osteogenesis imperfecta; clinical and genetic heterogeneity dissertation.pdf · osteogenesis...

179
Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity Fleur van Dijk

Upload: vothien

Post on 23-Mar-2019

235 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity

Fleur van Dijk

Osteogenesis Im

perfecta; Clinical and Genetic H

eterogeneity

Fleur van D

ijk

Page 2: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity

Fleur van Dijk

Dijk_Opmaak.indd 1 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 1 T1 - Black CyanMagentaYellow

Page 3: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Osteogenesis Imperfecta; clinical and genetic heterogeneity

PhD thesis, VU University Medical Center, Amsterdam, the NetherlandsISBN: 978-90-533-5479-7

Thesis review committee:Prof. Dr. P. Coucke, Ghent University, BelgiumProf. Dr. V. Everts, VU University Medical Center, Amsterdam, the NetherlandsProf. Dr. G. Mortier, University of Antwerp, BelgiumDr. P.G.J. Nikkels, University Medical Center Utrecht, the NetherlandsDr. E.A. Sistermans, VU University Medical Center, Amsterdam, the Netherlands

Photo cover: Broken glass, Wikimedia CommonsCover design: Nikki Vermeulen, Ridderprint BV, Ridderkerk, the NetherlandsLay out: Nikki Vermeulen, Ridderprint BV, Ridderkerk, the NetherlandsPrinted by: Ridderprint BV, Ridderkerk, the Netherlands

Copyright © 2011, Fleur S. van Dijk, Amsterdam.All rights reserved. No part of this publication may be reproduced or transmitted in any form by any means, electronical or mechanical, including photocopy, recording or any other information storage and retrieval system, without the written permission of the author.

Financial support for the publication of this thesis was kindly provided by-Anna Foundation | NOREF -MRC-holland

Dijk_Opmaak.indd 2 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 1 T1 - Black CyanMagentaYellow

Page 4: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

VRIJE UNIVERSITEIT

Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity

ACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad Doctor aan

de Vrije Universiteit Amsterdam,

op gezag van de rector magnificus

prof.dr. L.M. Bouter,

in het openbaar te verdedigen

ten overstaan van de promotiecommissie

van de faculteit der Geneeskunde

op 16 december 2011 om 11.45 uur

in de aula van de universiteit,

de Boelelaan 1105

door

Fleur Stephanie van Dijk

geboren te Amersfoort

Dijk_Opmaak.indd 3 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 2 T1 - Black CyanMagentaYellow

Page 5: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

promotor: prof.dr. H. Meijers-Heijboer

copromotor: dr. G. Pals

Dijk_Opmaak.indd 4 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 2 T1 - Black CyanMagentaYellow

Page 6: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

“That bone was broken!”

“It has been remade.”

From: The Lord of the Rings: The Return of the King

(dialogue between the King of the Dead and Aragorn, “blade” has been replaced by “bone”)

Voor mijn ouders

Dijk_Opmaak.indd 5 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 3 T1 - Black CyanMagentaYellow

Page 7: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 6 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 3 T1 - Black CyanMagentaYellow

Page 8: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CONTENTS

Scope of the thesis

Chapter I Introduction 11

I-1 Osteogenesis imperfecta: a review with clinical examples. Accepted for publication 13

in Molecular Syndromology

Chapter II Dominant Osteogenesis Imperfecta 41

II-1 Complete COL1A1 allele deletions in osteogenesis imperfecta. Genet Med. 2010 43

Nov;12(11):736-41

Chapter III Recessive Osteogenesis Imperfecta 59

III-1 CRTAP mutations in lethal and severe osteogenesis imperfecta: the importance 61

of combining biochemical and molecular genetic analysis. Eur J Hum Genet. 2009

Dec;17(12):1560-9.

III-2 PPIB mutations cause severe osteogenesis imperfecta. Am J Hum Genet. 2009 77

Oct;85(4):521-7.

III-3 Lethal/Severe Osteogenesis Imperfecta in a Large Family: a novel homozygous 93

LEPRE1 mutation and bone histological findings. Pediatr Dev Pathol. 2011 May-Jun;

14(3): 228-34.

III-4 A novel homozygous 5 bp deletion in FKBP10 causes clinically Bruck syndrome in 105

an Indonesian Patient. Submitted

Chapter IV Implications for clinical and laboratory diagnosis of Osteogenesis Imperfecta 117

IV-1 Classification of Osteogenesis Imperfecta revisited. Eur J Med Genet. 119

2010 Jan-Feb;53(1):1-5

IV-2 EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis 129

Imperfecta. Eur J Hum Genet. 2011 Aug 10. [Epub ahead of print]

Chapter V Discussion 165

Summary 169

Samenvatting voor de geïnteresseerde leek 171

List of Publications 173

Dankwoord 175

Curriculum Vitae 177

Dijk_Opmaak.indd 7 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 4 T1 - Black CyanMagentaYellow

Page 9: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 8 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 4 T1 - Black CyanMagentaYellow

Page 10: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

SCOPE Of THE THESIS

Since the first scientific description of Osteogenesis Imperfecta (OI) in 1788, OI is in the literature reported

to be clinically heterogeneous with a severity ranging from a mildly increased fracture frequency to

multiple prenatal fractures with a poor prognosis. In 1979, the Australian physician David Sillence wrote the

landmark publication, classifying OI in four types based on clinical/radiological features and inheritance.

This particular publication was entitled “Genetic heterogeneity in Osteogenesis Imperfecta” as Sillence believed,

based on the pedigrees of affected families, that both autosomal dominant and recessive causes of OI

would exist [Sillence et al., 1979]. After the discovery of COL1A1/2 mutations as causes in all types of OI,

it was the general opinion that OI was an autosomal dominant disorder with recurrence of lethal OI in

siblings due to germ line mosaicism. This general belief changed again in 2006, with the discovery of the

first autosomal recessive cause of OI [Barnes et al., 2006]. Over the past five years, eight genes have been

discovered to cause recessive OI including Bruck syndrome (bone fragility with congenital contractures of

the large joints). The assumption of genetic heterogeneity in OI made by Sillence in 1979 [Sillence et al.,

1979] is now, almost three decades later, confirmed with the use of molecular genetics.

The scope of this thesis is to study the genetic causes of OI and to incorporate this newly gained

knowledge in the clinical and laboratory diagnosis of OI. In chapter one, an overview is given concerning

the clinical/radiological/histological and biochemical features, genetic causes and treatment of OI. In the

second and third chapter we report our contributions to the knowledge of resepectively dominant and

recessive causes of OI with among others first descriptions of COL1A1 complete allele deletions causative

of mild OI and PPIB mutations as a cause of severe recessive OI. In the fourth chapter, the impact of the

clinical and genetic heterogeneity on the clinical diagnosis, classification and laboratory diagnosis of OI is

ellaborated. Chapter V-1 focuses on future perspectives in OI research.

Dijk_Opmaak.indd 9 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 5 T1 - Black CyanMagentaYellow

Page 11: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 10 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 5 T1 - Black CyanMagentaYellow

Page 12: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I

Introduction

Dijk_Opmaak.indd 11 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 6 T1 - Black CyanMagentaYellow

Page 13: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 12 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 6 T1 - Black CyanMagentaYellow

Page 14: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Osteogenesis Imperfecta: a review with clinical examples

Fleur S. van Dijk1*, Jan M. Cobben2, Ariana Kariminejad3, Alessandra Maugeri1,

Peter G.J. Nikkels4, Rick R. van Rijn5, Gerard Pals1

1 Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands;2 Department of Pediatric Genetics, Emma Children Hospital, AMC, Amsterdam, the Netherlands;

3 Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran;4 Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands;

5 Department of Pediatric Radiology, Academic Medical Centre, Amsterdam, the Netherlands

Accepted for publication in Molecular Syndromology

I-1

Dijk_Opmaak.indd 13 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 7 T1 - Black CyanMagentaYellow

Page 15: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I14

AbSTrACT

Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

ranging from subtle increase in fracture frequency to prenatal fractures. The first scientific description

of OI dates from 1788. Since then, important milestones in OI research and treatment have, among

others, been the classification of OI into 4 types (the “Sillence classification”), the discovery of defects

in collagen type I biosynthesis as a cause of most cases of OI and the use of bisphosphonate therapy.

Furthermore, in the past five years it has become clear that OI comprises a group of heterogeneous

disorders with an estimated 90% of cases due to a causative variant in the COL1A1 or COL1A2 genes

and with the remaining 10% due to causative recessive variants in the 8 genes known so far, or

in other currently unknown genes. This review aims to highlight the current knowledge around

the history, epidemiology, pathogenesis, clinical/radiological features, management, and future

prospects of OI. The text will be illustrated with clinical descriptions, including radiographs and where

possible photographs of patients with OI.

Dijk_Opmaak.indd 14 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 7 T1 - Black CyanMagentaYellow

Page 16: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 15

INTrODuCTION

Osteogenesis Imperfecta (OI) comprises a heterogeneous group of diseases characterized by

susceptibility to bone fractures with variable severity and in most cases with presumed or proven

defects in collagen type I biosynthesis [Van Dijk et al., 2010c] (chapter IV-1). Other clinical manifestations

include short stature, blue sclerae, dentinogenesis imperfecta and hearing loss.

Epidemiology

OI has a birth prevalence of approximately 6-7/100,000 [Steiner et al., 1993]. The prevalence and

incidence of the OI types are different from each other, with OI type I and OI type IV accounting for

considerably more than half of all OI cases [Steiner et al., 1993]. In 1979, Sillence et al. [1979] reported a

prevalence of 3-4/100,000 and an incidence of 3.5/100,000 for OI type I in Victoria, Australia. For OI type

II the incidence is about 1-2/100,000 [Steiner et al., 1993], prevalence data are not available due to early

lethality [Steiner et al., 1993]. OI type III has a prevalence of 1-2/100,000 [Steiner et al., 1993]. An incidence

of 1.6/100,000 has been reported in Australia [Sillence et al., 1979]. OI type IV was believed to be a rare

entity [Sillence et al., 1979] but this proved not to be the case [Steiner et al., 1993].

History

The earliest known patient with OI probably dates from about 1000 B.C and appears to be an Egyptian

infant. This conclusion can be drawn after studying the remains of an Egyptian mummy [Lowenstein,

2009].

The first scientific description of OI was given by the Swedish army Surgeon Olaus Jakob Ekman (1788)

who in his thesis on “congenital osteomalacia” described a three generation family with hereditary

bone fragility [Peltier, 1981; Baljet, 2002]. Since then, many names have been used to describe familial

bone fragility. Willem Vrolik (1801-1863), a Dutch professor at the Athenaeum Illustre of Amsterdam,

introduced the term “osteogenesis imperfecta”[Baljet, 1984] and was one of the first to realize that OI

might be due to insufficient intrinsic “generative energy” [Baljet, 2002] as opposed to the result of an

acquired disease.

In the twentieth century it became clear that OI was a disease showing remarkable clinical variability

with severity ranging from death in the perinatal period to subtle increase in fracture frequency. Looser

[1906] made the first classification of OI into congenital and tarda. Attempts were made to further

classify OI, and in 1979 the “Sillence classification” [Sillence et al., 1979] was proposed which, though in

an adjusted form, is still in use today.

In 1974 bone collagen aggregation abnormalities were described by scanning electron microscopy of

bone collagen in three patients with OI [Teitelbaum et al., 1974]. Sykes et al. [1977] reported altered

relation of two collagen types in pepsin digests of skin of OI patients using a method of interrupted

polyacrylamide-gel electrophoresis. In 1983 the presence of an internal deletion of about 0.5 kb in one

allele for the pro- α1(I) chain in a patient with OI [Chu et al., 1983] was discovered. This finding was the

beginning of the unraveling of the collagen type I biosynthesis and the pathogenic mechanisms of OI

which is still not finished today.

Dijk_Opmaak.indd 15 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 8 T1 - Black CyanMagentaYellow

Page 17: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I16

Classification

In 1979 Sillence et al.[1979] proposed a numerical classification of OI into four types based on clinical

and genetic findings in OI patients ascertained in Victoria, Australia. This classification distinguished type

I (mild OI, blue sclerae, autosomal dominant inheritance), type II (lethal perinatal OI, autosomal recessive

inheritance, later subdivided in II-A,-B,-C based on radiographic features [Sillence et al., 1984], type III

(progressively deforming, autosomal recessive inheritance), and type IV (dominantly inherited OI with

normal sclerae). It is evident that Sillence et al. [1979] assumed that OI was a heterogeneous condition.

OI was considered for some time to be an autosomal dominant disease with recurrence due to germ

line mosaicism since heterozygous collagen type I mutations (COL1A1 or COL1A2 mutations) were

discovered in all OI types. The recurrence risk of OI type II was observed to be less than 10% [Cohn

et al., 1990; Cohen-Solal et al., 1991; Byers et al., 1988; Pepin et al., 1997]. The Sillence classification was

used only for the clinical/radiological classification of OI. Thereafter, studies were reported of families, in

some cases consanguineous, with OI not caused by pathogenic variants in the COL1A1 or COL1A2 genes

[Wallis et al., 1993]. The original Sillence classification was extended with OI types V-VII based on OI cases

with unknown genetic etiology and/or distinctive clinical manifestations [Rauch et al., 2004].

In 2006, the first genetic cause of autosomal recessive lethal OI was discovered, i.e. bi-allelic variants in

the CRTAP gene causing complete loss of protein function [Barnes et al., 2006]. Partial loss of function

CRTAP variantsencoding cartilage-associated protein (CRTAP) were found to cause OI type VII [Morello

et al., 2006] . Presently, six more causes of recessive OI (causative variants in LEPRE1 [Cabral et al., 2007],

PPIB [Van Dijk et al., 2009b (chapter III-2); Barnes et al., 2010], SERPINH1 [Christiansen et al., 2010],

FKBP10 [Alanay et al., 2010], SP7 [Lapunzina et al., 2010] and SERPINF1 [Becker et al., 2011] have been

described, all but two (SP7 and SERPINF1) concerning genes encoding proteins involved in collagen

type I biosynthesis.

There is some debate in the literature about how to involve this newly discovered heterogeneity of OI in

the classification. One research group proposes to have OI caused by recessive causative variants in PPIB,

SERPINH1, FKBP10, SP7 and SERPINF1 added to the current classification as OI type IX [Barnes et al., 2010]

and presumably X, XI , XII, and XIII, respectively. Another viewpoint expressed by our research group is

that the clinical/radiological characteristics of recessive OI do not substantially differ from OI type II-B,

III or IV caused by dominant OI, so we propose to have recessive OI classified as OI type II-B/III/IV- PPIB/

SERPINH1 /FKBP10 /SP7 /SERPINF1- related. OI type I and OI type II-A appear to be exclusively caused by

causative COL1A1/2 variants [Van Dijk et al., 2009a] (chapter III-1). We also proposed to only add a new

numerical type when the phenotype of the patients differs from the numerical types described so far

[Van Dijk et al., 2010c] (chapter IV-2).

There is also debate as to whether Bruck syndrome type 1 [Breslau-Siderius et al., 1998] and type 2 [van

der Slot et al., 2003], characterized clinically by bone fragility and congenital contractures of the large

joints, should be classified as subtypes of OI.

The International Nomenclature group for Constitutional disorders of the Skeleton, among others

concerning the classification of OI, proposed to retain the Sillence classification into five types and free

the classification from direct molecular reference. Bruck syndrome 1 and 2 were not listed as subtypes

of OI [Warman et al., 2011].

Dijk_Opmaak.indd 16 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 8 T1 - Black CyanMagentaYellow

Page 18: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 17

Collagen type I biosynthesis

Collagen type I biosynthesis is depicted in figure 1.

Nucleus

Approximately 90% of individuals affected with OI are heterozygous for a causative variant in one of the

two genes, COL1A1 or COL1A2 [Sykes et al., 1990; Korkko et al., 1998], which encode the pro-α1(I) and

pro-α2(I) chains of type I procollagen, respectively.

Cytoplasm

Procollagen type I is cotranslationally translocated into the lumen of the endoplasmic reticulum [Canty

and Kadler, 2005].

Rough Endoplasmic Reticulum (rER)

Procollagen type I contains C- and N-terminal propeptides and a large “triple helix” domain comprising

predominantly Gly-X-Y triplets. In the rER, two a1(I)- collagen chains encoded by COL1A1 and one

a2(I)-collagen chain encoded by COL1A2 align. Interactions between the C propeptides are largely

stabilized by interchain disulphide bounds to ensure correct alignment [Prockop et al., 1989]. Protein

disulphide isomerase (PDI) has also been implicated in the formation of inter-chain disulphide bonds

[Canty & Kadler, 2005]. The two pro-a1 chains and one pro-a2 chain then assemble in the C- to N-

direction to form a triple helix. During folding, collagen is modified by, among others, specific enzymes

that hydroxylate lysine and proline residues and glycosylate hydroxylysyl residues. This post-translational

modification stops when triple helix assembly is complete [Engel et al., 1991]. The CRTAP/P3H1/CyPB

complex encoded by the CRTAP, LEPRE1 and PPIB genes, is responsible for the 3-hydroxylation of P986

(p.P1164 counting from the methionine which initiates translation), but the complex most likely also

acts as a proline cis-trans isomerase and a molecular chaperone [Ishikawa et al., 2009]. FKBP65 encoded

by FKBP10 also acts as a molecular chaperone for type I (pro)collagen [Alanay et al., 2010]. The protein

product of PLOD2 hydroxylates telopeptide lysines in the rER [van der Slot et al., 2003]. HSP47 encoded

by SERPINH1 is thought to maintain the stability of the triple helix [Christiansen et al., 2010].

Dijk_Opmaak.indd 17 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 9 T1 - Black CyanMagentaYellow

Page 19: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I18

Golgi

After folding, the procollagen molecules are transported through the Golgi apparatus and plasma

membrane (PM) into the extracellular matrix.

Extracellular matrix (ECM)

In the ECM cleavage of the N-and C-terminal propeptides occurs and collagen molecules aggregate

to form fibrils. Covalent crosslinks occur within and between triple-helical collagen molecules in fibrils.

These fibrils converge into collagen type I fibers.

Genotype-phenotype

Autosomal dominant OI types I, II-A, II-B, III, IV

At present more than 1000 distinct variants in the COL1A1 and COL1A2 genes have been identified

that give rise to OI (https://oi.gene.le.ac.uk) [Dalgleish, 1997, 1998]. Type of mutation as well as position

appear to influence the phenotype.

OI type I is mostly characterized by a 50% reduction of the amount of collagen type I (quantitative

quantitative or haploinsufficiency effect) usually resulting from variants in one COL1A1 allele (frameshift,

nonsense, and splice-site alterations) that lead to mRNA instability and haploinsufficiency [Marini et al.,

2007b] and sometimes from a deletion of the complete COL1A1 allele [van Dijk et al., 2010a] (chapter

II-1) or from substitutions for glycine by small amino acids (cysteine, alanine and serine) near the amino

terminal ends of the triple helical domains in either one COL1A1 or COL1A2 allele.

OI types II-IV are characterized by intertwining of mutated and normal collagen type I chains resulting in

production of abnormal collagen type I (dominant-negative effect). This occurs usually due to causative

variants in either COL1A1 or COL1A2 that result in substitutions for glycine. Less common causative

variants include splice site alterations, insertion/deletion/duplication events that lead to in-frame

sequence alterations and variants in the carboxyl-terminal propeptide coding-domains. Most of these

variants result in synthesis of an abnormal type I procollagen molecule which has to intertwine with

normal pro-α chain(s) [Marini et al., 2007b]. This assembly leads to disturbed helical folding resulting in

overprocessing by the enzymes responsible for post-translational modification of (pro)collagen type

I. Post-translational overmodification of the triple-helical domain results in alterations visible by SDS-

polyacrylamide gel electrophoresis.

Autosomal recessive OI types II-B, III, IV

It is estimated that 10% of OI cases will be caused by recessive causative variants in one of the currently

known genes (CRTAP, LEPRE1, PPIB, SERPINH1, FKBP10, PLOD2, SP7, SERPINF1) or in genes that have yet to

be discovered.

CRTAP

Cartilage associated protein (CRTAP) forms a complex with Prolyl-3-hydroxylase-1 (P3H1) and cyclophilin

B (CyPB). One known function of the complex is the 3-hydroxylation of a single proline residue at

position 986 (P986) in the proα1(I)-collagen chain [Marini et al., 2007a]. O. It is highly likely that the

complex also acts as a cis-trans isomerase and a molecular chaperone [Ishikawa et al., 2009], initiating

chain recognition and helical folding [Pyott et al., 2011b]. Most CRTAP variants are reported to cause

Dijk_Opmaak.indd 18 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 9 T1 - Black CyanMagentaYellow

Page 20: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 19

autosomal recessive lethal/severe OI [Barnes et al., 2006] types II-B and III but CRTAP variants do not seem

to cause OI type II-A [Van Dijk et al., 2009a]. Most causative variants result in null alleles with absence or

severe reduction of gene transcripts and proteins [Marini et al., 2010a]. Biochemically, decreased prolyl

3-hydroxylation of P986 is evident as well as post-translational overmodification on (pro)collagen gel

electrophoresis [Barnes et al., 2006].

LEPRE1

LEPRE1 encodes P3H1 and pathogenic variants in LEPRE1 cause autosomal recessive lethal/severe

OI[Cabral et al., 2007; Baldridge et al., 2008], types II-B/ III [Van Dijk et al., 2010b] (chapter III-3). In

most patients, the causative variants result in null alleles with absence or severe reduction of gene

transcripts and proteins [Cabral et al., 2007; Willaert et al., 2009; Marini et al., 2010a]. As in CRTAP related

OI, decreased prolyl 3-hydroxylation of P986 in the pro-α1(I)-collagen chains as well as post-translational

overmodification are observed [Cabral et al., 2007].

PPIB

PPIB encodes the protein cyclophilin B, a collagen-specific proline cis-trans isomerase. CyPB is bound in

a complex with P3H1 and CRTAP. Recessive variants in PPIB were described to cause OI with decreased

P986 3-hydroxylation and post-translational overmodification of type I (pro)collagen in two families

[Van Dijk et al., 2009b] (chapter III-2). These findings were confirmed in other patients with OI types

II, III and IV due to PPIB mutations [Pyott et al., 2011a]. Interestingly, in another family a homozygous

PPIB variant in the presumed start codon of PPIB (c.2T>G in the sequence of PPIB described by Price

et al. [1991], c.26T>G in the current reference sequence of PPIB (NM_000942.4))appeared to cause

only a moderately deforming type of OI without decreased P986 3-hydroxylation and detected post-

translational overmodification but with absence of CyPB on western blot [Barnes et al., 2010]. At this

point, it would be correct to state that recessive variants in the PPIB gene can cause OI type II-B, III, IV

with, in the majority of cases, decreased P986 3-hydroxylation in the proα1(I)-collagen chains and post-

translational overmodification, as is seen in CRTAP and LEPRE1 related OI.

SERPINH1

First detected in Dachshund pedigrees with OI [Drogemuller et al., 2009], bi-allelic causative missense

variants in the SERPINH1 gene encoding collagen chaperone protein HSP47 appear to result in severe

recessive OI. HSP47 monitors the integrity of the triple helix of type I procollagen at the endoplasmic

reticulum (ER) /cis-Golgi boundary. When absent, the rate of transit of procollagen type I chains from

ER to Golgi appears to be increased and the triple helical structure is compromised. Since the action

of HSP47 appears not to be required for procollagen type I chain modification, no post-translational

overmodification is visible on (pro)collagen gel electrophoresis [Christiansen et al., 2010].

SERPINF1

Four individuals (two related and two unrelated individuals) with OI type III, and without collagen type

I overmodification on electrophoresis, were recently described to harbor bi-allelic causative variants in

SERPINF1 leading to a loss of Pigment-Epithelium-Derived Factor (PEDF). The causative SERPINF1 variants

in the index patient were found by next generation sequencing. PEDF is known mainly for its strong

inhibition of angiogenesis. However, expression analyses in bone tissue from wild-type mice and in vitro

Dijk_Opmaak.indd 19 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 10 T1 - Black CyanMagentaYellow

Page 21: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I20

experiments with murine cell systems support a role for PEDF in bone formation and remodeling. It is

speculated that a loss of PEDF causes OI independent from collagen type I biosynthesis [Becker et al.,

2011]. Interestingly, SERPINF1 mutations appear to cause OI type VI [Homan et al., 2011].

SP7Recently, a homozygous causative variant in SP7 was described in a patient with a moderately severe

recessive form of OI with recurrent fractures, mild bone deformities, delayed tooth eruption, normal

hearing and white sclerae. SP7 does not seem to encode a protein involved in the collagen type I

biosynthesis pathway. However, it should be noted that in vivo studies to assess the effect on collagen

type I production could not be performed [Lapunzina et al., 2010]. SP7 encodes Osterix, an osteoblast-

specific transcription factor that has been shown to be essential for bone formation in mice [Sun et al.,

2010; Zhou et al., 2010].

Autosomal recessive OI/ Bruck syndromePLOD2 and FKBP10In 1998 a family was reported with bone fragility, congenital contractures of the large joints and aberrant

cross linking of bone collagen [Breslau-Siderius et al., 1998]. The affected individuals were diagnosed

as having Bruck syndrome (BS) which is characterized by bone fragility in combination with congenital

joint contractures. A locus responsible for Bruck syndrome in this family was mapped to 17p12 [Bank

et al., 1999]. ]. In other families with similar clinical and biochemical features, recessive variants in PLOD2,

encoding a bone-specific telopeptidase lysyl hydroxylase-2, appeared to be causative [van der Slot et

al., 2003; Ha-Vinh et al., 2004]. All three causative variants identified in PLOD2 are homozygote missense

mutations affecting highly conserved residues in exon 17 of PLOD2. These variants possibly alterate

folding and isomerization of the protein, leading to severe reduction of the enzymatic activity [Hyry et

al., 2009], which results in aberrant cross-linking of bone collagen due to underhydroxylation of lysine

residues in the telopeptides.

However, in the family described by Breslau-Siderius et al. [1998] no recessive variants in PLOD2 were

found, leading to a classification of Bruck syndrome in type I (with unknown genetic cause) and II (with

recessive variants in PLOD2) [van der Slot et al., 2003]. Recently however, causative variants in FKBP10

(locus 17q12) encoding FKBP65, were described to cause (a) recessive OI without post-translational

overmodification of (pro)collagen type I most closely resembling OI type III [Alanay et al., 2010] or (b)

Bruck syndrome [Shaheen et al., 2010, Kelley et al., 2011] (chapter III-4). The genetic cause of Bruck

syndrome type I in the originally described family has not been reported yet.

bone formation and OIAlthough causative variants in many genes are now known to result in OI, the pathogenic mechanism

leading from causative variant(s) to OI is still largely unknown, which emphasizes the need to study

normal bone formation.

Bone consists of cells and mineralized extracellular matrix, it provides support and protection, is an

insertion site for muscles and serves as a storage site for calcium and phosphate. Four cell types are active

in bone tissue: (1)osteoprogenitor cells (resting cells that can transform into osteoblasts, chondrocytes

Dijk_Opmaak.indd 20 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 10 T1 - Black CyanMagentaYellow

Page 22: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 21

and adipocytes) (2) osteoblasts (3) osteocytes (mature osteoblasts) and (4) osteoclasts. The bone matrix

(osteoid) is secreted by osteoblasts and consists of type I collagen and ground substance containing

proteoglycans and non-collageneous glycoproteins. Resorption of the bone matrix is performed by

macrophage-related osteoclasts and is important for adaption to growth, repair and mineral mobilization

[Ross et al., 1995]

Bone consists of an outer layer of mature compact bone (cortex) largely composed of cylindrical units

called osteons or Haversian systems. The inner layer of adult spongy bone is arranged as trabeculae or

spicules (Figure 2-I). Bone is formed by endochondral (long bones, ribs and vertebrae) (figure 2-II) or

intramembranous (flat bones of skull and face, mandible, clavicle, ileum) ossification. Intramembranous

ossification takes place by direct differentiation of mesenchymal cells into osteoblasts that secrete

osteoid. Osteoblasts retreat or become entrapped as osteocytes in the osteoid. The osteoid calcifies to

form spicules of spongy bone, the spicules unite and form trabeculae. Endochondral bone formation is

characterized by the presence of a cartilaginous model in which chondrocytes differentiate [Ross et al.,

1995] (figure 2-II).

figure 2-I: Compact and spongy bone

figure II-2: Endochondral ossificationEndochondral ossification occurs when mesenchymal cells differentiate into chondroblasts that produce a cartilage matrix. This cartilage acquires the shape of the bone that will be formed. A periosteal collar of bone forms around the diaphysis. Osteoblasts in this region are engaged in periosteal bone formation, which is responsible for the growth in thickness of long bones. In the centre of the diaphysis chondroblasts hypertrophy, the cartilage matrix becomes calcified, blood vessels and connective tissue cells evade the calcified cartilage, creating a marrow cavity (primary ossification center). Trabeculae of calcified cartilage (primary and secondary spongiosa) remain at the two ends of the cavity on which endochondral bone forms (secondary ossification centres).

Dijk_Opmaak.indd 21 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 11 T1 - Black CyanMagentaYellow

Page 23: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I22

figure 2-III: Histologic abnormalities in OI compared to a controlFigure A, C and E show histology of a normal control femur at 18 weeks of gestational age (GA). Figure B, D and F are from an 18 weeks GA type II OI case. The transition zone of cartilage to primary spongiosa is sharp in both cases (A, B). Minor disruption can occur due to fractures and scar formation in the primary spongiosa (not shown). Figure D shows extensive metaplastic cartilage formation at the sight of a fracture. The bony trabeculae are hypercellular and the marrow is fibrotic. Panel E shows normocellular trabeculae and hematopoietic marrow in between these trabeculae. In the severe OI cases, the trabeculae are thin, irregular and hypercellular in comparison with normal trabeculae (F). The marrow in between is fibrotic with hardly any hematopoiesis (D and F).

Paracrine factors and transcription factors appear to be active in the transition of cartilage to bone.

For example, SOX9is important for differentiation of mesenchymal cells into chondroblast (mutations

in SOX9 cause campomelic dysplasia [Unger et al., 1993]) whereas RUNX2 (mutations in RUNX2

cause cleidocranial dysplasia [Mundlos et al., 1997]) is critical for differentiation of prehypertrophic

chondrocytes into preosteoblasts and activation of Osterix (mutations in SP7, encoding Osterix, cause OI

[Lapunzina et al., 2010]), which activates bone-specific proteins [Gilbert 2010].

Dijk_Opmaak.indd 22 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 11 T1 - Black CyanMagentaYellow

Page 24: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 23

Mineralized cartilage is replaced by bone through remodeling. During life, bone is remodeled through

the action of osteoblasts and osteoclasts, replacing old bone with new bone. Normally, remodeling is

coupled such that the level of resorption is equal to the level of formation and bone density remains

constant. The continued growth of the long bones is dependent on the presence of epiphyseal cartilage

(epiphyseal plate) between the primary and secondary ossification centers. This continues to form new

cartilage which is replaced by bone, resulting in increased length of the bone. Growth continues until

the cartilage in the plate is replaced by bone.

A study of iliac bone specimens of 70 children with OI type I, III and IV compared to 27 age-matched

controls reported that in OI bone thickness is reduced because of delayed periosteal bone formation.

Trabeculae are reduced in number and abnormally thin. The overall bone formation is increased but

counteracted by increased activity of bone resorption [Rauch and Glorieux, 2004]. Figure 2-III compares

bone tissue of an individual affected with OI to a control. There appears to be no difference in bone

histology of individuals with OI types I, II-B, III, IV due to dominant causative COL1A1/2 variants or bone

histology of individuals with recessive OI due to causative variants in CRTAP, LEPRE1 and PPIB [van Dijk

et al., 2010b] (chapter III-3).

Prenatal and postnatal diagnosis of OI with clinical examples

The severity of clinical features of OI at birth ranges from no clinical features to prenatally lethal skeletal

abnormalities. The clinical variability of OI led to a classification of OI originally in four types [Sillence et al.,

1979]. In 2004, OI type V, VI and VII were added to this classification. Important for the prenatal as well as

postnatal diagnosis of OI is that a continuum of severity is observed in OI with clinical overlap between

OI types I and IV, II and III, III and IV. Tables 1 and 2 represent an overview of the clinical and radiological

characteristics of OI type I-VI. In our opinion, the addition of OI type VII and the newly proposed OI types

VII-XII are unnecessary [Van Dijk et al., 2010c]. Figures 3-7 are clinical pictures and radiographs of patients

with OI type I, II, III, IV with case descriptions in the legend.

Prenatal diagnosis of OI

OI type II and III (figures 4-6) can prenatally be diagnosed with ultrasonography since they are likely

to have prenatal fractures that can be observed. Increased nuchal translucency can be the earliest

(nonspecific) sonographic sign of OI type II [Viora et al., 2002]. Sonographic signs of OI type II can be

detected as early as 14 weeks [Marini et al., 2007b] due to reduced echogenicity of the fetal bones,

followed by multiple fractures at various stages of healing and deformity of the long bones, ribs and

skull [Morgan and Marcus, 2010]. Sonographic details of OI type III are usually visible from 18 weeks

[Marini et al., 2007b] whereas OI type IV may occasionally be detected after 20 weeks [Marini et al.,

2007b] and may consist of bowing of the long bones with or without shortening and without evidence

of fractures or osteopenia. Prenatal ultrasonographic diagnosis of OI type I is unreliable. For differential

diagnosis, campomelic dysplasia and perinatal lethal hypophosphatasia can be considered [Morgan

and Marcus., 2010]. Pathognomonic radiological/ultrasonographic features of campomelic dysplasia are

cervical spine abnormalities, scapular hypoplasia, narrow iliac wings, bowing of the femora and the

Dijk_Opmaak.indd 23 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 12 T1 - Black CyanMagentaYellow

Page 25: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I24

tibiae, and club feet [Unger et al., 1993]. Distinguishing features of perinatal lethal hypophosphatasia

are (i) deficient ossification of the spine notably in the thoracic region with patchy ossification of ribs and

vertebrae and (ii) deep cupping of the metaphyses of the long bones [Zankl et al., 2008].

The ultrasonographic prenatal diagnosis of OI can be confirmed by laboratory investigations either by

(i) performing a chorion villus biopsy with cultured chorion villi cells showing abnormal production of

collagen type I, visible as post-translational overmodification on procollagen electrophoresis or (ii) by

performing a chorion villus biopsy/amniocentesis to obtain fetal DNA for molecular analysis of genes

involved in OI. Recently guidelines have been established for the laboratory diagnosis of OI . Molecular

analysis of the COL1A1/2 genes will be the first step, followed by DNA analysis of the other causative

genes upon re-evaluation and confirmation of the diagnosis OI [van Dijk et al., 2011] (chapter IV-2).

Prenatal or preimplantation genetic diagnosis with the intention of terminating a pregnancy or not

selecting embryos carrying the causative variant(s) is possible in the case of identification of known

disease-causing variants [van Dijk et al., 2011].

Type I1 II-A2 II-b2 III1 IV1 V3 VI4

Inheritance AD AD AD/AR5 AD/AR6 AD/AR7 AD AR

Severity mild Perinatal lethal

Perinatal lethal-severe

severe Moderate-mild

Moderate Moderate

Congenital fractures

No Yes Yes Usually Rarely No No

bone deformity Rarely Vey severe severe Moderate-severe

Mild to moderate

Moderate Moderate

sclerae Predominantly blue8

Dark blue9 Dark blue9 Blue/grey/white10

Normal to grey11

Normal Normal

Stature Normal12 Severely short13

Severely short14

Very short 15 Variable short16

Variable Mild short stature

Joint hypermobility

Yes 17 Yes Yes Yes 17 Variable Variable Variable18

Hearing loss Present in about 60% of cases19

NA NA Common20 Present in~ 42% of cases21

No No

Dentinogenesis Imperfecta (DI)

Variable22 NA NA Yes23 Variable No No

respiratory complications

No Yes24 Yes24 Yes25 No No No

Neurological complications

No NA NA Yes26 No No No

Table 1: clinical characteristics of OI.I OI type I, II, III, IV constitute the original Sillence classification [Sillence et al.,1979]2 OI type II has been divided in OI type II-A ,II-B and II-C on the base of radiological characteristics [Sillence et al., 1984]. II-C is an uncertain entity that is extremely rare, it is left out of consideration in this table.3 OI type V was added to the Sillence classification based on distinct clinical/radiological (limitations in the range of pronation/supination in one or both forearms associated with a radiologically apparent calcification of the interosseous membrane) and histological features (irregular arrangement or meshlike appearance of lamellae) in patients originally diagnosed with OI type IV in the absence of COL1A1/2 mutations[Glorieux et al., 2000].4 OI type VI was added to the Sillence classification because of distinct histological features (increase in both osteoid surface and thickness pointing to a mineralization defect) in the absence of COL1A1/2 mutations in patients originally diagnosed with OI type IV, but without abnormalities of collagen type I on electrophoresis. Autosomal recessive inheritance is presumed because of two consanguineous families with recurrence of OI in one of these families [Glorieux et al., 2002].

Dijk_Opmaak.indd 24 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 12 T1 - Black CyanMagentaYellow

Page 26: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 25

5recessive variants in CRTAP, LEPRE1 and PPIB have been described to result in a clinical/radiological phenotype indistinguishable from OI type II-B ([Van Dijk et al., 2009a; van et al., 2010; Van Dijk et al., 2009b])6 Recessive variants in CRTAP, LEPRE1, PPIB, SERPINH1, SERPINF1, FKBP10 can result in a clinical/radiological phenotype of OI type III [Alanay et al., 2010; Baldridge et al., 2008; Van Dijk et al., 2009a; Van Dijk et al., 2009b; Pyott et al., 2011; Christiansen et al., 2010; Becker et al., 2011]7 Recessive variants in CRTAP, PPIB, SP7 can result in a clinical/radiological phenotype of OI type IV [Morello et al., 2006; Barnes et al., 2010; Lapunzina et al., 2010; Pyott et al., 2011] 8 In infants < 1 year blue sclerae can be observed as a normal phenomenon due to a thin, scleral envelope and the underlying darkly pigmented choroid layer [Aase 1990]. This normal blue sclera usually disappears by 1 year of age. In OI, the presence of blue sclera is not associated with any significant ocular pathology [Zack et al., 2007]. A correlation between blue sclerae and reduced corneal thickness has been described [Evereklioglu et al., 2002] but has also been challenged by opposite findings [Sarathchandra et al., 1999]. In spite of also other explanations [Lanting et al., 1985;Eichholtz et al., 1972] the etiology of the blue sclerae in OI type I is unclear [Zack et al., 2007].9-11 The blue sclera in OI type II, as well as the blue sclera that can be observed in OI type III (and sometimes in OI type IV) are probably the result of the light reflected from the pigmented layers of the eye because of abnormal slender collagen fibrils and reduced tissue thickness [Zack et al., 2007; Chan et al., 1982; Pedersen et al., 1984] 12 Short stature has been described in OI type I but most patients do not meet the criteria of growth deficiency (> -2 SD). However, in most instances they will be shorter than family members [Marini et al., 1995].13-16 The cause of short stature in OI is unclear. Children with OI type III will typically have a final adult stature in the range of a prepubertal child. The final stature of a child with OI type IV approximates that of an early teenager [Marini 2010a]. 17 Generalized hypermobility is described in OI type I [Engelbert et al., 1997]. Joint hypermobility (ligamentous laxity) is also a feature of OI types II-A, B, and III [Spranger et al., 2003].18 Of the first 16 patients with OI type VI 50% had ligamentous laxity [Glorieux et al., 2002]19-21 According to a Finnish study in adult patients with OI, conductive or mixed hearing loss in late adolescence is observed in 60,4% of patients with OI type I, 42,3% of patients with OI type IV and is common in OI type III. The hearing loss in OI resembles that of otosclerosis. In most cases, the hearing loss is initially conductive and later mixed or sensorineural. It is common in adults and usually progressive [Kuurila et al., 2002].22 OI type I and OI type IV have been subdivided in the past based on absence of DI (1A,IVA )and presence of DI(IB, IVB) [Levin et al., 1978] 23 In DI observed in patients affected with OI (figure 9), the dentine structure of both primary and secondary dentitions are abnormal with teeth appearing typically amber and translucent and showing significant attrition [Barron et al., 2008].24-25 Infants with OI type II die mostly perinatally of respiratory insufficiency or pneumonias. Children with OI type III develop later in life vertebral collapse and kyphoscoliosis, which contribute to restrictive lung disease. They are at risk for developing multiple pneumonias. Lung disease can progress into cor pulmonale [Marini, 2010a]

26 There is a risk of basilar invagination in patients with OI type III [Marini, 2010a]

Postnatal diagnosis of OI

OI types I-V are clinically diagnosed peri- (types II, III and sometimes IV) and postnatally (all types). A flow

diagram for the postnatal diagnosis of OI is presented in figure 8. Clinical case examples are presented

in figures 3, 6 and 7. For differential diagnosis, rare genetic conditions such as Bruck syndrome (MIM

%259450, #609220), Osteoporosis pseudoglioma syndrome (MIM #259770), Cole-Carpenter syndrome

(MIM 112240), Hajdu-Cheney (MIM %102500), gerodermia osteodysplastica (MIM #231070) can be

considered. Furthermore, idiopathic juvenile osteoporosis (MIM 259750) and isolated dentinogenesis

imperfecta (#125490) can be important differential diagnostic considerations. The most common to

be considered is non-accidental injury (NAI), frrequently in cases of suspected OI type I or IV [Bilo et al.,

2010]. Fractures resulting from NAI occur in 24/ 10,000 children under three years of age whereas the OI

prevalence is 1: 10,000 -20,000 [Marlowe et al., 2002]. In the study by Marlowe et al. [2002] the reported

incidence of OI among children evaluated for NAI was 2-5%. Differentiation between OI and NAI is aided

by an experienced clinician familiar with OI [Ablin et al., 1990]. A positive family history, the presence of

blue sclerae, osteoporosis, multiple wormian bones in the skull, and platyspondyly point to OI [Chapman

and Hall., 1997]. The laboratory confirmation of OI is made preferably by DNA analysis of the genes

involved in OI or by decreased or abnormal production of (pro)collagen type I by fibroblasts measured

on procollagen electrophoresis (chapter IV-2).

Dijk_Opmaak.indd 25 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 13 T1 - Black CyanMagentaYellow

Page 27: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I26 T

able

2: r

adio

logi

cal c

hara

cter

istic

s of

OI t

ypes

I-VI

[Si

llenc

e et

al.,

1979

; Sill

ence

et a

l., 19

84; S

pran

ger e

t al.,

2003

;Glo

rieux

et a

l., 20

00; G

lorie

ux e

t al.,

2002

].

