verification
We screened genomic DNA (gDNA) of dermal fibroblasts,
leukocytes, or both, from the control, patients, parents or unaf-
fected siblings. Sequencing of complementary DNA (cDNA) and
gDNA from the patients revealed no mutations in COL1A1,
COL1A2,CRTAP,LEPRE1,PPIB,SERPINH1,FKBP10 or
SERPINF1. All patient skin and bone biopsies were obtained
with informed consent under a protocol approved by the
NICHD IRB.
The two exons and flanking intronic sequences of IFITM5
gDNA from leukocytes of control, patients, sibling and parents
were amplified by PCR, as previously described (25) and se-
quenced. Patient and control IFITM5 cDNA from fibroblasts
was also sequenced and the mutation was confirmed by BsmAI
restriction enzyme digestion.
Cell culture
Dermal fibroblast (FB) cultures were established from skin
punch biopsies. FB were grown in Dulbecco’s Modified Eagle
Medium (DMEM) (Life Technologies, Grand Island, NY) con-
taining 10% fetal bovine serum (FBS), 100 U/ml penicillin and
100
!
g/ml streptomycin. Osteoblast (OB) primary cultures were
established from surgical bone chips (26) of normal control and
OI patients who had not received bisphosphonates. OB were
cultured in MEM alpha (Life Technologies) supplemented with
10% FBS, penicillin and streptomycin at 37°C and 8% CO
2
.
Osteoblast cultures were grown to confluence, then treated with
osteoblast differentiation medium (containing 25
!
g/ml L-ascor-
bic acid, 10
-8
M Dexamethasone and 2.5 mM 2-glycerophos-
phate, with or without 100 ng/ml human recombinant BMP2
(the generous gift of Wyeth, Dallas, TX)).
RT-PCR
Total RNA was extracted from control and patient primary
osteoblast and fibroblast cultures using TriReagent (Molecular
Research Center, Cincinnati, OH). cDNA was reverse tran-
scribed from 5
!
g RNA using a High-Capacity cDNA Archive
Kit or MuLV reverse transcriptase and Oligo d(T)
16
(Life Tech-
nologies, Grand Island, NY). Transcript levels of IFITM5
(Hs00942485 g1), RUNX2 (Hs00231692 m1), COL1A1
(Hs00164004 m1), ALPL (Hs01029144 m1), BGLAP
(Hs01587814 g1), SPP1 (Hs00959010 m1), SERPINF1
(Hs01106934 m1), and IBSP (Hs00173720 m1) (Life Tech-
nologies) were determined with TaqMan Gene Expression As-
says on a 7500 Fast Real-Time PCR System (Applied Biosys-
tems). Relative expression of each gene of interest was performed
in triplicate and normalized to expression of GAPDH
(Hs99999905 m1). Expression levels were compared with age-
2Increased mineralization of type V OI Osteoblasts J Clin Endocrinol Metab
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
matched control osteoblasts (Day 1 or 5). Expression of IFITM5
in osteoblasts and fibroblasts by PCR (as shown in Figure S1C)
was demonstrated using primers in exon 1 (F: 5!-TTGATCTG-
GTCGGTGTTCAG-3!) and exon 2 (5!-GTCAGTCATAGTC-
CGCGTCA-3!) as previously described (21) and generated the
expected 292 bp product.
Western Blot
Cell lysates were collected in RIPA buffer supplemented with
a protease inhibitor cocktail (
!
–Aldrich, St. Louis, MO). Osteo-
blast conditioned media was collected after incubation for 24
hours in serum-free media and supplemented with protease in-
hibitors. Media was concentrated using centrifugal filters (EMD
Millipore, Billerica, MA) and normalized to cell counts for equal
loading.
Proteins were separated on precast 4%–15% Ready Gels
(Bio-Rad, Hercules, CA), transferred to 0.2
"
m nitrocellulose
membranes, and blocked with 5% bovine serum albumin (BSA)
plus 1"casein in PBS before probing with antibody overnight in
2.5% BSA and 0.5"casein. Antibodies used were as follows:
IFITM5 (Abcam, Cambridge, MA), COL1A1 LF-68 (the gener-
ous gift of Dr. Larry W. Fisher, NIH), and actin (
!
–Aldrich).
Blots were washed, incubated with secondary IR-conjugated an-
tibodies for 1 hour, washed, and visualized on a LI-COR Odys-
sey infrared imager (LI-COR, Lincoln, NE).
Mineralization Assay
Osteoblast cultures were grown to confluence in 12-well cul-
ture dishes and stimulated with mineralization medium contain-
ing 25
"
g/ml L-ascorbic acid, 10
-8
M Dexamethasone and 2.5
mM 2-glycerophosphate. Mineralization of the nodules was de-
termined by Alizarin Red-S staining. Cells were washed with PBS
and fixed in 4% paraformaldehyde in PBS for 1 hour, washed
with water and incubated in 1% Alizarin Red-S in 2% ethanol,
followed by a final water wash. Alizarin Red was solubilized with
0.5 ml of 0.5N HCl/5% SDS and absorbance was measured with
spectrophotometer at 405nm (27).
Steady-state Collagen
Control and patient fibroblasts or osteoblasts were grown to
confluence in 6-well culture dishes. Steady-state type I collagen
analysis was conducted as previously described (28). Cells were
incubated with 437.5
"
Ci/ml L-[2,3,4,5-
3
H] proline for 16–18
hours, prior to collection and ammonium sulfate precipitation,
then pepsin-digested and analyzed by 6% SDS-urea-PAGE.
Matrix Deposition
Postconfluent osteoblasts were stimulated with 100
"
g/ml
ascorbic acid three times a week for 14 days then labeled for 24
hours with 400
"
Ci/ml L-[2,3,4,5-
3
H] proline. The media and
extracellular matrix were harvested as described previously (29).
The
3
H-proline-labeled collagens were sequentially extracted
from the matrix fraction with neutral salt, acetic acid and pepsin,
then electrophoresed on 6% SDS-urea-PAGE. Collagen content
of each fraction was measured by densitometry and normalized
to total sample volume.
Immunofluorescence Microscopy
Fibroblasts and osteoblasts were grown to confluence on
chamber slides, then treated with 200
"
M ascorbic acid every
day for 5 days and fixed in 4% paraformaldehyde. Staining of
extracellular matrix was performed essentially as described (30).
The matrix was blocked in 1% BSA in PBS plus 0.02% Tween-
20, washed with PBS and incubated with primary antibody (LF-
68,
#
1(I) C-telopeptide) in 1% BSA/PBS. After washing, the ma-
trix was incubated with secondary antibody, then washed slides
were mounted with DAPI (Vector Laboratories, Burlingame,
CA), and imaged using a Zeiss LSM 510 Inverted Meta micro-
scope and LSM510 software.
Statistical analysis
The data were analyzed by Excel using repeated-measures T
test. Values are presented as mean #SD unless noted. Signifi-
cance was achieved at P$.05.
Results
Identification of Type V OI patients and
phenotypic variability
We identified 8 affected individuals from 5 families
who are heterozygous for the IFITM5 c.-14C$T mutation
found in all individuals with type V OI. Seven patients
were classified clinically as type V OI prior to sequencing,
while patient 8, an adult with progressive deforming OI
not previously classified as type V OI, was found to have
the same mutation. For each patient, the mutation was
confirmed by BsmAI restriction digest; the mutation elim-
inates a BsmAI restriction site (Figure S1C). Sequencing of
patients and family members, as well as patients’ Case
Reports are detailed in the Supplemental material (Figure
S1A, B; Table 1, Table S1).
Type V OI is understood to have variability of clinical
manifestations and of the timing of their appearance (17,
18). Patients 2, 3 and 5 (Figure S3–5) have characteristic
bone histology and radiographic findings. Patients 1, 4, 6
Table 1. Patients Characteristics
Patient Age Sex
Hypertrophic
callus
Radial head
dislocation
Forearm interosseous
membrane calcification Sclera Scoliosis
Age at first
fracture Height (50th centile for age)
Age of diagnosis
of OI type V
1Deceased at 44 yr M %Unknown Unknown Normal Yes Unknown 7-year-old Post mortem
227 yr F %%% Blue Mild in utero, 7 months 6.5-year-old 11-years-old
326 yr M %%% Normal Yes birth 12.5-year-old 10-years-old
450 yr F %Unknown Unknown Blue Yes Unknown 3.5-year-old 33-years-old
522 yr M %%% Blue Mild in utero, birth 2.5-year-old 6-years-old
615 yr M %Unknown Unknown Blue No 1 month 4.5 months at age 30 months before 3 months
75 yr M %%% Blue Yes in utero At CA 3.8 HA 19 months 5-years-old
8Deceased at 73 yr F - %Unknown Normal Yes 18 months 3-year-old Post mortem
doi: 10.1210/jc.2014-3082 jcem.endojournals.org 3
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
and 7 (Figure S6) have typical radiographic findings, but
bone histology was not examined. The patients in all five
families developed at least one of the classic characteristics
of type V OI. Patients 1, 2, 3, 5, 6 and 7 developed hy-
pertrophic callus. Radial head dislocation was noted in
patients 2, 3, 5, 7 and 8, and ossification of the forearm
interrosseous membrane was documented in patients 2, 5
and 7. Although the initial clinical description of type V OI
reported white sclerae (17, 18), patients 2, 5, 6 and 7 have
blue sclerae (Table 1, Table S1).