Type

III-

AII-

BIII

IVV

VI

Radi

ogra

phic

feat

ures

in

peri

nata

l/in

fanti

le p

erio

d

Skul

lW

orm

ian

bone

sSe

vere

ly d

imin

ishe

d m

iner

aliz

ation

, w

orm

ian

bone

s

Dim

inis

hed

min

eral

izati

on,

wor

mia

n bo

nes

Dim

inis

hed

min

eral

izati

on ,

wor

mia

n bo

nes

Dim

inis

hed

min

eral

izati

on ,

som

etim

es w

ith

wor

mia

n bo

nes

Poss

ibly

dim

inis

hed

min

eral

izati

on ,

som

etim

es w

ith

wor

mia

n bo

nes

8/11

pati

ents

re

port

ed h

ad

no w

orm

ian

bone

s

Ribs

No

frac

ture

sSh

ort,

bro

aden

ed

ribs

with

co

ntinu

ous

bead

ing/

frac

ture

s

Thin

rib

s w

ith

disc

ontin

uous

be

adin

g/ fr

actu

res

Thin

rib

s w

ith

disc

ontin

uous

bea

ding

/fr

actu

res

Noc

onge

nita

l fr

actu

res

No

cong

enita

l fr

actu

res

No

con

geni

tal

frac

trur

es

Vert

ebra

eN

orm

al a

t bir

thPl

atys

pond

yly

at

birt

hPl

atys

pond

yly

at

birt

hPl

atys

pond

yly

at b

irth

Nor

mal

at b

irth

Nor

mal

at b

irth

Nor

mal

at b

irth

Extr

emiti

esN

orm

al

mod

elin

g at

bi

rth

Thic

k, s

hort

, cr

umpl

ed s

haft

s of

th

e lo

ng b

ones

Shor

t, d

efor

med

lo

ng tu

bula

r bo

nes

Shor

t, d

efor

med

long

tu

bula

r bo

nes

Bow

ing

of lo

ng

bone

s-C

alci

ficati

on

of o

sseo

us

mem

bran

e in

fo

rear

ms

-Bow

ing

of lo

ng

bone

s

Bow

ing

of lo

ng

bone

s

Oth

erU

sual

ly n

o co

ngen

ital

frac

ures

or

oste

open

ia

-Gen

eral

ized

os

teop

enia

-Mul

tiple

frac

ture

s w

ith c

allu

s fo

rmati

on

-Gen

eral

ized

os

teop

enia

-Mul

tiple

frac

ture

s w

ith c

allu

s fo

rmati

on

-Gen

eral

ized

ost

eope

nia

- Mul

tiple

frac

ture

s w

ith

callu

s fo

rmati

on

Gen

eral

ized

os

teop

enia

Gen

eral

ized

os

teop

enia

Gen

eral

ized

os

teop

enia

Radi

ogra

phic

feat

ures

la

ter

in li

fe

-Sle

nder

sha

fts

of tu

bula

r bo

nes

with

th

in c

orte

x an

d po

orly

tr

abec

ulat

ed

spon

gios

a-V

erte

bral

co

mpr

essi

on

frac

ture

s

Not

app

licab

leN

ot a

pplic

able

-Kyp

hosc

olio

sis

with

co

mpr

esse

d ve

rteb

ral

bodi

es-T

hin,

sev

erel

y de

-fo

rmed

long

tubu

lar

bone

s oft

en w

ith p

op-

corn

cal

cific

ation

s-d

efor

med

sku

ll w

ith

tem

pora

l bul

ge a

nd

plat

ybas

ia-C

oxa

vara

-Pro

gres

sive

bo

win

g of

long

bo

nes

in s

ome

patie

nts

-Ver

tebr

al

com

pres

sion

fr

actu

res

-Cox

a va

ra

-Pro

gres

sive

bow

ing

of lo

ng b

ones

in

som

e pa

tient

s-V

erte

bral

co

mpr

essi

on

frac

ture

s-C

oxa

vara

-Pro

gres

sive

bo

win

g of

long

bo

nes

in s

ome

patie

nts

-Ver

tebr

al

com

pres

sion

fr

actu

res

-Cox

a va

ra

Dijk_Opmaak.indd 26 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 13 T1 - Black CyanMagentaYellow

Page 28: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 27

figure 3-I: clinical pictures of patients with OI type I. Blue sclerae in a mother and daughter with OI type I.

Clinical synopsis: An Iranian mother and daughter were referred because of suspected OI. The mother, 28 years old, has had 3 fractures in the wrist, ankle and femur occurring between 2-3 years of age. She developed hearing loss at the age of 13-14. She has strikingly blue sclerae, her height is 160 cm (-1.5 SD) and she has no evidence of dentinogenesis imperfecta (DI). The daughter, 8 years old, has had no fractures, a normal height of 125cm (-1 SD) and no DI or hearing loss. Only deep blue sclerae can be observed. Coincidentally, she appears to have decreased vision due to deterioration of cone cells. Radiographs and bone densitometry of both mother and daughter were normal. There were no family members affected with OI. Molecular analyses of the COL1A1/2 genes revealed a heterozygous variant in the COL1A1 gene creating a premature stopcodon (c.1081C>T; p.Arg361X).

figure 3-II: Radiographs of a patient with OI type I A: Lateral radiograph showing osteopenia and mid thoracic vertebral compression fracture (open arrow). Note a general decrease in height, compared to adjacent vertebral bodies, and some wedging. B: Oblique mid-diaphyseal fibula fracture (open arrow) and a communitive diaphyseal tibia fracture (arrow). Clinical synopsis: The patient is a 10-year-old girl with OI type I due to a deletion of the whole COL1A1 allele, who had eight fractures so far. A fracture of the femur was noted shortly after birth. Her length is 128.5 cm (-2.5 SD), head circumference is 53.5 cm (0 SD) and bone density of the hips and lumbar vertebral column is between -4 and -7 SD, as measured by Dual X-ray absorptiometry (DXA). She has Wormian bones, a scoliosis and grey-blue sclerae typical for OI. She has no dentinogenesis imperfecta (van Dijk et al., 2010).

Tab

le 2

: rad

iolo

gica

l cha

ract

eris

tics

of O

I typ

es I-

VI [

Sille

nce

et a

l., 19

79; S

illen

ce e

t al.,

1984

; Spr

ange

r et a

l., 20

03;G

lorie

ux e

t al.,

2000

; Glo

rieux

et a

l., 20

02].

Type

III-

AII-

BIII

IVV

VI

Radi

ogra

phic

feat

ures

in

peri

nata

l/in

fanti

le p

erio

d

Skul

lW

orm

ian

bone

sSe

vere

ly d

imin

ishe

d m

iner

aliz

ation

, w

orm

ian

bone

s

Dim

inis

hed

min

eral

izati

on,

wor

mia

n bo

nes

Dim

inis

hed

min

eral

izati

on ,

wor

mia

n bo

nes

Dim

inis

hed

min

eral

izati

on ,

som

etim

es w

ith

wor

mia

n bo

nes

Poss

ibly

dim

inis

hed

min

eral

izati

on ,

som

etim

es w

ith

wor

mia

n bo

nes

8/11

pati

ents

re

port

ed h

ad

no w

orm

ian

bone

s

Ribs

No

frac

ture

sSh

ort,

bro

aden

ed

ribs

with

co

ntinu

ous

bead

ing/

frac

ture

s

Thin

rib

s w

ith

disc

ontin

uous

be

adin

g/ fr

actu

res

Thin

rib

s w

ith

disc

ontin

uous

bea

ding

/fr

actu

res

Noc

onge

nita

l fr

actu

res

No

cong

enita

l fr

actu

res

No

con

geni

tal

frac

trur

es

Vert

ebra

eN

orm

al a

t bir

thPl

atys

pond

yly

at

birt

hPl

atys

pond

yly

at

birt

hPl

atys

pond

yly

at b

irth

Nor

mal

at b

irth

Nor

mal

at b

irth

Nor

mal

at b

irth

Extr

emiti

esN

orm

al

mod

elin

g at

bi

rth

Thic

k, s

hort

, cr

umpl

ed s

haft

s of

th

e lo

ng b

ones

Shor

t, d

efor

med

lo

ng tu

bula

r bo

nes

Shor

t, d

efor

med

long

tu

bula

r bo

nes

Bow

ing

of lo

ng

bone

s-C

alci

ficati

on

of o

sseo

us

mem

bran

e in

fo

rear

ms

-Bow

ing

of lo

ng

bone

s

Bow

ing

of lo

ng

bone

s

Oth

erU

sual

ly n

o co

ngen

ital

frac

ures

or

oste

open

ia

-Gen

eral

ized

os

teop

enia

-Mul

tiple

frac

ture

s w

ith c

allu

s fo

rmati

on

-Gen

eral

ized

os

teop

enia

-Mul

tiple

frac

ture

s w

ith c

allu

s fo

rmati

on

-Gen

eral

ized

ost

eope

nia

- Mul

tiple

frac

ture

s w

ith

callu

s fo

rmati

on

Gen

eral

ized

os

teop

enia

Gen

eral

ized

os

teop

enia

Gen

eral

ized

os

teop

enia

Radi

ogra

phic

feat

ures

la

ter

in li

fe

-Sle

nder

sha

fts

of tu

bula

r bo

nes

with

th

in c

orte

x an

d po

orly

tr

abec

ulat

ed

spon

gios

a-V

erte

bral

co

mpr

essi

on

frac

ture

s

Not

app

licab

leN

ot a

pplic

able

-Kyp

hosc

olio

sis

with

co

mpr

esse

d ve

rteb

ral

bodi

es-T

hin,

sev

erel

y de

-fo

rmed

long

tubu

lar

bone

s oft

en w

ith p

op-

corn

cal

cific

ation

s-d

efor

med

sku

ll w

ith

tem

pora

l bul

ge a

nd

plat

ybas

ia-C

oxa

vara

-Pro

gres

sive

bo

win

g of

long

bo

nes

in s

ome

patie

nts

-Ver

tebr

al

com

pres

sion

fr

actu

res

-Cox

a va

ra

-Pro

gres

sive

bow

ing

of lo

ng b

ones

in

som

e pa

tient

s-V

erte

bral

co

mpr

essi

on

frac

ture

s-C

oxa

vara

-Pro

gres

sive

bo

win

g of

long

bo

nes

in s

ome

patie

nts

-Ver

tebr

al

com

pres

sion

fr

actu

res

-Cox

a va

ra

Dijk_Opmaak.indd 27 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 14 T1 - Black CyanMagentaYellow

Page 29: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I28

figure 4-I: OI type II-A in a preterm infantSkeletal overviews without (A) and with (B) silver nitrate impregnation show generalized osteopenia with diminished ossification of the calvarian bones and gross skeletal deformation. No vertebral anomalies are seen. The ribs are broad with continuous fractures (continuous beading). The long bones show multiple fractures and are shortened, deformed and broadened.

Clinical synopsis: It concerned the second pregnancy of a nonconsanguineous Caucasian couple. At a gestational age of 21 weeks ultrasonographic abnormalities were seen. All long bones were severely shortened (<p5). Bowing of the humeri and the femora was observed. The skull appeared somewhat doligocephalic with a remarkable clear imaging of the cerebrum and could be deformed by pressure of the ultrasound transducer. The parents decided to terminate the pregnancy and a child was born at a gestational age of 22+3 weeks with a birth weight of 160 gram. A skeletal overview revealed an almost total absence of skull mineralization and multiple fractures of the ribs and the long bones consistent with a diagnosis of OI type II-A. Bone histology showed hypercellular irregular trabeculae with multiple fractures, fibrotic marrow and metaplastic cartilage formation. On collagen electrophoresis post-translational overmodification was observed. A causative variant in the COL1A1 gene was found c.2300G>A; p.Gly767Asp.

figure 4-II: perinatal OI type II-A A: Skeletal overview of a 35 4/7 week old fetus with OI type IIA shows multiple fractures of both the long bones as well as the ribs. Note the under mineralisation of the skull and its deformity on the left side. B: Detail of the lower extremities shows multiple consolidated fractures resulting in deformed growth of the femurs, tibiae and fibulae. C: Lateral radiograph of the lumbar spine shows mild

Clinical synopsis: It concerned the first pregnancy of a non-consanguineous Caucasian couple. At a pregnancy duration of 35+4 weeks a boy was delivered by caesarean section. Apgar scores were 4/4/4 after 1,5 and 10 minutes

Dijk_Opmaak.indd 28 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 14 T1 - Black CyanMagentaYellow

Page 30: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 29

respectively. Birth weight was 1100 gram. Blue sclerae with proptosis were visible. The thorax was small and bell-shaped. Fractures of the ribs were felt. Gasping breath with insufficient thorax excursions was apparent. Auscultation of the heart revealed bradycardia with no other anomalies. The skin was thin and fragile with a large skin defect temporo-occipital. The extremities were short and deformed. The child died 30 minutes after birth due to severe respiratory and circulatory insufficiency. A skeletal overview was consistent with a diagnosis of osteogenesis imperfecta. Histology showed decreased osteoid formation at the level of the primary spongiosis in combination with decreased numbers of osteoblasts and osteoclasts. A causative variant in the COL1A1 gene c.1804G>A; p.Gly602Arg was found.

figure 5-I: OI type II-B in a preterm infantFetal anteroposterior radiographs of proband 1 from family 1 at 21+2 weeks of gestation show: normal skull mineralization for gestational age, slender ribs without fractures, incomplete ossification of T5 and T12, somewhat irregular proximal metaphyses of the humeri, radii and ulnae and bowing of the ulnae. Bowed femora with fractures and some loss of modeling, bowed tibiae and fibula are apparent, possibly with fractures.Clinical synopsis: The affected individual was delivered after termination of pregnancy at 22+1 weeks of gestation. She was the second child of nonconsanguineous North European parents. During pregnancy the diagnosis OI was suspected based on advanced ultrasounds. Bone histology was indicative of OI. Overmodification of collagen type I in fibroblasts was evident on electrophoresis. A homozygous causative variant c.556_559delAAGA in exon 5, resulting in p.Lys186GlnfsX8, was detected in the PPIB gene [van Dijk et al., 2009b].

figure 5-II: perinatal OI type II-BA: Radiograph shows diminished but visible mineralization of the calvarium. The ribs show multiple fractures in a discontinuous pattern with normal rib parts in between callus formation (discontinuous beading). There are bilateral clavicular fractures.B: The long bones of the lower extremities are broadened, deformed and shortened as a result of multiple fractures. There is no complete loss of modeling of the femora.

Clinical synopsis: It concerned the first pregnancy of a nonconsanguineous Caucasian couple. A boy was born a term. A severe skeletal dysplasia was suspected. A skeletal overview showed decreased skull mineralization and multiple fractures of the ribs and the long bones. A causative variant in the COL1A2 gene was found c.1720G>A; p.Gly574Ser.

Dijk_Opmaak.indd 29 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 15 T1 - Black CyanMagentaYellow

Page 31: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I30

figure 6-I: clinical pictures of a patient with OI type IIIA-C: White sclerae, severe kyphoscoliosis with thoracal deformation, severe shortening and bowing of arms and legs.

Clinical synopsis: a 10-year-old Iranian girl was born with fractures of the humerus and the tibia. According to the parents, their daughter has had about 100 fractures up to now. Her length is 88 cm (<<-3 SD, 0 SD for a 2-year old child), weight is 13 kg (0.5 SD (weight for length) and her head circumference (HC) is 49 cm (+2 SD (HC for age)). She has white sclerae. Dentinogenesis imperfecta was apparent. The skin was soft. Cognition was normal. Molecular analysis of the COL1A1/2 genes is currently being performed.

figure 6-II: radiographs of a patient with OI type III Clinical synopsis: In the first pregnancy abnormalities of the extremities were observed in the fetus at 23 weeks of gestation. At the 24th week of gestation short upper and lower extremities were observed indicative of a severe skeletal dysplasia. At a gestation of 40+4 weeks a caucasian boy was born. He had a round face with shallow orbits,

Dijk_Opmaak.indd 30 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 15 T1 - Black CyanMagentaYellow

Page 32: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 31

greyish sclerae, small thorax, rhizomelic shortening of the upper and lower extremities, abducted position of the legs and normocephaly. A skeletal overview showed multiple fractures suggestive of OI type II-B/III. The child is alive at the age of 4 years with a current diagnosis of OI type III due to a homozygous CRTAP mutation (intron 1, c.471+2C>A) [van Dijk et al., 2009a]. A. The skull shows normal mineralization. The spinal column shows normal development and no fractures. The ribs are slender without fractures. No fractures of humeri, radii and ulnae are visible. Multiple fractures of femora with loss of modeling (arrow) can be observed in combination with fracture and bowing of right tibia (arrow head). B. Wormian bones (see insert), broad skull C: At the age of five years radiographs of the lower extremities show osteopenia and multiple fractures for which surgical intervention, using intramedullary rods, has been performed. No popcorn epiphyses are observed. Multiple growth acceleration lines are visible due to intravenous biphosphonate treatment and calcium suppletion (arrow). D: Radiograph of the left arm shows normal epiphyses, with a broad metaphysis of the distal humerus. Note middiaphyseal fractures of the radius and ulna. Dislocation of the radial head is observed. E: AP Spine shows platypondyly and scoliosis

figure 7: clinical pictures and radiographs of a patient with OI type IV

Clinical synopsis: A 33-year- old Iranian man consulted a clinical geneticist when his wife was pregnant as he wanted to be informed about the chance of recurrence of OI in his unborn child. His height and head circumference are respectively 145 cm (-5.5 SD) and 57 cm (-0.5 SD). He claims he has had multiple fractures first occurring at two years of age. Unfortunately, no documented medical history is available. His sclerae are greyish. No hearing loss or dentinogenesis imperfecta is present. His father and sister were also known to be affected wit(h OI. MLPA analysis of the COL1A1 gene showed a partial deletion of COL1A1 (exon 6-51). A,B: White sclerae, muscular upper extremities, wheelchair bound.C: AP radiograph of the abdomen, of poor quality, shows a compression fracture of T10 (between arrows). D-E: Radiographs of the lower extremities show reduced bone density and thin tibia shafts with both fibula being very thin and tortuous. Intramedullary rods are in position.

Dijk_Opmaak.indd 31 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 16 T1 - Black CyanMagentaYellow

Page 33: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I32

Abnormal modeling of long bones?

Multiple prenatal congenital rib fractures and/or short narrow thorax with respiratory insufficiency

Blue sclerae?***

Yes No

Yes

No Yes No

OI type I Wormian bones and/or osteopenia?

Short, crumpled femora with continuously beaded (fractured) ribs and almost no skull mineralization?

Some modeling of femora with discontinuously beaded (fractured) ribs and decreased skull mineralization?

Consider differential diagnosis of OI

No

OI unlikely, cave NAI

Uni-or bilateral calcification of osseous membrane between forearms?

Progressively deforming?

OI type III

OI type IV OI type V

OI type II-A

OI type II-B

Yes

Yes

No

No

Yes No

Yes No

clinical suspicion of OI (non-familial)*

Skeletal overview**

Yes

figure 8: Flow schedule for postnatal diagnosis of OI*recurrent fractures, shortening of limbs, deformation of bones, short stature, early osteoporosis, blue sclerae, hearing loss, dental problems, and joint laxity ** Particularly in case of short stature and/or disproportiate stature and/or clinical deformity of long bones*** In infants < 1 year blue sclerae can be a normal phenomenon

Genetic Counselling

In a large majority of patients, OI type I is caused by dominant (de novo or recurrent) causative variants

in the COL1A1/2 genes.

In case of autosomal dominant inheritance, the causative variant is either de novo or, with clinically

unaffected parents, recurrent due to germ line mosaicism in a parent. The empirical recurrence risk is a

mixture of recurrence risk due to gonadal mosaicism and autosomal recessive inheritance.

Pepin et al.[1997] reported a 2% empirical recurrence risk of lethal OI for families with one previous

affected child. In a recent study [Pyott et al., 2011b], a recurrence rate of 1.3% was observed for lethal

OI (based on 1 recurrence in 76 families with 1 previous affected child). Interestingly, it was reported

that approximately 16% of parents with one affected child due to a causative variant in COL1A1/2, was

mosaic in somatic cells [Pyott et al., 2011b] resulting in a higher risk of recurrence of OI in these families.

Identifying the causative variant(s) and DNA analysis of the parents in case of a causative variant in

COL1A1/2 is necessary for an accurate estimate of the recurrence risk, which is important for genetic

counseling in case of early prenatal diagnosis and preimplantation genetic diagnosis [Pyott et al., 2011b].

Dijk_Opmaak.indd 32 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 16 T1 - Black CyanMagentaYellow

Page 34: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 33

Management

When the diagnosis OI has been established, the affected individual should preferentially be evaluated

by a multidisciplinary team [Steiner et al., 1993]. Important members of the team would be orthopedic

surgeons, rehabilitation physicians, endocrinologists, physical therapists and pediatricians. Referral to

other disciplines can take place upon individual needs and for routine surveillance such as dental controls.

Management consists of pharmacological treatment, orthopaedic treatment, physical medicine, dental

treatment, treatment for hearing loss, and prevention of primary (e.g. basilar impression) and secondary

(e.g. problems due to general anaesthesia) complications [Steiner et al., 1993].

Pharmacological treatment

Oral and intravenous bisphosphonates are commonly prescribed for all OI types, adults and children.

The main rationale for bisphosphonate therapy is based on several (not placebo-controlled) clinical

trials that showed improvements of bone mineral density (BMD) in individuals with OI. Nitrogenous

bisphosphonates disrupt osteoclast formation, survival and cytoskeletal dynamics and non-nitrogenous

bisphosphonates initiate osteoclast apoptosis. A recently published systematic review of bisphosponate

treatment in OI OI [Philippi et al., 2009] concluded, that in a relatively small group of patients, there is

significant improvement in BMD in individuals affected with OI and treated with oral or intravenous

bisphosphonates. However, the most important question arising is whether increase in BMD leads to

fracture reduction and functional improvement; this has not been answered yet and warrants further

investigations [Philippi et al., 2009]. The use of growth hormone to affect short stature in types III and IV

OI [Marini et al., 2003] is still under active investigation [Marini et al., 2010a].

Orthopaedic treatment

In case of decreased bone mineralization, high fracture frequency and/or bone deformities,

intramedullary (IM) rods will be placed in the majority of patients with OI types III and IV and sometimes

with OI type I [Monti et al., 2010]. These rods are inserted in the bone marrow canal in the centre of the

long bones and are used to align and stabilize fractures (fig. 6II-A and 7 C-D). Severe scoliosis occurs

most often in patients with OI type III (figure 6-IIE) and sometimes IV and appears not to be related to the

number of vertebral compression fractures. Since severe scoliosis can lead to pulmonary insufficiency,

corrective surgery is often performed when the curvature is less than 60º [Marini et al., 2010a]. In case of

anaesthesia, precautions should be undertaken during intubation because of possible cervical fragility

and the patient should be carefully monitored during surgery, because of the (possibly weak) association

with hyperthermia during anaesthesia [Oakley and Reece, 2010]. Non-surgical management consists of

bracing and splinting interventions [Monti et al., 2010].

Physical medicine treatment (rehabilitation)

An intensive rehabilitation program is necessary especially in OI types III and IV [Moni et al., 2010]

with early intervention such as correct positioning of the child and proper head support, muscle

strengthening (isotonic) and aerobic conditioning [Marini et al., 2010a].

Dijk_Opmaak.indd 33 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 17 T1 - Black CyanMagentaYellow

Page 35: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I34

Dental treatment

In patients with dentinogenesis imperfecta (DI), fractures and excessive wear of fragile teeth often

occurs (figure 9). This can be treated by capping teeth with hard polymers in order to prevent infections,

facial deformities due to the loss of (parts of ) teeth and/or malocclusion [Monti et al., 2010].

figure 9: Dentinogenesis imperfecta in a patient with OI type III

Treatment for hearing loss

Hearing loss often occurs in adults with OI. Initially it concerns conductive hearing loss, but as the

hearing loss progresses, a significant sensorineural component emerges. Surveillance for hearing loss is

advised after adolescence every 3-5 years [Steiner et al., 1993]. Initially hearing aids will be sufficient. As

the hearing loss progresses, stapedectomy can be considered for which successful outcomes have been

reported, however long term hearing restoration may be unsatisfactory due to fragility of the ossicular

middle ear structures. Cochlear implantation has been reported because of the sensorineural hearing

loss but data are too limited to draw conclusions on effectiveness [Marini et al., 2010a].

Basilar invagination

Basilar invagination is a rare complication occurring in adults with OI type III when the top of the

C2 vertebra migrates upward which may lead to (partial) closure of the foramen magnum with

hydrocephalus, pressure on the brain stem, syrinx formation and hindbrain herniation, requiring

ventricular shunt placement or surgery. Only prolonged orthotic immobilization has been proven to

stabilize symptoms and arrest progression [Monti et al., 2010].

Pregnancy and mode of delivery

Pregnant women with OI who have significant skeletal deformity and short stature should be monitored

in high-risk prenatal care centers, not only for maternal reasons but also because of the risk that the

fetus is affected with OI as well [Steiner et al., 1993]. In a large retrospective study of 167 pregnancies

in which a child was affected with OI, there was an unusually high rate of breech presentation at term

(37%). Cesarean delivery did not decrease fracture rates at birth in infants with OI types I, III and IV nor

did it prolong survival in OI type II. Prenatal diagnosis did not influence the mode of delivery in most

instances [Cubert et al., 2001].

Dijk_Opmaak.indd 34 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 17 T1 - Black CyanMagentaYellow

Page 36: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 35

future prospects for OI therapy

Gene therapy: silencing or replacement of the allele containing the causative variant

90% of OI patients have a causative variant in the COL1A1/2 genes encoding either the (pro)-α1 chain

or the (pro)-α2 chain of collagen type I. OI types II-IV result from intertwining of mutated and normal

collagen type I chains resulting in production of abnormal collagen type I (dominant-negative effect)

whereas OI type I is mostly due to decreased or nonexpression of one COL1A1 allele (haploinsufficiency

effect). It was therefore thought that the future treatment of OI should consist of so-called antisense

suppression therapy, directed towards decreasing or silencing of the allele containing the causative

variant. This would transform a severe type of OI into a mild OI type. Various murine models (oim [Saban

et al., 1996], brtl IV [Kozloff et al., 2004], crtap-/- [Morello et al., 2006], Mov-13 [Bonadio et al., 1990],

OASIS-/- [Sekiya et al., 2010]) have been developed, which can be used for the purpose of investigating

gene therapy in OI. However, the various antisense techniques (short oligonucleotides [Wang et al.,

1996], ribozymes [Grassi et al., 1997], silencing RNA [Millington-Ward et al., 2004]) all lack true specificity

against the mutant transcript. Others problems are stability of the antisense molecules apart from

specific problems which each technique. As such, antisense suppression therapy is currently limited to

in vitro studies [Monti et al., 2010].

Another approach is the replacement of cells harbouring the causative variant with normal cells by bone

marrow transplantation (BMT) leading to engraftment of functional mesenchymal stem cells (MSC) that

differentiate into bone cells [Undale et al., 2009]. This approach may hold promises for OI treatment

[Niyibizi & Li, 2009] as is illustrated by various in vitro studies and in vivo studies (children with severe OI)

[Horwitz et al., 1999; 2001; 2002; Panaroni et al., 2009]. Further studies are warranted to evaluate whether

gene therapy, BMT, mesenchymal stem cell transplantation or even de-and redifferentiation of somatic

cells [Masaki et al., 2010] are possible treatment options for patients with OI (chapter V-1).

CONCLuSION

Osteogenesis Imperfecta is a complex hereditary disease with; (i) a remarkable clinical variability

warranting a logical classification system; (ii) causative recessive or dominant variants in 8 different

genes with 6 of 8 genes encoding proteins involved in collagen type I biosynthesis; (iii) a need

for multidisciplinary management and further investigations of therapeutic approaches such as

bisphosphonates, growth hormone therapy and gene therapy.

Dijk_Opmaak.indd 35 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 18 T1 - Black CyanMagentaYellow

Page 37: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I36

rEfErENCES

Aase JM. Diagnostic dysmorphology. Plenum medical book company. New York and London (1990).

Ablin DS, Greenspan A, Reinhart M, Grix A. Differentiation of child abuse from osteogenesis imperfecta. AJR Am.J.Roentgenol. 154: 1035-1046 (1990).

Alanay Y, Avaygan H, Camacho N, Utine GE, Boduroglu K, Aktas D,et al: Mutations in the gene encoding the RER protein FKBP65 cause autosomal-recessive osteogenesis imperfecta. Am.J.Hum.Genet. 86: 551-559 (2010).

Baldridge D, Schwarze U, Morello R, Lennington J, Bertin TK, Pace JM, et al: CRTAP and LEPRE1 mutations in recessive osteogenesis imperfecta. Hum.Mutat. 29: 1435-1442. (2008).

Baljet B. Willem Vrolik as a teratologist. Ned.Tijdschr.Geneeskd. 128: 1530-1534 (1984).

Baljet B. Aspects of the history of osteogenesis imperfecta (Vrolik’s syndrome). Ann.Anat. 184: 1-7 (2002).

Bank RA, Robins SP, Wijmenga C, Breslau-Siderius LJ, Bardoel AF, van der Sluijs HA, et al: Defective collagen crosslinking in bone, but not in ligament or cartilage, in Bruck syndrome: indications for a bone-specific telopeptide lysyl hydroxylase on chromosome 17. Proc.Natl.Acad.Sci.U.S.A 96: 1054-1058 (1999).

Barnes AM, Carter EM, Cabral WA, Weis M, Chang W, Makareeva E, et al: Lack of cyclophilin B in osteogenesis imperfecta with normal collagen folding. N.Engl.J.Med. 362: 521-528 (2010).

Barnes AM, Chang W, Morello R, Cabral WA, Weis M, Eyre DR, et al: Deficiency of cartilage-associated protein in recessive lethal osteogenesis imperfecta. N.Engl.J.Med. 355: 2757-2764 (2006).

Barron MJ, McDonnell ST, Mackie I, Dixon MJ. Hereditary dentine disorders: dentinogenesis imperfecta and dentine dysplasia. Orphanet.J.Rare.Dis. 3: 31 (2008).

Becker J, Semler O, Gilissen C, Li Y, Bolz HJ, Giunta C, et al: Exome Sequencing Identifies Truncating Mutations in Human SERPINF1 in Autosomal-Recessive Osteogenesis Imperfecta. Am.J.Hum.Genet. (2011).

Bilo RAC, Robben, SGF, van Rijn RR. Forensic Aspects of Paediatric Fractures; Differentiating Accidental Trauma from Child Abuse. 1st edition. 2010. Heidelberg Dordrecht London New York, Springer.

Bonadio J, Saunders TL, Tsai E, Goldstein SA, Morris-Wiman J, Brinkley L, et al: Transgenic mouse model of the mild dominant form of osteogenesis imperfecta. Proc.Natl.Acad.Sci.U.S.A 87: 7145-7149 (1990).

Breslau-Siderius EJ, Engelbert RH, Pals G, van der Sluijs JA. Bruck syndrome: a rare combination of bone fragility and multiple congenital joint contractures. J.Pediatr.Orthop.B 7: 35-38 (1998).

Byers PH, Bonadio JF, Cohn DH, Starman BJ, Wenstrup RJ, Willing MC. Osteogenesis imperfecta: the molecular basis of clinical heterogeneity. Ann.N.Y.Acad.Sci. 543: 117-128 (1988).

Cabral WA, Chang W, Barnes AM, Weis M, Scott MA, Leikin S, et al: Prolyl 3-hydroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfecta. Nat.Genet. 39: 359-365 (2007).

Canty EG, Kadler KE. Procollagen trafficking, processing and fibrillogenesis. J.Cell Sci. 118: 1341-1353 (2005).

Chan CC, Green WR, de la Cruz ZC, Hillis A. Ocular findings in osteogenesis imperfecta congenita. Arch.Ophthalmol. 100: 1458-1463 (1982).

Chapman S, Hall CM. Non-accidental injury or brittle bones. Pediatr. Radiol 27: 106-110 (1997).

Christiansen HE, Schwarze U, Pyott SM, AlSwaid A, Al BM, Alrasheed S, et al: Homozygosity for a missense mutation in SERPINH1, which encodes the collagen chaperone protein HSP47, results in severe recessive osteogenesis imperfecta. Am.J.Hum.Genet. 86: 389-398 (2010).

Chu ML, Williams CJ, Pepe G, Hirsch JL, Prockop DJ, Ramirez F. Internal deletion in a collagen gene in a perinatal lethal form of osteogenesis imperfecta. Nature 304: 78-80 (1983).

Cohen-Solal L, Bonaventure J, Maroteaux P. Dominant mutations in familial lethal and severe osteogenesis imperfecta. Hum.Genet. 87: 297-301 (1991).

Cohn DH, Starman BJ, Blumberg B, Byers PH. Recurrence of lethal osteogenesis imperfecta due to parental mosaicism for a dominant mutation in a human type I collagen gene (COL1A1). Am.J.Hum.Genet. 46: 591-601 (1990).

Cubert R, Cheng EY, Mack S, Pepin MG, Byers PH. Osteogenesis imperfecta: mode of delivery and neonatal outcome. Obstet.Gynecol. 97: 66-69 (2001).

Dalgleish R. The human type I collagen mutation database. Nucleic Acids Res. 25: 181-187 (1997).

Dalgleish R. The Human Collagen Mutation Database 1998. Nucleic Acids Res. 26: 253-255 (1998).

Drogemuller C, Becker D, Brunner A, Haase B, Kircher P, Seeliger F, et al: A missense mutation in the SERPINH1 gene in Dachshunds with osteogenesis imperfecta. PLoS.Genet. 5: e1000579 (2009).

Eichholtz W, Muller D. Electron microscopy findings on the cornea and sclera in osteogenesis imperfecta. Klin.Monbl.Augenheilkd. 161: 646-653 (1972).

Dijk_Opmaak.indd 36 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 18 T1 - Black CyanMagentaYellow

Page 38: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 37

Engelbert RH, van der Graaf Y, van ER, Beemer FA, Helders PJ. Osteogenesis imperfecta in childhood: impairment and disability. Pediatrics 99: E3 (1997).

Engel J, Prockop DJ. The zipper-like folding of collagen triple helices and the effects of mutations that disrupt the zipper. Annu.Rev.Biophys.Biophys.Chem. 20: 137-152 (1991).

Evereklioglu C, Madenci E, Bayazit YA, Yilmaz K, Balat A, Bekir NA. Central corneal thickness is lower in osteogenesis imperfecta and negatively correlates with the presence of blue sclera. Ophthalmic Physiol Opt. 22: 511-515 (2002).

Gilbert SF. Developmental biology, 9th edition. Published by Sinauer Associates, Inc. Sunderland, Massachusetts USA (2010).

Glorieux FH, Bishop NJ, Plotkin H, Chabot G, Lanoue G, Travers R. Cyclic administration of pamidronate in children with severe osteogenesis imperfecta. N.Engl.J.Med. 339: 947-952 (1998).

Glorieux FH, Rauch F, Plotkin H, Ward L, Travers R, Roughley P, et al: Type V osteogenesis imperfecta: a new form of brittle bone disease. J.Bone Miner.Res. 15: 1650-1658 (2000).

Glorieux FH, Ward LM, Rauch F, Lalic L, Roughley PJ, Travers R. Osteogenesis imperfecta type VI: a form of brittle bone disease with a mineralization defect. J.Bone Miner.Res. 17: 30-38 (2002).

Grassi G, Forlino A, Marini JC. Cleavage of collagen RNA transcripts by hammerhead ribozymes in vitro is mutation-specific and shows competitive binding effects. Nucleic Acids Res. 25: 3451-3458 (1997).

Ha-Vinh R, Alanay Y, Bank RA, Campos-Xavier AB, Zankl A, Superti-Furga A, Bonafe L. Phenotypic and molecular characterization of Bruck syndrome (osteogenesis imperfecta with contractures of the large joints) caused by a recessive mutation in PLOD2. Am.J.Med.Genet.A 131: 115-120 (2004).

Homan EP, Rauch F, Grafe I, Lietman C, Doll JA, Dawson B, et al: Mutations in SERPINF1 cause Osteogenesis imperfecta Type VI. J Bone Miner Res. E-pub ahead of print.

Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, et al: Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc.Natl.Acad.Sci.U.S.A 99: 8932-8937 (2002).

Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, et al: Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat.Med. 5: 309-313 (1999).

Horwitz EM, Prockop DJ, Gordon PL, Koo WW, Fitzpatrick LA, Neel MD, et al: Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta. Blood 97: 1227-1231 (2001).

Hyry M, Lantto J, Myllyharju J. Missense mutations that cause Bruck syndrome affect enzymatic activity, folding, and oligomerization of lysyl hydroxylase 2. J.Biol.Chem. 284: 30917-30924 (2009).

Ishikawa Y, Wirz J, Vranka JA, Nagata K, Bachinger HP. Biochemical characterization of the prolyl 3-hydroxylase 1.cartilage-associated protein.cyclophilin B complex. J.Biol.Chem. 284: 17641-17647 (2009).

Kelley BP, Malfait F, Bonafe L, Baldridge D, Homan E, Symoens S, et al: Mutations in FKBP10 cause recessive osteogenesis imperfecta and bruck syndrome. J.Bone Miner.Res. 26: 666-672 (2011).

Korkko J, Ala-Kokko L, De Paepe A, Nuytinck L, Earley J, Prockop DJ. Analysis of the COL1A1 and COL1A2 genes by PCR amplification and scanning by conformation-sensitive gel electrophoresis identifies only COL1A1 mutations in 15 patients with osteogenesis imperfecta type I: identification of common sequences of null-allele mutations. Am.J.Hum.Genet. 62: 98-110 (1998).

Kozloff KM, Carden A, Bergwitz C, Forlino A, Uveges TE, Morris MD, et al: Brittle IV mouse model for osteogenesis imperfecta IV demonstrates postpubertal adaptations to improve whole bone strength. J.Bone Miner.Res. 19: 614-622 (2004).

Kuurila K, Kaitila I, Johansson R, Grenman R. Hearing loss in Finnish adults with osteogenesis imperfecta: a nationwide survey. Ann.Otol.Rhinol.Laryngol. 111: 939-946 (2002).

Lanting PJ, Borsboom PC, te Meerman GJ, ten Kate LP. Decreased scattering coefficient of blue sclerae. Clin.Genet. 27: 187-190 (1985).

Lapunzina P, Aglan M, Temtamy S, Caparros-Martin JA, Valencia M, Leton R, et al: Identification of a frameshift mutation in Osterix in a patient with recessive osteogenesis imperfecta. Am.J.Hum.Genet. 87: 110-114 (2010).

Levin LS, Salinas CF, Jorgenson RJ. Classification of osteogenesis imperfecta by dental characteristics. Lancet 1: 332-333 (1978).

Lowenstein EJ. Osteogenesis imperfecta in a 3,000-year-old mummy. Childs Nerv.Syst. 25: 515-516 (2009).

Looser E. Zur Kenntnis der Osteogenesis Imperfecta Congenita et Tarda (sogenannte idiopathische Osteopsatyrosis. Mittlg Grenzgebiete Med Chir 15:161-207(1906).

Marini JC.Osteogenesis Imperfecta (2010a). Available at https://www.endotext.org/parathyroid/parathyroid17/parathyroidframe17.htm

Dijk_Opmaak.indd 37 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 19 T1 - Black CyanMagentaYellow

Page 39: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter I38

Marini JC, Bordenick S, Chrousos GP. Endocrine aspects of growth deficiency in OI. Connect.Tissue Res. 31: S55-S57 (1995).

Marini JC, Cabral WA, Barnes AM. Null mutations in LEPRE1 and CRTAP cause severe recessive osteogenesis imperfecta. Cell Tissue Res. 339: 59-70. (2010b)

Marini JC, Cabral WA, Barnes AM, Chang W. Components of the collagen prolyl 3-hydroxylation complex are crucial for normal bone development. Cell Cycle 6: 1675-1681 (2007a).

Marini JC, Forlino A, Cabral WA, Barnes AM, San Antonio JD, Milgrom S, et al: Consortium 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. 28: 209-221 (2007b).

Marini JC, Hopkins E, Glorieux FH, Chrousos GP, Reynolds JC, Gundberg CM, Reing CM. Positive linear growth and bone responses to growth hormone treatment in children with types III and IV osteogenesis imperfecta: high predictive value of the carboxyterminal propeptide of type I procollagen. J.Bone Miner.Res. 18: 237-243 (2003).

Marlowe A, Pepin MG, Byers PH. Testing for osteogenesis imperfecta in cases of suspected non-accidental injury. J.Med.Genet. 39: 382-386 (2002).

Masaki I, Ji-Dong F, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, Srivastava. Direct repogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142: 375-386 (2010).

Millington-Ward S, McMahon HP, Allen D, Tuohy G, Kiang AS, Palfi A, Kenna PF, et al: RNAi of COL1A1 in mesenchymal progenitor cells. Eur.J.Hum.Genet. 12: 864-866 (2004).

Morello R, Bertin TK, Chen Y, Hicks J, Tonachini L, Monticone M, et al: CRTAP is required for prolyl 3- hydroxylation and mutations cause recessive osteogenesis imperfecta. Cell 127: 291-304 (2006).

Monti E, Mottes M, Fraschini P, Brunelli P, Forlino A, Venturi G, et al: Current and emerging treatments for the management of osteogenesis imperfecta. Ther.Clin.Risk Manag. 6: 367-381 (2010).

Morgan JA, Marcus PS. Prenatal diagnosis and management of intrauterine fracture. Obstet.Gynecol.Surv. 65: 249-259 (2010).

Mundlos S, Otto F, Mundlos C, Mulliken JB, Aylsworth AS, Albright S, et al: Mutations involving the transcription factor CBFA1 cause cleidocranial dysplasia. Cell 89: 773-779 (1997).

Niyibizi C and Li F: Potential implications of cell therapy for osteogenesis imperfect. Int J Clin Rheumatol: 57-66 (2009).

Oakley I, Reece LP. Anesthetic implications for the patient with osteogenesis imperfecta. AANA.J. 78: 47-53 (2010).

Panaroni C, Gioia R, Lupi A, Besio R, Goldstein SA, Kreider J, et al: In utero transplantation of adult bone marrow decreases perinatal lethality and rescues the bone phenotype in the knockin murine model for classical, dominant osteogenesis imperfecta. Blood 114: 459-468 (2009).

Pedersen U, Bramsen T. Central corneal thickness in osteogenesis imperfecta and otosclerosis. ORL J.Otorhinolaryngol.Relat Spec. 46: 38-41 (1984).

Peltier LF. The classic: congenital osteomalacia. Olaus Jacob Ekman. Clin.Orthop.Relat Res. 3-5 (1981).

Pepin M, Atkinson M, Starman BJ, Byers PH. Strategies and outcomes of prenatal diagnosis for osteogenesis imperfecta: a review of biochemical and molecular studies completed in 129 pregnancies. Prenat.Diagn. 17: 559-570 (1997).

Philippi CA et al. Bisphosphonate therapy for osteogenesis imperfecta. The Cochrane Collaboration. Published by John Wiley&Sons,Ltd (2009).

Price ER, Zydowsky LD, Jin MJ, Baker CH, McKeon FD, Walsh CT. Human cyclophilin B: a second cyclophilin gene encodes a peptidyl-prolyl isomerase with a signal sequence. Proc.Natl.Acad.Sci.U.S.A 88: 1903-1907 (1991).

Prockop DJ, Constantinos D. et al. Type I Procollagen: the gene-protein system that harbors most of the mutations causing osteogenesis imperfecta and probably more common heritable disorders of connective tissue. Am. J. Med. Gen. 34: 60-67 (1989).

Pyott SM, Schwarze U, Christiansen HE, Pepin MG, Leistritz DF, Dineen R, et al: Mutations in PPIB (cyclophilin B) delay type I procollagen chain association and result in perinatal lethal to moderate osteogenesis imperfecta phenotypes. Hum.Mol.Genet. (2011a).

Pyott, SM, Pepin MG, Schwarze U,Yang K, Gretchen S, Byers PH. Recurrence of perinatal lethal osteogenesis imperfect in sibships: Parsing the risk between parental mosaicism for dominant mutations and autosomal recessive inheritance. Genetics in Medicine 13: 125-130 (2011b).

Rauch F, Glorieux FH. Osteogenesis imperfecta. Lancet 363: 1377-1385 (2004).

Ross MH, Romrell RJ, Kaye GI. Histology, a text and atlas. Third edition. Lippincott Williams & Wilkins, Baltimore, Maryland. 21202, USA (1995).