IFITM5 expression in osteoblasts and fibroblasts
IFITM5 transcripts were detected by RT-PCR in both
fibroblasts and osteoblasts from normal controls, with the
expected higher expression in osteoblasts than fibroblasts
(Figure S1D). IFITM5 transcripts were also detected in
cultured cells from patients 3 and 7 (Figure S1D). Sanger
sequencing was used to confirm expression of mutant IF-
ITM5 transcripts containing the 5!-terminal addition.
These transcripts could be detected in total RNA from
untreated cells, indicating they have normal stability (Fig-
ure S1E).
In osteoblasts from type V OI patients and control,
IFITM5 expression was barely detectable by real-time RT-
PCR throughout differentiation
with osteogenic media, but addition
of BMP2 significantly and compara-
bly increased IFITM5 expression in
both control and type V osteoblasts
(Figure 1A, left). In differentiated os-
teoblasts, BRIL protein level was
generally comparable in type V OI
and control cells (Figure 1A, right).
IFITM5 mutation affects
osteoblast markers and
mineralization
Multiple markers of osteoblast
differentiation have significantly in-
creased expression in type V OI os-
teoblasts from patient 3 during a dif-
ferentiation timecourse (Figure 2A).
While the preosteoblast marker
RUNX2 had equivalent expression
in mutant and control osteoblasts,
early osteoblast marker alkaline
phosphatase (ALPL) expression was
3 to 6-fold greater than control
throughout differentiation, and the
mid-differentiation marker bone sia-
loprotein (IBSP) (31) peaked on Day
12 at "3-fold greater than control.
Osteocalcin (BGLAP/OCN) and os-
teopontin (SPP1/OPN) are markers of late osteoblast mat-
uration (31). Their peak expression in type V osteoblasts
is about 4-fold greater than control and came late in the
timecourse (Day 15), with BGLAP/OCN expression ris-
ing over time while SPP1/OPN had a late sharp peak. The
addition of BMP2 to maintain IFITM5 expression during
osteoblast differentiation increased transcript levels of dif-
ferentiation markers but maintained their relative level in
type V and control osteoblasts (Figure 2B). Type V OI
osteoblast ALPL, BGLAP and SERPINF1 levels re-
mained greater than control on Day 10 in BMP2 treated
cells.
This acceleration of osteoblast markers was coordi-
nated with increased osteoblast mineralization. Mineral-
ization of differentiating osteoblasts was measured by aliz-
arin red staining. Type V OI osteoblasts displayed a more
brisk mineralization response than did control, increasing
earlier in differentiation and remaining elevated through
Day 15 (Figure 1B).
IFITM5 mutation affects type I collagen expression,
secretion and matrix incorporation
Expression of COL1A1 in type V OI osteoblasts from
2 typical patients (patients 3 and 5), with hyperplastic
Figure 1. IFITM5 expression and osteoblast mineralization A) Left, Relative IFITM5 expression in
control (C) and patient 3 (OI V) osteoblasts (OB) stimulated with osteogenic media for 10 days
with or without addition of BMP2. patient 3 shows normal IFITM5 expression levels by qPCR (n #
3). Right, Representative BRIL western blot from OB stimulated with osteogenic media showing
protein stability in patient (n #3). B) In vitro mineralization assays of control (C) and patient 3
(OI V) OB assessed by alizarin red staining shows increased mineralization of OI V OB at earlier
time points than control (n #3). *P$.05
4Increased mineralization of type V OI Osteoblasts J Clin Endocrinol Metab
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
callus, calcification of the forearm interosseous mem-
brane, and radial-head dislocation, peaked at Day 5, and
was significantly (2–3 fold) lower than control during mid
to late differentiation (Figure 2A; Figure S2A). The relative
decrease in COL1A1 expression on day 10 was partially
rescued by addition of BMP2 but was still significantly
lower than control (Figure 2B). Electrophoretic migration
of steady-state collagen synthesized by osteoblasts of a
type V OI patient (patient 3) with a typical phenotype
(Figure 3C) is normal. Fibroblast collagen from several,
but not all, type V patients displayed reproducible migra-
tion anomalies which did not correlate with phenotype
(Figure S2B).
Patient collagen protein secreted during osteoblast dif-
ferentiation reflected transcript levels and was reduced
compared to control (Figure 3A). Secreted collagen pro-
tein was quantified as total secreted
!
1(I) chain, by com-
bining measurements of all detected
!
1(I) forms at each
timepoint. Interestingly, collagen from type V osteoblasts
appears to be processed more rapidly in the pericellular
space than does collagen from control osteoblasts, with
relatively more proband mature
!
1(I), or partially pro-
cessed pC
!
1(I), than pro
!
1(I). We do not currently un-
derstand the mechanism of this difference.
Type V OI cultured osteoblasts deposit substantially
less (!4-fold) collagen into matrix fractions containing
crosslinks, with notable reduction in crosslinked
"
-forms
(Figure 3B). Matrix deposited by Type V OI fibroblasts
(Figure 3D, left) and osteoblasts (Figure 3D, right) in long-
term culture was stained for collagen. Type V OI matrix
had fewer well-delineated fibrils and more amorphous
ground material. Collagen fibril packing in patient matrix
was patchy compared to control, with parallel fibril bun-
dles rather than the network appearance of control.
Figure 2. Expression of osteoblast differentiation markers A) Relative expression of Runt-related transcription factor 2 (RUNX2), Alkaline
Phosphatase (ALPL), Bone sialoprotein (IBSP), type I collagen
!
1(COL1A1), Osteopontin (SPP1/OPN) and Osteocalcin (BGLAP/OCN) in control (C)
and patient 3 (OI V) OB stimulated with osteogenic media for up to 15 days. patient 3 shows increased expression of ALPL, IBSP, OPN, and OCN
genes throughout the timecourse (n "3). *P#.05 B) Relative expression of RUNX2,ALPL,COL1A1,SERPINF1 and OCN, in control (C) and
patient 3 (OI V) OB on Day 10 of stimulation with osteogenic media with or without addition of BMP2. patient 3 shows significantly increased
expression of ALPL,SERPINF1 and OCN $BMP2, however, COL1A1 expression $BMP2 is significantly decreased.
doi: 10.1210/jc.2014-3082 jcem.endojournals.org 5
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
Discussion
Type V OI is an autosomal dominant form of OI (1, 17).
It is the only rare type of OI which shares the inheritance
pattern of classical OI types I-IV, which are caused by
mutations in the genes that code for type I collagen,
COL1A1 and COL1A2 (1, 2, 4). Type V OI was first
described with clinical criteria over a decade ago, based on
distinctive radiographic and bone histology features (17).
In 2012, two teams of investigators reported that a unique
heterozygous mutation in IFITM5 (21, 22), which en-
codes the protein BRIL, was responsible for all cases of
type V OI (24, 32, 33). BRIL is a transmembrane protein
expressed predominantly in osteoblasts (19, 20), where it
has a Type II orientation in the plasma membrane with the
N-terminal end in the cytoplasm (23). BRIL is also teth-
ered to the membrane by S-palmitoylation modifications
at Cys52 and Cys53, which insert into the cytoplasmic side
of the membrane (23, 34). In the osteoblast membrane, the
association of BRIL with FKBP11 is promoted by S-pal-
mitoylation of the cysteine residues and leads to a larger
complex containing CD81 and CD9 (34).
Two distinct heterozygous IFITM5 mutations have
been reported in OI patients: 1) the c.-14C!T mutation at
the 5"-end of IFITM5 which generates a new start codon,
adding 5 residues to the N-terminal end of BRIL but leav-
ing membrane insertion intact (21, 22): 2) a missense mu-
tation c.119C!T resulting in an p.S40L substitution (25,
35, 36) that interferes with palmitoylation and membrane
insertion (23). All cases of type V OI are caused by the
5"-end mutation, while the p.S40L substitution causes an
atypical progressive deforming OI with the typical bone
histology of type VI OI (25).
The IFITM5 mutation causing type V OI was quickly
speculated to have a gain-of-function mechanism because
of its N-terminal location and the hyperactive mineraliza-
tion traits of the type V OI phenotype (22, 23). Additional
indirect support for this interpretation was provided by
functional studies of normal BRIL, in which adenovirus-
mediated BRIL overexpression in UMR106 cells stimu-
lated mineralization and lentivirus-mediated Ifitm5
shRNA knockdown in MC3T3 cells inhibited mineraliza-
tion (19). Furthermore, comparison of the two IFITM5
mutations in primary patient osteoblasts in studies focus-
ing not on mineralization, but on pigment epithelium de-
Figure 3. Type I Collagen secretion and deposition A) Left, Representative western blot of type I procollagen secretion from control (C) or
patient 3 (OI V) OB stimulated with osteogenic media for up to 15 days, or Right, for 10 days with BMP2 treatment. patient 3 has decreased
collagen secretion and was equal with BMP2 treatment. B) Extracellular collagen matrix deposited by control (C) or patient 3 (OI V) OB. Normalized
densitometry of control and OI V
!
1(I) chains after balanced loading revealed a 3.5-fold decrease in mature cross-linked collagen, as well as a
decrease in crosslinked
"
-forms in acetic acid (AA) and pepsin (P) fractions. C) Steady-state type I collagen protein in osteoblasts from control (C)
and patient 3 (OI V). Migration of the
!
1(I) and
!
2(I) chains is normal in the patient. D) Immunofluorescence microscopy of collagen matrix
deposited in culture by control (C) and patient 3 (OI V) fibroblasts and osteoblasts. Type V OI matrix has patchy, parallel fibers in comparison to the
networked, well-formed fibers in control.
6Increased mineralization of type V OI Osteoblasts J Clin Endocrinol Metab
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
rived factor (PEDF), also implicated a gain-of-function
mechanism for the IFITM5 mutation causing type V OI
(25). We demonstrated that the type V OI mutation had
complimentarity with the BRIL p.S40L substitution func-
tion, in that the BRIL p.S40L substitution decreased ex-
pression of PEDF by an unknown mechanism, while pri-
mary osteoblasts from type V OI patients have increased
PEDF expression and secretion (25), again supporting a
putative gain-of-function mechanism in type V OI. The
increased mineralization of type V OI has not been dem-
onstrated directly at the cellular level of primary patient
osteoblasts, nor has a relationship of the type V OI phe-
notype to type I collagen been demonstrated.
We report here 8 additional patients with type V OI and
the first studies of type V OI primary osteoblasts focused
on mineralization and differentiation. We confirmed ex-
pression of the mutant allele in patient osteoblasts and
fibroblasts, and the stability of the mutant transcript. Pa-
tients in all 5 families developed some of the clinical fea-
tures of type V OI, but there was considerable variation in
the features manifested and in their timing of appearance.
Even the elderly adult with severe OI, who died prior to
delineation of her OI type and mutation, had a history of
radial head dislocation. This clinical variability among
individuals heterozygous for the IFITM5 c.-14C!T mu-
tation has also been noted by others (24, 32).
IFITM5 transcripts were below the level of detectibility
by real-time RT-PCR in primary human osteoblasts, while
BRIL protein was detectable by Western blot at a compa-
rable level in type V and in control. Treatment with BMP2
to stimulate the osteogenic differentiation increased IF-
ITM5 transcripts in Type V OI and control osteoblasts to
a comparable level, and did not alter the relative levels of
transcripts for differentiation markers, providing reassur-
ance that BMP2 treatment had not fundamentally altered
type V OI osteoblast differentiation. In primary type V OI
osteoblasts, multiple markers of bone differentiation were
increased, including early (alkaline phosphatase), mid
(bone sialoprotein) and late (osteocalcin and osteopontin)
markers, consistent with a generalized acceleration in type
V bone cell differentiation.
Furthermore, the type V primary osteoblast studies
support a mechanism for type V OI which is specifically
collagen-related. The type V OI IFITM5 mutation is
shown here to cause decreased type I collagen expression
and secretion. Given the general increase in other osteo-
blast maturation markers, the decrease in type I collagen
expression stands out as reflecting a specific pathway
which is altered in the opposite direction to the generalized
increase in cell maturation. Consequent to the decreased
collagen secretion, Type V OI osteoblasts have decreased
collagen deposition into matrix and an altered deposition
pattern when fibrils are compared to control by fluores-
cent microscopy. This data supports the collagen-related
character of BRIL defects. The full pathway for BRIL func-
tioning in bone cells is an active area of investigation.
Finally, we demonstrated increased mineral deposition
by primary type V osteoblasts in culture. These data in-
dicate that increased osteoblast mineralization activity is
the underlying cause of the hyperactive mineralization
seen in patient traits such as hyperplastic callus and dense
metaphyseal bands. The studies with type V OI primary
osteoblasts presented here provide direct confirmation for
a cell-based gain-of-function mechanism, and support the
collagen-related character of BRIL defects.
In summary, these data demonstrate a gain-of-function
mechanism for the BRIL mutation causing type V OI at the
cellular level. In combination with the dominant inheri-
tance of type V OI, this data indicates that a BRIL protein
replacement strategy for treating type V OI is unlikely to
be successful. Instead, targeted therapy will need to focus
on the intracellular signaling pathways and partners of
BRIL to interfere with the mechanisms activating cell dif-
ferentiation and mineralization. Delineation of these path-
ways and their connection to PEDF (25) is under
investigation.
Acknowledgments
We thank the patients and their families for their dedicated long
term support of OI research. We thank the NICHD Microscopy
and Imaging Core in which the confocal microscopy was con-
ducted. This work was supported by NICHD intramural funding
(JCM). Authors’ roles: Study design: AR, ASB, AMB, and JCM.
Data collection: AR, ASB, AMB, WAC, AH, DC. Data analysis
and interpretation: AR, ASB, AMB, WAC, AH, DC, JC, SCH
and JCM. Drafting manuscript: JCM, AR, ASB. Revising man-
uscript content: AR, ASB, AMB, WAC, AH, DC, and JCM. Ap-
proving final version of manuscript: AR, ASB, AMB, WAC, AH,
DC, JC, SCH and JCM. JCM takes responsibility for the integrity
of the data analysis.
Address all correspondence and requests for reprints to: Com-
municating author: Joan C. Marini, MD, PhD, Chief, Bone and
Extracellular Matrix Branch, Building 10; Rm 10D39, 9000
Rockville Pike, Bethesda, MD 20 892, (T) 301–594-3418, (F)
301–480-3188, (e) oidoc@helix.nih.gov.
Disclosures Statement: The authors have nothing to disclose.
This work was supported by .
References
1. Marini JC, Blissett AR. New Genes in Bone Development: What’s
New in Osteogenesis Imperfecta. J Clin Endocrinol Metab. 2013;
98(8):3095–3103.
doi: 10.1210/jc.2014-3082 jcem.endojournals.org 7
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
2. Forlino A, Cabral WA, Barnes AM, Marini JC. New perspectives on
osteogenesis imperfecta. Nat Rev Endocrinol. 2011;7(9):540–557.
3. Marini JC, Forlino A, Cabral WA, Barnes AM, San Antonio JD,
Milgrom S, Hyland JC, Korkko J, Prockop DJ, De Paepe A, Coucke
P, Symoens S, Glorieux FH, Roughley PJ, Lund AM, Kuurila-Svahn
K, Hartikka H, Cohn DH, Krakow D, Mottes M, Schwarze U, Chen
D, Yang K, Kuslich C, Troendle J, Dalgleish R, Byers PH. Consor-
tium for osteogenesis imperfecta mutations in the helical domain of
type I collagen: regions rich in lethal mutations align with collagen
binding sites for integrins and proteoglycans. Hum Mutat. 2007;
28(3):209–221.
4. Marini JC, Reich A, Smith SM. Osteogenesis imperfecta due to mu-
tations in non-collagenous genes: lessons in the biology of bone
formation. Current opinion in pediatrics. 2014;26(4):500–507.
5. Barnes AM, Carter EM, Cabral WA, Weis M, Chang W, Makareeva
E, Leikin S, Rotimi CN, Eyre DR, Raggio CL, Marini JC. Lack of
cyclophilin B in osteogenesis imperfecta with normal collagen fold-
ing. N Engl J Med. 2010;362(6):521–528.