Saban J, Zussman MA, Havey R, Patwardhan AG, Schneider GB, King D. Heterozygous oim mice exhibit a mild form of osteogenesis imperfecta. Bone 19: 575-579 (1996).

Sarathchandra P, Pope FM, Ali SY. Morphometric analysis of type I collagen fibrils in the osteoid of osteogenesis imperfecta. Calcif.Tissue Int. 65: 390-395 (1999).

Dijk_Opmaak.indd 38 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 19 T1 - Black CyanMagentaYellow

Page 40: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Introduction 39

Sekiya H, Murakami T, Saito A, Hino S, Tsumagari K, Ochiai K, Imaizumi K. Effects of the bisphosphonate risedronate on osteopenia in OASIS-deficient mice. J.Bone Miner.Metab 28: 384-394 (2010).

Shaheen R, Al-Owain M, Sakati N, Alzayed ZS, Alkuraya FS. FKBP10 and Bruck syndrome: phenotypic heterogeneity or call for reclassification? Am.J.Hum.Genet. 87: 306-307 (2010).

Sillence DO, Barlow KK, Garber AP, Hall JG, Rimoin DL. Osteogenesis imperfecta type II delineation of the phenotype with reference to genetic heterogeneity. Am.J.Med.Genet. 17: 407-423 (1984).

Sillence DO, Senn A, Danks DM. Genetic heterogeneity in osteogenesis imperfecta. J.Med.Genet. 16: 101-116 (1979).

Spranger J., Brill P., Poznanski A. Bone dysplasias. An atlas of genetic disorders of skeletal development, Oxford University Press, Oxford (2003).

Steiner RD, Pepin MG, Byers PH. Osteogenesis Imperfecta. Genereviews (1993).

Sun HB, Chen JC, Liu Q, Guo MF, Zhang HP. Substance P stimulates differentiation of mice osteoblast through up-regulating Osterix expression. Chin J.Traumatol. 13: 46-50 (2010).

Sykes B, Francis MJ, Smith R. Altered relation of two collagen types in osteogenesis imperfecta. N.Engl.J.Med. 296: 1200-1203 (1977).

Sykes B, Ogilvie D, Wordsworth P, Wallis G, Mathew C, Beighton P, et al: Consistent linkage of dominantly inherited osteogenesis imperfecta to the type I collagen loci: COL1A1 and COL1A2. Am.J.Hum.Genet. 46: 293-307 (1990).

Teitelbaum SL, Kraft WJ, Lang R, Avioli LV. Bone collagen aggregation abnormalities in osteogenesis imperfecta. Calcif.Tissue Res. 17: 75-79 (1974).

Undale AH, Westendorf JJ, Yaszemski MJ, Khosla S. Mesenchymal stem cells for bone repair and metabolic bone diseases. Mayo Clin.Proc. 84: 893-902 (2009).

Unger S, Scherer G, Superti-Furga A. Campomelic dysplasia. Genereviews (1993).

van der Slot AJ, Zuurmond AM, Bardoel AF, Wijmenga C, Pruijs HE, Sillence DO, et al: Identification of PLOD2 as telopeptide lysyl hydroxylase, an important enzyme in fibrosis. J.Biol.Chem. 278: 40967-40972 (2003).

Van Dijk FS, Nesbitt IM, Nikkels PG, Dalton A, Bongers EM, van de Kamp JM, et al: CRTAP mutations in lethal and severe osteogenesis imperfecta: the importance of combining biochemical and molecular genetic analysis. Eur.J.Hum.Genet. 17: 1560-1569. (2009a)

Van Dijk FS, Nesbitt IM, Zwikstra EH, Nikkels PG, Piersma SR, Fratantoni SA, et al: PPIB mutations cause severe osteogenesis imperfecta. Am.J.Hum.Genet. 85: 521-527 (2009b).

van Dijk FS, Huizer M, Kariminejad A, Marcelis CL, Plomp AS, Terhal PA, et al: Complete COL1A1 allele deletions in Osteogenesis Imperfecta. Genet. Med. 12:736-741 (2010a).

van Dijk F, Nikkels PG, den Hollander NS, Nesbitt IM, van Rijn RR, Cobben JM, Pals G. Lethal/ Severe Osteogenesis Imperfecta in a Large Family: A Novel Homozygous LEPRE1 Mutation and Bone Histological Findings. Pediatr.Dev.Pathol (2010b).

Van Dijk FS, Pals G, van Rijn RR, Nikkels PG, Cobben JM. Classification of Osteogenesis Imperfecta revisited. Eur.J.Med.Genet. 53: 1-5 (2010c).

Van Dijk FS, Byers PH, Dalgleish R, Malfait F, Maugeri A, Rohrbach M. EMQN best practice guidelines for the laboratory diagnosis of osteogenesis imperfecta. Accepted in: Eur. J. Hum. Genet. E-pub ahead of print.

Viora E, Sciarrone A, Bastonero S, Errante G, Campogrande M, Botta G, Franceschini P. Increased nuchal translucency in the first trimester as a sign of osteogenesis imperfecta. Am.J.Med.Genet. 109: 336-337 (2002).

Wallis GA, Sykes B, Byers PH, Mathew CG, Viljoen D, Beighton P. Osteogenesis imperfecta type III: mutations in the type I collagen structural genes, COL1A1 and COL1A2, are not necessarily responsible. J.Med.Genet. 30: 492-496 (1993).

Wang Q, Marini JC. Antisense oligodeoxynucleotides selectively suppress expression of the mutant alpha 2(I) collagen allele in type IV osteogenesis imperfecta fibroblasts. A molecular approach to therapeutics of dominant negative disorders. J.Clin.Invest 97: 448-454 (1996).

Warman ML, Cormier-Daire V, Hall C, Krakow D, Lachman R, LeMerrer M. Nosology and classification of genetic skeletal disorders: 2010 revision. Am. J. Med. Gen 2011: 943-968 (2011).

Willaert A, Malfait F, Symoens S, Gevaert K, Kayserili H, Megarbane A, et al: Recessive osteogenesis imperfecta caused by LEPRE1 mutations: clinical documentation and identification of the splice form responsible for prolyl 3-hydroxylation. J.Med.Genet. 46: 233-241 (2009).

Zack P, Zack LR, Surtees R, Neville BG. A standardized tool to measure and describe scleral colour in osteogenesis imperfecta. Ophthalmic Physiol Opt. 27: 174-178 (2007).

Zankl A, Mornet E, Wong S. Specific ultrasonographic features of perinatal lethal hypophosphatasia. Am. J. Med. Gen. 146A: 1200-1204 (2008).

Zhou X, Zhang Z, Feng JQ, Dusevich VM, Sinha K, Zhang H, et al: Multiple functions of Osterix are required for bone growth and homeostasis in postnatal mice. Proc.Natl.Acad.Sci.U.S.A 107: 12919-12924 (2010).

Dijk_Opmaak.indd 39 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 20 T1 - Black CyanMagentaYellow

Page 41: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 40 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 20 T1 - Black CyanMagentaYellow

Page 42: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II

Dominant Osteogenesis Imperfecta

Dijk_Opmaak.indd 41 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 21 T1 - Black CyanMagentaYellow

Page 43: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 42 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 21 T1 - Black CyanMagentaYellow

Page 44: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta

van Dijk FS1, Huizer M1, Kariminejad A2, Marcelis CL3, Plomp AS4, Terhal PA5,

Meijers-Heijboer H1, Weiss MM1, van Rijn RR6, Cobben JM7, Pals G8*.

1 Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands 2 Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran

3 Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands 4 Department of Clinical Genetics, Academic Medical Centre, Amsterdam, the Netherlands

5 Department of Clinical Genetics, University Medical Centre Utrecht, the Netherlands6 Department of Pediatric Radiology, Academic Medical Centre, Amsterdam, the Netherlands

7 Department of Pediatrics, Academic Medical Centre, Amsterdam, the Netherlands8Centre for Connective Tissue Research, Department of Clinical Genetics, VU University Medical Centre,

Amsterdam, The Netherlands

Genet Med. 2010 Nov;12(11):736-41

II-1

Dijk_Opmaak.indd 43 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 22 T1 - Black CyanMagentaYellow

Page 45: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II44

AbSTrACT

Purpose: To indentify a molecular genetic cause in patients with a clinical diagnosis of Osteogenesis

Imperfecta (OI) type I/ IV .

Methods: We performed Multiplex Ligation Dependent Probe Amplification (MLPA) analysis of the

COL1A1 gene in a group of 106 index patients.

results: In four families with mild OI and no other phenotypic abnormalities a deletion of the

complete COL1A1 gene on one allele was detected, a molecular finding which to our knowledge has

not been described beforeapart from a larger chromosomal deletion detected by FISH encompassing

the COL1A1 gene in a patient with mild OI and other phenotypic abnormalities. Microarray analysis in

three of the four families showed that it did not concern a founder mutation.

Conclusion: The clinical picture of complete COL1A1 allele deletions is a comparatively mild type

of OI. As such, MLPA analysis of the COL1A1 gene is a useful additional approach to defining the

mutation in cases of suspected OI type I with no detectable mutation.

Dijk_Opmaak.indd 44 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 22 T1 - Black CyanMagentaYellow

Page 46: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta 45

INTrODuCTION

Osteogenesis Imperfecta (OI) constitutes a heterogeneous group of diseases characterized by

susceptibility to bone fractures with variable severity and presumed or proven defects in collagen type

I biosynthesis 1. The severity of OI ranges from perinatally lethal to occasional fractures 2. The apparent

clinical variability in OI has led to the development of the Sillence classification, initially in OI type I

(mild, dominantly inherited OI with bone fragility and blue sclerae), II (perinatal lethal), III (progressive

deforming) and IV (dominant with normal sclerae and mild deformity) 3. Depending on the age of

presentation, OI can be difficult to distinguish from some other genetic and non-genetic causes of

fractures including nonaccidental injury (NAI) 2, 4-6. Recently, rare autosomal recessive causes of lethal and

severe OI have been described 7-9 but in the majority of affected individuals OI is dominantly inherited

and caused by a heterozygous mutation in either of the two genes COL1A1 and COL1A2 encoding the

chains of type I collagen. Type I collagen is the major structural protein in bone, tendon, and ligamen. It

is first synthesized in the rough endoplasmic reticulum (rER) as type I procollagen, containing C and N

terminal propeptides. In the rER, the two alpha1 chains and the one alpha2 chain consisting of Gly-X-Y

triplets will fold in the C- to-N direction to form a triple helix. During folding, collagen is modified by,

among others, specific enzymes that hydroxylate lysine and proline residues and glycosylate hydroxylysyl

residues. This process is called posttranslational modification and it stops as soon as the chain in which

the residues are located is folded 10. After folding the procollagen molecules are transported through the

Golgi apparatus in the pericellular environment where cleavage of the N-and C-terminal propeptides

occurs and collagen molecules aggregate to form fibrils 11. At present more than 800 distinct mutations

in the COL1A1 and COL1A2 genes have been described to cause OI type II-IV 12. The two mildest forms

of OI, OI type I and IV account for considerably more than half of all OI cases13 . OI type II-IV cases are

mostly caused by glycine substitution mutations and splice site mutations, resulting in post-translational

overmodification and synthesis of abnormal collagen type I molecules. In contrast, OI type I is often

caused by a non-functional COL1A1 allele (null-allele) 14 due to mutations generating destabilization

and rapid degeneration of the mutant COL1A1 mRNA resulting in decreased amount of normal collagen

type I molecules 15,16. Both types of abnormalities (abnormal or decreased synthesis of collagen type

I) may be detected by electrophoresis of type I collagen synthesized by cultured dermal fibroblasts 17.

The presence of normal collagen type I molecules explains the fact that OI type I is the mildest type

of OI. OI type I is characterized clinically by increased bone fragility often leading to fractures, ranging

from few to 100 18, without secondary deformities in combination with blue sclera, conductive or mixed

hearing loss in late adolescence (approximaterly 50% of cases), short but also often normal height and

dentinogenesis imperfecta (approximately 60% of cases). Radiologically, in OI type I, bone fragility in

combination with generalized demineralization, slender shafts of tubular bones with thin cortex and

poorly trabeculated spongiose are evident. Furthermore, ossification of the cranial vault is often retarded

leading to a mosaic pattern of Wormian bones 19. Here, we describe four families with OI type I due to

a deletion of the complete COL1A1 allele detected by MLPA, which to our knowledge has not been

described in the literature before.

Dijk_Opmaak.indd 45 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 23 T1 - Black CyanMagentaYellow

Page 47: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II46

MATErIALS AND METHODS

Patients

Our DNA laboratory currently has material of 803 index patients with a clinical suspicion of OI. 430 of

these index patients have been referred with a clinical diagnosis of OI type I, or OI type I/IV. In 100

index patients a COL1A1 or COL1A2 mutation has been detected. In the remaining 330 index patients,

sequence analysis of the COL1A1/2 genes has not yet been completed, or no mutation has been found.

From those 330 index patients we performed MLPA analysis of the COL1A1 gene in 106 patients. In these

106 patients, four index patients with normal COL1A1 and COL1A2 sequencing results were found to

have a deletion of the COL1A1 gene. Written informed consent for publication was obtained from these

4 families according to the journal standard for patient consent. Physical examination was carried out in

combination with radiological surveys. Skin biopsies were available from individuals 1.II.1, 2.I.2, 3.I.2 and

4.I.1. 90 clinically normal individuals were used as a control group.

Collagen Type I Electrophoresis

Fibroblast cultures were established under standard conditions. Cells were seeded at 35,000 cells/ cm2.

Labelling of the fibroblasts and purification of the collagen molecules were performed as reported

elsewhere 20 with the exception of performing SDS-electrophoresis without 3% stacking gel. Dried gels

were analyzed on a phosphor imager (Molecular Dynamics). Collagen type I electrophoresis was carried

out in 39 healthy controls and in affected individuals 1.II.1, 2.I.2, 3.I.2 and 4.I.1.

Genomic DNA Analysis

Genomic DNA sequencing of the COL1A1 (NG_007400.1) and COL1A2 (NG_007405.1) genes was

performed in individuals 1.II.1, 2.I.2 and 3.I.2. Primer sequences are available on request.

Complementary DNA Analysis

RNA was isolated from primary fibroblasts cultured in the presence or absence of cycloheximide (5mg/

ml) using the mini RNA isolation II kit (Baseclear) according to the manufacturers instructions. To identify

alternatively spliced transcript, cDNA was prepared with oligo-dT-primer and SuperscriptTMII RT reverse

transcriptase (Invitrogen). PCR products were sequenced using a 3730 sequencing system (Applied

Biosystems). cDNA analysis was performed in individuals 2.I.2, 3.I.2 and 4.I.1

MLPA

MLPA analysis was performed as described elsewhere 21 with the SALSA MLPA kit P271-B1 COL1A1 that

contains 33 probes in the COL1A1 gene of which 26 were used in this study and 7 reference probes

widely spread over the genome (MRC-Holland, see www.mlpa.com).

Microarray

The COL1A1 deletions in families 2-4 were further characterized by high-resolution microarray using

Array CGH.No permission was obtained from family 1 to perform Microarray. Labelling and hybridization

Dijk_Opmaak.indd 46 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 23 T1 - Black CyanMagentaYellow

Page 48: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta 47

procedures were performed according to Buffart et al. 22 Briefly, 500 ng genomic DNA was labelled

according to the manufacturer’s instructions using the ENZO Genomic DNA Labelling kit (Enzo Life

Sciences). Cy3 label was used for the patient’s DNA and Cy5 label for reference DNA (DNA isolated from

blood obtained from eighteen healthy females). Hybridization of the patient was performed onto 180k

arrays with each array containing approximately 170000 in situ synthesized 60-mer oligonucleotides,

representing 13 KB overall median probe spacing (Agilent Technologies, Palo Alto, CA, genome

browser build hg 18). Images of the arrays were acquired using a micro array scanner G2505B (Agilent

technologies) and image analysis was performed using feature extraction software (version 10.5; Agilent

Technologies). The Agilent CGH-v4_91 protocol was applied using default settings. Data analysis was

performed with Agilent analysis software (Agilent Genomic Workbench 5.0, Agilent Technologies, Inc.),

with threshold 4.0, algorithm ADM-2 and aberration filter settings minimum number of probes in region

2 and minimum absolute average log ratio for region 0.3.

Marker Analysis

Genetic analysis (Poweplex ® 16 System, Promega, Madison, Wisconsin, United States) was performed to

investigate monozygosity in twin sisters II:1 and II:2 from family 1.

rESuLTS

Phenotypical Analysisfamily 1

Patient II.1

Individual II.1 (figure 1) is a 10- year old girl with OI type I. A fracture of the femur was noted shortly

after birth. Until now she has had eight fractures. Her length is 128.5 cm (-2.5 SD), head circumference

is 53.5 cm (0 SD) and bone density of the hips and lumbar vertebral column is between -4 and -7 SD as

measured by Dual X-ray absorptometry (DXA). She has Wormian bones, a scoliosis and grey-blue sclerae

typical for OI. She has no dentinogenesis imperfecta.

Patient II.2

Individual II.2 (figure 1) is the monozygous twin sister of II.I. She never had fractures nor other signs of

osteogenesis imperfecta. Her length is 143.5 cm (-1 SD), head circumference is 53.2 cm ( 0 SD).

family 2

Patient I.2

Affected individual I.2 (figure 1) is a 44-year-old woman. She was diagnosed with OI type I because of

multiple fractures in childhood in combination with blue sclerae. She has had 17 fractures in total, with

her last fracture occurring at the age of 34 years. She has one-sided hearing loss. At the age of 43 years

she had a cerebrovascular accident which resulted in hemiparesis.

Patient II.1

The daughter II.1 (figure 1) has had 2 fractures from birth until the age of five years. Both fractures

occurred after trauma. She has grey-blue sclera but no other features of OI.

Dijk_Opmaak.indd 47 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 24 T1 - Black CyanMagentaYellow

Page 49: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II48

family 3

Patient I.2

Affected individual I:2 (figure 1) is a 37-year-old woman. Her length is 170 cm (0 SD). She has had

one fracture at the age of 7 years. Normal bone density measured by DXA was noted. She experiences

frequent luxations of her knee and ankle joints. Her face has a mild triangular shape and her sclerae

are blue. She has extensive caries in combination with enamel loss. Her two daughters have not had

any fractures, but they have blue sclerae, hypermobility, and enamel loss. No abnormalities on skeletal

radiographs of the daughters were noted. Bone density measured by DXA appeared to be reduced in

both children. No biochemical or molecular diagnostic testing has been performed in these children

family 4

Patient I.1

Affected individual I.1 (figure 1) is a 40-year-old male. His length is 168 cm (0 SD). He has had 7 fractures,

all occurring before the age of 12 years. His sclerae are blue. He has no hearing loss or dentinogenesis

imperfecta. No radiographs are available.

Patient II.1

Affected individual II.1 (figure 1E) is a 1-year-old boy. He has blue sclerae and he has had no fractures.

figure 1: Pedigrees of family 1-4.

Dijk_Opmaak.indd 48 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 24 T1 - Black CyanMagentaYellow

Page 50: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta 49

The individuals that have a COL1A1 allele deletion are marked with an asterisk and the individuals that

are clinically affected with OI are depicted in black. Individuals II:1 and II:2 from family 3 are marked

with a question mark since they have no clear diagnosis of OI and no molecular genetic diagnosis has

been performed.

Electrophoresis of Collagen Type I

Collagen electrophoresis revealed a reduced but somewhat variable level of type I collagen synthesis.

This is expressed in table 1 by A: the percentage collagen type I of total collagen production (=(collagen

alpha-1(I) + collagen alpha-2(I) /( collagen alpha-1(I) + collagen alpha-2(I) + collagen alpha-1(III) )) and b:

the percentage collagen alpha-1(I) of total collagen production (=collagen alpha-1(I) / (collagen alpha-

1(I) + collagen alpha-2(I) + collagen alpha-1(III)). In 39 healthy individuals, A was 91% and b was 63%,

both with a standard deviation of 3% .

Molecular Genetic Analysis

Molecular genetic analysis with complementary and genomic DNA sequencing revealed no mutations

in the COL1A1/2 genes.

MLPA

By MLPA a deletion of the complete COL1A1 allele (figure 2, supplementary table 1) was detected in

individuals II:1 and II:2 from family 1, I:2 and II:1 from family 2, I:2 from family 3, I:1 and II:1 from family 4

(see also figure 1). The average peak ratio of all COL1A1 probes combined in 1.II.2 (0.67) and of 3.I.2 (0.59)

may point to mosaicism (supplementary table 1). Complete COL1A1 allele deletions were not detected

in 180 chromosomes from clinically normal controls.

Microarray Analysis

Further characterization of the COL1A1 allele deletion using high resolution microarray revealed that

the deletions had different sizes (182-217 kb (family 2), 512-353 kb (family 3), 451-481 kb (family 4))

containing multiple other genes (figure 3), which were studied by using the UCSC browser at http://

genome.ucsc.edu/23.

The deletion in family 2 encompasses the following reference sequence genes: ITGA3(NM_002204),

PDK2(NM_002611.3), SAMD14(NM_174920.2), PPP1R9B(NM_032595.3), SGCA(NM_000023), HILS1

(NR_024193), COL1A1(NM_000088.3)

The deletion in family 3 encompasses the following reference sequence genes: TAC4 (NM_001077504.1)

DLX4(NM_001934), DLX3(NM_005220) ITGA3, PDK2, SAMD14,PPP1R9B, SGCA, HILS1, COL1A1, TMEM92

(NM_153229.2), XYLT2(NM_022167.2), MRPL27(NM_016504.2).

The deletion in family 4 encompasses the following reference sequence genes: DLX4, DLX3,

ITGA3,PDK2, SAMD14, PPP1R9B, SGCA, HILS1, COL1A1, TMEM92, XYLT2, MRPL27, EME1(NM_001166131.1),

LRRC59(NM_018509.2)

The DLX3, SGCA and XYLT2 genes (underlined) are disease-associated genes apart from COL1A1.

Dijk_Opmaak.indd 49 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 25 T1 - Black CyanMagentaYellow

Page 51: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II50

Tabl

e 1.

Phe

noty

pica

l, bo

ne m

orph

olog

ical

and

bio

chem

ical

cha

ract

eris

tics

of p

atie

nts

with

OI t

ype

I due

to a

del

etio

n of

the

com

plet

e CO

L1A1

alle

le.

In 3

9 he

alth

y in

divi

dual

s, A

was

91%

and

B w

as 6

3%, b

oth

with

a s

tand

ard

devi

atio

n of

3%

.

Patie

nt

IDA

geSe

xra

ceSc

lera

e bl

ue/g

rey

Hea

ring

loss

DI1

Ost

eope

nia

Shor

tbo

win

gW

orm

ian

bone

sfr

actu

res

(n)

% Colla

gen

alph

a-1(

I) of

tota

l col

lage

n (=

b)

% Colla

gen

type

I of

to

tal c

olla

gen

(=A

)

1.II.

110

FN

E2+

--

++

-+

844

.8%

62.9

%

1.II.

210

FN

E-

--

U3

--

-0

UU

2.I.2

44F

NE

++

-U

UU

U17

57.7

%82

.9%

2.II.

15

FN

E+

--

U-

-U

2U

U

3.I.2

37F

NE

+-

+-

--

U0

55.2

%77

.5%

4.I.1

40M

NE

+-

-U

UU

U7

52.9

%76

.3%

4.II.

11

MN

E+

--

--

-U

0U

U

1 D

entin

ogen

esis

impe

rfec

ta

2 N

orth

Eur

opea

n3 U

nkno

wn

Dijk_Opmaak.indd 50 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 25 T1 - Black CyanMagentaYellow

Page 52: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta 51

Marker Analysis

Genetic analysis (concordance in 16 short tandem repeats markers) confirmed monozygosity of 1.II.1

and 1.II.2 (Powerplex ® 16 System, Promega, Madison, Wisconsin, United States).

figure 2: MLPA results in individuals from family 1-4, showing a deletion of the complete COL1A1 allele. 26 probes in the COL1A1 gene and 7 reference probes are shown. The MLPA COL1A1 probes are listed on the X-axis according to their position in 5’>3’ orientation. The peak ratio is depicted on the Y-axis. A heterozygous deletion of the complete COL1A1 allele is visible as an approximately 50% reduced relative peak area of the amplification product of that probe compared to reference probes. The average peak ratio of 0.71 and 0.59 in respectively 1.II.2 and 3.I.2 points to mosaicism. The peak ratio values can be found in supplementary table 1.

Dijk_Opmaak.indd 51 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 26 T1 - Black CyanMagentaYellow

Page 53: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II52

figure 3: Results of microarray analysis

The results of microarray analysis in family 2-4, showing deletions of different sizes indicating that no

founder mutation exists.

DISCuSSION

We describe five families in which individuals are affected with OI due to a deletion of a complete

COL1A1 allele. Most affected individuals in our four families have a clinical phenotype consistent with

osteogenesis imperfecta type I. Interestingly, the molecularly affected individual II.2 from family 1

appears to have no symptoms of OI at all whereas her monozygous twin sister with the same deletion

has a phenotype fitting the diagnosis OI type I. No deletion was found in DNA from blood samples of

the parents. The most likely explanation is that the twin sisters are mosaic with a post-zygotic mutation

leading to unequal grades of somatic mosaicism. The average peak ratio of the COL1A1 MLPA probes in

Dijk_Opmaak.indd 52 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 26 T1 - Black CyanMagentaYellow

Page 54: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta 53

1.II.2 (0.67) may point to mosaicism in leukocytes. Unfortunately, we were not able to obtain cell samples

from different tissues in both twin sisters to further investigate this. In family 1 the clinical variability in

individuals with a COL1A1 deletion may be due to mosaicism for the COL1A1 deletion.

OI type I almost always results from mutations in one COL1A1 allele that introduce a premature

termination codon and decrease the stability of mRNA. These mutations may consist of nucleotide

substitutions, frame shifts and splice site mutations. Large deletions of the collagen type I genes are

infrequent in OI 24. Interestingly, the first mutation described to cause OI was a multi-exon COL1A1

deletion eliminating exon 23-25 resulting in OI type II 25. In 1996, another multi-exon deletion was

published namely a 562-bp exon to intron deletion extending from the last codon of exon 34 to intron

36 26. A genomic deletion of exon 40-48 was recently reported causing OI type II 27. In the COL1A2

gene three multi-exon deletions have been described to cause OI type II 28 and OI type IB 24. So it is

generally accepted, that in-frame partial deletions in the COL1A1 or COL1A2 genes can result in a lethal

or severe OI phenotype when the protein is not rapidly degraded but instead is incorporated in the

triple helix exerting a dominant negative effect. Interestingly, a 17q21.33 to q23.1deletion including

the whole COL1A1 gene was described to cause a mild OI phenotype with blue sclerae, osteopenia

and a fracture at birth in combination with micrognathia, cleft palate and marfanoid features 29. We

state that deletions of the complete COL1A1 allele do not have the above described dominant-negative

effect on COL1A1-expression and thereby result in a milder clinical picture. The clinical phenotype in

most of our patients with a complete COL1A1 deletion is comparable with the phenotype of OI type

I seen in patients with a non-functional COL1A1 allele. Microarray analysis revealed that no common

deletion of COL1A1 exists in our families. In family 3 and 4, the DLX3 gene was also deleted. A 4 bp

deletion in the DLX3 gene has been described to cause trichodentoosseous syndrome (TDO) ( OMIM

#190320) characterized by kinky or curly hair, dolichocephaly, enamel hypoplasia, increased dental

caries, radial dense bones, and occasionally brittle nails. The observed mutation was predicted to cause

a frameshift and premature termination codon, resulting in a functionally altered DLX3 protein with

a potential dominant negative effect. A 2 bp deletion in the DLX3 gene appeared to be responsible

for the characteristic dental phenotype without hair or bone abnormalities30. Interestingly, patient I.2

from family 3 has extensive caries in combination with enamel loss which is a feature of osteogenesis

imperfecta but has also been described in patients with a deletion in the DLX3 gene. However, patient

I.1 from family 4 has no teeth abnormalities. Therefore, it cannot be concluded whether a deletion of the

DLX3 gene affected the clinical phenotype in our patients. Given the function, when known, of the other

deleted reference sequence genes, it is unlikely that they contribute to the phenotype of the patients of

family 2-4 described in this article.

Sequence analysis of COL1A1 and COL1A2 cDNA, which detects mutations in the coding sequence ,

and of gDNA, which detects mutations that alter either alter the sequence or the stability of mRNA,

identifies mutations in approximately 90% of patients with OI type I. Measuring the amount and

structure of type I procollagen molecules synthesized by dermal fibroblasts also detects approximately

90% of individuals known to have OI 2,18. It has been stated that it is likely that the combination of

biochemical and molecular genetic analysis would identify a small number of additional individuals

with OI 4. Indeed, the patients we describe with deletions of a complete COL1A1 gene would not have

Dijk_Opmaak.indd 53 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 27 T1 - Black CyanMagentaYellow

Page 55: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II54

been diagnosed with OI if only cDNA and/or gDNA sequencing would have been performed. As such,

in cases of suspected OI type I without a detectable mutation, MLPA analysis of the COL1A1 gene is an

useful additional approach to defining the mutation. This has important implications for the prenatal

diagnostic approach, prior to a pregnancy, in patients with suspected OI type I and reduced type I

procollagen synthesis, since the reduced synthesis of type I collagen chains cannot be detected reliably

in chorionic villi cells 2. Interestingly, in our families, three patients (2.I.2, 3.I.2, 4.I.1) had minimally reduced

amount of collagen type I making the diagnosis OI purely based on biochemical testing unreliable. The

observation that minimally diminished collagen type I production, as measured in fibroblasts, still causes

OI type I might be due to tissue-specific expression of collagen type I with the regulation of expression

in skin, or dermal fibroblasts, and bone 31 being different.

In conclusion, in patients with suspected OI type I, performing collagen type I electrophoresis and

sequencing of the COL1A1/2 genes is insufficient to rule out OI type I when only gDNA is available and no

heterozygous polymorphic variants are detected. In view of the clinical and biochemical characteristics

of complete COL1A1 allele deletions, MLPA should be performed as well, even in the presence of near-

normal or normal collagen type I production as determined by collagen type I electrophoresis.

ACKNOWLEDGEMENTS

We thank Isabel M. Nesbitt from the Sheffield Molecular Genetics Service, Sheffield Children’s National

Health Service Foundation Trust, Sheffield Children’s Hospital, United Kingdom for her technical

assistance and we thank the patients for their kind cooperation.

Dijk_Opmaak.indd 54 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 27 T1 - Black CyanMagentaYellow

Page 56: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta 55

rEfErENCES

1. van Dijk FS, Pals G, Rijn van RR, Nikkels PGJ, Cobben JM. Classification of osteogenesis imperfecta revisited. Eur. J. Med. Genet. 2010;53:1-5.

2. Byers PH, Krakow D, Nunes ME, Pepin M. Genetic evaluation of suspected osteogenesis imperfecta. Genetics in Medicine 2006 ; 8: 383-388.

3. Sillence DO, Senn A, Danks DM. Genetic heterogeneity in Osteogenesis Imperfecta. J. Med. Genet. 1979;16:101-116.

4. Marlowe A, Pepin MG, Byers PH. Testing for osteogenesis imperfecta in cases of suspected non-accidental injury. J. Med. Gen. 2002;39:282-286.

5. Ablin DS, Greenspan A, Reinhart M, Grix A. 1990. Differentiation of Child Abuse from Osteogenesis Imperfecta. AJR 1990;154:1035-1046.

6. Steiner RD, Pepin M, Beyers PH. Studies of collagen synthesis and structure in the differentiation of child abuse from osteogenesis imperfecta. J. Pediatr. 1996;128:542-547.

7. Barnes AM, Chang W, Morello R, et al. Deficiency of cartilage-associated protein in recessive lethal osteogenesis imperfecta. NEJM 2006; 355:2757-2764.

8. Cabral WA, Chang W, Barnes AM, et al. Prolyl 3-hyrdroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfecta. Nat. Genet. 2007:39;359-365

9. van Dijk FS, Nesbitt IM, Zwikstra EH, et al. PPIB mutations cause severe osteogenesis imperfecta. Am. J. Hum. Genet. 2009;85:521-527.

10. Engel J, Prockop DJ. The zipper-like folding of collagen triple helices and the effects of mutations that might disrupt the zipper. Annual reviews 1991;20:137-152.

11. Byers PH, WallisGA, Willing MC. Osteogenesis Imperfecta: translation of mutation to phenotype. J. Med. Genet. 1991;28:433-442

12. Marini JC, Forlino A, CabralWA, et al. Consortium 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. Human Mutation 2007;28:209-221.

13. Steiner RD, Pepin MG, Byers PH. Osteogenesis Imperfecta. Available at http://www.genetests.org. Accessed April 4, 2010.

14. Willing MC, Pruchno CJ, Atkinson M, Byers PH. Osteogenesis imperfecta type I is commonly due to a COL1A1 null allele of Type I collagen. Am. J. Hum. Genet. 1992;51:508-515.

15. Willing MC, Deschenes SP, Scott DA, et al. Osteogenesis Imperfecta type I: Molecular heterogeneity for COL1A1 null alleles of type I collagen. Am. J. Hum. Genet. 1994;55:638-647

16. Nuytinck L, Sayli BS, Wettinck K, De Paepe A. Prenatal diagnosis of osteogenesis imperfecta type I by COL1A1 null-allele testing. Prenat. Diagn. 1999;19:873-875.

17. Wenstrup RJ, Willing MC, Starman BJ, Byers PH. Distinct biochemical phenotypes predict clinical severity in nonlethal variants of osteogenesis imperfecta. Am. J. Hum. Genet. 1990; 46:975-982.

18. Steiner RD, Pepin MG, Byers PH. 2005. Osteogenesis Imperfecta. www.genetests.com.

19. Spranger J, Brill P, Poznanski A. Bone dysplasias. An atlas of genetic disorders of skeletal development. In: Spranger J, Brill P, Poznanski A, editors.Section X: skeletal dysplasias with prominent diaphyseal involvement, Osteogenesis Imperfecta, type III/ IIB. Oxford:OxfordUniversity Press, 2003:440-441.

15. Körkkö J, Ala-Kokko L, De Paepe A, Nuytinck L, Earley J, Prockop DJ. Analysis of the COL1A1 and COL1A2 genes by PCR amplification and scanning by conformation-sensitive gel electrophoresis identifies only COL1A1 mutations in 15 patients with osteogenesis imperfect type I: identification of common sequences of null-allele mutations. Am. J. Hum. Genet. 1998;62:98-110.

16. Schouten JP, McWlgunn CJ, Waaijer R, Zwijnenburg D, Diepvens F, Pals G. Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acid Research 2002; 30:e57.

17. Buffart TE, Israeli D, Tijssen M, et al. Across Arraycomparative genomic hybridization: a strategy to reduce reference channel hybridizations. Genes Chromosomes Cancer2008;47: 994-1004.

18. Kent WJ, Sugnet CW, Furey TS et al. The human genome browser at UCSC. Genome Res. 2002;12:996-1006.

19. Mundlos S, Chan D, Weng YM, Sillence DO, Cole WG, Bateman JF. Multiexon deletions in the type I collagen COL1A2 gene in Osteogenesis Imperfecta type 1B. JBC1996; 271:21068-21074.

20. Barsh GS, Roush CL., Bonadio J., Byers PH, Gelinas RE. Intron-mediated recombination may cause a deletion in an alpha1 type I collagen chain in a lethal form of osteogenesis imperfecta. Proc. Natl.Acad.Sci. USA 1985; 82: 2870-2874.

Dijk_Opmaak.indd 55 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 28 T1 - Black CyanMagentaYellow

Page 57: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter II56

21. Wang O, Forlino A, Marini JC. Alternative splicing in COL1A1 mRNA leads to a partial null allele and two in-frame forms with structural defects in non-lethal osteogenesis imperfecta. JBC 1996;271:28617-28623.

15. Bodian DL, Chan T, Poon A, et al. Mutation and polymorphism spectrum in osteogenesis imperfecta type II. Implications for genotype-phenotype relationships. Human Molecular Genetics 2009;18:463-471.

16. Willing MC, Cohn DH, Starman B, Holbrook KA, Greenberg CR, Byers PH. Heterozygosity for a large deletion in the alpha2(I) collagen gene has a dramatic effect on type I collagen secretion and produces perinatal lethal osteogenesis imperfecta. JBC 1988;263: 8398-8404.

17. Pollitt R, McMahon R, Nunn J, et al. Mutation analysis of COL1A1 and COL1A2 in patients diagnosed with osteogenesis imperfecta type I-IV. Human Mutation 2006. Mutation in brief

18. OMIM (Online Mendelian Inheritance in man) Baltimore: John Hopkins University, Center for Medical Genetics, 1996. http://www3.ncbi.nlm.nih.gov/omim/(September)

19. Lichtler A, Stover M, Angilly J, Kream B, Rowe DW. Isolation and characterization of the rat alpha (I) collagen promoter. J. Biol. Chemistry 1989;264:3072-3077.

Dijk_Opmaak.indd 56 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 28 T1 - Black CyanMagentaYellow

Page 58: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Complete COL1A1 allele deletions in osteogenesis imperfecta 57

Supplementary table 1: The peak ratio values of 7 internal reference probes and 26 COL1A1 MLPA probes are shown for individuals from families 1-4 with a COL1A1 deletion and a healthy control. The average peak ratio of 1.II.1 (0.51) compared to the average peak ratio value of 1.II.2 (0.67) points to mosaicism in leukocytes in 1.II.2. This might also be the case in 3.I.2 who has an average peak ratio value of 0.59.

1.II.1 1.II.2 2.I.2 2.II.1 3.I.2 4.I.1 4.II.1 Con

Con1 1 1.083 1 1.229 1 1 0.787 1

Con2 1.053 1 0.626 1.091 0.787 1.119 0.933 1.005

Con3 0.939 0.931 1.202 1.043 1.282 0.949 0.825 0.965

Con4 0.892 1.047 1.006 1.047 1.006 1.057 1.123 0.97

Con5 0.969 0.999 0.977 0.0965 1.014 1 1.105 1.019

Con6 1.007 1.099 0.781 1.05 0.93 0.979 0.982 1.007

Con7 1 1.103 0.572 1.174 0.637 1 0.896 1

ex02 0.487 0.641 0.413 0.544 0.561 0.528 0.403 0.939

ex04 0.503 0.662 0.507 0.579 0.699 0.54 0.412 0.983

ex05 0.482 0.69 0.452 0.577 0.644 0.558 0.484 0.991

ex06 0.459 0.713 0.299 0.588 0.481 0.534 0.377 1.065

ex07 0.511 0.649 0.464 0.592 0.652 0.548 0.53 0.969

ex09 0.428 0.691 0.462 0.061 0.625 0.576 0.479 0.931

ex11 0.536 0.694 0.412 0.507 0.648 0.546 0.523 0.947

ex12 0.687 0.699 0.313 0.552 0.562 0.542 0.489 0.878

ex14 0.543 0.666 0.39 0.51 0.575 0.553 0.568 0.942

ex18 0.503 0.7 0.469 0.572 0.667 0.586 0.513 0.94

ex21 0.516 0.634 0.457 0.539 0.588 0.587 0.523 0.914

ex25 0.474 0.655 0.466 0.599 0.649 0.542 0.462 0.938

ex27 0.448 0.628 0.35 0.532 0.41 0.649 0.45 0.89

ex29 0.477 0.614 0.406 0.505 0.609 0.561 0.627 0.947

ex30 0.525 0.658 0.497 0.569 0.637 0.565 0.477 0.999

ex32 0.549 0.675 0.365 0.583 0.489 0.553 0.476 0.936

ex34 0.517 0.685 0.345 0.505 0.495 0.524 0.477 0.952

ex36 0.512 0.69 0.511 0.612 0.73 0.577 0.5 0.954

ex38 0.533 0.701 0.477 0.563 0.622 0.546 0.532 0.938

ex40 0.488 0.658 0.426 0.58 0.638 0.579 0.566 0.99

ex42 0.537 0.724 0.357 0.617 0.549 0.557 0.48 0.957

ex43 0.491 0.661 0.484 0.588 0.661 0.58 0.522 0.935

ex45 0.568 0.692 0.397 0.539 0.531 0.53 0.57 0.971

ex49 0.456 0.671 0.491 0.631 0.602 0.607 0.5 0.915

ex50 0.562 0.692 0.395 0.559 0.5 0.577 0.557 0.949

ex51 0.569 0.705 0.342 0.559 0.49 0.556 0.493 0.979

Dijk_Opmaak.indd 57 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 29 T1 - Black CyanMagentaYellow

Page 59: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 58 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 29 T1 - Black CyanMagentaYellow

Page 60: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III

recessive Osteogenesis Imperfecta

Dijk_Opmaak.indd 59 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 30 T1 - Black CyanMagentaYellow

Page 61: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 60 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 30 T1 - Black CyanMagentaYellow

Page 62: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta: the importance of combining biochemical and molecular genetic analysis

Fleur S. van Dijk1*, Isabel M. Nesbitt 2, Peter G.J. Nikkels3, Ann Dalton2, Ernie MHF Bongers4,

Jiddeke M.van de Kamp1, Y. Hilhorst-Hofstee5, Nicolette S. Den Hollander 5, Guus Lachmeijer1,

Carlo L. Marcelis4, Gita B. Tan-Sindhunata1, Rick R. van Rijn6 , Hanne Meijers-Heijboer1,

Jan M Cobben7 , Gerard Pals G1

1Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands;2Sheffield Molecular Genetics Service, Sheffield Children’s Hospital NHS Foundation Trust, Sheffield S10 2TH, UK

3Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands4Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands

5Department of Clinical Genetics, Leiden University Medical Centre, Leiden, the Netherlands

6Department of Pediatric Radiology, Academic Medical Centre, Amsterdam, the Netherlands7Department of Pediatrics, Academic Medical Centre, Amsterdam, the Netherlands

Eur J Hum Genet. 2009 Dec;17(12):1560-9.

III-1

Dijk_Opmaak.indd 61 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 31 T1 - Black CyanMagentaYellow

Page 63: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-162

AbSTrACT

Autosomal recessive lethal and severe Osteogenesis Imperfecta (OI) is caused by deficiency of

Cartilage Associated Protein (CRTAP) and prolyl-3-hydroxylase due to CRTAP and LEPRE1 mutations.

We analyzed 5 families in which 10 individuals had a clinical diagnosis of lethal and severe OI with

overmodification of collagen type 1 on biochemical testing and without a mutation in the collagen

type 1 genes. CRTAP mutations not previously described were identified in the affected individuals.

Although it appears that one important feature of autosomal recessive OI due to CRTAP mutations

is that the radiological features are more consistent with OI type II-B/ III, differentiation between

autosomal dominant and autosomal recessive OI based on clinical, radiological and biochemical

investigations proves difficult in the affected individuals reported here. These observations

confirm that once a clinical diagnosis of OI has been made in a proband, biochemical testing for

overmodifcation of collagen type 1 should always be combined with molecular genetic analysis of

the collagen type I genes. If no mutations in the collagen type I genes are found, additional molecular

genetic analysis of the CRTAP and LEPRE1 genes should follow. This approach will allow proper

identification of the genetic cause of lethal or severe OI, which is important in providing prenatal

diagnosis, preimplantation genetic diagnosis and estimating recurrence risk.

Dijk_Opmaak.indd 62 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 31 T1 - Black CyanMagentaYellow

Page 64: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta 63

INTrODuCTION

Osteogenesis imperfecta (OI) was first described by the Swedish surgeon Olaus Jakob Ekman in a family

with hereditary bone fragility (1788) 1. Because of the extreme variability in the presentation of OI, a

classification in four types (OI type I (mild), II (lethal), III (severe), and IV (moderate) was introduced by

Sillence et al (1979) (the “Sillence classification”). Type II OI was further subdivided in type A, B and C on

the basis of radiological features. The earlier assumption based on pedigree findings that OI type II was

inherited in an autosomal recessive way shifted towards the view that OI type II was primarily inherited

as an autosomal dominant disease 2-5 with the discovery of heterozygous mutations in the collagen type

I genes COL1A1 and COL1A2 for all types of OI. Also, it was recorded that most recurrences in OI type II

and III were caused by gonadal mosaicism in one of the parents. The empiric recurrence rate of lethal OI

was estimated at 6% 6-10. However, it appeared that not all patients with lethal or severe OI had a collagen

type 1 mutation 11,12. Hypomorphic loss of function of CRTAP encoding cartilage-associated protein

(CRTAP) was shown to cause the rhizomelic form of moderate-severe recessive OI originally described as

OI type VII 13. Furthermore, complete loss of function mutations of CRTAP and of LEPRE1, which encodes

prolyl-1-hydroxylase (P3H1), appeared to cause autosomal recessive lethal and severe OI 14,15. CRTAP, P3H1

and cyclophilin B (CYPB) encoded by PPIB form a protein complex in the rough endoplasmic reticulum

(rER) that is responsible for 3- hydroxylation of proline at position 986 in the alpha1 chain of collagen

type I. In individuals affected with lethal or severe OI due to mutations in COL1A1, COL1A2, CRTAP and

LEPRE1, comparable delayed migration of collagen type I is evident, which is visible as backstreaking,

increased band width, or baseline shift on electrophoresis (see figure 1). This phenomenon is due to

posttranslational overmodification and can be explained by the structure of collagen type I, which is first

synthesized as procollagen type I consisting of two proα1(I) chains and one proα2(I) chain, containing

N- and C terminal propeptides. Posttranslational modification necessary to reach intertwining of the

chains resulting in triple helical conformation takes place in the rER. Currently, two posttranslational

modification systems of collagen type I are known namely (i) hydroxylation of multiple lysine and proline

residues by lysyl hydroxylase and prolyl-4-hydroxylase and (ii) 3-hydroxylation of a single residue in the

alpha1 chain, proline at position 986 by the protein complex in the rER consisting of P3H1, CRTAP and

CYPB. Triple helical folding occurs in the C-to-N direction making lysines and prolines inaccessible to

posttranslational modification as soon as the chain in which they are located is folded. Posttranslational

overmodification takes place when a delay in triple helical folding results in overprocessing of the

constituent collagen type I chains by the enzymes involved in posttranslational modification. This delay

can be caused by collagen type 1 mutations disturbing the primary structure of collagen type I or by

defects in two of the components of the prolyl-3-hydroxylation complex, CRTAP and LEPRE1 16-19. Here,

we describe 10 individuals affected with lethal or severe OI due to novel CRTAP mutations.

MATErIALS AND METHODS

Patients

We selected 13 individuals from 7 families for screening based on clinical diagnosis of OI type II/III,

collagen type 1 overmodification on electrophoresis and absence of collagen type I mutations.

Dijk_Opmaak.indd 63 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 32 T1 - Black CyanMagentaYellow

Page 65: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-164

Protein analysis

Skin biopsies were obtained from the affected individuals, and fibroblast cultures were established under

standard conditions. Cells were seeded at 35,000 cells/ cm2. Labelling of the fibroblasts and purification

of the collagen molecules were performed as reported elsewhere with the exception of performing

SDS-electrophoresis without 3% stacking gel and exposing the dried gels to phosphor imager screen

(Molecular Dynamics) 20..

DNA analysis of the collagen type 1 genes

cDNA of the COL1A1 and COL1A2 genes was prepared from total RNA from cultured fibroblasts. RNA

was isolated using the RNA isolation minikit (Qiagen) according to the manufacturers instructions.

Full-length single-stranded cDNA was prepared with oligo-dT-primer and SuperscriptTMII RT reverse

transcriptase (Invitrogen). The complete coding sequences of the COL1A1 and COL1A2 genes (reference

sequence NM_000088 and NM_000089) were both analyzed by direct sequencing in 11 overlapping

PCR fragments (primer sequences available on request).

Molecular analysis of the CRTAP and LEPRE1 genes.

Molecular analysis of CRTAP (reference sequence NM_006371), LEPRE1 (reference sequence NM_022356)

and PPIB was performed as reported elsewhere 14,15,22 .

rESuLTS

In all 13 individuals from 7 families posttranslational overmodification on electrophoresis was evident.

In 2 individuals from one family LEPRE1 mutations were detected, in one individual no mutations in

CRTAP, LEPRE1 and PPIB were detected and 10 individuals from 5 families appeared to have novel CRTAP

mutations. Here we describe the clinical features and mutation analysis of patients with OI due to CRTAP

mutations only.

family 1 [figure 2]

Clinical findings The parents were consanguineous (Caucasian) and mother was pregnant with male monozygotic twins.

An advanced ultrasound screening at 21 weeks of gestation showed bowed and short femura <p5 of

both fetuses (VI:3 and VI:4). In one fetus a possible fracture of the femur was seen. Because of the strong

suspicion of OI type II/III, the pregnancy was terminated at 23 +2 weeks of gestation. No permission

was given for autopsy. On the babygram, fractures of the femora were detected in both fetuses with no

apparent rib fractures [Supplementary Appendix figures 3, 4]. The fetuses were diagnosed with OI type

II-B.

In the following pregnancy an advanced ultrasound in the 19th week of gestation showed again skeletal

abnormalities consisting of a fracture of the femur, a bowed tibia and short osseous elements. The

parents decided to proceed with the pregnancy and a daughter (VI:5) was born by caesarean section at

39 weeks of gestation with an Apgar score of 9/9/10 after respectively 1, 5 and 10 minutes. Birth weight

Dijk_Opmaak.indd 64 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 32 T1 - Black CyanMagentaYellow

Page 66: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta 65

was 3245 gram. The neonate had normocephaly, a round face with shallow orbits, white sclerae, long

philtrum, small thorax, rhizomelic shortening of the upper and lower extremities and abducted position

of the legs [Figure 3]. A skeletal overview showed evidence of multiple fractures [figure 4]. The girl was

diagnosed with OI type III and is alive at the age of two years.

figure 1: Biochemical testing in families 1-5Overmodification on electrophoresis is evident by either backstreaking and/or baseline shift and/or increased band width. Three examples of these biochemical features are numbered in the electrophoresis images of the families

figure 2: Pedigrees families 1-5

Dijk_Opmaak.indd 65 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 33 T1 - Black CyanMagentaYellow

Page 67: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-166

family 1 [figure 2]

Clinical findings The parents were consanguineous (Caucasian) and mother was pregnant with male monozygotic twins.

An advanced ultrasound screening at 21 weeks of gestation showed bowed and short femura <p5 of

both fetuses (VI:3 and VI:4). In one fetus a possible fracture of the femur was seen. Because of the strong

suspicion of OI type II/III, the pregnancy was terminated at 23 +2 weeks of gestation. No permission

was given for autopsy. On the babygram, fractures of the femora were detected in both fetuses with no

apparent rib fractures [Supplementary Appendix figures 3, 4]. The fetuses were diagnosed with OI type II-B.

In the following pregnancy an advanced ultrasound in the 19th week of gestation showed again skeletal

abnormalities consisting of a fracture of the femur, a bowed tibia and short osseous elements. The parents

decided to proceed with the pregnancy and a daughter (VI:5) was born by caesarean section at 39 weeks

of gestation with an Apgar score of 9/9/10 after respectively 1, 5 and 10 minutes. Birth weight was 3245

gram. The neonate had normocephaly, a round face with shallow orbits, white sclerae, long philtrum,

small thorax, rhizomelic shortening of the upper and lower extremities and abducted position of the legs

[Figure 3]. A skeletal overview showed evidence of multiple fractures [figure 4]. The girl was diagnosed

with OI type III and is alive at the age of two years.

figure 3: clinical pictures of family 1 VI: 5 and family 5 II:1.Note round face with shallow orbits, white or greyish sclerae, long philtrum, small thorax, rhizomelic shortening of the upper and lower extremities and abducted position of the legs.

Mutation analysis

In the twin monozygosity was confirmed by marker studies. Mutation screening of the COL1A1 and

COL1A2 genes in the fetuses VI:3 and VI:4 identified a variant c.613 C>G; p.pro205Ala in the COL1A1 gene.

This variant has been reported in literature before and is probably associated with osteopenia21. Mutation

analysis of the ALPL gene responsible for hypophosphatasia was negative. In the third affected child no

mutations in the COL1A1, COL1A2 and LEPRE1 genes were found.

Sequencing of the CRTAP gene in the three affected children revealed homozygosity for the mutation

c.21_22dupGG introducing a premature termination codon at p.Ala8fs in exon 1. The parents were

heterozygous for this mutation

Dijk_Opmaak.indd 66 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 33 T1 - Black CyanMagentaYellow

Page 68: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta 67

figure 4: radiographs of individual VI:5 from family 1A. Apparent normal mineralization of the skull, no Wormian bones b. Mild brachycephaly C. Normal aspect of the right arm. A rhizomelic shortening may exist that may also be due to projection. D. Oblique mid diafyseal fracture of left humerus (arrow). E. Normal chest radiograph without rib fractures. f. Platyspondyly of thoracic vertebrae 5, 7, 8, 10, 11 (arrow heads). G. Bowing of femora (arrow head), tibiae and fibulae (arrow). H. Bowing of femora (arrow head), tibiae and fibulae (arrow).

figure 5: radiographs of individual IV:4 from family 2A. Small thorax. Slender ribs. Lateral fracture of 8th left rib (see insert). Asymmetric platyspondyly Th5 and Th7 (visible on original radiograph). b. Slight flattening of the skull (arrow). C. Consolidation of right humerus fracture (arrow). Humerus measures 4.38 cm (normal range=6.0-7.7 cm.), ulna 4.9 cm (normal range =5.8-7.2 cm.). Broad distal metaphyses of left humerus and radius (arrow head). Possible fracture of left radius. D. Broad distal metaphyses of left humerus and ulna (arrow head). E. Bilateral proximal femur fracture with consolidation and angulation (arrows). Bilateral bowing of tibia and fibula (arrow heads).

family 2 [figure 2]

Clinical findings

The parents were consanguineous (Moroccan). In the third trimester of pregnancy extensive skeletal

abnormalities were observed in the fetus. A boy (IV:1) was born with multiple fractures consistent with

the diagnosis OI type II/III. He died one month after birth.

In the fourth pregnancy ultrasound images around the 20th week of gestation displayed skeletal

abnormalities suggestive of OI. At 23+2 weeks of gestation shortened and deformed extremities, a

Dijk_Opmaak.indd 67 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 34 T1 - Black CyanMagentaYellow

Page 69: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-168

fracture of the left femur and a bell shaped thorax were observed on ultrasound screening. A girl (IV:4)

was born at 38+3 weeks. A skeletal overview showed gracile bones and multiple fractures with callus

formation (figure 5). The girl died one week later because of respiratory insufficiency. A femur was taken

for histologic examination (figure 6).

Mutation analysis

Fibroblast cultures of the first child became infected and no mutations in the COL1A1, COL1A2 and

LEPRE1 genes were identified in the second affected child. Sequencing of the CRTAP gene showed

homozygosity for the mutation c.404delG introducing a premature termination codon at p. Ser135fs in

exon 1 of the CRTAP gene. The parents were both heterozygous for this mutation.

figure 6: bone histology of family 2 IV:4A. H and E stain of epiphysial growth zone of femur. Note the sharp demarcation between cartilage and primary spongiosa. The epiphyseal growth zone and primary spongiosa are normal. The resting cartilage shows no abnormalities. b. H and E stain of the metaphysis with the for OI typical small, slender and hypercellular bony trabeculae and fibrotic medulla. The bony trabeculae consist of woven bone. C. Low power PAS acian blue stain of the midshaft of the femur with metaplastic cartilage formation (blue) in the area of a fracture with fibrotic marrow. This formation of cartilage is quite typical for the severe forms of OI

family 3 [figure 2]

Clinical findings

A caucasian girl (II:1) was born at 40+6 weeks of gestation with Apgar scores of 7 and 9 at respectively

1 and 5 minutes. On physical examination fragmentation of the skull bones was felt and a short thorax

with short and bowed extremities was noticed suggestive of OI. The child had no overt blue sclerae. A

skeletal overview showed multiple rib fractures and fractures of both upper and lower extremities. The

neonate was diagnosed with OI type II-C/III. At the age of 2,5 months she died of respiratory failure.

In the following pregnancy an ultrasound in the 17th week showed no evidence of fractured or bowed

Dijk_Opmaak.indd 68 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 34 T1 - Black CyanMagentaYellow

Page 70: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta 69

extremities. However, at 18+4 weeks of gestation fractures of both femora were observed. Recurrence

of OI was suspected and the pregnancy was terminated at 20+2 weeks. Multiple fractures were evident

[figure 7] and the fetus (II:2) was diagnosed with OI type II.

The fifth pregnancy (II:5) was terminated because of evident posttranslational overmodification of

collagen type 1 in cultured chorion villi.

figure 7: radiographs of II:2 from family 3A. Slender ribs. No rib fractures, two fractures of right femur with callus formation. b. Underexposed radiograph which makes evaluation for mineralization difficult. The calvaria seem to be thin for gestational age. C. No vertebral abnormalities. D. Two fractures of left femur with callus formation.

Mutation analysisMutations in the COL1A1, COL1A2 and LEPRE1 genes were not detected in the three affected children.

Sequencing of the CRTAP gene showed that they were all compound heterozygous for a c.198C>A

mutation predicted to result in a p. Tyr66X amino acid change in exon 1 and a c.471+2 C>A splice site

change in intron 1 of the CRTAP gene. The mother was heterozygous for the c.471+2 C>A mutation and

the father was heterozygous for the c.198C>A mutation.

family 4 [figure 2]Clinical findingsAt a pregnancy duration of 20 weeks ultrasound abnormalities were observed consisting of short and

bowed extremities and a short thorax. Because of suspicion of OI a caesarean section was performed

at 39+4 weeks of gestation. A caucasian boy (II:1) was born with Apgar scores of 2-3 and 8-9 after

respectively 1 and 5 minutes. He had a large anterior fontanel, short thorax, short and bowed extremities

and glandular hypospadia. A skeletal overview showed wormian bones as well as multiple fractures of

ribs and both upper and lower extremities. [figure 8] He was diagnosed with OI type III and died five

days after birth.

Mutation analysisMutations in the COL1A1, COL1A2 and LEPRE1 genes were not detected in the affected child. Sequencing

of the CRTAP gene showed that the affected child was compound heterozygous for a splice site mutation

c.923-2A>G in intron 4 and two mutations on the other allele in exon 1 namely c.38C>A and c.469A>G.

Mother was heterozygous for the c.38C>A and c.469A>G mutation in exon 1. Father was not available.

Dijk_Opmaak.indd 69 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 35 T1 - Black CyanMagentaYellow

Page 71: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-170

figure 8: radiological images of II:2 from family 4A. No fractures of humeri, radii and ulnae. Multiple fractures of femora with loss of modeling (arrow). Fracture and bowing of right tibia (arrow head). b. Wormian bones (see insert), broad skull. C. Slender ribs, lateral rib fractures of left rib 2 (see insert) and right ribs 2-6 (arrow heads).

family 5 [figure 2]Clinical findingsIn the first pregnancy abnormalities of the extremities were observed in the fetus at 23 weeks of

gestation. At the 24th week of gestation short upper and lower extremities were observed indicative of

a severe skeletal dysplasia. At a gestation of 40+4 weeks a caucasian boy (II:1) was born. He had a round

face with shallow orbits, greyish sclerae, small thorax, rhizomelic shortening of the upper and lower

extremities, abducted position of the legs and normocephaly [figure 9]. A skeletal overview showed

multiple fractures suggestive of OI type II/III. The child is alive at the age of 4 years.

figure 9: radiological images of II:1 from family 5A. Fracture of left clavicula (arrow head), small thorax, normal vertebral column, fractures of left ribs 4-7 (see insert). Fractures of left humerus (solid arrow head) and radius. Consolidated fracture of right humerus. b. Fracture of right femur (arrow) and tibia (arrow head), severe bowing of right fibula and tibia. Multiple fractures of left femur (arrow), bowing and fracture of left tibia (solid arrow head). C. Wormian bones (see insert) and flattening of the occiput (arrow).

Mutation analysisMutations in the COL1A1, COL1A2, LEPRE1 genes were not detected in the affected child. Sequencing of

the CRTAP gene showed homozygosity for the c.471+2C>A mutation in intron 1 of CRTAP.

Dijk_Opmaak.indd 70 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 35 T1 - Black CyanMagentaYellow

Page 72: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta 71

DISCuSSION

We describe ten individuals with lethal or severe OI based on clinical (table 1 supplementary appendix),

radiological (table 1) and biochemical (figure 1) findings in whom novel CRTAP mutations (table 2)

were found. So far, six probands with lethal or severe OI due to CRTAP mutations have been reported

in literature. An important question is whether the phenotype of patients with lethal OI due to CRTAP

mutations differs from that of patients with OI due to mutations in the collagen type I genes.

Barnes et al., in 3 individuals with OI due to CRTAP mutations, described some clinical features in CRTAP-

related OI differing from the clinical features in individuals with OI due to collagen type I mutations,

namely (i) no relative macrocephaly, (ii) a round face with shallow orbits, (iii) white or greyish sclerae and

(iv) rhizomelic shortening of the upper and lower extremities 14. Indeed, our patients, VI:5 and II:1 from

family 1 and 5 depicted in figure 3 show no relative macrocephaly, a round face and no blue sclerae.

The clinical pictures of VI:5 suggest rhizomelic shortening of the arms. However, due to the absence

of sufficient clinical details of the other lifeborn children with CRTAP mutations, the specificity of these

clinical differences is uncertain. Therefore, a distinction between autosomal dominant OI due to collagen

type 1 mutations and autosomal recessive OI due to CRTAP mutations on clinical features proves difficult.

Radiologically, bowing of extremities and/or fractures could be observed in the prenatal and neonatal

period in all our patients with CRTAP mutations. The earliest appearance of features of OI was noted in

the 18th week of pregnancy (table 1 supplementary appendix). However, this pregnancy was monitored

extensively because of an affected sib. Recently, “popcorn” epiphyses have been suggested to be a

distinguishing radiological feature of autosomal recessive OI, reflecting a cartilaginous dysplasia at the

developing growth plate 22. However, histology of the femoral epihysis in proband IV:4 from family 2 did

not show a primary cartilaginous dysplasia (see figure 4). Furthermore, popcorn epiphysis have been

reported in a patient with OI due to a COL1A2 mutation and recently in other patients with OI type III and

IV due to COL1A1 and COL1A2 mutations 23,24. Popcorn epiphyses were absent in the 4-year-old proband

II:1 from family 5 (see figure 5 supplementary appendix). Considering the above, we hypothesize that

popcorn calcifications reflect a disturbance of enchondral ossification which can occur in CRTAP-related

OI as well as in COL1A1/2-related OI.

Importantly, upon revision of the available radiographs by a pediatric radiologist the classification of

OI differed in 3/8 cases (table 2) which stresses the need for evaluation of radiographs by a pediatric

radiologist in all cases of OI. Interestingly, a radiological diagnosis of OI type II-B or III was evident in

all affected individuals with CRTAP mutations in the literature 14,22 as well as in all our patients. One

important difference between OI type II-A, B and C is the extent and manner of rib involvement 3. In OI

II-A the ribs are very broad with contiguous fractures (contiguous beading), while in OI II-B the ribs are

rather small, with an irregular course and few fractures and in OI II-C, which is very rare, varying thickness

and discontinuous beading of the ribs is noted. In OI III rib fractures are less commonly observed.

Interestingly, a recurrence rate from 0 percent in OI II-A patients 4 to 7.7 percent in OI II-B patients 5 has

been described. It seems that CRTAP mutations cause OI type II-B/III and it might be that recurrence in OI

type II-A is due to gonadal mosaicism of COL1A1/2 mutations only. However, OI type II-B/III can also be

caused by COL1A1/2 mutations, which makes differentiation of CRTAP-related OI based on radiological

features difficult.

On biochemical testing by electrophoresis, full overmodification of collagen type 1 was observed

comparable with dominant inherited lethal and severe OI due to collagen type I mutations. However,

Dijk_Opmaak.indd 71 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 36 T1 - Black CyanMagentaYellow

Page 73: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-172

in some instances it can be possible to discriminate between autosomal recessive and dominantly

inherited lethal and severe OI since in the latter some normal molecules can be made and chains with

normal mobility can be visualized.

In the affected individuals genomic and complementary DNA sequencing of the COL1A1 and COL1A2

genes was negative. Mutation analysis of CRTAP and LEPRE1 genes identified 7 new CRTAP mutations in

the five families and 1 new homozygous LEPRE1 mutation in one family. An overview of (new) CRTAP

mutations is given in table 2 and figure 6 of the supplementary appendix. Pathogenicity of the identified

mutations was supported by in silico analysis. The c.21_22dupGG and c.404delG mutations in exon 1

are frameshift mutations leading to a stop codon respectively 6 and 39 amino acids downstream. The

c.471+2C>A mutation is a splice donor site mutation that changes the second nucleotide in intron 1,

which is the unusual nucleotide, C. (see Supplementary Appendix figure 2). cDNA sequencing proves

that this mutation causes a null allele (see Supplementary Appendix figure 1). The 198C>A mutation

replaces tyrosine at position 66 with a premature termination codon. Interstingly, II:1 form family 4

has three mutations. The c.923-2A>G mutation in intron 4 is a splice acceptor site mutation leading to

disruption of normal splicing of exon 5. The c.469A>G mutation in exon 1 and the c.38 C>A mutation

reside on the same allele. The c.469A>G mutation is a non-conservative change resulting in a p.Lys157Glu

amino acid change and could therefore be considered pathogenic. However, the c.38C>A missense

mutation resulting in a p.Ala13Glu might also be pathogenic since it disturbs the charge distribution

in the signal peptide and could lead to retainment of CRTAP in the rER. Currently, functional studies are

being performed to investigate pathogenicity of these mutations.

In conclusion, although a diagnosis of OI type II-A seems to point to autosomal dominant OI, in OI

types II-B and III clinical, radiological and biochemical differentiation between autosomal dominant OI

caused by collagen type 1 mutations and autosomal recessive OI caused by CRTAP mutations proves

difficult. Therefore, it is important to perform biochemical analysis in combination with preferably

genomic and complementary DNA sequencing of the collagen type 1 genes in individuals affected

with lethal or severe OI. Posttranslational overmodification without collagen type 1 mutations should

prompt CRTAP or LEPRE1 mutation analysis. This approach is important for providing accurate recurrence

risk, prenatal diagnosis and preimplantation genetic diagnosis. Also, therapeutic options might differ in

the future. Furthermore, knowledge of the proportion of cases of lethal and severe OI with recurrence

due to CRTAP and LEPRE1 mutations will lead to a more acccurate estimation of recurrence risk due

to germline mutations in the collagen type 1 genes. It may very well be that the greater part of the

empiric recurrence risk in OI type II and III, currently estimated at 6% for OI type II 9, can be contributed

to autosomal recessive causes.

Supplementary table 1: clinical features of probands affected with CRTAP mutations

family(pedigree nr) Sex

first appearance of features of OI

Gestational age

Head circum-ference(cm) at birth

Length (cm)

Weight (g) Status

Age at death

1 (VI:3) M 21 weeks 23+2 20 28 548 TAB

1 (VI:4) M 21 weeks 23+2 20,5 29,5 571 TAB

1 (VI:5) F 19 weeks 39 3245 LB alive

2 (IV:1) M Third trimester 42+2 2570 LB 1 month

2 (IV:4) F 20 weeks 38+3 34 2725 LB 24 days

3 (II: 1) F birth 40+6 24 2710 LB 2,5 month

3 (II: 2) M 18+4 20+2 16,5 275 TAB

Dijk_Opmaak.indd 72 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 36 T1 - Black CyanMagentaYellow

Page 74: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta 73

Supplementary table 1: (Continued)

family(pedigree nr) Sex

first appearance of features of OI

Gestational age

Head circum-ference(cm) at birth

Length (cm)

Weight (g) Status

Age at death

3 (II: 5) M TAB

4 (II: 1) M 20 39+4 LB 5 days

5 (II: 1) M 23 40+4 35 (at 3 weeks) 3110 LB alive

TAB= Therapeutic abortionLB= Life birth

Supplementary figure 1: the effect of the c.471+2C>A mutationIn affected individuals from family 3 a c.198 C>A nonsense mutation resulting in p.Y66X in combination with a presumed splice site mutation c.471+2 C>A and 2 heterozygous polymorphisms in exon 5 were found. Amplification of gDNA and cDNA across the mutations and polymorphisms with a forward 3’ primer of exon 1 confirmed loss of heterozygosity at the cDNA level suggesting a null allele. This null allele is indeed caused by the c.471+2 C>A mutation since the c.198 C>A mutation on the other allele appears homozygous on cDNA sequencing as opposed to heterozygous if the null allele would have been caused by the nonsense mutation c.198 C>A. The observation that the c.471+2 C>A allele causes a null allele is more likely to be the result of a splice error than of a missense mutation.

Dijk_Opmaak.indd 73 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 37 T1 - Black CyanMagentaYellow

Page 75: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-174

1

Supplementary Figure 1

.

Supplementary figure 2; The c.471+2 C>A mutation in exon 1 of the CRTAP geneInterestingly, upon viewing the human CRTAP genomic sequence, we noticed that the splice donor site of intron 1 does not start with GT. Comparison with the transcript (ENST00000320954) showed that the first four bases of exon 2 are identical with the first four bases of intron 1. A more likely splice donor site is the GT four bases into the proposed intron (figure 1). However, this would require the use of an unlikely splice acceptor site (AT). In the latter case the mutation c.471+2 C>A would be in the coding sequence and result in a missense:c.473C>A; p.Ala158Glu. Based on the genomic and cDNA sequences it is impossible to determine which splice donor and acceptor sites are actually being used.

Supplementary figure 3: radiological features of VI:3 of family 1A. No rib fractures. Mild platyspondyly vertrebrae 4-10. b. Normal mineralization for gestational age. C. No abnormalities. D. No abnormalities. E. Fractures of both femora (arrows) with minor loss of modeling.

Supplementary figure 4: radiological images of VI:4 from family 1A. Normal chest radiograph no rib fractures. b. Slight flattening of the skull (arrow head). C. Normal aspect of the right arm. D. Possible fracture of the proximal left ulna (see insert)E. Two fractures in left and in right femora (arrows), minor loss of femoral shape. Irregularity of the distal tibiae (arrow heads).

Dijk_Opmaak.indd 74 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 37 T1 - Black CyanMagentaYellow

Page 76: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

CRTAP mutations in lethal and severe osteogenesis imperfecta 75

Supplementary fi gure 5A. Radiographs of the lower extremities show multiple fractures for which surgical intervention has been performed. No popcorn epiphyses are observed. Multiple growth acceleration lines are visible due to intravenous biphosphonate treatment (arrow). b. Radiograph of the left arm shows normal epiphyses, with a broad metaphysic of the distal humerus. Note middiaphyseal fractures of the radius and ulna (arrow). Dislocation of the radial head is observed (arrow head). C. AP Spine shows platypondyly (arrow) and scoliosis

c.21_22dupGG

c. 198C>A

c.404delC

c.469A>G*

c.471+2C>A c.923-2A>G

c.471+1G>C15

c.879delT14

c.3G>A15

c.278_293dup15,22

New mutations

Previously reported mutations c.24_31del22

c.200T>C22

c.822_826delAATACinsT22

c.826C>T15

c.472-1021C>G14

c.38C>A*

Supplementary fi gure 6: new and previously described pathogenic CRTAP mutationsThe gene for human Cartilage Associated Protein (CRTAP) consists of 1.203 bp and is localized at 3p22. The 7 exons of the CRTAP gene are shown as boxes, with the exact localization of the mutations in the exons and the connecting introns not to scale. It is not certain which of the two mutations marked with an asterisk is pathogenic.

ACKNOWLEDGEMENTS

We thank the parents for their kind cooperation and we thank clinical geneticist J.J van der Smagt at

the Department of Clinical Genetics, University Medical Center Utrecht, The Netherlands for his advice.

Dijk_Opmaak.indd 75 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 38 T1 - Black CyanMagentaYellow

Page 77: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-176

rEfErENCES

(1) Baljet B: Aspects of the history of osteogenesis imperfecta (Vrolik’s syndrome). Ann Anat 2002; 184: 1-7.

(2) Sillence DO, Senn A, Danks DM: Genetic heterogeneity in osteogenesis imperfecta. J Med Genet 1979; 16:101-116.

(3) Sillence DO, Barlow KK, Garber AP, Hall JG, Rimoin DL: Osteogenesis imperfecta type II delineation of the phenotype with reference to genetic heterogeneity. Am J Med Genet 1984; 17: 407-423.

(4) Young ID, Thompson EM, Hall CM, Pembrey ME: Osteogenesis imperfecta type IIA: evidence for dominant inheritance. J Med Genet 1987; 24: 386-389.

(5) Thompson EM, Young ID, Hall CM, Pembrey ME: Recurrence risks and prognosis in severe sporadic osteogenesis imperfecta. J Med Genet 1987; 24: 390-405.

(6) Chu ML, Williams CJ, Pepe G, Hirsch JL, Prockop DJ, Ramirez F: Internal deletion in a collagen gene in a perinatal lethal form of osteogenesis imperfecta. Nature 1983; 304: 78-80.

(7) Cohn DH, Starman BJ, Blumberg B, Byers PH: Recurrence of lethal osteogenesis imperfecta due to parental mosaicism for a dominant mutation in a human type I collagen gene (COL1A1). Am J Hum Genet 1990; 46: 591-601.

(8) Cohen-Solal L, Bonaventure J, Maroteaux P: Dominant mutations in familial lethal and severe osteogenesis imperfecta. Hum Genet 1991; 87: 297-301.

(9) Byers PH, Tsipouras P, Bonadio JF, Starman BJ, Schwartz RC: Perinatal lethal osteogenesis imperfecta (OI type II): a biochemically heterogeneous disorder usually due to new mutations in the genes for type I collagen. Am J Hum Genet 1988; 42: 237-248.

(10) Byers PH, WallisGA, Willing MC: Osteogenesis imperfecta: translation of mutation to phenotype. J Med Genet 1991; 28: 433-442.

(11) Wallis GA, Sykes B, Byers PH, Mathew CG, Viljoen D, Beighton P: Osteogenesis imperfecta type III: mutations in the type I collagen structural genes, COL1A1 and COL1A2, are not necessarily responsible. J Med Genet 1993; 30: 492-496.

(12) Rauch F, Glorieux FH: Osteogenesis imperfecta. Lancet 2004; 363: 1377-1385.

(13) Ward LM, Rauch F, Travers R et al: Osteogenesis imperfecta type VII: an autosomal recessive form of brittle bone disease. Bone 2002; 31: 12-18.

(14) Barnes AM, Chang W, Morello R et al: Deficiency of cartilage-associated protein in recessive lethal osteogenesis imperfecta. N Engl J Med 2006; 355: 2757-2764.

(15) Cabral WA, Chang W, Barnes AM et al: Prolyl 3-hydroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfecta. Nat Genet 2007; 39: 359-65.

(16) Marini JC, CabralWA, Barnes AM, Chang W: Components of the collagen prolyl 3-hydroxylation complex are crucial for normal bone development. Cell Cycle 2007; 6:1675-1681.

(17) Prockop DJ, Constantinou CD, Dombrowski KE et al:Type I procollagen: the gene-protein system that harbors most of the mutations causing osteogenesis imperfecta and probably more common heritable disorders of connective tissue. Am J Med Genet 1989; 34: 60-67.

(18) Canty EG, Kadler KE: Procollagen trafficking, processing and fibrillogenesis. J Cell Sci 2005; 118:1341-1353.

(19) Trackman PC: Diverse biological functions of extracellular collagen processing enzymes. J Cell Biochem 2005; 96: 927-937.

(20) Korkko J, la-Kokko L, De PA, Nuytinck L, Earley J, Prockop DJ: Analysis of the COL1A1 and COL1A2 genes by PCR amplification and scanning by conformation-sensitive gel electrophoresis identifies only COL1A1 mutations in 15 patients with osteogenesis imperfecta type I: identification of common sequences of null-allele mutations. Am J Hum Genet 1998; 62: 98-110.

(21) Spotila LD, Colige A, Sereda L et al: Mutation analysis of coding sequences for type I procollagen in individuals with low bone density. J Bone Miner Res 1994; 9: 923-932.

(22) Baldridge D, Schwarze U, Morello R et al: CRTAP and LEPRE1 mutations in recessive osteogenesis imperfecta. Hum Mutat 2008 Jun 19.

(23) Cole WG, Chan D, Chow CW, Rogers JG, Bateman JF: Disrupted growth plates and progressive deformities in osteogenesis imperfecta as a result of the substitution of glycine 585 by valine in the alpha 2 (I) chain of type I collagen. J Med Genet 1996; 33: 968-71.

(24) Obafemi AA, Bulas DI, Troendle J, Marini JC: Popcorn calcifications in osteogenesis imperfecta: Incidence, progression and molecular correlation. Am J Med Gen A Sept 2008.

(25) Nikkels PGJ. The skeletal system. In: Keeling JW, Kong TY, editors. Fetal and neonatal pathology. 4 ed. Springer; 2008.

Dijk_Opmaak.indd 76 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 38 T1 - Black CyanMagentaYellow

Page 78: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIb mutations cause severe Osteogenesis Imperfecta

Van Dijk FS1, Nesbitt IM2, Zwikstra EH1, Nikkels PGJ3, Piersma SR4, Fratantoni SA4, Jimenez CR4 , Huizer M1,

Morsman AC2, Cobben JM5, van Roij MHH1, Elting MW1, Verbeke JIML6 , Wijnaendts LCD7, Shaw NJ8,

Högler W8, McKeown C9, Sistermans EA1, Dalton A2, Meijers-Heijboer H1, Pals G10

1 Department of Clinical Genetics, VU University Medical Centre, De Boelelaan 1117,

P.O. box 7057, 1007 MB Amsterdam ,The Netherlands; 2 Sheffield Molecular Genetics Service, Sheffield Children’s NHS Foundation Trust, Sheffield Children’s Hospital,

Western Bank Sheffield, South Yorkshire, S10 2TH, United Kingdom; 3 Department of Pathology, University Medical Centre Utrecht, Heidelberglaan 100,

P.O. box 85500, 3508 GA, Utrecht, the Netherlands; 4 Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Centre,

De Boelelaan 1117, P.O. box 7057, 1007 MB Amsterdam, The Netherlands; 5 Department of Pediatric genetics, Emma Children Hospital, AMC, Meibergdreef 9,

P.O. box 22660, 1100 DD Amsterdam, the Netherlands; 6 Department of Radiology, VU University Medical Centre, De Boelelaan 1117,

P.O. box 7057, 1007 MB Amsterdam, The Netherlands; 7 Department of Pathology, VU University Medical Centre, De Boelelaan 1117,

P.O. box 7057, 1007 MB Amsterdam, The Netherlands; 8 Department of Pediatric Endocrinology, Birmingham Children’s Hospital, Steelhouse Lane,

Birmingham, West Midlands. B4 6NH, United Kingdom; 9 West Midlands Regional Genetic Service, Birmingham Women’s Hospital, Metchley Park Rd,

Birmingham, B15, United Kingdom ; 10 Centre for Connective Tissue Research, Department of Clinical Genetics, VU University Medical Centre,

De Boelelaan 1117, P.O. box 7057, 1007 MB Amsterdam, The Netherlands

Am J Hum Genet. 2009 Oct;85(4):521-7.

III-2

Dijk_Opmaak.indd 77 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 39 T1 - Black CyanMagentaYellow

Page 79: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-278

Deficiency of cartilage associated protein (CrTAP) or prolyl 3-hydroxylase 1(P3H1) has been

reported in autosomal recessive lethal/severe Osteogenesis Imperfecta (OI). CrTAP, P3H1 and

Cyclophylin b(CyPb) form an intracellular collagen-modifying complex that 3-hydroxylates

proline at position 986 (P986) in the alpha1 chains of collagen type I. This prolyl-

3-hydroxylation is decreased in patients with CrTAP and P3H1 deficiency. It was suspected

that mutations in the PPIB gene encoding CyPb would also cause OI with decreased collagen

3-prolyl hydroxylation.

To our knowledge we present the first two families with recessive OI caused by PPIB gene

mutations. The clinical phenotype is compatible with OI Sillence type II-b/III as seen with

COL1A1/2, CRTAP and LEPRE1 mutations. The percentage of 3-hydroxylated P986 residues in

patients with PPIB mutations is decreased compared to normal but higher than in patients with

CRTAP and LEPRE1 mutations. This result and the fact that CyPb is demonstrable independent

of CrTAP and P3H1, in combination with reported decreased 3-prolyl hydroxylation due to

deficiency of CrTAP lacking the catalytic hydroxylation domain and the known function of

CyPb as cis-trans isomerase,suggest that recessive OI is caused by a dysfunctional P3H1/

CrTAP/CyPb complex rather than by lack of 3-prolyl hydroxylation of a single proline residue

in the alpha1 chains of collagen type I.

Dijk_Opmaak.indd 78 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 39 T1 - Black CyanMagentaYellow

Page 80: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIB mutations cause severe Osteogenesis Imperfecta 79

Collagen type I is the major protein component of the extracellular matrix of bone and skin.1 It is a trimeric

molecule in which each chain consists of repeating Gly-X-Y triplets with proline and hydroxyproline

(4Hyp) usually occupying the X and Y positions, respectively. Collagen type I is first synthesized as

procollagen type I consisting of two proα1(I) chains and one proα2(I) chain, containing N- and C terminal

propeptides.2 After correct alignment of these three chains, propagation of triple helical folding in the

C-to-N direction occurs in the rough endoplasmic reticulum (rER) in a zipper like fashion 3, during post-

translational modification by specific rER proteins.2 In the literature two important post-translational

modification systems of collagen type I are known: (i) hydroxylation of multiple proline and lysine

residues by prolyl-4-hydroxylase, providing the triple helix with thermal stability, with lysyl-hydroxylase

providing attachment sites for carbohydrate units as well as stability of intermolecular cross links and

(ii) 3-hydroxylation of a single residue in the alpha1 chain, proline at position 986 (P986) by the protein

complex in the rER consisting of P3H1[MIM 610339], CRTAP [MIM 605497] and CyPB [MIM 123841].1

Osteogenesis Imperfecta (OI [MIM 166200/166210/259420/166220]) is an inherited connective tissue

disorder characterized clinically by bone fragility and increased susceptibility to fractures. Autosomal

dominant OI is usually caused by COL1A1 [MIM 120150] and COL1A2 [MIM 120160] mutations

disturbing the primary structure of collagen type I resulting in delayed triple helical folding and post-

translational overmodification. CRTAP 4 and LEPRE1 5mutations encoding CRTAP and P3H1 respectively,

cause autosomal recessive lethal/severe OI characterized by decreased 3- prolyl hydroxylation of P986

in the alpha1 chain of collagen type I and by post-translational overmodification.1 The function of

3-hydroxyproline in the alpha1 chains of collagen type I is not quite clear but it has been hypothesized

that 3-hydroxyproline takes part in protein-collagen interactions required for bone formation.6 In vitro

studies have shown that P3H1 is responsible for prolyl 3-hydroxylase activity since the presence of

CRTAP and CyPB is not required to reach full 3-prolyl hydroxylation activity.7 CRTAP and P3H1 share the

same amino terminal domain but CRTAP lacks the catalytic hydroxylation domain. Remarkably, CRTAP

mutations appear to cause decreased 3-prolyl hydroxylation as well. Therefore, it was hypothesized that

deficiency in collagen 3-Hyp might simply be a marker for the “dysfunctional” complex rather than an

actual cause of lethal/severe OI.8Also, the role of the third member CyPB in this 3-prolyl hydroxylation

complex remained obscure. CyPB is a 21 kDA protein encoded by the PPIB gene 9 and is known to belong

to the cyclophylins (Cyps), a conserved class of intracellular and/or secreted proteins originally identified

as cellular binding proteins for the immunosuppressive drug cyclosporin A (CsA).10 Cyps are a family

of peptidyl-prolyl cis-trans isomerases (PPIases), which catalyze the cis-trans isomerisation of peptide

bonds. CyPB has a signal sequence directing it to the rER 11 but it is also secreted extracellularly and

appears to play a role in inflammation, viral infection and cancer.10 Previous studies showed that CyPB

directly interacts with procollagen, which is necessary since procollagen contains many cis-conformers.

These have to be converted to trans-conformers since only trans peptide bonds can be incorporated

into the triple collagen helix.12 CyPB is also involved in procollagen export and secretion along with

HsP47, a collagen-binding heat-shock protein.11 It was assumed that, due to co-existence in the complex

with P3H1 and CRTAP, a CyPB deficiency would also result in decreased prolyl-3-hydroxylation, post-

translational overmodification and autosomal recessive OI 8, indicating PPIB as an important candidate

gene for OI. 13, 14

Dijk_Opmaak.indd 79 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 40 T1 - Black CyanMagentaYellow

Page 81: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-280

Therefore, we performed sequence analysis of the PPIB gene in 4 affected individuals from 2 families

in whom no COL1A1/2, CRTAP and LEPRE1 mutations were detected. The procedures followed were in

accordance with institutional and national standards on human experimentation. Appropriate informed

consent was obtained from the families. Proband 1 from family 1 (P1-1) (figure 1A) was delivered after

termination of pregnancy at 22+1 weeks of gestation. She was the second child of non-consanguineous

North European parents. During pregnancy the diagnosis OI was suspected, based on advanced

ultrasounds. Radiographs and autopsy showed no rib fractures, shortened, bowed and fractured

long bones without evident rhizomelia, consistent with a diagnosis of OI type II-B. Weight and head

circumference were normal for gestational age and no other abnormalities were noted. Bone histology

was indicative of OI. Overmodification of collagen type I in fibroblasts was evident on electrophoresis.

Proband 2 from family 1 (P1-2) was delivered after termination of pregnancy at 16+1 weeks of gestation

(Supplementary figure 1A). During pregnancy the diagnosis OI was made based on advanced

ultrasounds and overmodification of collagen type I in chorionic villi cells. Radiographs and autopsy

showed osteopenia, no apparent rib fractures and short long bones with fractures but without evident

rhizomelia. Weight and head circumference were normal for gestational age and no other abnormalities

were noted.Histology was indicative of OI. A diagnosis of OI type II-B was made.

Proband 1 from family 2 (P2-1) (supplementary figure 1B-H) was delivered at 38 weeks of gestation by

Caesarian section to consanguineous Pakistani parents (first cousins). Birth weight was 2.89 kg (p25).

Multiple long bone fractures had been detected at 20 weeks gestation on ultrasound scan and were

evident at birth. A large head with large anterior fontanelle was noted in combination with grey colored

sclera typical of severe OI, flexed and abducted hips and short bowed femurs with anterior bowing of

the tibiae. Hypermobility of the joints was seen, especially of the hips and finger joints. Based on clinical

features and X-ray findings P2-1 was diagnosed with OI type III. Treatment with intravenous Pamidronate

0.5mg/kg/day for 3 days every 6 weeks was started at age 2 weeks. Gross motor development was

delayed with unsupported sitting at age 2.5 years and standing with support at age 4.5 years. No

new fractures occurred between birth until the age of 3 years. There were no signs of dentinogenesis

imperfecta. Kyphoscoliosis of the thoracic and lumbar spine was evident. P2-1 never walked and at the

age of 7 years P2-1 uses a wheelchair. Current height at the age of 8 years is 79.9 cm (SDS -8.4), which is

at the 50 percentile of a 17-month-old child.