6. Barnes AM, Chang W, Morello R, Cabral WA, Weis M, Eyre DR,
Leikin S, Makareeva E, Kuznetsova N, Uveges TE, Ashok A, Flor
AW, Mulvihill JJ, Wilson PL, Sundaram UT, Lee B, Marini JC.
Deficiency of cartilage-associated protein in recessive lethal osteo-
genesis imperfecta. N Engl J Med. 2006;355(26):2757–2764.
7. Cabral WA, Chang W, Barnes AM, Weis M, Scott MA, Leikin S,
Makareeva E, Kuznetsova NV, Rosenbaum KN, Tifft CJ, Bulas DI,
Kozma C, Smith PA, Eyre DR, Marini JC. Prolyl 3-hydroxylase 1
deficiency causes a recessive metabolic bone disorder resembling
lethal/severe osteogenesis imperfecta. Nat Genet. 2007;39(3):359
365.
8. Barnes AM, Cabral WA, Weis M, Makareeva E, Mertz EL, Leikin
S, Eyre D, Trujillo C, Marini JC. Absence of FKBP10 in recessive
type XI osteogenesis imperfecta leads to diminished collagen cross-
linking and reduced collagen deposition in extracellular matrix.
Hum Mutat. 2012;33(11):1589–1598.
9. Christiansen HE, Schwarze U, Pyott SM, AlSwaid A, Al Balwi M,
Alrasheed S, Pepin MG, Weis MA, Eyre DR, Byers PH. Homozy-
gosity for a missense mutation in SERPINH1, which encodes the
collagen chaperone protein HSP47, results in severe recessive osteo-
genesis imperfecta. Am J Hum Genet. 2010;86(3):389–398.
10. Alanay Y, Avaygan H, Camacho N, Utine GE, Boduroglu K, Aktas
D, Alikasifoglu M, Tuncbilek E, Orhan D, Bakar FT, Zabel B,
Superti-Furga A, Bruckner-Tuderman L, Curry CJ, Pyott S, Byers
PH, Eyre DR, Baldridge D, Lee B, Merrill AE, Davis EC, Cohn DH,
Akarsu N, Krakow D. Mutations in the gene encoding the RER
protein FKBP65 cause autosomal-recessive osteogenesis imperfecta.
Am J Hum Genet. 2010;86(4):551–559.
11. Drogemuller C, Becker D, Brunner A, Haase B, Kircher P, Seeliger
F, Fehr M, Baumann U, Lindblad-Toh K, Leeb T. A missense mu-
tation in the SERPINH1 gene in Dachshunds with osteogenesis im-
perfecta. PLoS Genet. 2009;5(7):e1000579.
12. Becker J, Semler O, Gilissen C, Li Y, Bolz HJ, Giunta C, Bergmann
C, Rohrbach M, Koerber F, Zimmermann K, de Vries P, Wirth B,
Schoenau E, Wollnik B, Veltman JA, Hoischen A, Netzer C. Exome
sequencing identifies truncating mutations in human SERPINF1 in
autosomal-recessive osteogenesis imperfecta. Am J Hum Genet.
2011;88(3):362–371.
13. Homan EP, Rauch F, Grafe I, Lietman C, Doll JA, Dawson B, Bertin
T, Napierala D, Morello R, Gibbs R, White L, Miki R, Cohn DH,
Crawford S, Travers R, Glorieux FH, Lee B. Mutations in SER-
PINF1 cause osteogenesis imperfecta type VI. J Bone Miner Res.
2011;26(12):2798–2803.
14. Martinez-Glez V, Valencia M, Caparros-Martin JA, Aglan M,
Temtamy S, Tenorio J, Pulido V, Lindert U, Rohrbach M, Eyre D,
Giunta C, Lapunzina P, Ruiz-Perez VL. Identification of a mutation
causing deficient BMP1/mTLD proteolytic activity in autosomal re-
cessive osteogenesis imperfecta. Hum Mutat. 2012;33(2):343–350.
15. Keupp K, Beleggia F, Kayserili H, Barnes AM, Steiner M, Semler O,
Fischer B, Yigit G, Janda CY, Becker J, Breer S, Altunoglu U, Grun-
hagen J, Krawitz P, Hecht J, Schinke T, Makareeva E, Lausch E,
Cankaya T, Caparros-Martin JA, Lapunzina P, Temtamy S, Aglan
M, Zabel B, Eysel P, Koerber F, Leikin S, Garcia KC, Netzer C,
Schonau E, Ruiz-Perez VL, Mundlos S, Amling M, Kornak U,
Marini J, Wollnik B. Mutations in WNT1 cause different forms of
bone fragility. Am J Hum Genet. 2013;92(4):565–574.
16. Laine CM, Joeng KS, Campeau PM, Kiviranta R, Tarkkonen K,
Grover M, Lu JT, Pekkinen M, Wessman M, Heino TJ, Nieminen-
Pihala V, Aronen M, Laine T, Kroger H, Cole WG, Lehesjoki AE,
Nevarez L, Krakow D, Curry CJ, Cohn DH, Gibbs RA, Lee BH,
Makitie O. WNT1 mutations in early-onset osteoporosis and os-
teogenesis imperfecta. N Engl J Med. 2013;368(19):1809–1816.
17. Glorieux FH, Rauch F, Plotkin H, Ward L, Travers R, Roughley P,
Lalic L, Glorieux DF, Fassier F, Bishop NJ. Type V osteogenesis
imperfecta: a new form of brittle bone disease. J Bone Miner Res.
2000;15(9):1650–1658.
18. Cheung MS, Glorieux FH, Rauch F. Natural history of hyperplastic
callus formation in osteogenesis imperfecta type V. J Bone Miner
Res. 2007;22(8):1181–1186.
19. Moffatt P, Gaumond MH, Salois P, Sellin K, Bessette MC, Godin E,
de Oliveira PT, Atkins GJ, Nanci A, Thomas G. Bril: a novel bone-
specific modulator of mineralization. J Bone Miner Res. 2008;23(9):
1497–1508.
20. Hanagata N, Li X, Morita H, Takemura T, Li J, Minowa T. Char-
acterization of the osteoblast-specific transmembrane protein IF-
ITM5 and analysis of IFITM5-deficient mice. J Bone Miner Metab.
2011;29(3):279–290.
21. Cho TJ, Lee KE, Lee SK, Song SJ, Kim KJ, Jeon D, Lee G, Kim HN,
Lee HR, Eom HH, Lee ZH, Kim OH, Park WY, Park SS, Ikegawa
S, Yoo WJ, Choi IH, Kim JW. A single recurrent mutation in the
5!-UTR of IFITM5 causes osteogenesis imperfecta type V. Am J
Hum Genet. 2012;91(2):343–348.
22. Semler O, Garbes L, Keupp K, Swan D, Zimmermann K, Becker J,
Iden S, Wirth B, Eysel P, Koerber F, Schoenau E, Bohlander SK,
Wollnik B, Netzer C. A mutation in the 5!-UTR of IFITM5 creates
an in-frame start codon and causes autosomal-dominant osteogen-
esis imperfecta type V with hyperplastic callus. Am J Hum Genet.
2012;91(2):349–357.
23. Patoine A, Gaumond MH, Jaiswal PK, Fassier F, Rauch F, Moffatt
P. Topological Mapping of BRIL Reveals a Type II Orientation and
Effects of Osteogenesis Imperfecta Mutations on Its Cellular Des-
tination. J Bone Miner Res. 2014:10.1002/jbmr. 2243.
24. Rauch F, Moffatt P, Cheung M, Roughley P, Lalic L, Lund AM,
Ramirez N, Fahiminiya S, Majewski J, Glorieux FH. Osteogenesis
imperfecta type V: marked phenotypic variability despite the pres-
ence of the IFITM5 c.-14C"T mutation in all patients. J Med Genet.
2013;50(1):21–24.
25. Farber CR, Reich A, Barnes AM, Becerra P, Rauch F, Cabral WA,
Bae A, Quinlan A, Glorieux FH, Clemens TL, Marini JC. A novel
IFITM5 mutation in severe atypical osteogenesis imperfecta type VI
impairs osteoblast production of pigment epithelium-derived factor.
J Bone Miner Res. 2014;29(6):1402–1411.
26. Robey PG, Termine JD. Human bone cells in vitro. Calcif Tissue Int.
1985;37(5):453–460.
27. Gregory CA, Gunn WG, Peister A, Prockop DJ. An Alizarin red-
based assay of mineralization by adherent cells in culture: compar-
ison with cetylpyridinium chloride extraction. Analytical biochem-
istry. 2004;329(1):77–84.
28. Cabral WA, Makareeva E, Colige A, Letocha AD, Ty JM, Yeowell
HN, Pals G, Leikin S, Marini JC. Mutations near amino end of
alpha1(I) collagen cause combined osteogenesis imperfecta/Ehlers-
Danlos syndrome by interference with N-propeptide processing.
J Biol Chem. 2005;280(19):19259–19269.
29. Bateman JF, Golub SB. Deposition and selective degradation of
structurally-abnormal type I collagen in a collagen matrix produced
by osteogenesis imperfecta fibroblasts in vitro. Matrix biology : jour-
nal of the International Society for Matrix Biology. 1994;14(3):
251–262.
8Increased mineralization of type V OI Osteoblasts J Clin Endocrinol Metab
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
30. Venturi G, Gandini A, Monti E, Dalle Carbonare L, Corradi M,
Vincenzi M, Valenti MT, Valli M, Pelilli E, Boner A, Mottes M,
Antoniazzi F. Lack of expression of SERPINF1, the gene coding for
pigment epithelium-derived factor, causes progressively deforming
osteogenesis imperfecta with normal type I collagen. J Bone Miner
Res. 2012;27(3):723–728.
31. Miron RJ, Zhang YF. Osteoinduction: a review of old concepts with
new standards. Journal of dental research. 2012;91(8):736–744.
32. Balasubramanian M, Parker MJ, Dalton A, Giunta C, Lindert U,
Peres LC, Wagner BE, Arundel P, Offiah A, Bishop NJ. Genotype-
phenotype study in type V osteogenesis imperfecta. Clin Dysmor-
phol. 2013.
33. Shapiro JR, Lietman C, Grover M, Lu JT, Nagamani SC, Dawson
BC, Baldridge DM, Bainbridge MN, Cohn DH, Blazo M, Roberts
TT, Brennen FS, Wu Y, Gibbs RA, Melvin P, Campeau PM, Lee BH.
Phenotypic variability of osteogenesis imperfecta type V caused by
an IFITM5 mutation. J Bone Miner Res. 2013.
34. Tsukamoto T, Li X, Morita H, Minowa T, Aizawa T, Hanagata N,
Demura M. Role of S-Palmitoylation on IFITM5 for the Interaction
with FKBP11 in Osteoblast Cells. PLoS One. 2013;8(9):e75831.
35. Guillen-Navarro E, Ballesta-Martinez MJ, Valencia M, Bueno AM,
Martinez-Glez V, Lopez-Gonzalez V, Burnyte B, Utkus A, Lapun-
zina P, Ruiz-Perez VL. Two mutations in IFITM5 causing distinct
forms of osteogenesis imperfecta. Am J Med Genet A. 2014;164(5):
1136–1142.
36. Hoyer-Kuhn H, Semler O, Garbes L, Zimmermann K, Becker J,
Wollnik B, Schoenau E, Netzer C. A Non-Classical IFITM5 Muta-
tion Located in the Coding Region Causes Severe Osteogenesis Im-
perfecta with Prenatal Onset. J Bone Miner Res. 2013:10.1002/
jbmr. 2156.
doi: 10.1210/jc.2014-3082 jcem.endojournals.org 9
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 18 November 2014. at 10:00 For personal use only. No other uses without permission. . All rights reserved.
... The patients that bear this mutation are classified as atypical OI type VI, given the bone histology that resembles the one found in patients with OI type VI. 6 Recently, a third variant, c.143A>G (p.N48S), was described in the IFITM5 gene, leading to a phenotype different from the one seen in type V, with fish-scale pattern in lamellar bone, osteoporosis, and calvarial doughnut lesions. 7 The disorders caused by variants in the IFITM5 gene are rare, with the most common (c.-14C> ...
... T) variant in the IFITM5 gene. However, two other variants have been published to be related to OI in this same gene: the c.119C> T (p.S40L) 6 and, the recently described c.143A>G (p.N48S). 7 The latter was reported in a mother and her daughter with a novel form of OI named osteoporosis with calvarial doughnut lesions (OP-CDL) that present with skeletal fragility, multiple fractures (especially long bones), scoliosis, bone deformities, and sclerotic skull lesions; a phenotype different from those arising from the classical (c.-14C> T) variant. ...
Article
Full-text available
Osteogenesis imperfecta (OI) is a group of genetic skeletal disorders, with a prevalence of 1 in 15,000–20,000 births. OI type V has been described in approximately 150 cases and all patients carry the variant (c.-14C> T) in the IFITM5 gene. However, two other variants, p.S40L and p.N48S have been reported in this gene, leading to clinical phenotypes different from OI type V. Here we described a patient with multiple bone fractures, scoliosis, skull alteration (plagiocephaly), bone deformation, bone rickets, and intramedullary epithelioid osteosarcoma that bears the recently reported heterozygous variant c.143A>G (p.N48S) in the IFITM5 gene. This case supports the pathogenicity of this new variant in the IFITM5 gene and adds information regarding its clinical phenotype.
... (25) Intriguingly, our previous studies found that IFITM5 (c.119C>T; p.S40L) results in decreased SERPINF1 transcript expression, whereas the type V OI IFITM5 mutation (c.-14C>T) increased SERPINF1 transcript expression and PEDF protein secretion. (26) These results suggest a potential positive feedback relationship between SERPINF1 (PEDF) and IFITM5 (BRIL) in bone formation. We generated CRISPR clones with IFITM5 (c.119C>T, p.Ser40Leu) mutation, a cellular model of atypical type VI OI. ...
... The same decrease in type I collagen transcripts and protein was found in osteoblasts with atypical type VI OI when examined on day 10 of differentiation. (25,26) Since type I collagen expression was also shown to increase upon stimulation with recombinant PEDF, (46) it is not unexpected that Serpinf1 (À/À) murine calvarial osteoblasts have reduced Col1a1 expression at days 7, 14, and 21, although not day 0, of differentiation. Furthermore, Serpinf1 (À/À) undifferentiated osteoblasts had decreased Col1a1 expression versus wild type after treatment with either BMP2 or TGF-β. ...
Article
Full-text available
Osteogenesis imperfecta (OI) is a heterogeneous genetic disorder of bone and connective tissue, also known as brittle bone disease. Null mutations in SERPINF1, which encodes pigment epithelium‐derived factor (PEDF), cause severe type VI OI, characterized by accumulation of unmineralized osteoid and a fish‐scale pattern of bone lamellae. Although the potent anti‐angiogenic activity of PEDF has been extensively studied, the disease mechanism of type VI OI is not well‐understood. Using Serpinf1(‐/‐) mice and primary osteoblasts, we demonstrate that loss of PEDF delays osteoblast maturation as well as extracellular matrix (ECM) mineralization. Barium sulfate perfusion reveals significantly increased vessel density in the tibial periosteum of Serpinf1(‐/‐) mouse compared to wild‐type littermates. The increased bone vascularization in Serpinf1(‐/‐) mice correlated with increased number of CD31(+)/Endomucin(+) endothelial cells, which are involved in the coupling angiogenesis and osteogenesis. Global transcriptome analysis by RNA‐Seq of Serpinf1(‐/‐) mouse osteoblasts reveals osteogenesis and angiogenesis as the biological processes most impacted by loss of PEDF. Intriguingly, TGF‐β signaling is activated in type VI OI cells, and Serpinf1(‐/‐) osteoblasts are more sensitive to TGF‐β stimulation than wild‐type osteoblasts. TGF‐β stimulation and PEDF deficiency showed additive effects on transcription suppression of osteogenic markers and stimulation of pro‐angiogenic factors. Furthermore, PEDF attenuated TGF‐β‐induced expression of pro‐angiogenic factors. These data suggest that functional antagonism between PEDF and TGF‐β pathways controls osteogenesis and bone vascularization and is implicated in type VI OI pathogenesis. This antagonism may be exploited in developing therapeutics for type VI OI utilizing PEDF and TGF‐β antibody. This article is protected by copyright. All rights reserved.
... OI type V is estimated to be the second most prevalent form of OI [12], with about 200 cases reported worldwide [4,5,[13][14][15]. OI type V is caused by the single recurrent mutation (c.-14C>T) in the 5' untranslated region of BRIL, leading to the creation of an inframe ATG with the addition of 5 amino acids (MALEP) at the N-terminus of BRIL [15,16]. ...
... Although highly variable, the clinical manifestations in OI type V patients are associated with low bone density, fractures, spinal deformities, radial head dislocation, hyperplastic callus formation, and paradoxical expanded periosteal bone formation that can be accompanied by interosseous membrane calcification [4,5,[13][14][15]17]. Although very well described from the clinical perspective, the onset and progression of those events in OI type V are much less well understood at the molecular level. ...