Proband 2 from family 2 (P2-2) (Figure 1B)was born by elective Caesarian section at 37 weeks of gestation.

On an antenatal ultrasound scan at 20 weeks of gestation multiple fractures were observed indicative of

OI. Birth weight was 2.32 kg (p10) and a skeletal survey confirmed multiple fractures of ribs and limbs. A

large head with a large anterior fontanelle was observed in combination with some occipital flattening.

There was no chest deformity. The legs were abducted at the hips in frog leg position. Bowing of the

femora and anterior bowing of the tibae was noted. At the age of 6 months her length is 47.4 cm (SDS

-8.8). Treatment with intravenous Pamidronate 0.5 mg/kg/day on 3 consecutive days every 8 weeks was

commenced shortly after birth.

Dijk_Opmaak.indd 80 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 40 T1 - Black CyanMagentaYellow

Page 82: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIB mutations cause severe Osteogenesis Imperfecta 81

Sequencing of the 5 exons of PPIB and adjacent intronic sequences with M13 tailed primers

(Supplementary table 1) yielded a homozygous mutation c.556-559delAAGA in exon 5 resulting in

p.Lys186GlnfsX8 (Figure 2A) in both affected sibs from family 1 (P1-1 and P1-2). This mutation leads to

a frameshift, replacing 31 amino acids at the C-terminal, which are highly conserved in evolution, and

is therefore expected to affect protein function. The parents proved to be heterozygous carriers. We

detected a homozygous mutation c.451 C>T in exon 4 of the PPIB gene, resulting in p.Gln151X (Figure

2B) in both affected sibs from family 2 (P2-1 and P2-2). The parents were heterozygous carriers. This

mutation removes the last 65 amino acids at the C-terminal which may either lead to diminished protein

function or to nonsense mediated decay (NMD). These PPIB mutations were not found in 192 alleles

from clinically normal controls.

1A

1B -1

1B -2

figure 1: Radiological features of affected probands from family 1 and 2(A) Fetal anteroposterior radiographs of proband 1 from family 1 at 21+2 weeks of gestation show: normal skull mineralization for gestational age, slender ribs without fractures, incomplete ossification of T5 and T12, somewhat irregular proximal metaphyses of the humeri, radii and ulnae and bowing of the ulnae. Bowed femora with fractures and some loss of modeling, bowed tibiae and fibula are apparent, possibly with fractures. The radiological features are compatible with Sillence OI type II-B/III.(b1-2) Radiographs of proband 2 from family 2 show normal mineralization of skull, fractures of both humeri, radii and left ulna with callus formation and some loss of modelling of the humeri. Discontinuously beaded ribs are noted with a small bell-shaped thorax. Multiple fractures with callus formation of femora, tibiae and fibula exist with moderate loss of modelling of the femora. The radiological features are compatible with Sillence OI type II-B/III.

Dijk_Opmaak.indd 81 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 41 T1 - Black CyanMagentaYellow

Page 83: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-282

Control

Parent

Patient

2A 2B

figure 2: PPIB mutations in severe OI(A) A homozygous c.556-559delAAGA in exon 5 resulting in p.Lys186GlnfsX8 is shown in probands of family 1. (b) A homozygous c.451 C>T mutation in exon 4 resulting in p.Gln151X is shown in probands of family 2.

The homozygous mutation c.556-559delAAGA in exon 5 leads to a premature stop in the last

exon, therefore NMD is unlikely to occur. Quantitative RT-PCR (Lightcycler480® Roche, Nutley, USA)

(Supplementary table 2) showed equal expression of both wild type and mutant PPIB mRNA in the

parents of P1-1 and P1-2 indicating that no NMD occurs, as was expected. The total mRNA level of mutant

and wild type PPIB in fibroblasts form the parents as well as the level of mutant mRNA in P1-1 were

comparable to the level of wild type PPIB mRNA in healthy control fibroblasts. Interestingly, intracellular

CyPB was detectable in fibroblasts from the control cell line and from the father of P1-1 and P1-2 but

was undetectable in fibroblasts from P1-1 (Figure 3), as measured by Western blot with a monoclonal

CyPB antibody raised against the C-terminal tail (HPA012720, Atlas Antibodies, Stockholm, Sweden)

and two different polyclonal antibodies raised against the whole protein (11607-1-AP, Proteintech,

Manchester, United Kingdom & AF5410, R&D systems, Minneapolis, USA). The absence of truncated CyPB

in the proband P1-1 might be due to the following: (i) truncated protein is generated but the amount is

insufficient to be detected by Western blot, or (ii) truncated protein is rapidly degraded.

Dijk_Opmaak.indd 82 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 41 T1 - Black CyanMagentaYellow

Page 84: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIB mutations cause severe Osteogenesis Imperfecta 83

38

28

1914

49

62

reco

mbi

nant

CyP

B

cont

rol

pare

nt fa

mily

1

pati

ent f

amily

1

CRTAP

CyPB

kD

figure 3: Western blot in family 1A Western blot is shown of fibroblast CyPB, which is lacking in the affected infant P1-1 and present in the parent and the control (polyclonal antibodies raised against the whole protein were used in this Western Blot, 11607-1-AP, Proteintech, Manchester, United Kingdom). CRTAP is present in P1-1, the parent and the control.

To investigate the function of CyPB in the complex and the interactions of CyPB with P3H1 and CRTAP in

complex assembly we performed immunohistochemistry as reported elsewhere 15 with CRTAP antibody

(sc-100920, Santa Cruz, Santa Cruz, USA), P3H1 antibody (H00064175-B01P, Abnova, Taipei, Taiwan)

and the CyPB antibodies raised against the C-terminal tail ((HPA012720, Atlas Antibodies, Stockholm,

Sweden) and the whole CyPB protein (11607-1-AP, Proteintech, Manchester, United Kingdom) on bone

tissue from P1-1 and P1-2 and from two probands with autosomal recessive OI due to CRTAP and LEPRE1

mutations (figure 4A-O). Interestingly, in bone tissue of patients with CRTAP or LEPRE1 mutations, no

antibody staining of both CRTAP and P3H1 was observed (4G-M) whereas in P1-2 with PPIB mutations

no signal of CyPB was noted and a positive signal of CRTAP and P3H1 was identified (4M-O). Apparently,

presence of both CRTAP and P3H1 is needed for a stable protein complex, whereas CyPB is demonstrably

independent of presence of either CRTAP or P3H1. This has been demonstrated previously (W. Chang

et al., 2008, ASBMR, Abstract). Recently, it was reported that the P3H1/CRTAP/CyPB complex has three

distinct activities: it is a prolyl 3-hydroxylase, a PPIase and a molecular chaperone. Consequently, it was

hypothesized that autosomal recessive OI due to LEPRE1 and CRTAP mutations did not only result from

lack of 3-Hyp residues in the collagen type I chains but also from a disturbance in the additional function

of the P3H1/ CRTAP /CyPB complex as a chaperone and PPIase.16 To further investigate this hypothesis

Dijk_Opmaak.indd 83 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 42 T1 - Black CyanMagentaYellow

Page 85: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-284

we performed tandem mass-spectrometry of collagen tryptic peptides 17 in individuals with OI due

to LEPRE1, CRTAP, COL1A1 and PPIB mutations (technical details can be found in Supplementary figure

3). In P1-2, homozygous for a PPIB mutation, P986 3-hydroxylation level is lower (33 %) than control

levels (93-100%) but still higher than observed in patients with CRTAP (16%) and LEPRE1 (22%)mutations

(figure 5). This result and the fact that CyPB is stable itself and does not require presence of CRTAP or

P3H1, in combination with reported decreased 3-prolyl hydroxylation due to deficiency of CRTAP 4

lacking the catalytic hydroxylation domain and the known function of CyPB,suggest that OI is caused

by a dysfunctional CRTAP/P3H1/CyPB- complex rather than by lack of 3-hydroxylation of a single proline

residue in the alpha1 chains of collagen type I. As such, PPIB mutations might also disrupt the folding

of the type I collagen helix due to interference with the other recently reported two functions of the

P3H1/CRTAP/CyPB complex, namely PPIase activity and/or chaperone function. To shed more light on

this, further investigations are warranted. It is already known, that inhibition of CyPB in cultured dermal

fibroblasts with cyclosporine A increases modification of the type I collagen chains, which indicates

that overmodification of collagen type I in autosomal recessive OI due to PPIB mutations results

from decreased rate of cis-trans isomerization by CyPB.18 Collagen type I electrophoresis performed

as reported elsewhere 19, showed overmodification of collagen type I chains comparable to collagen

type I overmodification observed in patients with CRTAP and LEPRE1 mutations and in patients with

COL1A1 mutations causing lethal/severe OI (figure 6) . In conclusion, we describe mutations in the PPIB

gene encoding the third member, CyPB, of the collagen-modifying complex. PPIB mutations result in

autosomal recessive OI with a clinical picture compatible with OI type II-B/III as is the case with CRTAP4,

13, 20 and LEPRE1 5, 13mutations. Our findings in combination with recent literature support the hypothesis

that OI due to PPIB mutations results from a disruption of the P3H1/CRTAP/CyPB complex functioning as

a prolyl 3-hydroxylase, a PPIase and a molecular chaperone.16

Dijk_Opmaak.indd 84 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Back - 42 T1 - Black CyanMagentaYellow

Page 86: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIB mutations cause severe Osteogenesis Imperfecta 85

A B C

D E F

G H I

J K L

M N O

CRTAP AB P3H1 AB CyPB AB

Patient without OI

Patient with OI dueto COL1A1 mutation

Patient with OI dueto CRTAP mutations

Patient with OI dueto LEPRE1 mutations

Patient with OI dueto PPIB mutations

figure 4: Results of immunohistochemistry with antibodies (AB) against CRTAP, P3H1 and CyPB Immunohistochemistry performed on bone tissue with CyPB-CRTAP- and- P3H1- antibodies shows in:(A-C) a control without OI, a positive signal of CyPB, CRTAP and P3H1 (D-F) a patient with a COL1A1 mutation (c.1238G>A [p.Gly413Asp]), a positive signal of CyPB, CRTAP and P3H1 (G-I) a patient with a homozygous c.404delG CRTAP mutation,no signal of either CRTAP or P3H1 whereas a signal of CyPB is present. (J-L) a patient with a homozygous c.401_402delAALEPRE1 mutation, no signal of either P3H1 or CRTAP whereas a signal of CyPB is present.(M-O) P1-2 with a homozygous PPIB mutation, no signal of CyPB whereas signals of both CRTAP and P3H1 antibodies are present.

Dijk_Opmaak.indd 85 17-10-11 16:54

Dijk_PROEF_16x24 (all).ps Front - 43 T1 - Black CyanMagentaYellow

Page 87: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-286

figure 5: Collagen I alpha-1 P986 3-hydroxylation level in patients with PPIB (A), LEPRE1 (B), CRTAP (C) mutations and a COL1A1 mutation (D) compared to parents of the patient with PPIB mutations (E,F) and a control (G).Figure 5 shows mass spectrometry analysis of the C1A1 P986-containing tryptic peptide, modified with either 2 or 3 hydroxyl groups (as indicated by * below the modified proline residue in supplementary Figure X). The left panel shows extracted ion chromatograms (XIC), generated at m/z 773.4 and 781.4 +/- 0.05 and the right panel shows the corresponding summed mass spectra (40-50 min.). XIC Peak areas were determined for double and triple hydroxylated peptides. The C1A1 P986 3-hydroxylation level (%) was calculated as triple hydroxylation area/ (triple hydroxylation area+double hydroxylation area). Collagen I alpha-1 peptides that show normal P986 3-hydroxylation have an m/z ratio of 781.4 whereas peptides lacking 3-hydroxylation have an m/z ratio of 773.4. Controls (D-F) show normal 3-hydroxylation (>90%). P1-1 with homozygous PPIB mutations (A) shows decreased 3-hydroxylation (33%). Patients with homozygous LEPRE1 (p.Glu134fs) or CRTAP (p.Ala8fs) mutations (B and C, respectively), however, show even more decreased 3-hydroxylation levels (22% and 16%, respectively).

Dijk_Opmaak.indd 86 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 43 T1 - Black CyanMagentaYellow

Page 88: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIB mutations cause severe Osteogenesis Imperfecta 87

CRTAP-OI LEPRE1 -OI PPIB -OI COL1A1 -OI Control

Chorionic villi

Fibroblasts

c m c m

mc

c m

c m

c m

c m mc

c m

mc

COL3A1

COL5A1

COL5A2

COL1A1

COL1A2

COL3A1

COL5A1

COL5A2COL1A1

COL1A2

figure 6: Electrophoresis of collagen type I chains in fibroblasts and chorionic villi of patients with CRTAP, LEPRE1, PPIB or COL1A1 mutations. Backstreaking and/or increased band width is visible in cell (c) and media (m), indicating overmodification of collagen type I chains in chorionic villi cells and fibroblasts of patients with lethal/ severe OI due to homoygous CRTAP(p.Ala8fs), LEPRE1 (p.Glu134fs), PPIB (p.Lys186GlnfsX8 ) mutations or a COL1A1 (p.G965S in chorionic villi cells and p.G662V in fibroblasts) mutation. The slight apparent overmodification of other collagens appears to be an effect typically evident in chorionic villi.

Supplementary table 1: PCR primers for DNA and cDNA sequencing

M13 Tailed Primer DNA Sequence

1F

1R

2F

2R

3F

3R

4F

4R

5F

5R

5’-GTAAAACGACGGCCAGCCGCGAGCAACCCCAGTC-3’

5’-CAGGAAACAGCTATGAACGCCACGAGGCCACAGACAGAA-3’

5’-GTAAAACGACGGCCAGCCGTCACCAAAATCAGATTCAGAACCA-3’

5’-CAGGAAACAGCTATGAGGTGCTCAGTGAGGTCCACGCTACAGA-3’

5’-GTAAAACGACGGCCAGCCTGCTGCCCCATACCAC-3’

5’-CAGGAAACAGCTATGAACCATCAGGTCCACCCCATTAT-3’

5’-GTAAAACGACGGCCAGTCTGCTTGGTTTGGCGCAATC-3’

5’-CAGGAAACAGCTATGAACTGGGCTGCATCTGTGGGTTGGGT-3’

5’-GTAAAACGACGGCCAGGCCTTTCTCCTGAGCGGTGGAC-3’

5’-CAGGAAACAGCTATGAGGGGAGGTGCTGCAGGCTCAAGAAC-3’

M13 Tailed Primer cDNA Sequence

1F

1R

2F

2R

5’-GTAAAACGACGGCCAGCCCGCGAGCAACCCCAGT-3’

5’-CAGGAAACAGCTATGAGGGGAAGCGCTCACCGTAGATG-3’

5’-GTAAAACGACGGCCAGCATGATCCAGGGCGGAGACTTC-3’

5’-CAGGAAACAGCTATGACATGGGCCTGTGGAATGTGAG-3’

Dijk_Opmaak.indd 87 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 44 T1 - Black CyanMagentaYellow

Page 89: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-288

Supplementary figure 1: radiological features of affected individuals from family 1 and 2.A) Fetal anteroposterior radiopgraphs of P1-2 at 16+1 weeks of gestation show normal skull mineralization for gestational age, fractures of humeri, normal ribs without fractures, bilateral bowed and fractured femora with moderate loss of modeling, slight bowing of tibiae and fibula with fractures in both proximal tibiae. All long bones show irregular metaphyses. b) Radiograph of skull of P2-1 at two years of age shows brachycephaly with normal mineralization of the skull with the exception of an open anterior fontanel and multiple wormian bones in the occipital region. C) Radiograph of right forearm of P2-1 at two years of age shows bowing of radius and ulnaD) Radiograph of P2-1 at five years of age show osteopenia with bowing and fracture of the humerus and bowing of the radius with midshaft callus formation E) Radiograph of lower legs of P2-1 at two years of age shows osteopenia and bilateral bowing of tibiae and fibula. f) Radiograph of proband P2-1 at five years of age shows osteopenia with severe bowing of left femur. G) Radiograph of P2-1 at 8 years of age shows calcifications in the distal metaphysis of the femur and the proximal metaphysis of the tibia. H) Radiograph of P2-1 at 6 years of age shows platyspondyly of L2 and L3

Supplementary table 2: PCR primers for quantitative RT-PCR

Primer cDNA sequence

Wild type sense

Mutant sense

Wild type antisense

Mutant antisense

5’-GATCCAGGGCGGAGACTT-3’

5’-TGATCCAGGGCGGAGACTTC-3’

5’-TTATCCCGGCTGTCTGTCTT-3’

5’-TTATCCCGGCTGTCTGGGT-3’

Dijk_Opmaak.indd 88 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 44 T1 - Black CyanMagentaYellow

Page 90: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIB mutations cause severe Osteogenesis Imperfecta 89

Supplementary figure 2: Tandem mass-spectometry of collagen tryptic peptides.MS/MS spectra of the P986 containing tryptic peptide DGLNGLPGPIGPPGPR with hydroxyproline assignment (* below modified proline) are shown. MS/MS spectra for the double hydroxylated peptide (2 HyP) ([M+2H]2+=773.4) are shown for CyPB (A), P3H1 (B) and CRTAP (C). The y10 (976.5) and y8 (806.5) ions support hydroxylation of P981 and not P983. The y6 (596.3)/b10 (950.5) ions localize the other hydroxyproline to the C-terminal -GPPGPR fragment (including P986), however, fragment ions discriminating between P986, P987 and P989 (b12, b13 , y3 and y4) are in all three cases detected at very low intensity and prohibit more detailed localisation. For the triple-hydroxylated peptides (3 HyP), shown for Control (D) and CyPB (E), the y10 (992.5) and y8 (822.4) ions support P981 hydroxylation. Furthermore, the y6 (612.3) supports double hydroxylation in the C-terminal –GPPGPR fragment, including P986. The minor y3 (329.2) fragment ion supports hydroxylation of both P986 and P987 and not P989.

Collagen hydroxylation analysis (technical details)Fibroblast culture lysates were preincubated with pepsin/HAc for 24 h. at 4˚C. After concentration (Amicon Utra 10 kDa, Millipore) proteins were separated by 5% PA-Urea-SDS gel. Collagen 1A1 bands were excised from the urea SDS PAGE gel and processed for in-gel digestion with trypsin 18. Tryptic peptides were separated by nanoLC (Ultimate 3000, Dionex) using a 75 µm ID x 30 cm C18 (ReproSil pur C18 5 µm) reversed phase column in an acetonitrile gradient (10-40% B in 60 min, A = 0.05% aqueous formic acid, B = 80% acetonitrile in 0.05% formic acid) at 300 nl/min. Peptides eluted from the column were analysed on-line by nanoESI-MS on a hybrid LTQ-FT mass spectrometer (ThermoFisher). Intact peptide masses were measured in the ICR cell at resolution 50000 and MS/MS spectra (top 5) were measured in the linear ion trap at unit resolution. LC-MS data was searched against the human IPI database (version 3.59) using Sequest 3.3 (ThermoFisher) allowing a maximum mass error of 10 PPM and variable modifications for cysteine (carboxamidomethylation), methionine (oxidation) and proline (hydroxylation). Sequest peptide identifications were validated using the PeptideProphet (>95%) and ProteinProphet (>99%) algorithms (Scaffold 2.0, Proteomesoftware).

Dijk_Opmaak.indd 89 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 45 T1 - Black CyanMagentaYellow

Page 91: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-290

ACKNOWLEDGEMENTSWe thank the families of the affected individuals for their kind cooperation. Y. Moutaouakil established

fibroblast cell lines of affected individuals and parents, E. van den Akker conducted prior collagen

electrophoretic studies on affected individuals from family 1, S. Elshof and K.van der Ven performed

immunohistochemistry. We thank B. Zandiehdoulabi for his help and advice, L. Scheffer for her technical

support and J. Hewitt for providing additional data on family 2.

WEb rESOurCES

The URLs for data presented herein as follows:

Online Mendelian Inheritance in Man (OMIM), http://www.ncbi.nlm.nih.gov

Dijk_Opmaak.indd 90 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 45 T1 - Black CyanMagentaYellow

Page 92: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

PPIB mutations cause severe Osteogenesis Imperfecta 91

rEfErENCES

1. Marini JC, Cabral WA, Barnes AM, and Chang W (2007) Components of the collagen prolyl 3-hydroxylation complex are crucial for normal bone development. Cell Cycle 6 (14):1675-1681

2. Canty EG and Kadler KE (2005) Procollagen trafficking, processing and fibrillogenesis. J Cell Sci 118 (Pt 7):1341-1353

3. Engel J and Prockop DJ (1991) The zipper-like folding of collagen triple helices and the effects of mutations that disrupt the zipper. Annu Rev Biophys Biophys Chem 20:137-152

4. Barnes AM, Chang W, Morello R, Cabral WA, Weis M, Eyre DR, Leikin S, Makareeva E, Kuznetsova N, Uveges TE, et al. (2006) Deficiency of cartilage-associated protein in recessive lethal osteogenesis imperfecta. N Engl J Med 355 (26):2757-2764

5. Cabral WA, Chang W, Barnes AM, Weis M, Scott MA, Leikin S, Makareeva E, Kuznetsova NV, Rosenbaum KN, Tifft CJ, et al. (2007) Prolyl 3-hydroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfecta. Nat Genet 39 (3):359-365

6. Mizuno K, Peyton DH, Hayashi T, Engel J, and Bächinger HP (2008) Effect of the -Gly-3(S)-hydroxyprolyl-4(R)-hydroxyprolyl- tripeptide unit on the stability of collagen model peptides. FEBS J 275 (23):5830-5840

7. Vranka JA, Sakai LY, and Bächinger HP (2004) Prolyl 3-hydroxylase 1, enzyme characterization and identification of a novel family of enzymes. J Biol Chem 279 (22):23615-23621

8. Krane SM (2008) The importance of proline residues in the structure, stability and susceptibility to proteolytic degradation of collagens. Amino Acids 35 (4):703-710

9. Price ER, Zydowsky LD, Jin MJ, Baker CH, McKeon FD, and Walsh CT (1991) Human cyclophilin B: a second cyclophilin gene encodes a peptidyl-prolyl isomerase with a signal sequence. Proc Natl Acad Sci U S A 88 (5):1903-1907

10. Yao Q, Li M, Yang H, Chai H, Fisher W, and Chen C (2005) Roles of cyclophilins in cancers and other organ systems. World J Surg 29 (3):276-280

11. Smith T, Ferreira LR, Hebert C, Norris K, and Sauk JJ (1995) Hsp47 and cyclophilin B traverse the endoplasmic reticulum with procollagen into pre-Golgi intermediate vesicles. A role for Hsp47 and cyclophilin B in the export of procollagen from the endoplasmic reticulum. J Biol Chem 270 (31):18323-18328

12. Zeng B, MacDonald JR, Bann JG, Beck K, Gambee JE, Boswell BA, and Bächinger HP (1998) Chicken FK506-binding protein, FKBP65, a member of the FKBP family of peptidylprolyl cis-trans isomerases, is only partially inhibited by FK506. Biochem J 330 ( Pt 1):109-114

13. Baldridge D, Schwarze U, Morello R, Lennington J, Bertin TK, Pace JM, Pepin MG, Weis M, Eyre DR, Walsh J, et al. (2008) CRTAP and LEPRE1 mutations in recessive osteogenesis imperfecta. Hum Mutat 29 (12):1435-1442

14. Willaert A, Malfait F, Symoens S, Gevaert K, Kayserili H, Megarbane A, Mortier G, Leroy JG, Coucke PJ, and De Paepe A (2009) Recessive osteogenesis imperfecta caused by LEPRE1 mutations: clinical documentation and identification of the splice form responsible for prolyl 3-hydroxylation. J Med Genet 46 (4):233-241

15. Groenendaal F, Vles J, Lammers H, De VJ, Smit D, and Nikkels PG (2008) Nitrotyrosine in human neonatal spinal cord after perinatal asphyxia. Neonatology 93 (1):1-6

16. Ishikawa Y, Wirz J, Vranka JA, Nagata K, and Bächinger HP (2009) Biochemical characterization of the prolyl 3-hydroxylase 1/CRTAP/cyclophilin B complex. J Biol Chem (Epub ahead of print)

17. Shevchenko A, Wilm M, Vorm O, and Mann M (1996) Mass spectrometric sequencing of proteins silver-stained polyacrylamide gels. Anal Chem 68 (5):850-858

18. Steinmann B, Bruckner P, and Superti-Furga A (1991) Cyclosporin A slows collagen triple-helix formation in vivo: indirect evidence for a physiologic role of peptidyl-prolyl cis-trans-isomerase. J Biol Chem 266 (2):1299-1303

19. Korkko J, la-Kokko L, De PA, Nuytinck L, Earley J, and Prockop DJ (1998) Analysis of the COL1A1 and COL1A2 genes by PCR amplification and scanning by conformation-sensitive gel electrophoresis identifies only COL1A1 mutations in 15 patients with osteogenesis imperfecta type I: identification of common sequences of null-allele mutations. Am J Hum Genet 62 (1):98-110

20. Van Dijk FS, Nesbitt IM, Nikkels PG, Dalton A, Bongers EM, van de Kamp JM, Hilhorst-Hofstee Y, Den Hollander NS, Lachmeijer AM, Marcelis CL, et al. (2009) CRTAP mutations in lethal and severe osteogenesis imperfecta: the importance of combining biochemical and molecular genetic analysis. Eur J Hum Genet (Epub ahead of print)

Dijk_Opmaak.indd 91 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 46 T1 - Black CyanMagentaYellow

Page 93: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 92 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 46 T1 - Black CyanMagentaYellow

Page 94: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large family: a novel homozygous LEPRE1 mutation and bone histological findings.

van Dijk FS1*, Nikkels PGJ2, den Hollander NS3, Nesbitt IM4, van Rijn RR5, Cobben JM6, Pals G7.

1 Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands.2 Department of Pathology, University Medical Centre Utrecht, the Netherlands.

3 Department of Clinical Genetics, Leiden University Medical Centre, Leiden, the Netherlands. 4 Sheffield Molecular Genetics Service, Sheffield Children’s Hospital NHS Foundation Trust, United Kingdom.

5 Department of Pediatric Radiology, Academic Medical Centre, Amsterdam, the Netherlands, 6 Department

of Pediatrics, Academic Medical Centre, Amsterdam, the Netherlands.7 Centre for Connective Tissue Research, Department of Clinical Genetics, VU University Medical Centre,

Amsterdam, The Netherlands.

Pediatr Dev Pathol. 2011 May-Jun;14(3):228-34.

III-3

Dijk_Opmaak.indd 93 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 47 T1 - Black CyanMagentaYellow

Page 95: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-394

AbSTrACTWe report a large consanguineous Turkish family in which multiple individuals are affected with

autosomal recessive lethal or severe osteogenesis imperfecta (OI) due to a novel homozygous LEPRE1

mutation. In one affected individual histological studies of bone tissue were performed, which may

indicate that the histology of LEPRE1 associated OI is indistinguishable from COL1A1/2, CRTAP and

PPIB related OI.

Dijk_Opmaak.indd 94 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 47 T1 - Black CyanMagentaYellow

Page 96: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 95

INTrODuCTION

Osteogenesis Imperfecta (OI) is a heterogeneous group of diseases characterized by susceptibility to

bone fractures with variable severity and with presumed or proven defects in collagen type I biosynthesis

(1). Because of the variable clinical phenotype, a numerical classification in OI types I-IV was proposed

in 1979 (2) based on (i) clinical findings and (ii) mode of inheritance, thus assuming heterogeneity in OI.

However, after the characterization of heterozygous mutations in the collagen type I genes as a cause

of all types of OI and subsequent publications delivering evidence for germline mosaicism as a cause of

recurrence in OI, a predominantly autosomal dominant mode of inheritance in OI was proposed (3). The

Sillence criteria were still used to describe the severity continuum and differences of clinical expression

in OI. Mild, lethal, severe and moderate deforming OI were classified as OI type I, II, III and IV, respectively.

Later it appeared that not all patient with lethal or severe OI had a collagen type I mutation which led to

expansion of the Sillence classification with OI types V-VIII because of distinct clinical, histological and/or

genetic findings (4). In our recent proposed classification of OI, only OI types I-VI are listed (1).

Deficiency of prolyl 3-hydroxylase 1 (P3H1) (5), Cartilage Associated Protein (CRTAP) (6,7) and cyclophilin

B (CyPB) (8-10) due to LEPRE1, CRTAP and PPIB mutations respectively, have been reported as causes of

autosomal recessive OI types II-IV with posttranslational overmodification of collagen type I (1). These

three proteins are known to form a complex, responsible for posttranslational modification of collagen

type I, II and III. One known function of the complex affecting collagen type I is the 3-hydroxylation of a

single proline residue at position 986 in the pro a1(I) collagen chain and it is highly likely that the complex

also acts as cis-trans isomerase and as a molecular chaperone (11). However, the exact mechanism by

which deficiency of the various components of the complex cause OI remains to be further investigated

(5,7,8,11).

In any case, it is evident that deficiency of CRTAP, P3H1 or CyPB leads to overprocessing of the constituent

collagen type I chains (5,7,8). This phenomenon is known as posttranslational overmodification of

collagen type I and it is visible as delayed migration of collagen type I on electrophoresis. Comparable

delayed migration of collagen I can be observed due to certain mutations in the COL1A1/2 genes (12).

We report a large consanguineous Turkish family in which multiple individuals were affected with lethal

or severe OI due to a novel homozygous LEPRE1 mutation.

Clinical report

Proband VI:1 was a female born to Turkish consanguineous parents (first cousins) at a pregnancy

duration of 40 weeks. The pedigree is depicted in figure 1. On physical examination blue sclerae and

wide fontanels were noted. No apparent abnormalities of the thorax were evident on visual examination.

The lower extremities were short and deformed. Multiple fractures were observed on the skeletal survey

(figure 2G-I) . The girl was diagnosed with OI type III. Posttranslational overmodification of collagen type

I was evident on electrophoresis, confirming the diagnosis (figure 3). The girl died in the first year of life

due to pneumonia. A third pregnancy (VI:3) was terminated after demonstration of overmodification of

collagen type I in cultured chorionic villus cells (figure 3). Complementary DNA sequencing in VI:1 and

VI:3 revealed no mutations in the COL1A1 and COL1A2 genes.

Dijk_Opmaak.indd 95 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 48 T1 - Black CyanMagentaYellow

Page 97: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-396

The family history is remarkable since the maternal and paternal grandfather (III:6) of V:17 and V:18

respectively had a brother III:4 and a sister III:3 whose children IV:9 and IV:10 married each other (first

cousin marriage) and had multiple children affected with OI. The first child was born at 40 weeks of

gestation with multiple fractures suggestive of OI. He was diagnosed with OI type II-B and he died shortly

after birth. The second child was affected with OI type III. She died 6 months after birth. Two pregnancies

were terminated after 22 and 20 weeks respectively because of abnormalities on ultrasound suggestive

of OI. A spontaneous miscarriage occurred three times. One healthy son was born. Histology of V:8 was

in keeping with a diagnosis of severe OI (figure 4).

I:1 I:2

II:1 II:2 II:3 II:4

III:1 III:2 III:3 III:4 III:5 III:6 III:7

IV: 1‐4 IV:5 IV:6‐8 IV:9 IV:10 IV:11 IV:12 IV:13 IV:14‐15 IV:16 IV:17 IV:18

V:1 V:2 V:3 V:4 V:5‐7 V:8 V:9‐12 V:13‐16 V:17 V:18 V:19 V:20

VI:1 VI:2 VI:3

figure 1: family pedigree

figure 3: Collagen type I electrophoresisOvermodification of the collagen type I chains is observed in cells (c) and media (m) of affected individuals (marked with an asterisk) VI:1 and VI:3.

Dijk_Opmaak.indd 96 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 48 T1 - Black CyanMagentaYellow

Page 98: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 97

figure 2: Radiographs of patients with OI type II-A or II-B due to collagen type I mutations and OI type II-B or III due to CRTAP, LEPRE1 or PPIB mutations.A: Radiograph of fetus at 22 weeks of gestation with OI II-A due to a heterozygous c.2300 G>A COL1A1 mutation.Diminished skull mineralization for gestational age, broad continuously beaded ribs, Platyspondyly of vertebral bodies, fractures of claviculae and shortening, bowing and fractures of all long bones with severe undermodeling of humeri and femora. B: Radiograph of fetus at 23 weeks of gestation with OI II-B due to a heterozygous c.3043 G>A COL1A1 mutation.Normal skull mineralization for gestational age, slender ribs without fractures, slight undermodeling of the femora, bowing and fractures of the femora , bowing of the left tibiae and fibula possibly due to fractures, bowing of the right tibia, the left fibula is not visible at this radiograph. C: Radiograph of fetus at 21+2 weeks of gestation with OI II-B/III due to a homozygous c.556-559delAAGA PPIB mutation (8). Normal skull mineralization for gestational age, slender ribs without fractures, incomplete ossification of T5 and T12, somewhat irregular proximal metaphyses of the humeri, radii and ulnae and bowing of the ulnae. Bowed femora with fractures and some loss of modeling, bowed tibiae and fibula are apparent, possibly with fractures.D-F: Postnatal radiographs of individual (three weeks old) affected with OI due to a homozygous c.404delG CRTAP mutation (12)D. Small thorax. Slender ribs. Lateral fracture of 8th left rib (see insert). Asymmetric platyspondyly Th5 and Th7 (visible on original radiograph). E. Consolidation of right humerus fracture (arrow). Humerus measures 4.38 cm (normal range=6.0-7.7 cm.), ulna 4.9 cm (normal range =5.8-7.2 cm.). Broad distal metaphyses of left humerus and radius (arrow head). Possible fracture of left radius.F. Bilateral proximal femur fracture with consolidation and angulation (arrows). Bilateral bowing of tibia and fibula (arrow heads).G-I: Postnatal radiographs of individual VI:1 affected with OI due to a homozygous c.401_402delAA LEPRE1 mutation G. Chest radiograph shows slender ribs without apparent fractures (see insert). The costochondral junctions show cupping.H. Radiograph shows deformation of the right femur (open arrow) with diaphyseal broadening. The left femur shows bowing (arrow) and broadening, the metaphysis are ill defined (a sign of under-mineralization). The tibia shows an acute angle (arrowhead) in keeping with a previous fracture.I. Radiograph of the right arm shows deformation of the right arm (open arrow) in keeping with consolidated fractures. The radius shows shortening in the distal aspect (arrow) with disruption of the distal radio-ulnar joint.

Dijk_Opmaak.indd 97 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 49 T1 - Black CyanMagentaYellow

Page 99: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-398

METHODS

PatientsThe family depicted in figure 1 was part of a panel of families in which we searched for mutations

in candidate genes for OI. In total, we selected 13 individuals from 7 families for screening based on

clinical/radiological diagnosis of OI type II/III, collagen type I overmodification on electrophoresis and

absence of collagen type I mutations.

For comparison of the radiologic and histologic findings, we also selected two individuals affected with

OI type II-A and II-B due to collagen type I mutations and individuals previously published with CRTAP

(12) and PPIB mutations (8).

Collagen modification analysisSkin fibroblasts and chorionic villus cells were obtained from respectively VI:1 and VI:3, and cultures were

established under standard conditions. Cells were seeded at 35,000 cells/ cm2. Collagen modification

analysis was performed as reported elsewhere (12).

HistologyBone tissue from V:8 and from individuals with collagen type I mutations, CRTAP and PPIB mutations

was obtained at postmortem examination and fixed using 4% buffered paraformaldehyde. Routine

samples for histology were taken from the femur. The samples were embedded in paraffin by standard

histological procedures after decalcification with buffered formic acid according to Kristensen.

Molecular analysiscDNA of the COL1A1 and COL1A2 genes was prepared from total RNA from cultured fibroblasts. RNA was

isolated using the RNA isolation minikit (Qiagen) according to the manufacturers instructions. Full-length

single-stranded cDNA was prepared with oligo-dT-primer and SuperscriptTMII RT reverse transcriptase

(Invitrogen). The complete coding sequences of the COL1A1 and COL1A2 genes were both analyzed by

direct sequencing in 11 overlapping PCR fragments (primer sequences available on request).

Molecular analysis of the CRTAP and LEPRE1genes was performed as reported elsewhere (5,7).

rESuLTS

PatientsIn all 13 individuals from 7 families posttranslational overmodification of collagen type I on electrophoresis

was evident. In 1 individual no mutations in CRTAP, LEPRE1 and PPIB were detected and 10 individuals

from 5 families appeared to have novel CRTAP mutations (12). In 2 individuals VI:1 and VI:3 from our

reported family, LEPRE1 mutations were detected. Here we discuss the results of the family with OI due

to LEPRE1 mutations only.

Dijk_Opmaak.indd 98 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 49 T1 - Black CyanMagentaYellow

Page 100: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 99

Collagen modification analysisOn biochemical analysis of collagen type I, posttranslational overmodification was evident in affected

individuals VI:1 and VI:3. As a coincidental finding, in VI:3 overmodification of collagen type V was

apparent as was the case in the unaffected individual VI:2.

HistologyThe histology of the affected individuals with OI type II-A and OI type II-B (figure 4 A-F) caused by

collagen type I mutations and the histology of the affected individuals with OI due to CRTAP, LEPRE1

or PPIB mutations (figure 4 G-O) shows a normal resting cartilage with similar sparse vascular channels

surrounded by loose connective tissue. The epiphyseal growth zone shows a normal organization of

the chondrocyte columns in all affected individuals including the individual diagnosed with OI type

II-A. In some cases of OI type II-A, however, one can occasionally observe a disturbed growth zone due

to fractures close to the growth zone and the formation of metaplastic cartilage in this fractured area.

Periosteal ossification is only partially present and most severely affected in the patient with OI type II-A

(A-C), especially immediately below the cartilage growth plate. Periosteal bone formation is less affected

in the patient with OI type II-B due to a COL1A1 mutation (D-F). The individuals with OI due to CRTAP,

LEPRE1 or PPIB mutations show a periosteal ossification in between severely diminished as is observed

in the patient with OI type II-A (A-C) and milder affected as is observed in the patient with OI II-B due

to a COL1A1 mutation (D-F). The periosteal bone lamellae show some fractures in all cases (again more

severe in the patient with OI type II-A) and a comparable accumulation of osteclasts. The perichondreal

groove of Ranvier, a stem cell niche for articular cartilage, has a similar aspect in all cases (13). The bony

band in the perichondreal ring of LaCroix shows comparable features as was described for the periosteal

bone formation; i.e. the least pronounced in the OI type II-A case (A-C) and better formed in the other

cases (D-O). The primary and secondary spongiosa consists of osteoid in combination with hypercellular

and thin bony trabeculae, more slender in the patient with OI type II-A (A-C). These trabeculae all consist

of woven bone with almost complete absence of lamellar bone. In the affected individual with OI

type II-A (A-C) multiple fractures can be seen with ample metaplastic cartilage. The bony trabeculae in

the other affected individuals (D-O) are broader and also consist mainly of woven bone. Occasionally

fractures can be seen and only in the affected individual with OI due to PPIB mutations (J-L) a very small

island of metaplastic cartilage was observed (not shown). The medullary cavity in all cases is fibrotic. The

transition zone between cartilage and bone is in general sharply demarcated

Molecular analysis

DNA analysis of the CRTAP and LEPRE1 genes revealed a novel homozygous c.401_402delAA mutation

in exon 1 of the LEPRE1 gene in affected individuals VI:1 and VI:3. This mutation results in a p.Glu134fs

amino acid change in exon 1 of the LEPRE1 gene. The parents V:17 and V:18 were heterozygous for this

mutation. No DNA of affected individuals V:1-3 and V:8 could be retrieved but the parents IV:9 and IV:10

of these affected individuals were also heterozygous for the above mentioned LEPRE1 mutation.

Dijk_Opmaak.indd 99 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 50 T1 - Black CyanMagentaYellow

Page 101: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-3100

figure 4: Histology of individuals with OI due to different genetic causes. Immunohistochemical studies published elsewhere (8) confirmed the molecular defect by showing absence of the particular protein in F-O. A-C Histology of a fetus with OI type II-A due to a heterozygous c.2300 G>A COL1A1 mutation D-F: Histology of a fetus with OI type II-B due to a heterozygous c.3043 G>A COL1A1 mutationG-I : Histology of a newborn with OI type II-B/ III due to a homozygous c.404delG CRTAP mutation (12).J-L: Histology of a fetus with OI type II-B/ III due to a homozygous c.556-559delAAGA PPIB mutation (8).M-O: Histology of a fetus (V:8) with OI type II-B/III due to a homozygous c.401_402delAA LEPRE1 mutation.A,D,G,JM (overview): H and E stain showing a sharp demarcation between cartilage and bone. The resting cartilage and epiphyseal growth zone shows no abnormalities.B,E,H,K,N (detail): H and E stain shows typical small, slender and hypercellular bony trabeculae and fibrotic medulla. The bony trabeculae consist of woven bone. OI type IIA (B) shows severest abnormalities. E,H,K,N are comparable.C,F,I,L,O (detail): H and E stain shows osteoclastic giant cells in C,F,I,L,O and metaplastic cartilage matrix with chondrocytes in C.

Dijk_Opmaak.indd 100 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 50 T1 - Black CyanMagentaYellow

Page 102: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 101

DISCuSSION

The affected individuals in our family have recessive OI due to a novel homozygous LEPRE1 mutation.

They were all diagnosed with OI type II-B or III based on the skeletal surveys, which is in line with the fact

that although some differentiating clinical/radiological characteristics of patients with LEPRE1 mutations

have been claimed (5), a distinct genotype-phenotype correlation has not emerged at present (1).

However, all cases of autosomal recessive OI caused by CRTAP, LEPRE1 or PPIB mutations presented in

literature and observed by the authors, can be radiologically classified as OI type II-B or milder. OI type

II-A has to our knowledge so far only been observed in patients with OI due to a COL1A1/2 mutation.

The radiological subclassification of OI type II was proposed in 1984. OI type II-A is characterized by

broad ribs with continuous fractures (continuous beading) and severe undermodeling of the femora

whereas OI type II-B is characterized by thinner ribs with some fractures (discontinuous beading) and

some undermodeling of the femora (14). Figure 2 shows radiographs from affected individuals with OI

type II-A and II-B due to collagen type I mutations and OI type II-B or III due to CRTAP and PPIB mutations.

Bone histology of individuals with OI due to different genetic causes has never been compared in the

literature. The bone histology of affected individual V:8 appears to be indistinguishable from bone

histology of patients with OI due to mutations in other genes (figure 4). As is known, CRTAP -/- mice show

abnormalities in growth columns of proliferating chondrocytes in the growthplate and decreased prolyl-

3-hydroxylation in cartilage consistent with the knowledge that cartilage contains collagen type II which

has also one proline residue at position 986 at the alpha1 chain that is subject to 3-prolyl hydroxylation

(6). However, histology of affected fetus V:8 with LEPRE1 mutations (4M-O) does not show abnormalities

of chondrocytes in the growthplate as was the case with patients with OI due to a COL1A1 mutation and

PPIB mutations. This might be due to the fact that our case concerns fetal material. However, in another

newborn with OI due to CRTAP mutations no abnormalities of chondrocytes are apparent (figure 4G-

I)) (12). So although the CRTAP -/- mice show histological abnormalities of the chondrocytes in the

growthplate, this appears not to be the case in humans with autosomal dominant OI or recessive OI due

to CRTAP, LEPRE1 or PPIB mutations.

In the individuals with OI due to LEPRE1 mutations, Cabral et al. reported overmodification of collagen

type V besides overmodification of collagen type I suggesting that P3H1 may directly function as a

chaperone for other types of collagen as well (5). In our patients, on biochemical analysis of VI:1 and

VI:3 posttranslational overmodification of collagen type I was evident (figure 3). No overmodification of

collagen type V was observed in the affected individual VI:1. Overmodification of collagen type V seems

to be visible in VI:2 and VI:3 which is strange since V:2 is not affected with OI. However, in VI:2 and VI:3

electrophoresis was performed on chorionic villi. Overmodification of collagen type V was also noted

in electrophoresis performed on control chorionic villi and therefore it appears to be an effect typically

evident in electrophoresis performed on chorionic villi (8) and is not pointing to a more general collagen

chaperone effect.

Mutation analysis revealed a c.401_402del AA in exon 1 of the LEPRE1 gene in affected individuals VI:1

and VI:3. This novel mutation results in a p.Glu134fs amino acid change which leads to a stop codon 41

aminoacids downstream. Eighteen different mutations in the LEPRE1 gene have been described so far

Dijk_Opmaak.indd 101 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 51 T1 - Black CyanMagentaYellow

Page 103: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-3102

in the literature (15). The c.1080+1 G>T LEPRE1 mutation is reported as the “West African Allele” with an

estimated carrier frequency of between 1/100 and 1/280 in populations from West Africa (5).

In conclusion, the individuals reported here with homozygous LEPRE1 mutations, all have a diagnosis of

OI type II-B/III based on clinical findings and skeletal surveys. Bone histology of a fetus with OI due to this

novel homozygous LEPRE1 mutation is indistinguishable from bone histology of patients with OI type

II-B/III due to a COL1A1 mutation or due to CRTAP and PPIB mutations. Posttranslational overmodifcation

of collagen type I is evident in all investigated individuals. The reported family illustrates the importance

of DNA analysis of the CRTAP, LEPRE1 and PPIB genes whenever one finds overmodification of collagen

type I on electrophoresis and absence of collagen type I mutations.

ACKNOWLEDGEMENTS

We thank the parents of the affected individuals in this family for their kind cooperation and we thank

pathologist H.H. Dahm for providing bone tissue of affected individual V:8.

Dijk_Opmaak.indd 102 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 51 T1 - Black CyanMagentaYellow

Page 104: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 103

rEfErENCES

1. van Dijk FS, Pals G, van Rijn RR, Nikkels PGJ, Cobben JM. Classification of Osteogenesis Imperfecta revisited. Eur J Med Genet 2009;53:1-5.

2. Sillence DO, Senn A, Danks DM. Genetic heterogeneity in osteogenesis imperfecta. J Med Genet 1979;16: 101-116.

3. Byers PH, Tsipouras P, Bonadio JF, et al. Perinatal lethal osteogenesis imperfecta (OI type II): a biochemically heterogeneous disorder usually due to new mutations in the genes for type I collagen. Am J Hum Genet 1988; 42: 237-248.

4. Rauch F, Glorieux FH. Osteogenesis Imperfecta. Lancet 2004; 363:1377-85.

5. Cabral WA, Chang W, Barnes AM, et al. Prolyl 3-hydroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfecta. Nat Genet 2007; 39: 359-365.

6. Morello R, Bertin TK, Chen Y, et al. CRTAP is required for prolyl 3-hydroxylation and mutations cause recessive osteogenesis imperfecta. Cell 2006; 127: 291-304.

7. Barnes AM, Chang W, Morello R, et al. Deficiency of cartilage-associated protein in recessive lethal osteogenesis imperfecta. N Engl J Med 2006; 355: 2757-2764.

8. van Dijk FS, Nesbitt IM, Zwikstra EH,et al. PPIB mutations cause severe Osteogenesis Imperfecta. Am J Hum Genet 2010; 85: 521-527.

9. Schwarze U, Pyott S, Russell D, et al. Mutations in PPIB, which encodes a prolyl cis-trans isomerase (cyclophilin B), in recessive forms of Osteogenesis Imperfecta (OI), ASHG, Honolulu, 2009, http://www.ashg.org/2009meeting/pdf/platforms_4up.pdf, page 73

10. Barnes AM, Carter EM, CabralWA, et al. Lack of cyclophilin B in osteogenesis imperfecta with normal collagen folding. N Eng J Med 2010; 362: 521-528.

11. Ishikawa Y, Wirz J, Vranka JA, et al. Biochemical characterization of the prolyl 3-hydroxylase 1, cartilage associated protein, cyclophilin B complex. J Biol Chem 2009; 284: 17641-17647.

12. van Dijk FS, Nesbitt IM, Nikkels PGJ, et al. CRTAP mutations in lethal and severe osteogenesis imperfecta: the importance of combining biochemical and molecular genetic analysis. Eur J Hum Genet 2009; 17: 1560-1569.

13. Karlsson C, Thomemo M, Henriksson HB, Lindahl A. Identification of a stem cell niche in the zone of Ranvier within the knee joint. J Anat 2009; 215: 355-363.

14. Sillence D, Barlow K, Garber A, Hall J, Rimoin D. Osteogenesis imperfecta type II delineation of the phenotype with reference to genetic heterogeneity. Am J Med Genet 1984; 17: 407-423.

15. Chang W, Barnes AM, CabralWA, Bodurtha JN, Marini JC. Prolyl 3-Hydroxase 1 and CRTAP are mutually stabilizing in the endoplasmic reticulum collagen prolyl 3-hydroxylation complex. Hum. Mol Genet. 2010;19: 223-234.

Dijk_Opmaak.indd 103 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 52 T1 - Black CyanMagentaYellow

Page 105: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 104 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 52 T1 - Black CyanMagentaYellow

Page 106: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

A Novel Homozygous 5 bp Deletion in FKBP10 Causes Clinically bruck Syndrome in an Indonesian patient

Setijowati ED1,2, van Dijk FS1, Cobben JM3, van Rijn RR4, Sistermans EA1, Faradz SMH2, Kawiyana S4, Pals G1*.

1Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands2Center of Biomolecular Research, Faculty of Medicine, Diponegoro University, Indonesia

3Department of Pediatric Genetics, Emma Children’s Hospital/ AMC, Amsterdam, the Netherlands4Departmeny of Orthopaedics and Traumatology, Sanglah Hospital, Denpasar, Indonesia

*Corresponding author: Centre for Connective Tissue Research, Department of Clinical Genetics, VU University

Medical Center, De Boelelaan 1117, P.O. box 7057, 1007 MB Amsterdam, The Netherlands.

Accepted for publication in Eur J Med Genet.

III- 4

Dijk_Opmaak.indd 105 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 53 T1 - Black CyanMagentaYellow

Page 107: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-4106

AbSTrACTWe report an Indonesian patient with bone fragility and congenital joint contractures. The initial

diagnosis was Osteogenesis Imperfecta type III (OI type III) based on clinical and radiological findings.

Because of (i) absence of COL1A1/2 mutations, (ii) a consanguineous pedigree with a similarly

affected sibling and (iii) the existence of congenital joint contractures with absence of recessive

variants in PLOD2, mutation analysis was performed of the FKBP10 gene, recently associated with

Bruck syndrome and/or recessive OI. A novel homozygous deletion in FKBP10 was discovered. Our

report of the first Indonesian patient with clinically Bruck syndrome, confirms the role of causative

recessive FKBP10 mutations in this syndrome.

Dijk_Opmaak.indd 106 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 53 T1 - Black CyanMagentaYellow

Page 108: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 107

1. INTrODuCTION

Bruck syndrome (BS) (OMIM %259450 and #609220) is characterized clinically by congenital contractures

with pterygia, onset of fractures in infancy or early childhood, postnatal short stature, severe limb

deformity, and progressive scoliosis [1] and biochemically by aberrant collagen type I cross-linking in

bone.

BS is named after the first physician (Alfred Bruck) who described a male patient with Osteogenesis

Imperfecta (OI) and arthrogrypsosis in 1897 [2]. Later it appeared that the condition described by Bruck

was in fact different from BS since the contractures in the initial patient were not congenital. In 2003

recessive variants in procollagen-lysine, 2-oxoglutarate 5-dioxygenase II (PLOD2) linked to 3q23-24,

encoding lysyl hydroxylase 2 (LH2) were identified as the cause of BS in two families [3]. However,

in a single consanguineous family with BS [4] linkage to 17p12 [5] had been found and no causative

variants in PLOD2 were detected [3] indicating genetic heterogeneity in Bruck syndrome. Therefore,

Bruck syndrome with linkage to 17p12 was classified as Bruck syndrome type 1(BS 1) and Bruck syndrome

caused by recessive variants in PLOD2 was classified as Bruck syndrome type 2 (BS 2) [3]. BS 1 and BS

2 are phenotypically and biochemically indistinguishable with their hallmarks congenital contractures

and aberrant cross-linking of bone collagen type I. Formation of inter-and intramolecular cross-links is

the final step in collagen biosynthesis necessary to generate fibrils with tensile properties. Cross links

are formed by two routes namely (i) the allysine route (predominating in the skin), characterized by the

conversion of a telopeptide lysine to allysine and (ii) the hydroxyallyisine route (predominating in the

stiff connective tissues), characterized by conversion of telopeptide hydroxylysine to hydroxyallysine.

The pyridinoline cross-links lysylpiridinolines (LP) and hydroxylysylpyridinolines (HP) are derived only via

the hydroxyallysine route and can be regarded as a measure of telopeptide lysyl hydroxylation. In Bruck

syndrome (1 and 2), decreased hydroxyallysine route derived cross-links (HP and LP) and increased

allysine route derived cross-links are observed only in collagen type I from bone [6] (figure 1). The

(HP+LP)/Hyp ratio of urinary collagen degradation products has been shown to be a good diagnostic

marker for BS patients with healthy children showing an average ratio of 2.80 (range:1.83-4.31) whereas

in BS patients, it is below 1[7].

Interestingly, causative variants in FKBP10 (gene map locus 17q21.2) were first described in OI most

closely resembling OI type III [8], but are recently reported also to cause BS [9] based on the presence of

congenital joint contractures, a clinical hallmark of BS. In another publication recessive variants in FKBP10

were described to cause BS as well and it was suggested that they may cause Bruck syndrome type I[10]

initially linked to 17p12. At present it is still unclear whether recessive variants in FKBP10 cause BS type

1 and whether aberrant cross-linking of bone collagen type I is present as observed in BS type 1 and

2. FKBP10 encodes FKBP65 which has been reported to have peptidylprolyl cis-trans isomerase (PPIase)

activity and rough endoplasmic reticulum (rER) chaperone capability [11].

Here, we report an Indonesian patient with a clinical diagnosis of BS due to a novel homozygous

deletion in FKBP10.

Dijk_Opmaak.indd 107 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 54 T1 - Black CyanMagentaYellow

Page 109: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-4108

figure 1 Procollagen type I consists of a triple helix domain, N-and-C-telopeptides and N-and C-propeptides. In the telopeptides, various lysine residues are hydroxylated by specific telopeptide lysyl hydroxylases such as PLOD2. To generate collagen type I fibrils, cross-linking of the collagen type I molecules takes place by the hydroxyallysine route which is characterized by conversion of hydroxylysine to hydroxyallysine. In case of Bruck syndrome due to PLOD2 mutations (Bruck syndrome type 2), lysyl hydroxylaltion of telopeptides is absent resulting in aberrant collagen type I cross-linking in bone. Clinically, PLOD2 mutations lead to bone fractures with presence of congenital contractures in all patients. The molecular genetic cause of Bruck syndrome type 1 is unknown but the clinical features are similar to that of Bruck syndrome caused by PLOD2 mutations. Patients with FKBP10 mutations appear to have bone fractures but congenital contractures are not always present. It remains to be investigated whether aberrant cross-linking of collagen type I in bone is also present in case of FKBP10 mutations.

2. CLINICAL rEPOrT

The proband was born at term. Birth weight, length and head circumference were not recorded. After

birth, unilateral talipes equinovarus, congenital joint flexion contractures of the knees and ankles, and

kyphoscoliosis were observed. There was no evidence of pterygia. In the following years it became

apparent that he could not walk and had to use a wheelchair. He had a normal cognitive development.

He had multiple fractures during childhood and adolescence. At the age of 26 he was operated on his

left radius and ulna because of pain due to an old fracture causing bowing. The orthopaedic surgeon

diagnosed him with OI type III because of a history of multiple fractures, clinical findings of short stature,

severe kyphoscoliosis, limb deformities (figure 2) and radiological abnormalities (bone demineralization,

multiple old healed fractures, deformed bones). Upon re-examination at the age of 30 years his length

was approximately 1.37 meter. Kyphoscoliosis and limb deformities of especially the lower limbs were

apparent. His muscle mass was minimal and he had no mobility of the lower limbs due to contractures.

He had a right pes equinovarus and a hallux valgus on the left foot. The range of motion of knees

and elbows was limited. He had a pectus carinatum and restrictive lung disease probably related to

the apparent kyphoscoliosis. Heart ausculation was normal. He had a normal facial appearance. His

sclerae were white and vision and hearing were normal. He had mild pitting of his dental enamel but no

evidence of dentinogenesis imperfecta. His skin texture and healing of the surgical incision were normal.

Review of recent radiographs (figure 3) showed generalized osteopenia and other abnormalities. At the

age of 31 years the proband died suddenly, possibly related to respiratory insufficiency, no permission

for autopsy was given.

The proband was the second child born to a consanguineous Indonesian (Balinese) couple (the parents

are second cousins) He had one older brother who died at the age of 25. The family history also reveals

Dijk_Opmaak.indd 108 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 54 T1 - Black CyanMagentaYellow

Page 110: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 109

three miscarriages. The older brother most likely had the same disorder as the proband with unilateral

talipes equinovarus, congenital joint contractures and severe kyphoscoliosis. He could not walk and had

to use a wheel chair. He probably died because of pneumonia causing respiratory failure.

figure 2: Clinical photograph of affected individualClinical photograph shows round face with white sclerae, short neck, kyphoscoliosis,short trunk, short and deformed limbs.

figure 3: Radiographs of affected individualA: AP Skull radiograph shows relatively thin calvarian bones. Normal pneumatization of the sinus. The teeth, although some elements are missing, show no radiological sign of dentinogenisis imperfecta.b: AP radiograph of the spine shows severe sinistroconvex torsion scoliosis. C: Pelvic AP radiograph shows severe acetabular protrusion with subsequent deformation of the pelvis D: Radiographs of the left radius and ulna. Plate osteosynthesis for mid shaft fractures. Note some irregularity of the radial cortex (arrow) E: Radiographs of the right radius and ulna shows bowing. There are no sequelae of fractures visible.

Dijk_Opmaak.indd 109 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 55 T1 - Black CyanMagentaYellow

Page 111: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-4110

3. METHODS

PCR and sequence analysis: the 10 exons of FKBP10 (NM_021939.3), as well as surrounding intronic

regions, were amplified by PCR from genomic DNA of the patient sample and analyzed by sequencing

on the ABI 3730 apparatus from Applied Biosystems (Life technology Corporation, Carlsbad, California)

Results were analyzed using Mutation Surveyor version 3.01. Interpretation of the results was performed

as described previously [10] and conventional coding sequence numbering was used to report the

homozygous variant as was the case in the publication of Alanay et al [8].

4. rESuLTS

A homozygous c.600-604del in exon 4 was identified, resulting in p.Tyr201HisfsX58. Parents were not

available for carrier testing.

5. DISCuSSION

Initially, the proband was diagnosed with OI type III. COL1A1/2 mutation analysis was negative. Ultimately,

the presence of congenital joint contractures, in combination with a consanguineous pedigree with

one other affected sibling, led to a clinical suspicion of BS. No recessive causative variants were detected

in PLOD2 responsible for BS 2. DNA analysis of FKBP10 was performed and a homozygous deletion in

FKBP10 was detected. Although formally it is possible that the mutation is heterozygous with a large

deletion on the other allele, this is not very likely given the fact that the parents were consanguineous.

Unfortunately the parents were not available for carrier testing to confirm this. This deletion of five

basepairs leads to a frame shift starting at codon 201 that if translated ends in a stop codon 57 positions

downstream and is as such expected to be causative. Most likely, nonsense mediated decay will prevent

any protein formation.

It appears that all causative FKBP10 variants found so far in BS (see table 1), are loss of function mutations.

The emerging clinical picture when comparing clinical data of the affected individuals is that in all

patients described so far, fractures are present that start in early infancy but are not present prenatally.

Bone deformation is described. The presence of congenital contractures and/or pterygia varies, even

in the same family. Most patients are wheelchair bound due to recurrent fractures and/or congenital

contractures with pterygia. There is severe short stature, often (kypho)scoliosis, and in none of the

patients blue sclerae or dentinogenesis imperfect are reported. Radiologically, generalized osteopenia,

wormian bones and evidence of fractures with bone deformation is frequently reported [8,9,10,12] There

were no reports on sudden death in other adult patients with Bruck syndrome due to FKBP10 mutations.

As the proband died suddenly we were not able to collect urine for measurement of the (HP+LP)/Hyp

ratio of urinary collagen degradation products. This might have been of particular interest since in two

publications it has already been reported that recessive variants in FKBP10 cause BS, whereas so far it

has not been investigated thoroughly whether the same biochemical abnormalities in collagen cross-

linking are present [9,10]. However, patient 1 described by Kelley et al.(see table 2)[10], is reported to

Dijk_Opmaak.indd 110 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 55 T1 - Black CyanMagentaYellow

Page 112: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 111

have a positive screen for urine lysyl hydroxylation defects suggesting that the mechanism by which

FKBP10 mutations cause Bruck syndrome is comparable with the effects of PLOD2 mutations, namely

underhydroxylation of (pro) collagen type I telopeptides resulting in aberrant cross-linking of collagen

type I in bone. To confirm this, in more patients with FKBP10 mutations urine should be screened for lysyl

hydroxylation defects and if possible, collagen type I cross-linking in bone should be studied.

In summary, our report confirms that the clinical hallmark of Bruck syndrome namely congenital joint

contractures in combination with bone fragility, can be caused by loss of function variants in the FKBP10

gene. At present, it remains unclear whether causative variants in FKBP10 (gene map locus 17q21.2) are

causative of BS type 1 (originally linked to 17p12).

Dijk_Opmaak.indd 111 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 56 T1 - Black CyanMagentaYellow

Page 113: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-4112

Table 1. Overview of patients with FKBP10 mutations.

Patient and reference

Family 1-5 (16 affected). (Alanay et al. 2010)[8]

Family 6 (3 affected). (Alanay et al. 2010)[8]

Patient 1 (Shaheen et al., 2010)[9]

Patient 2 (Shaheen et al.,2010)[9]

Age(s) (yr) U 12, 16, 18 9 13

Mutations Homozygous c.321_353del

Homozygous c.831dupC

Homozygous c.1023insGGAGAATT

Homozygous c.1023insGGAGAATT

fractures (#) # beginning in infancy

Yes -Multiple long bone #

-first #at 7 months of age

Frequent #

Congenital contractures

ND ND Yes, knees, ankles and elbows

Multiple joint contractures

Pterygia ND ND ND ND

blue sclerae No ND No ND

Stature <p5 ND ND

Joint hypermobility 2/16 affected individuals

Yes ND ND

Hearing loss No ND ND

Dentinogenesis Imperfecta (DI)

No no No ND

Other -Progressive kyphoscoliosis-Wheelchair bound

2/3 wheelchair bound

Unable to walk

radiographic abnormalities

-Severe osteopenia, -Fractures leading to deformities in the long bones

-Flattening and wedging of the vertebrae

-Severe osteopenia-Wormian bones-Scoliosis-Wedge vertebrae

-Old healed fractures-Severe flexion deformities of knees and ankles

-Wormian bones, -Generalized osteopenia

ND

ND= not describedU=unknown

Dijk_Opmaak.indd 112 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 56 T1 - Black CyanMagentaYellow

Page 114: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 113

Patient 1(Kelley et al. 2011)[10]

Patient 2(Kelley et al. 2011)[10]

Patient 3(Kelley et al. 2011)[10]

Patient 4(Kelley et al. 2011)[10]

Patient 5(Kelley et al. 2011)[10]

6 22 U 10 31

Homozygousc.831dupC

Homozygousc.122_156del

Homozygousc.831dupC

Homozygousc.831dupC

c.831dupCc.1276dupC

-Multiple femoral and costal #

-first # at 2 months of age

5 # at age 3 -Multiple # of the extremity long bones

–first # at 2 months of age

-Multiple # of the long bones

-first # at 2 months of age

Multiple # of the long bones

Yes, knees and ankles No Yes, elbows, wrists, knees, thumb in palm deformity

No Yes

ND ND Yes, pterygia of knees

No No No ND ND

ND -4 SD ND <p3 ND

ND ND ND ND

no ND ND No ND

no ND No No ND

-Wheelchair bound -Positive screen for urine lysyl hydroxylation defects

-Crutches and wheelchair

ND -Wormian bones-kyphoscoliosis-Gracile fibulas

-Wormian bones -Wormian bones

Dijk_Opmaak.indd 113 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 57 T1 - Black CyanMagentaYellow

Page 115: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter III-4114Ta

ble

2.

Cha

ract

eris

tics

of p

atie

nts

with

FKB

P10

mut

atio

ns

Patie

nt a

nd

refe

renc

ePa

tient

6 (K

elle

y et

al.,

2011

)[10]

Patie

nt 1

, fam

ily

1(Sh

ahee

n et

al.,

2011

Patie

nt 2

, fam

ily 1

(S

hahe

en e

t al.,

2011

) Pa

tient

3, f

amily

1

(Sha

heen

et a

l., 20

11)

Patie

nt 4

, fam

ily 1

(Sha

heen

et a

l., 20

11)

Patie

nt 5

, fam

ily 2

(Sha

heen

et a

l., 20

11)

This

pat

ient

Age

(s) (

yr)

56

14

17

12

10-m

onth

old

Dec

ease

d at

31

Mut

atio

nsc.

831d

upC

c.34

4G>

AH

omoz

ygou

sc.

743d

upC

Hom

ozyg

ous

c.74

3dup

CH

omoz

ygou

sc.

743d

upC

Hom

ozyg

ous

c.74

3dup

CH

omoz

ygou

s c.

831d

upC

Hom

ozyg

ous

c.60

0-60

4del

frac

ture

s (#

)M

ultip

le #

dur

ing

first

w

eeks

of l

ifeFi

rst #

at 1

wee

k of

ag

e-#

freq

uenc

e 5-

6 pe

r ye

ar

-Firs

t # a

t 6 m

onth

s of

age

-# fr

eque

nce

2-3

per

year

-Fem

ur #

dur

ing

labo

ur,

-# fr

eque

ncy

3 pe

r ye

ar

-Firs

t # a

t at a

ge

four

-# fr

eque

ncy

1-2

per y

ear

-Fem

ur #

at t

hree

w

eeks

of a

geM

ultip

le #

Cong

enita

l co

ntra

ctur

esM

ultip

le c

onge

nita

l co

ntra

ctur

es, b

ilate

ral

club

feet

No

ND

Cont

ract

ures

of t

he

knee

sN

oCo

ntra

ctur

es o

f hi

ps, k

nees

, elb

ows,

bila

tera

l tal

ipes

de

form

ity

Cont

ract

ures

of k

nees

an

d an

kles

, uni

late

ral

talip

es e

quin

ovar

us

Pter

ygia

ND

No

ND

Yes,

popl

iteal

pte

rygi

aN

oYe

s, m

ultip

le p

tery

gia

No

blue

scl

erae

No

No

ND

ND

ND

ND

No

Stat

ure

P3-p

10Se

vere

sho

rt s

tatu

re-6

SD

ND

-5 S

DN

D<

-3SD

Join

t hyp

erm

obili

tyN

DN

DN

DN

DN

DN

DU

Hea

ring

loss

ND

ND

ND

ND

ND

ND

U

Den

tinog

enes

is

Impe

rfec

ta (D

I)N

oN

DN

DN

DN

DN

DN

o

Oth

erN

ot m

obile

-Pro

gres

sive

sco

liosi

s-M

obile

-Whe

elch

air b

ound

-Slo

wly

pro

gres

sive

sc

olio

sis

-Sco

liosi

s-B

arel

y m

obile

Kyph

osco

liosi

s

radi

ogra

phic

ab

norm

aliti

esM

ultip

le w

orm

ian

bone

s-O

ld h

eale

d fra

ctur

es-S

ever

e de

form

ity o

f kn

ees

and

ankl

es-O

steo

peni

a

ND

ND

ND

-Gen

eral

ized

os

teop

enia

-Ver

tebr

al

com

pres

sion

s-B

owin

g w

ith

defo

rmity

of f

emor

a,

tibia

e, a

nd fi

bula

-Gen

eral

ized

os

teop

enia

-Thi

n ca

lvar

ian

bone

s-K

ypho

colio

sis

-ace

tabu

lar

prot

rusi

on a

nd

defo

rmat

ion

pelv

is-B

owin

g tib

iae

and

fibul

a

Dijk_Opmaak.indd 114 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 57 T1 - Black CyanMagentaYellow

Page 116: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Lethal/Severe Osteogenesis Imperfecta in a Large Family 115

ACKNOWLEDGEMENTS

The authors of this manuscript wish to thank the family of the patient for their support of this study.

Eva Setijowati is a recipient of a Beasiswa Unggulan Fellowship, Ministry of National Education,

Government of Indonesia

Dijk_Opmaak.indd 115 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 58 T1 - Black CyanMagentaYellow

Page 117: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

rEfErENCES

[1] E.McPherson, M.Clemens M. Bruck syndrome (osteogenesis imperfecta with congenital joint contractures): review and report on the first North American case. Am.J.Med.Genet. 70 (1997) 28-31.

[2] D.Viljoen, G.Versfeld, P.Beighton. Osteogenesis imperfecta with congenital joint contractures (Bruck syndrome). Clin.Genet. 36 (1989) 122-126.

[3] A.van der Slot, A.Zuurmond, A. Bardoel, C. Wijmenga, H. Pruijs, D. Sillence, J. Brinckmann, D. Abraham, C. Black, N. Verzijl, J. DeGroot, R. Hanemaaijer, J. TeKoppele, T. Huizinga, R. Bank . Identification of PLOD2 as telopeptide lysyl hydroxylase, an important enzyme in fibrosis. J.Biol.Chem. 278 (2003) 40967-40972.

[4] E.Breslau-Siderius, R. Engelbert, G. Pals, J. van der Sluijs J. Bruck syndrome: a rare combination of bone fragility and multiple congenital joint contractures. J.Pediatr.Orthop.B 7(1998) 35-38.

[5] R.Bank, S. Robins, C. Wijmenga, L. Breslau-Siderius, A. Bardoel, H. van der Sluijs, H. Pruijs, J. TeKoppele . Defective collagen crosslinking in bone, but not in ligament or cartilage, in Bruck syndrome: indications for a bone-specific telopeptide lysyl hydroxylase on chromosome 17. Proc.Natl.Acad.Sci.U.S.A 96 (1999) 1054-1058.

[6] M. Hyry, J. Lantto, J. Myllyharju. Missense mutations that cause Bruck syndrome affect enzymatic activity, folding, and oligomerization of lysyl hydroxylase 2. J.Biol.Chem. 284 (2009) 30917-30924.

[7] R. Ha-Vinh R, Y. Alanay, R.Bank, A. Campos-Xavier, A. Zankl, A.Superti-Furga, L. Bonafe. Phenotypic and molecular characterization of Bruck syndrome (osteogenesis imperfecta with contractures of the large joints) caused by a recessive mutation in PLOD2. Am.J.Med.Genet.A 131 (2004) 115-120.

[8] Y. Alanay, H. Avaygan, N. Camacho, G. Utine, K. Boduroglu, D. Aktas, M. Alikasifoglu, E. Tuncbilek, D. Orhan, F. Bakar, B. Zabel, A. Superti-Furga, L. Bruckner-Tuderman, C. Curry, S. Pyott, P. Byers, D. Eyre, D. Baldridge, B. Lee, A. Merrill, E. Davis, D. Cohn, N. Akarsu, D. Krakow. Mutations in the gene encoding the RER protein FKBP65 cause autosomal-recessive osteogenesis imperfecta. Am.J.Hum.Genet. 86 (2010) 551-559.

[9] R. Shaheen, M. Al-Owain, N. Sakati, Z. Alzayed, F. Alkuraya. FKBP10 and Bruck syndrome: phenotypic heterogeneity or call for reclassification? Am.J.Hum.Genet. 87 (2010) 306-307.

[10] B. Kelley, F. Malfait, L. Bonafe, D. Baldridge, E. Homan, S. Symoens, A. Willaert, N. Elcioglu, L. van Maldergem, C. Verellen-Dumoulin, Y. Gillerot, D. Napierala, D. Krakow, P. Beighton, A. Superti-Furga, A. De Paepe, B. Lee. Mutations in FKBP10 cause recessive osteogenesis imperfecta and bruck syndrome. J.Bone Miner.Res. 26 (2011): 666-672.

[11] Y. Ishikawa, J. Vranka, J. Wirz, K. Nagata, H. Bachinger. The rough endoplasmic reticulum-resident FK506-binding protein FKBP65 is a molecular chaperone that interacts with collagens. J.Biol.Chem. 283 (2008) 31584-31590.

[12] R. Shaheen, M. Al-Owain, E. Faqeih, N. Al-Hashmi, A. Awaji, Z. Al-Zayed, F. Alkuraya. Mutations in FKBP10 cause both bruck syndrome and isolated osteogenesis imperfecta in humans. Am. J. Med. Gen. A. 155 (2011) 1448-1452.

Dijk_Opmaak.indd 116 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 58 T1 - Black CyanMagentaYellow

Page 118: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV

Implications for clinical and laboratory diagnosis of Osteogenesis Imperfecta

Dijk_Opmaak.indd 117 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 59 T1 - Black CyanMagentaYellow

Page 119: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 118 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 59 T1 - Black CyanMagentaYellow

Page 120: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Classification of Osteogenesis Imperfecta revisited

Van Dijk FSa*, Pals G a, Van Rijn RRb, Nikkels PGJc, Cobben JMd

aDepartment of Medical Genetics, VUMC Hospital, Amsterdam, The NetherlandsbDepartment of Paediatric Radiology, Emma Children Hospital AMC, Amsterdam, The Netherlands

cDepartment of Pathology, University Medical Centre Utrecht, The NetherlandsdDepartment of Paediatric Genetics, Emma Children Hospital AMC, Amsterdam, The Netherlands

Eur J Med Genet. 2010 Jan-Feb;53(1):1-5.

IV-1

Dijk_Opmaak.indd 119 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 60 T1 - Black CyanMagentaYellow

Page 121: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-1120

AbSTrACT

In 1979 Sillence proposed a classification of Osteogenesis Imperfecta (OI) in OI types I, II, III and IV. In

2004 and 2007 this classification was expanded with OI types V-VIII because of distinct clinical features

and/or different causative gene mutations. We propose a revised classification of OI with exclusion

of OI type VII and VIII since these types have been added because of genetic criteria (autosomal

recessive inheritance) while the clinical and radiological features are indistinguishable from OI types

II-IV. Instead, we propose continued use of the Sillence criteria I, II-A, II-B, II-C, III and IV for clinical and

radiological classification of OI with additional mentioning of the causative mutated gene to this

classification. OI type V en VI are still separate entities in this revised classification, because of the

distinguishing clinical/radiological and histological features observed in these types.

Dijk_Opmaak.indd 120 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 60 T1 - Black CyanMagentaYellow

Page 122: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Classification of Osteogenesis Imperfecta revisited 121

1. INTrODuCTION

The definition and classification of Osteogenesis Imperfecta (OI) have been the subject of ongoing

debate in the literature. The most commonly used definition of OI is “a hereditary disorder with

osteopenia and increased bone fractures.” Secondary features such as short stature, blue sclerae,

dentinogenesis imperfecta and hearing loss may also exist in affected individuals. The prevalence of

OI is approximately 6-7/100,000 [26]. The prevalence of lethal OI in the Netherlands was reported to be

5/100,000 [7]. Because of the extreme clinical variability in OI various authors have attempted to classify

patients with this disease [3]. In 1979 a classification was introduced by the Australian physician David

Sillence (the “Sillence classification”) that is still in use today [24].

2. CLASSIfICATION Of OSTEOGENESIS IMPErfECTA

2.1 Original Sillence classificationThe original Sillence classification of OI in four types (OI type I-IV), was based on (i) clinical findings

with a radiological subclassification of OI type II in A, B, C and (ii) mode of inheritance, thus assuming

heterogeneity in OI [23,24]. However, after the discovery of heterozygous collagen type I gene mutations

in all OI types [5], the Sillence criteria were used mainly to describe the severity continuum and

differences in clinical expression of OI. Later it appeared that that not all patients with lethal or severe

OI had a collagen type I mutation [27] which led eventually to expansion of the Sillence classification.

2.2 Expanded Sillence classificationIn 2004 The Lancet published a Seminar on Osteogenesis Imperfecta (OI), containing among others an

“expanded Sillence classification” [22]. This classification recognizes seven types of OI, type I-VII. In 2007,

OI type VIII was proposed as an additional type [6] (see figure 1).

Sillence type

Clinical severity Mutated gene Mode of inheritance

I Mild-non deforming COL1A1/2 AD

II Perinatal lethal COL1A1/2 AD

III Severely deforming COL1A1/2 AD

IV Moderately deforming

COL1A1/2

AD

V Moderately deforming

Unknown AD

VI Moderately to severely deforming Unknown AR

VII Moderately deforming CRTAP AR

VIII Severely deforming to perinatal lethal

LEPRE1 AR

In 2006 Morello et al. described partial loss of CRTAP as a cause of moderately deforming recessive OI originally described as OI type VII [19]. Barnes et al. described complete loss of CRTAP to cause OI type II-III [2].

In 2007 Cabral et al. proposed to classify OI due to LEPRE1 mutations as OI type VIII [6].

In 1984 a radiological subclassification of OI type II was proposed in A, B and C[23], with short stature, lethality and multiple congenital long bone fractures present in all types: OI II-A: -broad ribs with multiple fractures -continuous beaded ribs -severe undermodeling of femur OI II-B: -normal/thin ribs with some fractures -discontinous beaded ribs -some undermodeling of femur OI II-C: -varying thickness of ribs -discontinuous beading of ribs. -malformed scapulae and ischiae -slender and twisted long bones

figure 1: Sillence Classification expanded with OI V-VIII as proposed by Rauch 2004 & Cabral 2007

Dijk_Opmaak.indd 121 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 61 T1 - Black CyanMagentaYellow

Page 123: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-1122

The addition of OI type V was based on distinct clinical/radiological and histological features in patients

originally diagnosed as having OI type IV in the absence of COL1A1/2 mutations [11]. At the moment

36 cases with OI type V have been reported in the literature and autosomal dominant inheritance is

presumed [10,15,29].

OI type VI was added to the classification because of distinct histological features in the absence of

COL1A1/2 mutations in patients originally diagnosed with OI type IV, but without abnormalities of

collagen type I on electrophoresis [12]. 11 patients with OI type VI have been reported in the literature

[14]. Autosomal recessive heritance is presumed because of two consanguineous families with

recurrence of OI in one of these families [12] .

The initial reason to distinguish OI type VII was autosomal recessive inheritance of moderate deforming OI

in 2 generations of 3 interrelated families [13, 28]. In 2006 partial loss of function CRTAP mutationsencoding

cartilage-associated protein (CRTAP) were found to cause OI type VII [19] and complete loss of function

CRTAP mutations appeared to cause lethal OI [2]. Apparently, the type of mutation in the CRTAP gene

can influence the severity of OI.

In 2007 Cabral et al. described autosomal recessive lethal and severe OI due to LEPRE1 mutations

encoding prolyl-3- hydroxylase 1 (P3H1) and proposed to classify this type of OI caused by LEPRE1

mutations as OI type VIII [6].

2.3 Distinguishing clinical features in OI types V-VIII?In OI type V distinct clinical/radiological features have been described. In OI type VI the main

distinguishing feature is histological and not clinical. An important question regarding the addition of OI

type VII and VIII to the expanded Sillence classification is whether clinically discriminating features of OI

caused by (partial) loss of function CRTAP mutations and LEPRE1 mutations exist. Indeed, some clinical

differences have been claimed [1,2,6] but the clinical/radiological features [20] are variable and are in

practice, especially at an early age, impossible to distinguish from the clinical/radiological features of OI

type II, III and IV, although with the description of more cases in the future, some phenotype-genotype

correlation might still emerge. OI types II, III and IV are usually caused by collagen type I gene mutations,

also the main cause of OI type I [16]. An interesting observation is that all affected individuals with CRTAP

and LEPRE1 mutations are classified as either OI type II-B or III and none can be classified as OI type II-A

(see figure 2) [8].

Dijk_Opmaak.indd 122 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 61 T1 - Black CyanMagentaYellow

Page 124: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Classification of Osteogenesis Imperfecta revisited 123

COL1A1/2 CRTAP LEPRE1

II-A

Not described

Not described

II-B

a

b c d

PPIB

Not described

e

COL1A1/2 CRTAP LEPRE1

III

IV Not described

g

j k

h f

PPIB

Not described

i

l

figure 2: Examples of OI Sillence types due to different causative genes.A.Full term neonate with OI II-A due to a COL1A1 mutation c.2120 G>T. Diminished skull mineralization. Platyspondyly Th11-L5. Small bell-shaped thorax with broad continuously beaded ribs (multiple fractures). Multiple fractures and bowing of all long bones with loss of modeling of femora.b. Full term neonate with OI II-B due to a COL1A2 mutation c.2967_2984dup. Normal skull mineralization. Platyspondyly Th10 – L1. Small thorax without apparent rib fractures. Bowing of both humeri, femora, tibiae and fibula with fractures of both humeri, right distal radius and ulna, left radius, both femora and possibly tibiae. Moderate loss of modeling of femora. Upper and lower radiographs were taken at different times (please note the umbilical line in the lower image).C. Full term neonate with OI II-B due to a homozygous CRTAP mutation c.826C>T [2]. No platyspondyly, discontinuously beaded ribs (arrow=callus formation). Bowing and fractures of both femora, tibiae and fibula. Loss of modeling of femora. The child was born to non-consanguineous healthy black parents and she had two healthy half-siblings. She died 18 days after birth [2]

Dijk_Opmaak.indd 123 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 62 T1 - Black CyanMagentaYellow

Page 125: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-1124

D. Full term neonate with OI II-B due to homozygous LEPRE1 mutation c.232delC [1]. Diminished skull mineralization. Due to poor radiological technique the spine is not evaluable. Small thorax. No obvious rib fractures, possible rib fractures lower left thorax. Fractures of left humerus and both femora, bowed humerus, femora and right tibia. Moderate loss of modeling femora. The child died at the age of 3.5 months. The parents were Irish Traveller consanguineous parents (fourth cousin) and no recurrence in sibs is known.[1]E. Fetus at 21+2 weeks of gestation with OI II-B/III due to a homozygous PPIB mutation c.556-559delAAGA . Normal skull mineralization for gestational age, slender ribs without fractures, incomplete ossification of T5 and T12, somewhat irregular proximal metaphyses of the humeri, radii and ulnae and bowing of the ulnae. Bowed femora with fractures and some loss of modeling, bowed tibiae and fibula are apparent, possibly with fractures [9]. f. Full term neonate with OI III due to a COL1A1 mutation c.1992 G>A. Normal skull mineralization. No platyspondyly. Small bell-shaped thorax with slender ribs without fractures. Fractures of both humeri, bowing of right radius with possible fracture. Bowing of both femora with fractures and to a lesser extent the tibia. Slight loss of modeling femora.G. Full term neonate with OI III due to CRTAP mutations c.38 C>A and c.469 A>G in exon 1 and c. 923-2A>G in intron 4. Normal skull mineralisation. No platyspondyly. Small thorax without rib fractures. Bowing and fractures (open arrow) of both femora and tibia (closed arrow). Moderate loss of modeling of femora. The child died 5 days after birth. The parents are not consanguineous and have no other children [8]. H. Full term neonate with OI III due to homozygous LEPRE1 mutation c.401_402delAA. Slender ribs without apparent fractures. Deformation of the right femur (open arrow) with diaphyseal broadening. The left femur shows bowing (arrow) and broadening, the metaphyses are ill defined (a sign of undermineralization). The tibia shows an acute angle (arrowhead) in keeping with a previous fracture. The child died in the first year of life due to pneumonia. The Turkish parents were consanguineous and recurrence in two sibs occurred.I. Full term neonate with OI type II-B/III due to a homozygous PPIB mutation c.451 C>T. Normal mineralization of skull, fractures of both humeri, radii and left ulna with callus formation and some loss of modelling of the humeri. Discontinuously beaded ribs are noted with a small bell-shaped thorax. Multiple fractures with callus formation of femora, tibiae and fibula exist with moderate loss of modelling of the femora [9]. The child is alive at the age of one year and has consanguineous Pakistani parents and one older affected sib who is alive at the age of 8 years.. J. Full term neonate with OI IV due to a COL1A1 mutation c.832G>A [18]. No platyspondyly. No small thorax. Gracile ribs and short femurs with bilateral midshaft fractures.K.Child, age 4 months, with OI IV due to a homozygous CRTAP mutations c.472-1021C>G in intron 1.Bilateral coxa vara and bowing of the lower extremities is evident [19, 27].L. A patient with OI type IV phenotype due to a homozygous PPIB mutation c.343+1G>A was described recently [U. Schwarze et al., 2009, ASHG, Abstract]. Unfortunately, no radiograph is available.

2.4 Problems with current classificationIt seems clear that the primary reason to distinguish OI type VII and VIII is genetic, with autosomal

recessive inheritance due to mutations in respectively the CRTAP and LEPRE1 genes whereas OI type I-IV

are mainly distinguished based upon differences in severity and/or clinical expression of OI.

The intermingling of a clinical and a genetic classification of OI poses serious problems, resulting in

confusing statements in the recent medical literature. Examples of this confusion are the following

statements in the literature: The widely used OMIM states: “autosomal recessive perinatal lethal

osteogenesis imperfecta caused by mutation in the CRTAP gene is here referred to as type IIB, to

distinguish it from the autosomal dominant form (type IIA;) caused by mutation in one of the genes

encoding subunits of type I collagen”[17]. A passage in an article in Human Mutation reads: “Mutations

in the CRTAP and LEPRE1 genes can each have a broad range of clinical presentation, from a very severe

and often lethal OI type II/III to a milder OI type VII seen with mutations in CRTAP that lead to production

of some normal protein”[1].

Dijk_Opmaak.indd 124 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 62 T1 - Black CyanMagentaYellow

Page 126: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Classification of Osteogenesis Imperfecta revisited 125

2.5 Classification proposalIn figure 3 we present a revised classification of OI which in our view is more logical and consistent

compared to the current one [16,22]. Instead of expansion of the original Sillence classification we

propose two things: (i) A format “OI type <I-VI><gene> related ”, implying continued use of the Sillence

criteria I, II (A,B,C), III and IV for clinical and radiological classification of OI and addition of the causative

mutated gene to this classification. OI type V is not excluded because of the specific clinical /radiological

and histological features observed in this type. OI type VI is placed between brackets since the main

distinguishing feature is histological and not clinical/radiological.