Article
Full-text available
BRIL (bone restricted ifitm-like; also known as IFITM5) is a transmembrane protein expressed in osteoblasts. Although its role in skeletal development and homeostasis is unknown, mutations in BRIL result in rare dominant forms of osteogenesis imperfecta. The pathogenic mechanism has been proposed to be a gain-of or neomorphic function. To understand the function of BRIL and its OI type V mutant (MALEP BRIL) and whether they could activate signaling pathways in osteoblasts, we performed a luciferase reporter assay screen based on the activity of 26 transcription factors. When overexpressed in MC3T3-E1 and MLO-A5 cells, the MALEP BRIL activated the reporters dependent on MEF2, NFATc, and NR4A significantly more. Additional co-transfection experiments with MEF2C and NFATc1 and a number of their modulators (HDAC4, calcineurin, RCAN, FK506) confirmed the additive or synergistic activation of the pathways by MALEP, and suggested a coordinated regulation involving calcineurin. Endogenous levels of Nr4a members, as well as Ptgs2, were upregulated by MALEP BRIL. Y2H and co-immunoprecipitation indicated that BRIL interacted with CAML, but its contribution as the most upstream stimulator of the Ca2+-calcineurin-MEF2/NFATc cascade was not confirmed convincingly. Altogether the data presented provide the first ever readout to monitor for BRIL activity and suggest a potential gain-of-function causative effect for MALEP BRIL in OI type V, leading to perturbed signaling events and gene expression.
... IFITM5 is expressed specifically in the initial stage of osteoblast mineralization, and overexpression of IFITM5 in osteoblasts promotes mineralization. Therefore, some studies speculated that HPC could be related to excessive mineralization induced by mutation in IFITM5 [19,20]. A study found that the differentiation and mineralization of osteoblasts were increased in OI type V [19]. ...
... Therefore, some studies speculated that HPC could be related to excessive mineralization induced by mutation in IFITM5 [19,20]. A study found that the differentiation and mineralization of osteoblasts were increased in OI type V [19]. Heterotopic ossification, HPC formation, and calcification of the interosseous membranes may be induced by the ossification potential of extra-skeletal tissue in patients with OI type V, and IFITM5 mutation seemed to promote the ectopic ossification and HPC formation. ...
Article
Full-text available
Hyperplastic callus (HPC) is the most conspicuous features of osteogenesis imperfecta (OI) type V, of which accurate diagnosis and treatment are facing challenges. We investigate the clinical features, and impact factors of HPC in OI type V patients. In this retrospective single-center study, a total of 21 patients with type V OI confirmed by IFITM5 mutation were included. Radiological characteristics of bone were evaluated by X-rays, dual-energy X-ray absorptiometry, and computed tomography scan. Bone biopsy specimens were performed and stained by routine hematoxylin–eosin. The effects of bisphosphonates on HPC were investigated. Eleven patients (52.3%) had HPCs at 19 skeletal sites, 11 of which affected the femur. Three patients developed four (21.1%) HPCs after fractures, and 15 (78.9%) HPCs occurred in absence of bone fracture. The progress of HPCs was variable, of which most HPCs enlarged in the initial phase and remained stable, and only one HPC dwindled in size. One patient had a rapidly growing mass on the right humerus, and biopsy showed irregular trabeculae of woven bone and immature bone and cartilage in the loose and edematous collagenous network without signs of tumor. Bisphosphonates treatment had no significant effects on HPC of OI patients. HPC is the specific characteristic of OI type V patients, and its location, shape, size, and progression are variable, and the femur is the most frequently involved site. It is very important to make a diagnosis of HPC through detecting IFITM5 mutation and completing pathological diagnosis if necessary. The treatment of HPC is worth further exploration.
... The elongated BRIL protein is stable in cells but does not display changes in topology, palmitoylation, or membrane localization, all providing additional evidence that the type V OI mutation is gain of function (68). Osteoblasts from patient bone samples cultured in vitro exhibit increased levels of transcripts for osteoblast maturation markers such as osteopontin (secreted phosphoprotein 1), alkaline phosphatase (ALPL), and integrin-binding sialoprotein (IBSP), suggestive of alteration of middle stages of differentiation (69). These data are in agreement with elevation of patient serum alkaline phosphatase levels during active hyperplastic callus formation (66), and chronically in some patients (70). ...
... collagen expression during osteoblast differentiation (69,81). A murine model for atypical type VI OI was recently generated with an Ifitm5 S42L mutation. ...
Article
Osteogenesis imperfecta (OI) is a phenotypically and genetically heterogeneous skeletal dysplasia characterized by bone fragility, growth deficiency and skeletal deformity. Previously known to be caused by defects in type I collagen, the major protein of extracellular matrix, it is now also understood to be a collagen-related disorder caused by defects in collagen folding, post-translational modification and processing, bone mineralization and osteoblast differentiation, with inheritance of OI types spanning autosomal dominant and recessive as well as X-linked recessive. This review provides the latest updates on OI, encompassing both classical OI and rare forms, their mechanism and the signaling pathways involved in their pathophysiology. There is a special emphasis on mutations in type I procollagen C-propeptide structure and processing, the later causing OI with strikingly high bone mass. Types V and VI OI, while notably different, are shown to be interrelated by the IFITM5 p.S40L mutation that reveals the connection between the BRIL and PEDF pathways. The function of Regulated Intramembrane Proteolysis has been extended beyond cholesterol metabolism to bone formation by defects in RIP components S2P and OASIS. Several recently proposed candidate genes for new types of OI are also presented. Discoveries of new OI genes add complexity to already-challenging OI management; current and potential approaches are summarized.
... Our results were in line with various studies that reported a greater ALP expression at 14 days post-culture than 7 days post-culture throughout osteoblast differentiation. The typical trend that is reported in these studies showed that the ALP started to be expressed at 7 days and it reached a peak at 14 days and decreased at 21 days [23][24][25]. Thus, as expected, osteocalcin is secreted when the expression of ALP reaches the highest level to maximize the mineralization of the extracellular matrix (ECM). ...
Article
Full-text available
The use of biomaterial for tissue repair involves the interaction between materials and cells, and the coagulum formation represents the first step of tissue healing. This process is particularly critical in the oral cavity, where the wounds are immediately subjected to the masticatory mechanical stress, saliva invasion, and bacterial attack. Therefore, the present study aimed to explore the structural features and the biological activities of a hemostatic collagen sponge on human gingival fibroblasts (HGFs) and human oral osteoblasts (HOBs). The microstructure of the collagen sponge was characterized by a scanning electron microscope (SEM) and histological analysis. The porosity was also calculated. To investigate biological activities, HGFs and HOBs were cultured on the collagen sponges, and their adhesion was observed at SEM on the third day, while cell viability was investigated at the third and seventh days by Tetrazolium (MTT) assay. For osteoblasts seeded on collagen sponge the mineralization ability was also evaluated by alkaline phosphatase (ALP) assay at the seventh day, and by Alizarin red staining on the 14th. Furthermore, the gene expression of ALP and osteocalcin (OCN) was investigated after 3, 7 and 14 days. SEM images of the sponge without cells showed a highly porous 3D structure, confirmed by the measurement of porosity that was more than 90%. The samples cultured were characterized by cells uniformly distributed and adhered to the sponge surface. Proliferation ended up being promoted, as well as the mineralization ability of the osteoblasts, mainly at the mature stage. In conclusion, this collagen sponge could have a potential use for tissue healing.
... . Whereas, IBSP is an osteoblast differentiation marker, which produces is considered to be whose increased expression indicates late stage of differentiation of osteoblasts [19,20]. This indicated that BD contains both early and late bone cells and is viable. ...
Article
Full-text available
Aims: To substantiate the osteogenic differentiation of human Umbilical cord Mesenchymal Stem Cells (hUMSCs) under the influence of Bone Dust (BD) by detecting expression of genes RUNX2 and IBSP in the differentiated cells. Study Design: In vitro study. Place and Duration of Study: MDRL Cell Culture Lab Ziauddin University and Dr.Zafar H.Zaidi Institute of Proteomics, University of Karachi, between July 2020 and July 2021. Methodology: We designed an innovative experiment to differentiate hUMSCs into osteoblasts under the influence of BD, without using any conventional differentiating medium. The hUMSCs were isolated from umbilical cord and cultured in a 6-well plate with 3μm inserts, in which BD was lodged, preventing direct contact of BD with hUMSCs. The plates with hUMSCs and BD-filled inserts were incubated at 37°C for 14 days after which RNA extraction, cDNA synthesis and qPCR for two genes was performed. Expression of Runt-related transcription factor-2(RUNX2) and Integrin-binding Sialoprotein (IBSP) were quantified. Results: Results revealed an increased expression of the late osteogenic marker IBSP(p=0.01) signifying that the hUMSCs had undergone differentiation into mature osteoblasts. Conclusion: This novel experiment identifies BD as a new source for osteogenic differentiation of hUMSCs in in vitro experiments.
Article
Introduction Osteogenesis imperfecta (OI) is a heterogenous group of heritable connective tissue disorders characterized by high bone fragility due to low bone mass and impaired bone material properties. Atypical type VI OI is an extremely rare and severe form of bone dysplasia resulting from a loss-of-function mutation (p.S40L) in IFITM5/BRIL, the causative gene of OI type V and decreased osteoblast secretion of pigment epithelium-derived factor (PEDF), as in OI type VI. It is not yet known which alterations at the material level might lead to such a severe phenotype. We therefore characterized bone tissue at the micrometer level in a novel heterozygous Ifitm5/BRIL p.S42L knock-in murine model at 4 and 8 weeks of age. Methods We evaluated in female mice, total body size, femoral and lumbar bone mineral density (BMD) by dual-energy X-ray absorptiometry. In the femoral bone we examined osteoid deposition by light microscopy, assessed bone histomorphometry and mineralization density distribution by quantitative backscattered electron imaging (qBEI). Osteocyte lacunae were examined by qBEI and the osteocyte lacuno-canalicular network by confocal laser scanning microscopy Vasculature was examined indirectly by qBEI as 2D porosity in cortex, and as 3D porosity by micro-CT in third trochanter. Collagen orientation was examined by second harmonic generation microscopy. Two-way ANOVA was used to discriminate the effect of age and genotype. Results Ifitm5/BRIL p.S42L female mice are viable, do not differ in body size, fat and lean mass from wild type (WT) littermates but have lower whole-body, lumbar and femoral BMD and multiple fractures. The average and most frequent calcium concentration, CaMean and CaPeak, increased with age in metaphyseal and cortical bone in both genotypes and were always higher in Ifitm5/BRIL p.S42L than in WT, except CaMean in metaphysis at 4 weeks of age. The fraction of highly mineralized bone area, CaHigh, was also increased in Ifitm5/BRIL p.S42L metaphyseal bone at 8 weeks of age and at both ages in cortical bone. The fraction of lowly mineralized bone area, CaLow, decreased with age and was not higher in Ifitm5/BRIL p.S42L, consistent with lack of hyperosteoidosis on histological sections by visual exam. Osteocyte lacunae density was higher in Ifitm5/BRIL p.S42L than WT, whereas canalicular density was decreased. Indirect measurements of vascularity revealed a higher pore density at 4 weeks in cortical bone of Ifitm5/BRIL p.S42L than in WT and at both ages in the third trochanter. Importantly, the proportion of bone area with disordered collagen fibrils was highly increased in Ifitm5/BRIL p.S42L at both ages. Conclusions Despite normal skeletal growth and the lack of a collagen gene mutation, the Ifitm5/BRIL p.S42L mouse shows major OI-related bone tissue alterations such as hypermineralization of the matrix and elevated osteocyte porosity. Together with the disordered lacuno-canalicular network and the disordered collagen fibril orientation, these abnormalities likely contribute to overall bone fragility.
Article
Full-text available
Recently, one of the interferon-induced transmembrane (IFITM) family proteins, IFITM3, has become an important target for the activity against influenza A (H1N1) virus infection. In this protein, a post-translational modification by fatty acids covalently attached to cysteine, termed S-palmitoylation, plays a crucial role for the antiviral activity. IFITM3 possesses three cysteine residues for the S-palmitoylation in the first transmembrane (TM1) domain and in the cytoplasmic (CP) loop. Because these cysteines are well conserved in the mammalian IFITM family proteins, the S-palmitoylation on these cysteines is significant for their functions. IFITM5 is another IFITM family protein and interacts with the FK506-binding protein 11 (FKBP11) to form a higher-order complex in osteoblast cells, which induces the expression of immunologically relevant genes. In this study, we investigated the role played by S-palmitoylation of IFITM5 in its interaction with FKBP11 in the cells, because this interaction is a key process for the gene expression. Our investigations using an established reporter, 17-octadecynoic acid (17-ODYA), and an inhibitor for the S-palmitoylation, 2-bromopalmitic acid (2BP), revealed that IFITM5 was S-palmitoylated in addition to IFITM3. Specifically, we found that cysteine residues in the TM1 domain and in the CP loop were S-palmitoylated in IFITM5. Then, we revealed by immunoprecipitation and western blot analyses that the interaction of IFITM5 with FKBP11 was inhibited in the presence of 2BP. The mutant lacking the S-palmitoylation site in the TM1 domain lost the interaction with FKBP11. These results indicate that the S-palmitoylation on IFITM5 promotes the interaction with FKBP11. Finally, we investigated bone nodule formation in osteoblast cells in the presence of 2BP, because IFITM5 was originally identified as a bone formation factor. The experiment resulted in a morphological aberration of the bone nodule. This also indicated that the S-palmitoylation contributes to bone formation.
Article
Full-text available
This report identifies human skeletal diseases associated with mutations in WNT1. In 10 family members with dominantly inherited, early-onset osteoporosis, we identified a heterozygous missense mutation in WNT1, c.652T→G (p.Cys218Gly). In a separate family with 2 siblings affected by recessive osteogenesis imperfecta, we identified a homozygous nonsense mutation, c.884C→A, p.Ser295*. In vitro, aberrant forms of the WNT1 protein showed impaired capacity to induce canonical WNT signaling, their target genes, and mineralization. In mice, Wnt1 was clearly expressed in bone marrow, especially in B-cell lineage and hematopoietic progenitors; lineage tracing identified the expression of the gene in a subset of osteocytes, suggesting the presence of altered cross-talk in WNT signaling between the hematopoietic and osteoblastic lineage cells in these diseases.
Article
Full-text available
Type V osteogenesis imperfecta (OI) presents with moderate-to-severe skeletal deformity and is characterized by hyperplastic callus formation at fracture sites and calcification of the interosseous membranes of the forearm and lower leg. The facial dysmorphism is not well characterized and has not been described in previous reports. Inheritance is autosomal dominant, although the genetic aetiology remained unknown until very recently. The aims of this study were to establish the genetic aetiology in patients with type V OI and further characterize patients with this condition, and to ascertain whether they have a similar clinical phenotype and facial dysmorphism. Three families (one mother-daughter pair and two singletons) were identified with the above features and further investigations (molecular genetic analysis and skin biopsy including electron microscopy, histology and collagen species analysis) were performed. Accurate phenotyping of patients with type V OI was performed. PCR amplification was performed using the Sheffield Diagnostic Genetics Service pyrosequencing assay for the interferon-induced transmembrane protein-5 (IFITM5) gene. All the patients had been confirmed to have a heterozygous variant, c.[-14C>T];[=], in the 5'-UTR of the IFITM5 gene, which is located in the transcribed region of this gene. This recurrent mutation, in IFITM5, also known as bone-restricted interferon-induced transmembrane protein-like protein or BRIL, encodes a protein with a function in bone formation and plays an important role in osteoblast formation. All four patients in this study appear to have similar clinical features and facial dysmorphism, including a short, up-turned nose, a small mouth, a prominent chin and greyish-blue sclerae. Skin biopsy in one patient showed clumping of elastic fibres and normal biochemical analysis of collagen. We have been able to characterize patients with type V OI further and confirm the genetic aetiology in this distinct form of OI. Accurate phenotyping (including describing the common facial features) before investigations is important to enable the useful interpretation of genetic results and/or target-specific gene testing.