(ii) Exclusion of OI types VII and VIII since these types have been added because of autosomal recessive

inheritance while the clinical and radiological features are in practice indistinguishable from OI types II-

IV. In the light of this, we would also like to refrain from classifying a recently discovered recessive genetic

cause of OI [9] as OI type IX since the clinical features of the affected individuals are indistinguishable

from OI types II-B/III or IV [U. Schwarze et al., 2009, ASHG, Abstract]. This last type of OI we clarify as “OI

type II-B, III or IV PPIB related.

1 No individuals with OI due to LEPRE1, CRTAP or PPIB mutations were diagnosed with OI type II-A. 2 II-B osteogenesis imperfecta with longer survival and OI type III with early death show considerable clinical and radiological overlap [25] 3 II-C OI is extremely rare and its existence even doubted [25] 4 No LEPRE1 mutations causing OI type IV have been described 5 OI type VI has been placed between brackets because the main distinguishing feature is histological

Type Subtype Gene

COL1A1/2-related

COL1A1/2-related

CRTAP-related

LEPRE1-related 4

PPIB-related

Osteogenesis Imperfecta

Osteogenesis Imperfecta

Type V [Type VI] 5

A 1 B 2 C 3

Type I Osteogenesis Imperfecta

Type II Type III Type IV

Unknown

figure 3: Proposed “Classification of Osteogenesis Imperfecta” (modified after Sillence 1979 & Rauch 2004)

3. DEfINITION Of OI

As mentioned above, the definition of OI has been a matter of discussion in the literature. In the past,

there has been a tendency to define OI as a “type I collagenopathy” [11] because of abnormal collagen

1 protein on electrophoresis and the discovery of mutations in the COL1A1/2 genes encoding collagen

type I in, so it seemed at that time, all patients with OI [4]. However, since eventually not all patients with

Dijk_Opmaak.indd 125 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 63 T1 - Black CyanMagentaYellow

Page 127: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-1126

OI appeared to have COL1A1/2 mutations [23], this definition of OI was not generally accepted. Based

on the current knowledge that all known genetic causes of OI affect collagen type I biosynthesis we

suggest to define OI primarily as “a heterogeneous group of diseases characterized by susceptibility to

bone fractures with variable severity and presumed or proven defects in collagen type I biosynthesis”.

We acknowledge the fact that it is unknown whether the genetic causes of OI types V and VI affect

collagen type I biosynthesis. However, if this appears not to be the case we propose to consider these

types as new syndromes instead of particular types of OI.

4. CONCLuSION

We propose a revised classification of OI, mentioning the causative gene and the clinical picture

indicated by OI type I-VI, with exclusion of OI type VII and VIII since these types have mainly been added

because of genetic criteria while the clinical and radiological features are indistinguishable from OI types

II-IV. This proposed classification leaves room for new genes being discovered as causes for OI until, as

already indicated by Sillence in 1979, the extent of heterogeneity is known [23]. Furthermore, since all

known genetic causes of OI affect collagen type I biosynthesis, we propose to define OI primarily as “a

heterogeneous group of diseases characterized by susceptibility to bone fractures with variable severity

and presumed or proven defects in collagen type I biosynthesis.”

ACKNOWLEDGEMENTS

We thank our colleagues dr. N.J. Shaw and dr. W. Högler from the Department of Pediatric Endocrinology,

Birmingham Children’s Hospital, Birmingham, United Kingdom and dr B.Lee from the department of

molecular and human genetics, Baylor College of Medicine, Houston, Texas, USA for their contribution

to figure 2.

Dijk_Opmaak.indd 126 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 63 T1 - Black CyanMagentaYellow

Page 128: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Classification of Osteogenesis Imperfecta revisited 127

rEfErENCES

[1] Baldridge D., Schwarze U., Morello R., Lennington .J, Bertin T., Pace J., Pepin M., Weis M., Eyre D., Walsh J., Lambert D., Green A., Robinson H., Michelson M., Houge G., Lindman C., Martin J., Ward J., Lemyre E., Mitchell J. Krakow D., Rimoin D., Cohn D., Byers P., Lee B., CRTAP and LEPRE1 mutations in recessive osteogenesis imperfecta, Hum Mutat 29 (2008) 1435-1442.

[2] Barnes A.., Chang W., Morello R.., Cabral W., Weis B., Eyre D., Leikin S., Makareeva E., Kuznetsova N., Uveges T., Ahok A., Flor A., Mulvihill., Wilson PL., Sundaram U., Lee B., Marini J., Deficiency of Cartilage-associated protein in recessive lethal osteogenesis imperfecta, NEJM 355 (2006) 2757-2764.

[3] Bauze R., Smith R., Francis M., An new look at osteogenesis imperfecta: a clinical, radiological and biochemical study of forty-two patients, J. Bone Joint Surg. 57-B (1975) 1415-1425.

[4] Byers P., Steiner R., Osteogenesis Imperfecta, Annu. Rev. Med. 43 (1992) 269-282.

[5] Byers P., Wallis G., Willing M., Osteogenesis Imperfecta: translation of mutation to phenotype, J. Med. Genet 28 (1991) 433-442.

[6] Cabral W, Chang W, Barnes A, Weis M, Scott M., Leikin S., Makareeva. E., Kuznetsova., Rosenbaum K., Tifft C., Bulas D., Kozma C., Smith P., Eyre D., & Marini J., Prolyl 3-hydroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfecta., Nat. Gen. 39 (2007) 359-365.

[7] Cobben J., Cornel M., Dijkstra I., Ten Kate L., Prevalence of lethal osteochondrodysplasias, AJMG 36 (1990) 377-378.

[8] van Dijk F., Nesbitt I., Nikkels P., Dalton A., Bongers E., van de Kamp J., Hilhorst-Hofstee Y., den Hollander N., Lachmeijer A., Marcelis C., Tan-Sindhunata M., van Rijn R., Meijers-Heijboer H., Cobben J., Pals G. CRTAP mutations in lethal and severe osteogenesis imperfecta: the importance of combining biochemical and molecular genetic analysis. EJHG. Epub ahead of print.

[9] van Dijk F., Nesbitt I., Zwikstra E., Nikkels P., Piersma S., Fratantoni S., Jimenez C., Huizer M., Morsman A., CobbenJ., van Roij M., Elting M., Verbeke J., Wijnaendts L., Shaw N., Högler W., McKeown C., Sistermans E., Dalton A., Meijers-Heijboer H., Pals G. PPIB mutations cause severe Osteogenesis Imperfecta..AJHG. Epub ahead of print.

[10] Fleming F., Woodhead H., Briody J., Hall J., Cowell C., Ault J., Kozlowski K., Sillence D., Cyclic biphosphonate therapy in osteogenesis imperfecta type V, J. Paediatr. Child Health 41 (2005) 147-151.

[11] Glorieux F., Rauch F., Plotkin H., Ward L., Travers R., Roughley., Lalic L., Glorieux D., Fassier F., Bishop N., Type V osteogenesis imperfecta: a new form of brittle bone disease., J. Bone Miner. Res. 15 (2000) 1650-1658.

[12] Glorieux F., Ward L., Rauch F., Lalic L., Roughley P., Travers R., Osteogenesis Imperfecta Type VI: a form of brittle bone disease with mineralization defect, J. Bone Miner Res 17 (2002) 30-37.

[13] Labuda M., Morissette, J., Ward., Rauch F., Lalic L., Roughley P., Glorieux F., Osteogenesis imperfecta type VII maps to the short arm of chromosome 3, Bone31 (2002) 19-25.

[14] Land C., Rauch F., Travers R., Glorieux F., Osteogenesis Imperfecta Type VI in childhood and adolescence: Effects of cyclical intravenous pamidronate treatment, Bone 40 (2007) 638-644.

[15] Lee D., Cho T., Choi I., Chung C., Yoo W., Kim J., Park Y., Clinical and radiological manifestations of Osteogenesis Imperfecta, J. Korean Med. Sci 21 (2006) 709-714.

[16] Marini J., Cabral W., Barnes A., Chang W., Components of the collagen prolyl-3-hydroxylation complex are crucial for normal bone development, Cell Cycle 6 (2007) 1675-1681.

[17] Marini J., Forlino A., Cabral W., Barnes A., San Antonio J., Milgrom S., Hyland J., Körkkö J., Prockop D., de Paepe A., Coucke P., Symoens S., Glorieux F., Roughely P., Lund A., Kuurila-Svahn K., Hartikka H., Cohn D., Krakow D., Mottes M., Schwarze U., Chen D., Yang K., Kuslich C., Troendle J., Dalgleish R., Byers P., Consortium for Osteogenesis Imperfecta Mutations in the Helical Domain of Type I Collagen: Regions Rich in Lethal Mutations align with collagen binding sides for integrins and proteins., Hum Mut 28 (2007) 209-221.

[18] Marini J., Grange D., Gottesman G., Lewis M., Koeplin D. Osteogenesis imperfecta type IV. Detection of a point mutation in one alpha1(I) collagen allele (Col1a1) by RNA/RNA hybrid analysis., Journal of biological chemistry 264 (1989) 11893-11900.

[19] Morello R., Bertin T., Chen Y., Hicks J., Tonachini L., Monticone M., Castagnola P., Rauch F., Glorieux F., Vranka J., Bächinger HP., Pace J., Schwarze U., Byers P., Weis M., Fernandes R., Eyre D., Yao Z., Buyce B., Lee B., CRTAP is required for prolyl 3-hydroxylation and mutations cause autosomal recessive osteogenesis imperfecta, Cell 127 (2006)291-304.

[20] Obafemi A., Bulas D., Troendle J., Marini C., Popcorn calcifications in osteogenesis imperfecta: incidence, progression, and molecular correlation, AJMG 146A (2008) 27-25-2732.

[21] Online Mendelian Inheritance in Man, OMIM (TM). Johns Hopkins University, Baltimore, MD. MIM Number:{ #610915}: {7/14/2008} and MIM number:{ #610854}:{3/19/2007}: World Wide Web URL: http://www.ncbi.nlm.nih.gov/omim/

Dijk_Opmaak.indd 127 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 64 T1 - Black CyanMagentaYellow

Page 129: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-1128

[22] Rauch F., Glorieux F., Osteogenesis Imperfecta, Lancet 363 (2004) 1377-1385.

[23] Sillence D., Barlow K., Garber A., Hall J., Rimoin D.,Osteogenesis imperfecta type II delineation of the phenotype with reference to genetic heterogeneity, Am J Med Genet 17 (1984 ) 407-423.

[24] Sillence D., Senn A., Danks D., Genetic heterogeneity in osteogenesis imperfecta, J Med Genet 16 (1979) 101-116.

[25]Spranger J., Brill P., Poznanski A., Section X: skeletal dysplasias with prominent diaphyseal involvement, Osteogenesis Imperfecta, type III/ IIB, in: Spranger J., Brill P., Poznanski A, Bone dysplasias. An atlas of genetic disorders of skeletal development, Oxford University Press, Oxford, 2003, pp 440-441.

[26] Steiner R., Pepin M., Byers P. Osteogenesis Imperfecta.www.genetests.com

[27] Wallis G., Sykes B., Byers P., Mathew C., Viljoen D., Beighton P. Osteogenesis imperfecta type III: mutations in the type I collagen structural genes, COL1A1 and COL1A2, are not necessarily responsible, J Med Genet 30 (1993) 492-496.

[28] Ward L., Rauch F., Travers R., Chabot G., Azouz E., Lalic L., Roughley P., Glorieux.F., Osteogenesis Imperfecta type VII an autosomal recessive form of brittle bone disease. Bone 31 (2002) 12-18.

[29] Zeitlin L., Rauch F., Travers R., Munns C., Glorieux F. The effect of cyclical intravenous pamidronate in children and adolescents with osteogenesis imperfecta type V, Bone 38 (2006) 13-20.

Dijk_Opmaak.indd 128 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 64 T1 - Black CyanMagentaYellow

Page 130: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta

Fleur S. van Dijk1, Peter H. Byers2, Raymond Dalgleish3, Fransiska Malfait4, Alessandra Maugeri1, Marianne

Rohrbach5, Sofie Symoens4, Erik A. Sistermans1,Gerard Pals1 following a EMQN Best Practice Meeting,

28-29 June 2010, Amsterdam, the Netherlands.

1 Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands2 Departments of Pathology and Medicine, University of Washington, Seattle, WA98195, USA

3 Department of Genetics, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom4 Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium

5 Division of Metabolism, University Children’s Hospital, Zurich, Switzerland

Eur J Hum Genet. 2011 Aug 10. [Epub ahead ofprint]

IV-2

Dijk_Opmaak.indd 129 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 65 T1 - Black CyanMagentaYellow

Page 131: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2130

AbSTrACT

Osteogenesis imperfecta (OI) comprises a group of inherited disorders characterized by bone fragility

and increased susceptibility to fractures. Historically, the laboratory confirmation of the diagnosis OI

rested on cultured dermal fibroblasts to identify decreased or abnormal production of abnormal

type I (pro)collagen molecules, measured by gel electrophoresis. With the discovery of COL1A1 and

COL1A2 gene variants as a cause of OI, sequence analysis of these genes was added to the diagnostic

process. Nowadays, OI is known to be genetically heterogeneous. About 90% of individuals with OI

are heterozygous for causative variants in the COL1A1 and COL1A2 genes. The majority of remaining

affected individuals have recessively inherited forms of OI with the causative variants in the more

recently discovered genes CRTAP, FKBP10, LEPRE1 ,PLOD2, PPIB, SERPINF1, SERPINH1 and SP7, or in

other yet undiscovered genes. These advances in the molecular genetic diagnosis of OI prompted

us to develop new guidelines for molecular testing and reporting of results in which we take into

account that testing is also used to “exclude” OI when there is suspicion of non-accidental injury (NAI).

Diagnostic flow, methods, and reporting scenarios were discussed during an international workshop

with 17 clinicians and scientists from 11 countries and converged in these best practice guidelines

for the laboratory diagnosis of OI.

Dijk_Opmaak.indd 130 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 65 T1 - Black CyanMagentaYellow

Page 132: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 131

1. INTrODuCTION

1.1. Classification of osteogenesis imperfectaOsteogenesis imperfecta (OI) comprises a heterogeneous group of disorders characterised by

susceptibility to bone fractures with severity that ranges from death in the perinatal period to subtle

increase in fracture frequency and in almost all cases presumed or proven defects in type I (pro)

collagen biosynthesis 1. In 1979, David Sillence developed a four-type classification which is still in use

for classification according to clinical/radiological features: OI type I (mild OI with bone fragility and

blue sclerae), II (perinatal lethal), III (progressive deforming) and IV (normal sclerae and mild deformity)

2,3. Dominant COL1A1/2 variants appeared to be causative in the majority of OI types. In 2004, OI types

V and VI were added to this classification because of specific clinical/radiological and/or histological

features, absence of abnormalities of type I (pro)collagen synthesis or structure on gel electrophoresis

and absence of causative variants in the COL1A1/2 genes 4,5. With the discovery of rare recessive genetic

causes of OI (see table 1), it was proposed to extend the classification with OI types VII and VIII 6.

However, the classification and subdivision into different types of OI is still under discussion 1 because

the phenotypic spectrum that results from mutations in some of these genes is almost as broad as that

with mutations in type I collagen genes. There is also debate as to whether Bruck syndrome type I 7 and

II 8, characterised clinically by bone fragility and congenital contractures of the large joints, should be

classified as a type of OI.

1.2. Biosynthesis of type I collagen Most individuals affected with OI are heterozygous for a causative variant in either of the two genes,

COL1A1 or COL1A2, which encode the proα1(I) and proα2(I) chains of type I procollagen, respectively.

Type I collagen is the major structural protein in bone, tendon, and ligament. Recently, rare recessive

genetic causes of OI have been described and, in all but one, the identified genes encode proteins

involved in the biosynthesis of type I procollagen (figure 1, table 1, table 2) 9,10.

figure 1: Critical steps in collagen type I biosynthesis and indication of genes known to be involved in OI.

Dijk_Opmaak.indd 131 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 66 T1 - Black CyanMagentaYellow

Page 133: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2132

Type I collagen is the major structural protein in bone, tendon, and ligament. It is first synthesized in the

rough endoplasmic reticulum (rER) as type I procollagen, containing C- and N-terminal propeptides. In

the rER, the two a1(I)- collagen chains encoded by COL1A1 and the one a2(I)-collagen chain encoded by

COL1A2 comprising predominantly Gly-X-Y triplets, align and assemble in the C- to-N direction to form

a triple helix. During folding, collagen is modified by, among others, specific enzymes that hydroxylate

lysine and proline residues and glycosylate hydroxylysyl residues. This process is called post-translational

modification and it stops when triple helix assembly is complete. The CRTAP/P3H1/CyPB complex

encoded by the CRTAP, LEPRE1 and PPIB genes, is responsible for the 3-hydroxylation of P986 (p.P1164

counting from the methionine which initiates translation) but will most likely also act as a cis-trans

isomerase and a molecular chaperone. FKBP65 encoded by FKBP10 also acts as a molecular chaperone

for type I procollagen. The protein product of PLOD2 (procollagen-lysine, 2-oxoglutarate 5-dioxygenase

2) hydroxylates telopeptide lysines in the rER. HSP47 encoded by SERPINH1 is thought to maintain the

stability of the triple helix. After folding, the procollagen molecules are transported through the Golgi

apparatus and plasma membrane (PM) into the extracellular matrix (ECM) where cleavage of the N-and

C-terminal propeptides occurs and collagen molecules aggregate to form fibrils.

Table 1. Genes in which sequence variants cause OI.

Gene OMIM (Gene)

refSeqGene genomic reference

refSeq mrNA reference refSeq protein reference

Locus reference Genomic (LrG)

COL1A1 120150 NG_007400.1 NM_000088.3 NP_000079.2 LRG_1

COL1A2 120160 NG_007405.1 NM_000089.3 NP_000080.2 LRG_2

CRTAP 605497 NG_008122.1 NM_006371.4 NP_006362.1 LRG_4

FKBP10 607063 NG_015860.1 NM_021939.3 NP_068758.3 LRG_12

LEPRE1 610339 NG_008123.1 NM_022356.3 (transcript variant 1)

NP_071751.3 (isoform 1, P3H1a, long)

LRG_5

NM_001146289.1 (transcript variant 2)

NP_001139761.1 (isoform 2, P3H1b, short)

PLOD2 601865 NG_009251.1 NM_182943.2 (transcript variant 1)

NP_891988.1 (isoform 1, LH2b, long)

*

NM_000935.2 (transcript variant 2)

NP_000926.2 (isoform 2, LH2a, short)

PPIB 123841 NG_012979.1 NM_000942.4 NP_000933.1 LRG_10

SERPINF1 172860 NG_028180.1 NM_002615.5 NP_002606.3 *

SERPINH1 600943 NG_012052.1 NM_001235.2 NP_001226.2 *

SP7 606633 NG_023391.1 NM_001173467.1 (transcript variant 1)

NP_001166938.1 # *

NM_152860.1 (transcript variant 2)

NP_690599.1 #

*LRG sequences have been developed to provide a stable genomic reference for each gene region annotated with transcripts, proteins and other associated data44. For some recently discovered genes in which sequence variation causes OI, no LRG is available yet. #The two transcripts of the SP7 gene are translated into identical proteins.

Dijk_Opmaak.indd 132 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 66 T1 - Black CyanMagentaYellow

Page 134: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 133Ta

ble

2. C

hara

cter

istic

s of

rece

ssiv

e O

I-rel

ated

gen

es

Gen

ePr

otei

n Pr

otei

n fu

nctio

nre

sults

of b

ioch

emic

al a

naly

sis

in c

ase

of c

ausa

tive

vari

ants

OI t

ype

CRTA

P 18

-21,

26Ca

rtila

ge-

asso

ciat

ed

prot

ein

(CRT

AP)

CRT

AP

form

s a

com

plex

with

the

prot

eins

enc

oded

by

LEP

RE1

and

PPIB

. Apa

rt fr

om 3

-pro

lyl h

ydro

xyla

tion

activ

ity, t

he c

ompl

ex a

ppea

rs to

hav

e ci

s-tr

ans

isom

eras

e ac

tivity

and

is th

ough

t to

act a

s a

mol

ecul

ar

chap

eron

e in

itiat

ing

chai

n re

cogn

ition

and

hel

ical

fo

ldin

g.

Post

-tra

nsla

tiona

l ove

rmod

ifica

tion

Dec

reas

ed 3

-pro

lyl h

ydro

xyla

tion

of P

986*

in th

e pr

oα1(

I)-co

llage

n ch

ains

II, II

I, IV

FKBP

10 27

-29

FKBP

65FK

BP65

has

pro

lyl c

is-tr

ans i

som

eras

e ac

tivity

, and

can

bi

nd c

olla

gen

No

post

- tra

nsla

tiona

l ove

rmod

ifica

tion

III/

Bruc

k sy

ndro

me

(Bon

e fra

gilit

y w

ith

cong

enita

l con

trac

-tu

res

of la

rge

join

ts)

LEPR

E1 18

-22,

23,2

6Pr

olyl

3-

hydr

oxyl

ase

1 (P

3H1)

P3H

1 fo

rms

a co

mpl

ex w

ith th

e pr

otei

ns e

ncod

ed

by C

RTAP

and

PPI

B. A

part

from

3-p

roly

l hyd

roxy

latio

n ac

tivity

, the

com

plex

is th

ough

t to

act a

s a

mol

ecul

ar

chap

eron

e an

d a

cis-

tran

s is

omer

ase.

Post

-tra

nsla

tiona

l ove

rmod

ifica

tion

Dec

reas

ed 3

-pro

lyl h

ydro

xyla

tion

of P

986*

in th

e pr

oα1(

I)-co

llage

n ch

ains

II, II

I

PLO

D2

8,30

Lysy

l hy

drox

ylas

e 2

Telo

pept

ide

lysy

l hyd

roxy

lase

-2 m

odifi

es ly

syl r

esid

ues

in th

e te

lope

ptid

es o

f typ

e I c

olla

gen

invo

lved

in

inte

rmol

ecul

ar c

ross

link

form

atio

n

Und

erhy

drox

ylat

ion

of ly

sine

resi

dues

in c

olla

gen

type

I te

lope

ptid

es re

sulti

ng in

abe

rran

t bon

e co

llage

n ty

pe I

cros

s lin

king

Bruc

k sy

ndro

me

PPIB

18,2

4-26

Cycl

ophi

lin B

P3H

1 fo

rms

a co

mpl

ex w

ith th

e pr

otei

ns e

ncod

ed b

y LE

PRE1

and

CRT

AP A

part

from

3-p

roly

l hyd

roxy

latio

n ac

tivity

, the

com

plex

is th

ough

t to

act a

s a

mol

ecul

ar

chap

eron

e an

d a

cis-

tran

s is

omer

ase

Post

-tra

nsla

tiona

l ove

r mod

ifica

tion

in m

ost c

ases

Dec

reas

ed 3

-pro

lyl h

ydro

xyla

tion

of P

986*

in th

e pr

oα1(

I)-co

llage

n ch

ains

in m

ost c

ases

II, II

I, IV

SERP

INF1

33PE

DF

PED

F is

kno

wn

mai

nly

for i

ts s

tron

g in

hibi

tion

of

angi

ogen

esis

. How

ever

, exp

ress

ion

anal

yses

in b

one

tissu

e fro

m w

ild-t

ype

mic

e an

d in

vitr

o ex

perim

ents

w

ith m

urin

e ce

ll sy

stem

s su

ppor

t a ro

le fo

r PED

F in

bo

ne fo

rmat

ion

and

rem

odel

ling

No

post

-tra

nsla

tiona

l ove

rmod

ifica

tion

III

SERP

INH

1 31

H

SP47

HSP

47 m

onito

rs th

e in

tegr

ity o

f the

trip

le h

elix

of t

he

type

I pr

ocol

lage

n at

the

ER/c

is-G

olgi

bou

ndar

yN

o po

st-t

rans

latio

nal o

verm

odifi

catio

nIII

SP7

32O

ster

ixO

ster

ix is

an

oste

obla

st-s

peci

fic tr

ansc

riptio

n fa

ctor

w

hich

has

bee

n sh

own

to b

e es

sent

ial f

or b

one

form

atio

n in

mic

e.

No

post

-tra

nsla

tiona

l ove

rmod

ifica

tion

IV

* p.

Pro1

164

coun

ting

from

the

met

hion

ine

whi

ch in

itiat

es tr

ansl

atio

n

Dijk_Opmaak.indd 133 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 67 T1 - Black CyanMagentaYellow

Page 135: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2134

1.3. Genes, proteins and causative variants1.3.1. Autosomal dominant OI

The most common form of OI in most populations is OI type I. Cultured dermal fibroblasts from affected

individuals produce about half the normal amount of type I procollagen molecules, which have normal

structure. OI type I usually results from variants in one COL1A1 allele (frameshift, nonsense, and splice-

site alterations) that lead to mRNA instability and haploinsufficiency. In a small subset of individuals

with OI type I, substitutions for glycine by small amino acids (cysteine, alanine and serine) near the

amino terminal ends of the triple helical domains of either COL1A1 or COL1A2 are found. In contrast,

OI types II, III and IV are usually caused by sequence variants in either COL1A1 or COL1A2 that result in

substitutions for glycine residues in the uninterrupted Gly-X-Y triplet repeat of the 1014-residue triple-

helical domains encoded by each gene. Less common causative variants include splice site alterations,

variants in the carboxyl-terminal propeptide coding-domains, or insertion/deletion events that lead to

in-frame sequence alterations. Most of these variants result in synthesis of abnormal type I procollagen

molecules characterized by post-translational overmodification of the triple-helical domain which

results in alterations visible by SDS-polyacrylamide gel electrophoresis. Loss of expression of either gene

due to large scale deletions appears to be rare 11-15. More than 1000 distinct variants in the COL1A1

and COL1A2 genes have been identified that cause OI (Dalgleish, R: Osteogenesis Imperfecta Variant

Database (https://oi.gene.le.ac.uk Accessed May 3 2011)16,17.

1.3.2. Autosomal recessive OI including Bruck syndrome

In the last 5 years, candidate-gene approaches have identified several loci in which causative variants

have been identified which result in the long sought causes for recessively inherited forms of OI (see

figure 1, table 2). We classify these genes in four groups—(i) one in which the genes encode proteins

that contribute to the initial phase of chain recognition and propagation of molecular folding (CRTAP,

LEPRE1, and PPIB 18-26) the second in which the genes encode proteins involved in the final quality control

of type I procollagen (FKBP10 27-29 and SERPINH1 31), the third which involves a gene encoding proteins

involved in late modification and crosslink formation (PLOD2 8,30), and the fourth group, which involves

recently recognized genes encoding proteins involved in bone cell differentiation (SP7 32 and possibly

SERPINF133).

1.4. Reasons for referral1.4.1. Prenatal

Prenatal ultrasounds showing abnormalities suggestive of OI (diminished mineralization of skull,

platyspondyly or bowing, shortening and/or fractures of long bones) are an important reason for referral

for molecular diagnostics of OI.

1.4.2. Postnatal

Radiographs at birth suggestive of OI, recurrent and/or unexplained fractures with or without suspicion

of non-accidental injury (NAI), primary (idiopathic) low bone mass, preferably with exclusion of

secondary causes, a family history of OI and request for confirmation of the clinical diagnosis are all

Dijk_Opmaak.indd 134 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 67 T1 - Black CyanMagentaYellow

Page 136: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 135

reasons for referral for molecular diagnostics of OI. However, in some cases dentinogenesis imperfecta

or even blue sclerae might be the only reason for referral. As such, physicians from many specialties

(gynaecologist, paediatrician, orthopaedic surgeon, clinical geneticist, general practitioner, dentist,

ear nose throat specialist, endocrinologist, internist, and ophthalmologist) might refer a patient for

molecular analysis. Depending on the age of presentation, OI can be difficult to distinguish from

some other genetic conditions, e.g. Ehlers-Danlos syndrome arthrochalasis type (former EDS VIIA and

B), isolated dentinogenesis imperfecta, blue sclerae and corneal fragility, hypophosphatasia, and non-

genetic causes of fractures including NAI and idiopathic juvenile osteoporosis 2, 34-36.

1.4.3. Non-accidental injury

Many referrals for OI diagnostics occur in the context of suspected NAI in an attempt to exclude a genetic

cause of fractures. Fractures resulting from NAI occur in 24 per 10,000 children under three years of age

whereas the OI prevalence is 1 per 10,000 to 20,000 34. The incidence of OI among children evaluated for

NAI is 2-5% 34. Differentiation is aided by an experienced clinician and radiologist familiar with OI 36 since

the nature and localisation of fractures in OI and NAI can often be distinguished 37. However, because

injuries may involve infants before many of the clinical features of OI are apparent, laboratory diagnostics

for OI can certainly be helpful in this differentiation.

2. METHODS

A group of clinicians and scientists involved in OI diagnostics met on 28-29 June 2010 at a workshop in

Amsterdam, the Netherlands, to formulate Best Practice Guidelines for the molecular genetic diagnosis

of OI supported by the European Molecular Genetics Quality Network (EMQN).

3. rESuLTS

Discussions focussed on diagnostic flow, methodologies, interpretation of results and reporting.

Consensus guidelines were established.

4. DISCuSSION

4.1. The diagnostic flowThe “traditional” way to establish or confirm the diagnosis of OI is based on studies of collagens synthesised

by cultured dermal fibroblasts. Previous studies 35 demonstrated that either quantitative or qualitative

alterations can be identified in about 90% 38 of individuals with clinically confirmed OI. Biochemical

analysis will separate individuals with quantitative defects (OI type I), from those with qualitative defects

(OI types II-IV) due to causative variants in the COL1A1/2, CRTAP, LEPRE1, PPIB genes and those with no

abnormalities detected. Electrophoretic analysis of type I procollagen may also identify other disorders

that can mimic some aspects of OI: EDS kyphoscoliotic type (type VIA), EDS arthrochalasia type (types

VIIA and VIIB), and EDS dermatosparaxis type (type VIIC).11Also, cultured fibroblasts provides a resource

for the analysis of RNA splicing and unclassified variants. However, biochemical analysis will not identify

some quantitative defects of type I procollagen, certain causative variants that alter sequences in some

Dijk_Opmaak.indd 135 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 68 T1 - Black CyanMagentaYellow

Page 137: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2136

coding regions of the COL1A1/COL1A2 genes and recessive forms of OI (including Bruck syndrome) that

result from variants in FKBP10, PLOD2, SERPINF1, SERPINH1 or SP7. Furthermore, prenatal diagnosis by

biochemical analysis is only possible in chorionic villus cells and is effective with qualitative alterations9

but unsuitable for quantitative defects and delays time of diagnosis by 2-4 weeks compared to genetic

analysis.

In contrast, direct genomic analysis(sequencing) of the known genes should identify causative variants

in more than 95% of affected individuals in most populations39,40. Next generation sequencing has

gained importance in the laboratory diagnosis of OI — in part because of shorter time to diagnosis and

in part because of the added value of the information as well as reduced costs, which is important for

the precise determination of recurrence risk (autosomal dominant versus recessive), prenatal diagnosis

and pre-implantation genetic diagnosis (PGD).

Given these considerations and current facilities, the consensus of the EMQN Best Practice in OI meeting

was to initiate laboratory-based diagnostic studies with direct genomic sequencing of the type I

procollagen genes, COL1A1 and COL1A2. The approach to diagnosis is detailed in figure 2. It was agreed

that the “traditional” approach can still be used in the context in which genomic DNA sequencing is

unavailable or the financial barriers are high. Moreover, analysis of proteins and mRNA/cDNA from

cultured fibroblasts remain valuable tools for follow up studies (see 3.2.4)

Sequence analysis and quantitative

analysis of COL1A1 and COL1A2 on genomic DNA

Report

Segregation and functional analysis

Re-evaluation of clinical diagnosis OI

Analysis of recessive genes

Report and banking of samples for research

Causative variant

No causative variant

Non-confirmed

Confirmed

Confirmed causative variant(s) or second causative variant found

No causative variant

Non-confirmed causative variant(s) or no second causative variant found

No further testing needed

figure 2: Preferred diagnostic flow in OI.The approach to diagnosis is designed to maximize the likelihood that causative variants will be identified in all affected individuals or assign those without causative variants to research pools. This flow assumes that the clinical diagnosis of OI is well established according to the traditional diagnostic criteria. With clear evidence of OI from radiological and clinical examination, further analysis should proceed according to the proposed strategy. Functional analysis consists of analysis of proteins and mRNA/cDNA from cultured fibroblasts and also includes COL1A1 null allele testing in certain selected cases.

Dijk_Opmaak.indd 136 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 68 T1 - Black CyanMagentaYellow

Page 138: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 137

4.2. Explanation of diagnostic work flow4.2.1. Sequencing of all type I procollagen gene exons

Procollagen type I gene sequencing should identify causative variants in 90% of affected individuals39,40,

provided that the clinical diagnosis of OI is accurate. In some cases a follow-up study is needed to

determine whether a variant is causative. This will often include genetic studies in the parents with

correlation to the phenotype observed in the parent, or analysis of mRNA splicing and protein-based

biochemical studies on cultured dermal fibroblasts.

If no causative variant in the COL1A1/2 genes is identified by sequence analysis, the next step is to

determine if a deletion or duplication of some or all of the coding regions of either gene has occurred.

Strategies such as array-based analysis, MLPA or qPCR if properly validated are considered equivalent by

the working group in their detection of such alterations. From currently available data in the represented

laboratories, the added causative variants expected from this approach should be about 1-2% . In case

of OI types II-IV, after identifying the causative variant in the index patient, determination of parental

mosaicism by sequence analysis of DNA from blood from each parent will provide data for genetic

counselling with respect to recurrence risk and care in subsequent pregnancies.

4.2.2. Re-review of clinical data and the question of non-accidental injury

When no causative COL1A1/2 variant is found, clinical priorities should be re-considered. Failure to

find a causative variant occurs if there is technical failure, or if no causative variant is present in these

genes (because of causative variants in other (un)known genes or because the patient does not have

OI). Therefore, referring physicians should be encouraged to provide a completed clinical checklist

(supplementary table 1) and X-rays of the patient. This clinical/radiological information should be

reviewed by a clinical geneticist or a clinician with experience in OI. This can help to determine the

likelihood of OI in the particular patient. With clear evidence of OI, further analysis should proceed (figure

2).

However, if the primary reason for genetic study is to identify children with OI among the larger group

suspected for NAI, analysis can reasonably stop after COL1A1/2 sequencing unless there is a strong

suggestion of consanguinity or recessive inheritance. Justification of this decision rests on the following

considerations. First, previous studies indicate that fewer than 5% of infants studied for suspicion of NAI

are found to have OI by biochemical or DNA-based studies 34. Second, DNA-based analysis will identify

a causative variant in more than 90% of all individuals with OI39, 40 so that the remaining risk that an

infant has OI, will be about 0.5%. Third, at present all infants with recessive OI have obvious radiographic

abnormalities fitting a diagnosis of OI and not of NAI.

4.2.3. Identification and characterization of causative variants in autosomal recessive OI

Variants in the genes causing recessive OI, are estimated to account for about 5 or 6% of individuals with

OI. This represents the pooled estimates from all the laboratories represented at the workshop. Although

laboratory context and clinical criteria may in some cases require otherwise, the preferred strategy is to

analyse all genes at the same time to minimize turnaround time. If no, or only one, causative variant is

identified by sequence analysis, the next step is to determine by array-based analysis, MLPA or qPCR if a

Dijk_Opmaak.indd 137 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 69 T1 - Black CyanMagentaYellow

Page 139: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2138

deletion or duplication of some or all of the coding regions of either allele has occurred. Once recessive

causative variants are identified, parental confirmation of carrier status should be completed, first to

confirm that the causative variants in the index case are inherited in trans and, secondly, to exclude the

possibility of uniparental disomy in the case of homozygosity in the infant.

4.2.4. Analysis of proteins and mRNA/cDNA from cultured fibroblasts (functional analysis)By following the guidelines to this point, virtually all causative variants in type I procollagen genes and

in the recessive OI-related genes will have been identified. However, in some cases analysis of proteins

and mRNA/cDNA from cultured fibroblasts can have an additive value. First of all, mRNA/cDNA analysis

provides a tool for studying the effect of unclassified variants suspected to alter splicing. Moreover, this

approach can unravel the effect of deep intronic variants that alter the mRNA sequences usually by

conversion of cryptic exons to active exons and which would not be detected by gDNA sequencing. In

these cases, biochemical analysis of type I procollagen chains can detect quantitative defects if the new

exon results in introduction of a premature stop codon and nonsense-mediated mRNA decay (NMD),

or qualitative defects if the mRNA is stable and translated into an abnormal protein. When OI type

I is suspected, but genomic DNA sequencing is unavailable or too costly, COL1A1 null-allele14 testing

on cDNA can be used to confirm the diagnosis. However, this approach relies on the availability of

informative markers and will not lead to the identification of the disease-causing variant.

4.2.5. Analysis of remaining samplesSome diagnostic laboratories maintain research arms and the identification of the additional OI-related

genes has profited from the repositories of cells and DNA samples from individuals with OI due to

unknown genetic causes. This practice should continue and cells and DNA samples should be banked

for future analysis. If the diagnostic laboratory does not maintain such a resource, banking with ones that

do, should be considered.

4.3 Laboratory Diagnosis of Osteogenesis Imperfecta 4.3.1. Molecular analysis for OI Molecular analysis for OI includes sequencing or mutation scanning and deletion/duplication testing

for COL1A1 and COL1A2, followed by sequencing of recessive OI-related genes (see table 1). Preferred

material for testing is genomic DNA from blood, but other sources can be used as well (see table

3A and 3B). For general issues regarding sample labelling and identification, sequence analysis and

interpretation see “Practice guidelines for Sanger Sequencing Analysis and Interpretation” (http://www.

cmgs.org/BPGs/pdfs%20current%20bpgs/Sequencingv2.pdf )

gDNA and cDNA sequencingCurrent practices in sequencing of the type I collagen genes differ among laboratories in that some use

exon-by-exon amplification followed by sequence determination (supplementary appendix table 3A

and 3B) and others use a multi-exon substrate for analysis. If properly validated, both will provide the

same mutation capture probability and their use should be determined by local preferences. To identify

splice variants, primer sequences for PCR used for direct sequencing should be designed far enough

Dijk_Opmaak.indd 138 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 69 T1 - Black CyanMagentaYellow

Page 140: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 139

from intron-exon boundaries to allow reading of the branch sites, consensus splice donor, and acceptor

sites or UTR sequences of acceptable quality. With regard to sequence analysis of cDNA, large fragments

should be analyzed to allow detection of multi-exon deletions, since these could be missed when

primers are located close to the exon junctions at multiple sites. The analysis of cDNA from fibroblasts

is warranted when an unknown variant is found that might affect splicing. In addition, cDNA sequence

analysis may detect null-alleles without a known cause (e.g. an unknown variant in the promoter region)

if care is taken to analyze regions in which coding sequence heterozygosity is known.

Quantitative analysisQuantitative analysis by MLPA (kits for COL1A1 and COL1A2 have been designed by MRC-Holland,

available at http://www.mlpa.com) and qPCR is particularly important in cases without a detected

molecular cause in which only genomic DNA is available and without heterozygous polymorphic

variants detected by sequencing. Recently, complete COL1A1 allele deletions have been reported to

cause OI type I 13. For reliable quantitative testing (MLPA/qPCR) patient and control materials from the

same source (e.g. blood or fibroblasts) should be used for comparison and the same DNA purification

protocol should be used for all samples within a test series.

Table 3A. Postnatal diagnostics for OI

Source Product Methods Comments

Blood (preferred) gDNA Direct SequencingMutation scanning (HRM*/QPCR)Deletion/Duplication Testing (MLPA)

Blood spots gDNA Direct Sequencing Very low yield

Saliva or buccal swabs gDNA Direct SequencingMutation scanning (HRM/QPCR)Deletion/Duplication Testing (MLPA)

High yield of DNA possible

Fibroblasts gDNA

mRNA/cDNAprotein

Direct SequencingMutation scanning (HRM/QPCR)Deletion/Duplication Testing (MLPA)**Sequencing for splice-site errors(pro)collagen type I electrophoresis

*HRM: high-resolution melting** Performing MLPA with DNA extracted from fibroblasts leads to unclear results in certain cases

Table 3b. Prenatal and preimplantation genetic diagnosis for OI

Source Product Methods

CVS gDNA

protein

Direct SequencingMutation scanning (HRM/QPCR)Deletion/Duplication Testing (MLPA)(pro)collagen type I electrophoresis

Amniocytes gDNA Direct SequencingMutation scanning (HRM/QPCR)Deletion/Duplication Testing (MLPA

Blastocysts gDNA Sequencing of known familial mutations

Dijk_Opmaak.indd 139 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 70 T1 - Black CyanMagentaYellow

Page 141: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2140

4.3.2. Protein analysis

Protein analysis of type I (pro)collagen (Supplementary Appendix figure 3A-D) is used to detect

quantitative and qualitative changes. The method used for this analysis is based on in vitro labelling of

collagen in cultured fibroblasts, followed by electrophoresis on 2M urea SDS-PAGE gels (the 2M urea

enhances chain separation) and autoradiography41. The analysis of procollagens is achieved by omitting

the pepsin digestion step from the collagen protocol. Procollagen electrophoresis enhances detection

sensitivity of quantitative type I (pro)collagen defects (OI type I), visualizes defects in type I (pro)collagen

located in the N-terminal region of the type I collagen triple helix more clearly and distinguishes

between EDS VIIC and EDS VIIA and B (if samples are analyzed after labeling in the presence and absence

of dextran sulfate to enhance proteolytic processing) 42.

4.3.3. Prenatal diagnosis

Prenatal diagnosis is possible in case of identification of known disease-causing variant(s) both

on genomic DNA extracted from chorionic villus sample (CVS) cells and amniocytes. “Testing for

Maternal Cell Contamination (MCC) in Prenatal Samples for Molecular Studies” (http://www.cmgs.org/

BPGs/pdfs%20current%20bpgs/MCC_08.pdf ) should be applied. In those rare instances when post-

translational over-modification of (pro)collagen type I is visible in, for example, cells from an affected

sibling but no disease-causing variant(s) have been detected in the genes involved in OI, electrophoresis

of type I (pro)collagen from cultured CVS cells can be used for prenatal diagnosis. Amniocytes are good

sources of DNA but they do not make type I procollagen in sufficient abundance to permit prenatal

diagnosis.

4.4. Interpretation of performed diagnostics

For the interpretation of an observed sequence variant it is essential to establish the causal role of the

variant in the pathogenesis of the disease. Textboxes 1 and 2 include descriptions of variants in the genes

involved in OI that are likely to be pathogenic or that should be considered as unclassified variants.

Dijk_Opmaak.indd 140 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 70 T1 - Black CyanMagentaYellow

Page 142: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 141

Textbox 1.

Title: Interpretation of variants in the COL1A1 and COL1A2 genes

COL1A1 and COL1A2 genes

1. Causative variants; considerations specific for the fibrillar collagen genes:

The following variants are likely to have pathological consequences for the protein function:

a. Substitution of the glycine residue of every Gly-X-Y triplet repeat within the triple-helical region of

the α1(I)- and α2(I)-collagen chains.

b. Sequence variants in consensus acceptor and donor splice sites (positions +1, +2, -1, -2) will lead

to splicing errors of the mRNA, but the outcome cannot be predicted without cDNA analysis,

with the exception of -1G>A acceptor-site splicing variants in the triple helical region of the α1(I)

collagen chains which result in a frameshift, nonsense-mediated mRNA decay, and a null-allele.