Article
Purpose of review: Osteogenesis imperfecta or 'brittle bone disease' has mainly been considered a bone disorder caused by collagen mutations. Within the last decade, however, a surge of genetic discoveries has created a new paradigm for osteogenesis imperfecta as a collagen-related disorder, where most cases are due to autosomal dominant type I collagen defects, while rare, mostly recessive, forms are due to defects in genes whose protein products interact with collagen protein. This review is both timely and relevant in outlining the genesis, development, and future of this paradigm shift in the understanding of osteogenesis imperfecta. Recent findings: Bone-restricted interferon-induced transmembrane (IFITM)-like protein (BRIL) and pigment epithelium-derived factor (PEDF) defects cause types V and VI osteogenesis imperfecta via defective bone mineralization, while defects in cartilage-associated protein (CRTAP), prolyl 3-hydroxylase 1 (P3H1), and cyclophilin B (CYPB) cause types VII-IX osteogenesis imperfecta via defective collagen post-translational modification. Heat shock protein 47 (HSP47) and FK506-binding protein-65 (FKBP65) defects cause types X and XI osteogenesis imperfecta via aberrant collagen crosslinking, folding, and chaperoning, while defects in SP7 transcription factor, wingless-type MMTV integration site family member 1 (WNT1), trimeric intracellular cation channel type b (TRIC-B), and old astrocyte specifically induced substance (OASIS) disrupt osteoblast development. Finally, absence of the type I collagen C-propeptidase bone morphogenetic protein 1 (BMP1) causes type XII osteogenesis imperfecta due to altered collagen maturation/processing. Summary: Identification of these multiple causative defects has provided crucial information for accurate genetic counseling, inspired a recently proposed functional grouping of osteogenesis imperfecta types by shared mechanism to simplify current nosology, and has prodded investigations into common pathways in osteogenesis imperfecta. Such investigations could yield critical information on cellular and bone tissue mechanisms and translate to new mechanistic insight into clinical therapies for patients.
Article
BRIL/IFITM5 is a membrane protein present almost exclusively in osteoblasts, which is believed to adopt a type III (N-out/C-out) topology. Mutations in IFITM5 cause OI type V, but the characteristics of the mutant protein and the mechanism involved are still unknown. The purpose of the current study was to re-assess the topology, localization, and biochemical properties of BRIL and compare it to the OI type V mutant in MC3T3 osteoblasts. Immunofluorescence labeling was performed with antibodies directed against BRIL N- or C-terminus. In intact cells, BRIL labeling was conspicuously detected at the plasma membrane only with the anti-C antibody. Detection of BRIL N-terminus was only possible after cell permeabilization, revealing both plasma membrane and Golgi labeling. Trypsinization of live cells expressing BRIL only cleaved off the C-terminus, confirming that it is a type II protein and that its N-terminus is intracellular. A truncated form of BRIL lacking the last 18 residues did not appear to affect localization, whereas mutation of a single leucine to arginine within the transmembrane segment abolished plasma membrane targeting. BRIL is first targeted to the endoplasmic reticulum as the entry point to the secretory pathway and rapidly traffics to the Golgi via a COPII-dependent pathway. BRIL was found to be palmitoylated and two conserved cysteine residues (C52 and C53) were critical for targeting to the plasma membrane. The OI type V mutant BRIL, having a five residue extension (MALEP) at its N-terminus, presented with exactly the same topological and biochemical characteristics as wild type BRIL. In contrast, the S42 > L mutant BRIL was trapped intracellularly in the Golgi. BRIL proteins and transcripts were equally detected in bone from a patient with OI type V, suggesting that the cause of the disease is a gain of function mediated by a faulty intracellular activity of the mutant BRIL. © 2014 American Society for Bone and Mineral Research
Article
Osteogenesis imperfecta (OI) types V and VI are caused, respectively, by a unique dominant mutation in IFITM5, encoding BRIL, a transmembrane ifitm-like protein most strongly expressed in the skeletal system, and recessive null mutations in SERPINF1, encoding pigment epithelium-derived factor (PEDF). We identified a 25-year-old woman with severe OI whose dermal fibroblasts and cultured osteoblasts displayed minimal secretion of PEDF, but whose serum PEDF level was in the normal range. SERPINF1 sequences were normal despite bone histomorphometry consistent with type VI OI, and elevated childhood serum alkaline phosphatase. We performed exome sequencing on the proband, both parents and an unaffected sibling. IFITM5 emerged as the candidate gene from bioinformatics analysis, and was corroborated by membership in a murine bone co-expression network module containing all currently known OI genes. The de novo IFITM5 mutation was confirmed in one allele of the proband, resulting in a p.S40L substitution in the intracellular domain of BRIL, but was absent in unaffected family members. IFITM5 expression was normal in proband fibroblasts and osteoblasts, and BRIL protein level was similar to control in differentiated proband osteoblasts on western blot, and in permeabilized mutant osteoblasts by microscopy. In contrast, SERPINF1 expression was decreased in proband osteoblasts; PEDF was barely detectable in conditioned media of proband cells. Expression and secretion of type I collagen was similarly decreased in proband osteoblasts; the expression pattern of several osteoblast markers largely overlapped reported values from cells with a primary PEDF defect. In contrast, osteoblasts from a typical case of type V OI, with an activating mutation at the 5'-terminus of BRIL, have increased SERPINF1 expression and PEDF secretion during osteoblast differentiation. Together, these data suggest that BRIL and PEDF have a relationship that connects the genes for types V and VI OI and their roles in bone mineralization. © 2014 American Society for Bone and Mineral Research.
Article
The IFITM5 gene has recently been found to be mutated in patients with autosomal dominant osteogenesis imperfecta (OI) type V. This form of OI is characterized by distinctive clinical manifestations, including hyperplastic callus formation at the site of fractures, calcification of the interosseous membrane of the forearm, and dislocation of the head of the radius. Notably, in spite of the fact that a considerable number of patients with IFITM5 mutations have been identified, to date all of them have been shown to have the same heterozygous mutation (c.-14C>T). Herein, we describe one patient with a de novo c.119C>T heterozygous mutation in IFITM5, which predicts p.Ser40Leu, and another with the recurrent c.-14C>T transition that was also apparently de novo. While the patient with the p.Ser40Leu mutation had none of the typical signs of OI type V and was diagnosed with limb shortening at prenatal stages, the patient with the c.-14C>T mutation developed hyperplastic calluses and had calcification of the forearm interosseous membrane. This study challenges the lack of allelic and clinical heterogeneity in IFITM5 mutations. © 2013 Wiley Periodicals, Inc.
Article
Osteogenesis imperfecta (OI) is a hereditary connective tissue disorder characterized by a wide range of skeletal symptoms. Most patients have dominantly inherited or de novo mutations in COL1A1 or COL1A2. Up to 5% of patients have OI type V, characterized by hyperplastic callus formation after fractures, calcification of the membrane interossea of the forearm, and a mesh-like lamellation pattern observed in bone histology. Recently, a heterozygous mutation in the 5'-untranslated region of IFITM5 (c.-14C > T) was identified as the underlying cause of OI type V, and only this specific mutation was subsequently identified in all patient cohorts with this OI subtype. We now present a case of a heterozygous mutation within the coding region of IFITM5 (c.119C > T; p.S40L). The mutation occurred de novo in the patient and resulted in severe OI with prenatal onset and extreme short stature. At the age of 19 months, the typical clinical hallmarks of OI type V were not present. Our finding has important consequences for the genetic "work-up" of patients suspected to have OI, both in pre- and in postnatal settings: The entire gene - and not only the 5'-UTR harbouring the "classical" OI type V mutation - has to be analyzed to exclude a causal role of IFITM5. We propose that this should be part of the initial diagnostic steps for genetic laboratories performing SANGER sequencing in OI patients. © 2013 American Society for Bone and Mineral Research.
Article
Osteogenesis Imperfecta (OI) is a heritable bone dysplasia characterized by bone fragility and deformity, and growth deficiency. Most cases of OI (Classical types) have autosomal dominant inheritance and are caused by mutations in the type I collagen genes. During the past several years, a number of noncollagenous genes whose protein products interact with OI have been identified as the cause(s) of rare forms of OI. This has led to a paradigm shift for OI as a collagen-related condition. The majority of the non-Classical OI types have autosomal recessive inheritance and null mutations in their respective genes. The exception is a unique dominant defect in IFITM5, which encodes Bril, and leads to hypertrophic callus and interosseus membrane ossification. Three recessive OI types arise from defects in any of the components of the collagen prolyl 3-hydroxylation complex (CRTAP, P3H1, CyPB), which modifies the collagen α1(I)Pro986 residue. Complex dysfunction leads to delayed folding of the procollagen triple helix and increased helical modification. Next, defects in collagen peptidly-prolyl isomerases, HSP47 and FKBP65, lead to improper procollagen folding, and deficient collagen crosslinking in matrix, respectively. A form of OI with a mineralization defect is caused by mutations in SERPINF1, whose protein product, PEDF, is a well-known anti-angiogenesis factor. Defects in the C-propeptide cleavage enzyme, BMP1, also cause recessive OI. Additional genes, including SP7 and TMEM38B, have been implicated in recessive OI, but are yet unclassified. Elucidating the mechanistic pathways common to dominant and recessive OI may lead to novel therapeutic approaches to improve clinical manifestations.