-1G>A splicing variants of exon 6 of the COL1A1 gene, results in skipping of exon 6, whereas for the

COL1A2 gene this results in use of a cryptic splice site in the exon and an in-frame deletion of 15

bases. Both are associated with EDS arthrochalasis type.

c. Nonsense mutations or deletions and insertions that result in interruption of the reading frame,

nonsense-mediated mRNA decay and a null-allele.

d. In-frame deletions and insertions that lengthen or shorten the α-chains.

e. Other variants* with experimental evidence (published or own data) of their impairment of the

protein’s function.

* Variants also should be checked against existing entries in the Osteogenesis Imperfecta Variant Database 16, 17 (https://oi.gene.le.ac.uk) which can be used for corroboration. However, it should be recognised that the data in the database are not warranted to be accurate or fit for any particular purpose.

2. unclassified variants (uV)

a. All other sequence variants must be considered as “unclassified” until segregation within the family

has been investigated and/or functional evidence becomes available.

b. It is strongly recommended that intronic nucleotide substitutions other than splice site mutations

affecting the -1, -2 or +1, +2 intronic nucleotide positions be examined by cDNA-analysis to

determine their splicing outcome.

c. It is strongly recommended that UVs be further investigated by in silico analysis (e.g. PolyPhen-2,

SIFT, Human Splicing Finder software, etc.). Of note: the new version of Polyphen classifies almost

all glycine substitutions as benign or tolerated.

Dijk_Opmaak.indd 141 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 71 T1 - Black CyanMagentaYellow

Page 143: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2142

Textbox 2

Title: Interpretation of variants in the CRTAP, FKBP10, LEPRE1, PLOD2, PPIB, SERPINF1, SERPINH1 and SP7

genes

CRTAP, LEPRE1, PPIB, FKBP10, SERPINH1, PLOD2 and SP7 genes

Causative variants

The following variants are likely to have pathological consequences for the protein function:

a. Sequence variants in consensus acceptor and donor splice sites that will lead to splicing errors of the

mRNA.

b. Nonsense mutations or deletions and insertions that result in interruption of the reading frame,

nonsense-mediated mRNA decay and a null-allele.

c. Most in frame deletions and insertions that lengthen or shorten the protein.

d. See 1e in textbox 1

Unclassified variants

See textbox 1.

4.5. Reporting General information on requirements for variant reporting can be found in the OECD Guidelines for

Quality Assurance in Molecular Genetic Testing (http://www.OECD.org/dataoecd/43/6/38839788.pdf )

and in the guidelines issued by the Swiss Medical Genetics Society (http://sgmg.ch/user_files/images/

SGMG_Reporting_Guidelines.pdf ).

4.6. Reporting scenarios4.6.1. Finding a causative variant in the COL1A1/COL1A2 genes in an affected index case

The report should state that a causative variant has been detected and that this confirms the clinical

diagnosis. The report should include a description of the reason why a particular variant is considered

causative. Determination of the parental origin of the detected variant(s) should be advised. Testing of

relatives at risk should be offered in conjunction with appropriate counselling.

In the case of severe/lethal OI due to a causative variant in COL1A1 or COL1A2, the measured recurrence

risk is 2% after the birth of one affected child. The recurrence risk is increased after the birth of two

affected individuals, presumably because the proportion of germ cells that carry the mutation

is higher43. This information may be mentioned in the results letter. If parental mosaicism can be

demonstrated in the father, the referring clinician could be encouraged to request a sperm sample

from the father to clarify the risk.

When the referring physician is not a clinical geneticist, it is recommended that the patient be referred

to a clinical genetics centre for counselling. Referral to OI centres for therapy can be proposed if

available.

Dijk_Opmaak.indd 142 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 71 T1 - Black CyanMagentaYellow

Page 144: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 143

4.6.2. Not finding a causative variant in COL1A1 and COL1A2 genes in an affected index case

If no causative variant is detected in the COL1A1 and COL1A2 genes after gDNA analysis and screening

for large gene deletions (e.g. MLPA), an interim report can be made with the recommendation for review

of the original patient diagnosis (detailed clinical information, e.g. with use of the clinical checklist,

radiographs, contact with the referring physician (see supplementary appendix table 1)). This review of

the clinical diagnosis should involve a clinician with expertise in diagnosis and classification of OI. The

outcome of this review might indicate the desirability for testing of additional OI genes and cDNA and

protein analysis of type I (pro)collagen. If applicable, a new request for additional testing should be sent

by the referring physician to the laboratory. Laboratories which do not offer analysis of the recessive

genes should suggest further analysis in another laboratory.

In the case of doubtful pathogenicity of the identified variant, the “Practice guidelines for the

Interpretation and Reporting of Unclassified Variants (UV’s) in Clinical Molecular Genetics” (http://www.

cmgs.org/BPGs/pdfs%20current%20bpgs/UV%20GUIDELINES%20ratified.pdf ) should be applied. If

the genetic testing procedure has identified an unclassified variant as the only sequence change, this

should be reported as such. However, the report should clearly state that the clinical significance of the

variant is unknown and that its identification does not provide an explanation for the clinical phenotype

of the patient.

4.6.3. Finding causative variants in the CRTAP, LEPRE1, PPIB, FKBP10, SERPINF1, SERPINH1, PLOD2,

SP7 genes in an affected index case

The final report should state that causative variants have been detected. It is necessary to investigate

the carrier status of the parents and siblings in order to determine whether a variant is causative. Testing

of relatives at risk may be offered in conjunction with appropriate counselling. Such testing might be

useful for prenatal diagnosis.

4.6.4. Finding one causative variant in the CRTAP, LEPRE1, PPIB, FKBP10, SERPINF1, SERPINH1,

PLOD2, SP7 genes in an affected index case

When one causative recessive variant has been detected, it cannot be excluded that a second causative

variant has been missed, for example when it concerns a deep intronic variant. Dependent on the

laboratory, a second interim report can be made indicating the desirability for cDNA and protein analysis

of type I (pro)collagen in the case of one causative variant in the CRTAP, LEPRE1 or PPIB genes. When

these additional tests are not informative or cannot be performed in the laboratory, the final report

should state that it is not possible to confirm the clinical diagnosis OI by biochemical and/or molecular

testing.

4.6.5. Not finding causative variants in the CRTAP, LEPRE1, PPIB, FKBP10, SERPINF1, SERPINH1,

PLOD2, SP7 genes in an affected index case

In the case of doubtful pathogenicity of the identified variants, the same procedure as described above

for the COL1A1 and COL1A2 genes should be followed. In the case where no causative variants in the

genes involved in recessive OI have been found, the final report should state that it is not possible to

confirm the clinical diagnosis OI by biochemical and/or molecular testing.

Dijk_Opmaak.indd 143 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 72 T1 - Black CyanMagentaYellow

Page 145: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2144

4.6.6. Not finding causative variant(s) in the genes known for autosomal dominant and autosomal

recessive OI; type I (pro)collagen electrophoresis

Reports of these studies are necessarily more descriptive than the DNA-based results. They should list

the types of genetic alterations that could not be identified. When low production or post-translational

over-modification of type I (pro)collagen is observed, the diagnosis of OI can be confirmed and in the

case of over-modification of type I (pro)collagen, prenatal diagnosis on chorionic villus cells is possible.

In reports concerning cells and samples with no molecular and/or biochemical diagnosis of OI it should

be stated that it is not possible to confirm the clinical diagnosis OI.

3.7. Prenatal or preimplantation genetic diagnosis

Prenatal or preimplantation genetic diagnosis with the intention of terminating a pregnancy or not

selecting embryos carrying the causative variant(s) is possible in the case of identification of known

disease-causing variant(s). This service should only be offered in a clinical genetics service and must be

accompanied by appropriate genetic counselling. Requests for prenatal or preimplantation diagnosis

should always be referred and announced in advance to a clinical genetics service. In addition, the

original result letter of the laboratory that identified the causative variant should accompany the request.

4. CONCLuSIONS

Best practice guidelines have been established for the molecular genetic diagnosis of osteogenesis

imperfecta. The most noteworthy issue is that molecular analysis of the COL1A1/2 genes is recommended

as the starting point in the diagnostic flow as opposed to protein analysis. It is to be expected that

molecular genetic testing in OI will only gain in importance as new OI-genes are discovered and with

the development of new technologies. However, in certain selected cases protein analysis will remain

important.

ACKNOWLEDGMENTS

Funding for the Best Practice Meeting was provided by the European Molecular Genetics Quality

Network (http://www.emqn.org).

CONfLICT Of INTErEST

The authors declare no conflict of interest.

Dijk_Opmaak.indd 144 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 72 T1 - Black CyanMagentaYellow

Page 146: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 145

SuPPLEMENTAry APPENDIx.

Table 1: Clinical check list for the diagnosis of OI

-Reason for referral

-Suspected type of OI

-Suspicion of non-accidental injury

-Family history/ pedigree

-Clinical information:

-Radiographs*

-Wormian bones

-Fractures (number of fractures, age at which first fracture occurred)

-Bone/skull/vertebral deformities

-Congenital contractures

-Low bone mineral density

-Large fontanel in infancy

-Ligamentous laxity

-Hernia(s)

-Easy bruising

-Respiratory insufficiency after birth

-Short stature

-Blue sclerae

-Dentinogenesis imperfecta

-Hearing loss (conductive/perceptive, age of onset)

* Physicians should be encouraged to send radiographs.

Table 2. Names and affiliations of other participants of the EMQN Best Practice Meeting OI

Javier Garcia-Planells, Instituto de Medicina Genomica, Valencia, Spain

Filomena Valentina Gentile, Rizzoli Orthopaedic Institute, Bologna, Italy

Anne-Sophie Lebre, Université Paris Descartes, AP-HP Hôpital Necker-Enfants Malades et Inserm U781,

Département de Génétique, Paris F-75015 France

Shirley McQuaid, National Centre for Medical Genetics, Our Lady’s Hospital for Sick Children, Dublin, Ireland

Rebecca Pollitt, Sheffield Children’s NHS Foundation Trust, Sheffield, United Kingdom

Agnieszka Rusinska, Department of Paediatric Propedeutics and Metabolic Bone Diseases, Medical University of

Lodz, Lodz, Poland

Thomas Schwarzbraun, Centogene GmbH, Rostock, Germany

Marjan Weiss, VU University Medical Centre, Amsterdam, the Netherlands

Dijk_Opmaak.indd 145 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 73 T1 - Black CyanMagentaYellow

Page 147: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2146

Table 3A: Examples of PCR Primers for gDNA sequencing of COL1A1

Example 1.

Exon Name Primer sequence (5’ > 3’)

01 C1A1E1F GTAAAACGACGGGCAGGGGTTGCAGGACGGGAGTTTCTCCTCGGGGTC

C1A1E1R CAGGAAACAGCTATGAAGAAACGAGTCTCCGGATCATCCACGTC

0203 C1A1-E02-M13-02F GTAAAACGACGGCCAGAGACCCGGGGGAAAGGTGGTTAAGC

C1A1-E02-M13-02R CAGGAAACAGCTATGAGTTTCTTGGTC GTGGGTGACTCTA

0304 C1A1E3F GTAAAACGACGGGCAGGGGTTGCAGGCTGGAGGCCTCTGCCGACGGGAGCAGC

C1A1E4R CAGGAAACAGCTATGAAGAAACAGGCTGTCCAGGGATGCCATC

05 C1A1E5F GTAAAACGACGGGCAGGGGTTGCAGACCTGGCCTCTTGTTTCTTCTC

C1A1E5R CAGGAAACAGCTATGAAGAAACCTGTAGGATTCTTGCAACTTTTCT

0608 C1A1E6F GTAAAACGACGGGCAGGGGTTGCAGCACACCAGGAAGTGCATGATGTCAG

C1A1E8R CAGGAAACAGCTATGAAGAAACAAGACCCAGGCCTGGGAGTTCTTC

09 C1A1E9F GTAAAACGACGGGCAGGGGTTGCAGCCCCTGGTGAGCCTGGCGAG

C1A1E9R CAGGAAACAGCTATGAAGAAACCTGAGTATCGTTCCCAAATGTG

1013 C1A1E10F GTAAAACGACGGGCAGGGGTTGCAGCTGGGGCCCCCCAAACCTGACCTGC

C1A1E13.vs2.R CAGGAAACAGCTATGAAGAAACGTCAGATGAGATGGGAGACAGC

1316 C1A1E13.vs2.F GTAAAACGACGGGCAGGGGTTGCAGGTAAGAGGCTGTCTGAACATC

C1A1E16R CAGGAAACAGCTATGAAGAAACTTTGGGGAACAGGGAGACATGAACC

16 COL1A1-E16-M13-01F GTAAAACGACGGCCAGTGTCCCAGGGTGTGACTT

COL1A1-E16-M13-01R CAGGAAACAGCTATGACCTTTGGTTTGGGGAACAG

1719 C1A1E17F GTAAAACGACGGGCAGGGGTTGCAGCTGATCATTGCTCTCCTGTCCCTGT

C1A1E19R CAGGAAACAGCTATGAAGAAACGAAGAGGATGAGCTGAGAGTC

2021 C1A1E20F GTAAAACGACGGGCAGGGGTTGCAGCAAGGGTAACAGCGTGAGTAC

C1A1E21R CAGGAAACAGCTATGAAGAAACGGAAACCACGGCTACCAGGTC

2224 C1A1E22F GTAAAACGACGGGCAGGGGTTGCAGCCGGACCCCCTGGCGAGCGTG

C1A1E24R CAGGAAACAGCTATGAAGAAACGTCCGGGGCGACCATCTTGAC

25 C1A1E25F GTAAAACGACGGGCAGGGGTTGCAGGCCCTGGCAGCCCTGGTCCTG

C1A1E25R CAGGAAACAGCTATGAAGAAACTAGGGAGGCTGAGGTCCAGAAAGTG

2627 C1A1E26F GTAAAACGACGGGCAGGGGTTGCAGAGGGCCCAGCAAGAAGCACCTGC

C1A1E27R CAGGAAACAGCTATGAAGAAACCACAGAGAGAACACTACAGTCAC

2829 C1A1E28F GTAAAACGACGGGCAGGGGTTGCAGCTGCTGTGAGTGTCCCTGATG

C1A1E29R CAGGAAACAGCTATGAAGAAACTGGCTGTCTGATTAGCTAGGAGGCGG

3031 C1A1E30F GTAAAACGACGGGCAGGGGTTGCAGGGGTTCCTCTCTAATCACGGCCAGAC

C1A1E31R CAGGAAACAGCTATGAAGAAACACCCCACACCCTATCTCCATG

32 C1A1E32F GTAAAACGACGGGCAGGGGTTGCAGTTTCTCAAGGCTTGTCGTTGGCCTTG

C1A1E32R CAGGAAACAGCTATGAAGAAACGATTCAAAGGAGGCAGAGATGGGAGC

3334 C1A1E33F GTAAAACGACGGGCAGGGGTTGCAGCCTCTCAGGAAACCCAGACACAAGCA

C1A1E34R CAGGAAACAGCTATGAAGAAACGTTCCCAGGTTGACAGCTCAG

Dijk_Opmaak.indd 146 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 73 T1 - Black CyanMagentaYellow

Page 148: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 147

3536 C1A1E35F GTAAAACGACGGGCAGGGGTTGCAGGTCCTGCCAAACTGAGCTGTC

C1A1E36R CAGGAAACAGCTATGAAGAAACCTTCTCCCCTGAGGATGGCTGAC

3739 C1A1E37F GTAAAACGACGGGCAGGGGTTGCAGTGCCTCCATTACTGCTCCTCC

C1A1E39R CAGGAAACAGCTATGAAGAAACTCAGTCAGCCCCACCATCCTTCTG

4042 C1A1E40F GTAAAACGACGGGCAGGGGTTGCAGGTGGGGGCTGCCAGAAGGATG

C1A1E42R CAGGAAACAGCTATGAAGAAACCAGGGGAACCTTCGGCACCAG

4344 C1A1E43F GTAAAACGACGGGCAGGGGTTGCAGCCCATGCCAGTACCCTCAGCATGGC

Exon Name Primer sequence (5’ > 3’)

4344 C1A1E43F GTAAAACGACGGGCAGGGGTTGCAGCCCATGCCAGTACCCTCAGCATGGC

C1A1E44R CAGGAAACAGCTATGAAGAAACCCTGCCTGGGTGAAGTCCGAC

4445 C1A1E44F GTAAAACGACGGGCAGGGGTTGCAGGCAACACTCCATGACCACAGC

C1A1E45R CAGGAAACAGCTATGAAGAAACGGGACAAACTGTCAGGCGGAAGTTC

4547 C1A1E45F GTAAAACGACGGGCAGGGGTTGCAGGGAGAGAGAGATCCAGCAGAGGGGA

C1A1E47R CAGGAAACAGCTATGAAGAAACAACCCTTCTCCAGAGAGGCAAAGGG

48 C1A1E48F GTAAAACGACGGGCAGGGGTTGCAGCCGTGGGGCCAGAGCCAGCAG

C1A1E48R CAGGAAACAGCTATGAAGAAACGCACAGAGAGGGAAGAGAGTGGGGA

4950 C1A1E49F GTAAAACGACGGGCAGGGGTTGCAGGCTGGTCCTGTTGTATGTAGC

C1A1E50R CAGGAAACAGCTATGAAGAAACCATGTCCCTTCTGAGCACTGGGCTA

50 COL1A1-E50-M13-01F GTAAAACGACGGCCAGATGCCCATATGTGCCCCTA

COL1A1-E50-M13-01R CAGGAAACAGCTATGAGACCTACTCCAGGACTTCA

51 C1A1E51F GTAAAACGACGGGCAGGGGTTGCAGGGACCCTGGACAGGAAGGCCAGCAGG

C1A1E51R CAGGAAACAGCTATGAAGAAACGATGGAGAGAGGGCACTATGGC

52 C1A1E52F GTAAAACGACGGGCAGGGGTTGCAGGGGCTTTTTGGCCAGGCCATAGTGCC

C1A1E52R CAGGAAACAGCTATGAAGAAACGAGGGGGTTCAGTTTGGGTTGCTTGTCTG

Example 2. COL1A1 Genomic DNA Plate Primers

Exon Name Sequence

Promoter - X1 1A1-PrOMO-S GCACCCCTACCACAGCACCT

1A1-IN1A GAGTCTCCGGATCATCCACGTC

X2 - 5 1A1-IN1Snew3 TTGCGGACTTTTTGGTTCTT

1A1-IN5Anew3 CGCAAAAGAGCCTGATGTTA

X6 - X9 1A1-IN5S CACACCAGGAAGTGCATGATGTCAG

1A1-IN9A CTGAGTATCGTTCCCAAATGTG

X10 - X13 1A1-IN9Snew CTGACCTGCAACAATCCAAA

1A1-IN13Anew GAGATGGGAGACAGCCTTGT

X14 - X18 new 1A1-IN13Snew GCAGGAGGCAGGGGCAAGAT

1A1-IN18Anew CGTGAGCCTAGGAGCAGAGGGAAA

Dijk_Opmaak.indd 147 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 74 T1 - Black CyanMagentaYellow

Page 149: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2148

X19 - X23 1A1-IN18Snew2 TAAGTATCCTGCCAGGCTTCAGT

1A1-IN23Anew2 TAGGGAGGCAGAGAGGTATGAGT

X24 - X25 1A1-IN23S GTCTGGGATGAGGCGTTCTGGATC

1A1-IN25A TAGGGAGGCTGAGGTCCAGAAAGTG

X26 - X29 1A1-IN25Snew GAGGAGTGGGCTCAGAAAGG

1A1-IN29Anew AGGGTTGAGGGAAAGTCACA

X30 - X32 1A1-IN29Snew GGGGAAGCCTTTGACACAT

1A1-IN32Anew CTGGACCACAGGGAGAGGAT

X33/34 - X37 1A1-IN32S CCTCTCAGGAAACCCAGACACAAGCA

1A1-x38A CTCCAGGAGCACCAACATTA

X38 - X41 1A1-IN37S TGAGTGGCTTGGCCCTCTGTG

1A1-IN41A GTCCGCTGGAGTCATCTCTAC

X42 - X45 1A1-IN41S GCGGAGCCAAGGAGAACAGAT

1A1-x46A TCGCCCTGTTCGCCTGTCTCACCCTTGTC

X46 - X48 1A1-x45S GTCCTGTCGGCCCTGTT

1A1-IN48A CAGAGAGGGAAGAGAGTGGGGA

X49 - X51 1A1IN48S GCTGGTCCTGTTGTATGTAGC

1A1-IN51A GATGGAGAGAGGGCACTATGGC

X52 1A1-IN51S GGGCATTTTGGCCAGGCCATAGTGCC

1A1-MnlI3’-A TTTTGGTTTTTGGTCATGTTCGGT

COL1A1 Individual Exon Primers

Exon Name Sequence

1A1-PrOMO-S GCACCCCTACCACAGCACCT

1A1-PrOMO-A CCGACCCCGAGGAGAAACT

 

1 1A1-5’uTr-S GACGGGAGTTTCTCCTCGGGGTC

1A1-IN1A GAGTCTCCGGATCATCCACGTC

 

2 new 1A1-IN1Snew* CGGGAAGTGAAAAATCCAAGT

1A1-IN2Anew

 

3/4 1A1-IN2S GACGGGAGCAGCATTAGCAA

1A1-IN4A AGGGGGAGAGGGCGGG

Dijk_Opmaak.indd 148 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 74 T1 - Black CyanMagentaYellow

Page 150: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 149

5 1A1-IN4Snew3 AAGCGTTGCACTCTGGGCTGT

1A1-IN5Anew TCCTGTAGGATTCTTGCAACTTT

 

6 1A1-IN5S CACACCAGGAAGTGCATGATGTCAG

1A1-IN6A* CTCCCAAGCTGTCTATACCAGCCGC

 

7 1A1-IN6S ATACGCGGCTGGTATAGACAG

1A1-IN7A* CCTCATCCCAGTCTTCCCTCCA

 

8 1A1-IN7S* TGGAGGGAAGACTGGGATGAG

1A1-IN8A AAGACCCCAGGCCTGGGAGTTCTTCT

 

9 1A1-x8S CCCCTGGTGAGCCTGGCGAG

1A1-IN9A CTGAGTATCGTTCCCAAATGTG

 

10 1A1-IN9S CTGGGCCCCCAAACCTGACCTGC

1A1-IN10A* GGCCATTAGAACACATCACTG

 

11 1A1-IN10S CTGAACCTGGGCTTCACTGCAC

1A1-IN11A GATGTCCACTCTCTGGCCCTTG

 

12 1A1-IN11S CAAAGGGATGGCGGTGATGA

1A1-IN12A GTGGGACTCTGGGGATGTGGA

 

13 1A1-IN12S CTCCACATCCCCAGAGTCCCA

1A1-IN13A CTCCATCTTGCCCCTGCCT

 

14 1A1-IN13S GAGGCAGGGGCAAGATGGA

1A1-IN14A CCCGTTAAGTCCACTGAGCACTG

 

15 1A1-IN14S GATCCCTGAGCTCTGGAAGGGGCTC

1A1-IN15A AGACCAACATAACCTGCCCCCA

 

16 1A1-IN15S GGGGGCAGGTTATGTTGGTCT

1A1-IN16A GGTTTGGGGAACAGGGAGACA

 

17 1A1-IN16S CTTCCTCCTGCCATCCCGA

1A1-IN17A GAGTGTCAGCAACAGGCAAGGA

 

Dijk_Opmaak.indd 149 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 75 T1 - Black CyanMagentaYellow

Page 151: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2150

18 1A1-IN17S GTCCCCTTTGCCACTTTCTAACCT

1A1-IN18A GCAGACAGCCAGGGCGTGA

 

19 1A1-IN18S CAGCCTGCCTGCCCCTTT

1A1-IN19A GCGTCTTCCTGCTCCCCA

 

20 1A1-IN19S CTGGGGAGCAGGAAGACGCA

1A1-IN20A* GGGGGTGTGGCAGGGACT

 

21 1A1-IN20S* GTCCCTGCCACACCCCCA

1A1-IN21A GCGACACACAGGCACCAGC

 

22 1A1-IN21Snew

1A1-IN22A CCAAAAGAGGAAGAAGATGCCCA

 

23 1A1-IN22S CCTGGGCATCTTCTTCCTCTTTT

1A1-IN23A GAGGTATGAGTGGGACTTGGGGA

 

24 1A1-IN23S GTCTGGGATGAGGCGTTCTGGATC

1A1-x25A GTCCGGGGCGACCATCTTGAC

 

25 1A1-x24S* GCCCTGGTCCTGATGGCAAAA

1A1-IN25A TAGGGAGGCTGAGGTCCAGAAAGTG

 

26 1A1-IN25S AGGGCCCAGCAAGAAGCACCTGC

1A1-x27A CTCCAGCCTCTCCATCTTTGCCA

 

27 1A1-IN26S CCTGCAGGAGGGGTGCTAGAG

1A1-x28A GCCTTGTTCACCTCTCTCGCCA

 

28 1A1-IN27S CTGCTGTGAGTGTCCCTGATG

1A1-IN28Anew

 

29 1A1-IN28S GGTGAGGCCTCATGGCTGTC

1A1-IN29A TGGCTGTCTGATTAGCTAGGAGGCGG

 

30 1A1-IN29S CAGACCCCAGGAGGAAGGACCGT

1A1-x31A CTCGCCAGGGAAACCTCTCTCG

 

Dijk_Opmaak.indd 150 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 75 T1 - Black CyanMagentaYellow

Page 152: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 151

31 1A1-x30S CCTCTGGAGCAAGAGTAAGTAG

1A1-IN31A ACCCCACACCCTATCTCCATG

 

32 1A1-IN31S CCTGGGAGATACCAAGCGGGG

1A1-IN32A GATTCAAAGCAGGCAGAGATGGGAGC

 

33/34 1A1-IN32S CCTCTCAGGAAACCCAGACACAAGCA

1A1-IN34A* GTTCCCAGGTTGACAGCTCAG

 

35 1A1-IN34S GAGGCTCTCCTGGGGTCATCTACTA

1A1-IN35A GAAGGCGGGGATGCGGT

 

36 1A1-IN35S CCCTGTCTGTGCCTTCACCCCTTGC

1A1-IN36A CTTCTCCCCTGAGGATGGCTGAC

 

37 1A1-IN36S* TGCCTCCATTACTGCTCCTCC

1A1-x38A CTCCAGGAGCACCAACATTA

 

38 1A1-IN37S* TGAGTGGCTTGGCCCTCTGTG

1A1-IN38A GAACATAGGAACAGTCAGAGGGAGAACA

 

39 1A1-x38S*

1A1-IN39A TCAGTCAGCCCCACCATCCTTCTG

 

40 1A1-IN39S GTGGGGGCTGCCAGAAGGATG

1A1-IN40A TGAGGTGCCAGACAGCAGCACAG

 

41 1A1-x40S* CGTCCTGGTGAAGTTGGTCCCC

1A1-IN41A GTCCGCTGGAGTCATCTCTAC

 

42 1A1-IN41S GCGGAGCCAAGGAGAACAGAT

1A1-IN42A GGGGAAGAGGGCTTAGGCAA

 

43 1A1-x42S CAAACAAGGTCCCTCTGGAGCAAGT

1A1-x44Aold CAGTCTCACCACGATCACCACTCT

 

44 1A1-IN43S* GCAACACTCCATGACCACAGC

1A1-IN44A CCTGCCTGGGTGAAGTCCGAC

 

Dijk_Opmaak.indd 151 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 76 T1 - Black CyanMagentaYellow

Page 153: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2152

45 1A1-IN44S GGAGAGAGAGATCCAGCAGAGGGGA

1A1-x46A* TCGCCCTGTTCGCCTGTCTCACCCTTGTC

 

46 1A1-x45S GTCCTGTCGGCCCTGTT

1A1-IN46A* GGGGAAAGAATGACTATCCAG

 

47 1A1-IN46S GTTGCCCACACTGCCCTTGTC

1A1-IN47A* CCACTCCCTGTCCCTGAACCCTT

 

48 1A1-IN47S GCTGGGGTTGTTCGGGCTT

1A1-IN48A CAGAGAGGGAAGAGAGTGGGGA

 

49 1A1IN48S* GCTGGTCCTGTTGTATGTAGC

1A1-IN49A CCAGCACCATATGGTAGGGGCACAT

 

50 1A1-IN49S CCAGGGTCCCCATGCCCATATGTGC

1A1-IN50A CATGTCCCTTCTGAGCACTGGGCTA

 

51 1A1-IN50S GGACCCTGGACAGGAGGCCAGCAGG

1A1-IN51A GATGGAGAGAGGGCACTATGGC

 

52 1A1-IN51S* GGGCATTTTGGCCAGGCCATAGTGCC

1A1-IN52A CAGTTTGGGTTGCTTGTCTGTTTCCG

* Better seq. primer

Table 3b: Examples of PCr Primers for gDNA sequencing of COL1A2

Example 1.

Exon Primer Primer sequence (5’ > 3’)

01 C1A2E1F GTAAAACGACGGGCAGGGGTTGCAGAGTTGGAGGTACTGGCCACGACTG

C1A2E1R CAGGAAACAGCTATGAAGAAACGCGTTTTCCCACATGCCTGAG

02 C1A2E2F GTAAAACGACGGGCAGGGGTTGCAGTGCTGATCCCTGCCATACTTTTGAC

C1A2E2R CAGGAAACAGCTATGAAGAAACTCTTCCCTTCCAAGAGAAGACATC

03 COL1A2-E03-M13-01F GTAAAACGACGGCCAGCTACACCAAAATGGAAGCTGTT

COL1A2-E03-M13-01R CAGGAAACAGCTATGACCTTTTGTATGTAAATCACCAGT

04 COL1A2-E04-M13-01F GTAAAACGACGGCCAGTTCACTGGAAATTACTTCTTAGGC

COL1A2-E04-M13-01R CAGGAAACAGCTATGACCAGTTCCTGTAGTTTCTAACATA

05 C1A2E5F GTAAAACGACGGGCAGGGGTTGCAGTCCACCCTACTTGCACATAGAAAGG

C1A2E5R CAGGAAACAGCTATGAAGAAACGACAAGGGCTCACAAAGAGAATGGG

Dijk_Opmaak.indd 152 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 76 T1 - Black CyanMagentaYellow

Page 154: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 153

06 C1A2E6F GTAAAACGACGGGCAGGGGTTGCAGTCGGCCAAGTTTTTGACGTACAGCT

C1A2E6R CAGGAAACAGCTATGAAGAAACTGGCGTGGTAAAATGTGACATAAAA

0709 C1A2E7F GTAAAACGACGGGCAGGGGTTGCAGGGGAGGAATAAAAACTATGGAATC

C1A2E9.vs2.R CAGGAAACAGCTATGAAGAAACTGTCAGGCATATTCAGCTTTTGGCA

0910 C1A2E9.vs2.F GTAAAACGACGGGCAGGGGTTGCAGTGAACCTGGTCAAACTGTGAGTAC

COL1A2-E10-M13-01R CAGGAAACAGCTATGAgttgcttatggtatgcttgctgtc

1112 COL1A2-E1112-M13-01F GTAAAACGACGGCCAGtactttggagggaagaagtcac

COL1A2-E1112-M13-01R CAGGAAACAGCTATGAggtcatggggaatttcaatca

1314 COL1A2-E1314-M13-01F GTAAAACGACGGCCAGatagagtaaaattgcactatcagga

COL1A2-E1314-M13-01R CAGGAAACAGCTATGAaacttaaatgacagccattagaaga

1516 COL1A2-E1516-M13-01F GTAAAACGACGGCCAGatcttctaatggctgtcatttaagt

COL1A2-E1516-M13-01R CAGGAAACAGCTATGAgttccaatctttcacatcacagta

1719 C1A2E17F GTAAAACGACGGGCAGGGGTTGCAGCAGTAGCCAAGATGGCAGAATC

C1A2E19R CAGGAAACAGCTATGAAGAAACCATATAGCAGACGGGAGTGTAC

2021 C1A2E20F GTAAAACGACGGGCAGGGGTTGCAGCTTGAGCTTCTCTTTACCTTGAC

C1A2E21R CAGGAAACAGCTATGAAGAAACAAGGCAGATGGAAAGCAGATG

2223 C1A2E22F GTAAAACGACGGGCAGGGGTTGCAGGGGTTGGGTGAAGTGTTTTGGCTTG

C1A2E23R CAGGAAACAGCTATGAAGAAACTCAAAAATGCAACTGTCAGCAAGAC

2425 C1A2E24F GTAAAACGACGGGCAGGGGTTGCAGAAAAAGTCGGGGGAAAAGGTGCCTT

C1A2E25R CAGGAAACAGCTATGAAGAAACCTGAGACTGGACTGATTCGCAG

2627 C1A2E26F GTAAAACGACGGGCAGGGGTTGCAGTGGAGCTGCATGGTGATGGATC

C1A2E27R CAGGAAACAGCTATGAAGAAACTAGCAACGTATGTCACCACTG

2829 COL1A2-E28-M13-01F GTA AAA CGA CGG CCA GGT GAC ATA CGT TGC TAT TTA TGC

COL1A2-E29-M13-01R CAG GAA ACA GCT ATG AGG GGA AAT TCT CAA GTT TAG TAG TT

30 COL1A2-E30-M13-01F GTA AAA CGA CGG CCA GCC AGG TTT ATT TCA CTC TTT CCA A

COL1A2-E30-M13-01R CAG GAA ACA GCT ATG AGC TTT AAG GAG AAA GCA CTA CTA C

31 COL1A2-E31-M13-01F GTAAAACGACGGCCAGaaaccagggctcggaagctacacaa

COL1A2-E31-M13-01R CAGGAAACAGCTATGAggtccactggaatcggattgctgtt

32 COL1A2-E32-M13-01F GTAAAACGACGGCCAGtctccctcctttcaatagcccagcc

COL1A2-E32-M13-01R CAGGAAACAGCTATGAgtgaaaacttgggcatccttgtgca

33 COL1A2-E33-M13-01F GTAAAACGACGGCCAGctgaaggtatcatagcatcttctgt

COL1A2-E33-M13-01R CAGGAAACAGCTATGAacccaggagagaaaggaatattat

34 COL1A2-E34-M13-01F GTAAAACGACGGCCAGaggttcacttttgatgatacgg

COL1A2-E34-M13-01R CAGGAAACAGCTATGAtctttgggactagggaaggc

3537 COL1A2-E3537-M13-01F GTAAAACGACGGCCAGccaaggcaactacagact

COL1A2-E3537-M13-01R CAGGAAACAGCTATGAcatagcaggcacttgacg

38 COL1A2-E38-M13-01F GTAAAACGACGGCCAGaaagagtagcatttacaagggt

COL1A2-E38-M13-01R CAGGAAACAGCTATGActcaaaagtttcctcatggtag

Dijk_Opmaak.indd 153 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 77 T1 - Black CyanMagentaYellow

Page 155: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2154

39 COL1A2-E39-M13-01F GTA AAA CGA CGG CCA GTA CCC AAA TTC TTG GAG TTG ATG

COL1A2-E39-M13-01R CAG GAA ACA GCT ATG ATC TTA CCA GTT TCC TAT CAG AAG C

40 COL1A2-E40-M13-01F GTA AAA CGA CGG CCA GCA GGC CCT TGG TGA TTA ACA

COL1A2-E40-M13-01R CAG GAA ACA GCT ATG AAC AAA TAG ATG CCA CTT GAA GAT

41 COL1A2-E41-M13-01F GTAAAACGACGGCCAGgccaacctgtgttatcaccta

COL1A2-E41-M13-01R CAGGAAACAGCTATGAtcacatttgaagtggcagc

42 COL1A2-E42-M13-01F GTAAAACGACGGCCAGttggaggggaaggttagc

COL1A2-E42-M13-01R CAGGAAACAGCTATGAtttggaagaagagaacctcagc

4345 C1A2E43F GTAAAACGACGGGCAGGGGTTGCAGAGGGTTCGTTACTGAGCACTG

C1A2E45R CAGGAAACAGCTATGAAGAAACGTATCAATTCTCAGCATGGACTG

46 COL1A2-E46-M13-01F GTAAAACGACGGCCAGCAGTATTTGTGGTGAAGTGAGT

COL1A2-E46-M13-01R CAGGAAACAGCTATGAAATATGAAAATCCTTCTGAGCTGA

4748 COL1A2-E4748-M13-01F GTAAAACGACGGCCAGTGTCTCTTGACATGTGCTCTG

COL1A2-E4748-M13-01R CAGGAAACAGCTATGACAAAGTTGTCTTGGTTTAGTCTGA

49 COL1A2-E49-M13-01F GTA AAA CGA CGG CCA GGT GTG AAG CTC AAC TGA AAA TCT

COL1A2-E49-M13-01R CAG GAA ACA GCT ATG ATT TGA AAA GCT CAA CTT GTG AGA A

50 COL1A2-E50-M13-01F GTA AAA CGA CGG CCA GTA GAA TCT GTG TTC TGC TCA ATG A

COL1A2-E50-M13-01R CAG GAA ACA GCT ATG ACC CAG GAA AGG AAC AGG TC

51 C1A2E51F GTAAAACGACGGGCAGGGGTTGCAGCCCTTTTCCTAAGCTTGGATCTGAG

COL1A2-E51-M13-01R CAGGAAACAGCTATGAttaacccccctttagacccccccttg

52 C1A2E52F GTAAAACGACGGGCAGGGGTTGCAGGGACAGACATCTTCAGAATGAC

C1A2E52R CAGGAAACAGCTATGAAGAAACTTGCCCACAATTTAAGCAAGTAG

Example 2.

Exons Name Sequence

Promoter - X1 1A2-PrOMO-S CCCCCATTCGCTCCCTCCT

1A2-IN1A CTGAGAGTCTGCCCTCCAAGTGTA

X2 - 4 1A2-IN1S ATGGGTGCCATACTTTTGACCTGC

1A2-IN4A GCTTCTTCTGCAGTGCATTACCT

X5 - 6 1A2-IN4S TCCACCCTACTTGCACATAGAAAGG

1A2-IN6A GCTAAATAAATGGCGTGGTAAAATGTGA

X7 - X10 1A2-IN6Snew2 GGACATGCTTCATCTGCTGT

1A2-IN10Anew2 CAGTGACTTCTTCCCTCCAAA

X11 - X12 1A2-IN10S TTTGTCGCTCTGTGCTTAGAGG

1A2-IN12A GAGGTCATGGGGAATTTCAATCA

Dijk_Opmaak.indd 154 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 77 T1 - Black CyanMagentaYellow

Page 156: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 155

X13 - X16 1A2-IN12S GAATCTAAGGGAGAAATTGGGGAGGA

1A2-IN16A AACACAGTTCCAATCTTTCACATCACA

X17 - X21 1A2-IN16S CAGTAGCCAAGATGGCAGAATC

1A2-IN21A ATAAGGCAGATGGAAAGCAGATG

X22 - X23 1A2-IN21Snew GGCCTGGAAACAATGTTGA

1A2-IN23Anew CCAAAAACCTCATAGCCATTG

X24 1A2-IN23S AAAAAGTCGGGGGAAAAGGTGCCTT

1A2-IN24A TCTCCCCTGCTCTGCTTTCAGTCCT

 

X25 1A2-IN24S TTTCATCCGTGGCAGCATCATAAGC

1A2-IN25A CTGAGACTGGACTGATTCGCAG

 

X26 1A2-IN25S TGGAGCTGCATGGTGATGGATC

1A2-IN26A AGTGTGGTTCTTAGATGAATGCTATGTGA

X27 - X29 1A2-IN26S CTTTGAGACATCTTAAACTACCTGCTT

1A2-IN29A TGGCTCATTCTCTCCATCAGCAC

X30 - X31 1A2-IN29S TGCACTCATGTAGATACTGCCAGGT

1A2-IN31A GGTCCACTGGAATCGGATTGCTGTT

 

X32 - X33 1A2-IN31S-DS TACTCTCTGCTTCCCATTGTCCTAT

1A2-IN33A-DS CCATGAGGCTGACATACGAGATAA

X34 1A2-IN33Snew2 AAAAAGAATGACAAGGTTCACTTTT

1A2-IN34Anew2 AGCATTTGTTTCCTGCTGCT

X35 - X38 1A2-IN34S ACTCTGTGAGATGTGCGTCAG

1A2-IN38A TCGGAATTGCTCTGAATAGAATGAA

X39 - X40 1A2-IN38S GAAATTCCCATCTTACCCAAATTCTTG

1A2-IN40A CATCAACAAATAGATGCCACTTG

X41 - X42 1A2-IN40S CTCACAATCTTCAAGCCAACCTGTG

1A2-IN42A AAAGCCCATTCTTTGGCCTAAGCAA

Dijk_Opmaak.indd 155 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 78 T1 - Black CyanMagentaYellow

Page 157: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2156

X43 - X46 1A2-IN42Snew AGCACTGGAAGTGATGAAGACA

1A2-IN47Anew TCGCTTTAGCCTCTATTTCAGA

X47 - X49 1A2-IN46Snew GGAGAAAAGAGCCCCACTTTACA

1A2-IN49A AGGGAAATGAGGTTGGGTGCTGGTT

X50 - X51 1A2-IN49S TGTTACTTATGAGAGTCAGTATCTTTC

1A2-IN51A TTAACCCCCCTTTAGACCCCCCTTG

X52 1A2-IN51S TCAAAGGTTCAGATATTATCAGATTCAGA

1A2-IN52A TTGCCCACAATTTAAGCAAGTAG

COL1A2 Individual Exon Primers

Exon Name Sequence

     

1A2-PrOMO-S CCCCCATTCGCTCCCTCCT

1A2-PrOMO-A AAGTCCGCGTATGGACAAAGCTGAGCAT

 

1 1A2-5’uTr-S GCACCACGGCAGCAGGAG

1A2-IN1A CTGAGAGTCTGCCCTCCAAGTGTA

 

2 1A2-IN1S ATGGGTGCCATACTTTTGACCTGC

1A2-IN2A* CCAAGAGAATACATCACTGTAGCCA

 

3 1A2-IN2S CGTATTGCTATGTATTTTTGTTCTGTA

1A2-IN3A CAAGTCTTTTTTTTTCCTTTTGTATGTA

 

4 1A2-IN3S* CATTGTAGTTACATCAGTCTTACC

1A2-IN4A GCTTCTTCTGCAGTGCATTACCT

 

5 1A2-IN4S* TCCACCCTACTTGCACATAGAAAGG

1A2-IN5A GACAAGGGCTCACAAAGAGAATGGG

 

6 1A2-IN5S GGGAAGTTCTCCATTTCAAAGAGGTGT

1A2-IN6A* GCTAAATAAATGGCGTGGTAAAATGTGA

 

7/8 1A2-IN6S GTCTGACCCCAGCCAACACCAT

1A2-IN8A GGAGACCCATCATTTCACTAAGG

Dijk_Opmaak.indd 156 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 78 T1 - Black CyanMagentaYellow

Page 158: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 157

9 1A2-IN8S TGAACCTGGTCAAACTGTGAGTAC

1A2-IN9A TGTCAGGCATATTCAGCTTTTGGCA

 

10 1A2-IN9S CCCCATTTTGTCTGATAGTTTACCA

1A2-IN10A GTTGCTTATGGTATGCTTGCTGTC

 

11 1A2-IN10S TTTGTCGCTCTGTGCTTAGAGG

1A2-IN11A CCAGGGATTTGAAAGTGAGTATATGTCA

 

12 1A2-IN11S CTGGGACCTGGAACACTGGACT

1A2-IN12A GAGGTCATGGGGAATTTCAATCA

 

13 1A2-IN12S GAATCTAAGGGAGAAATTGGGGAGGA

1A2-IN13A GCAAAAACATTAAAGAAGTTCCATCTCAT

 

14 1A2-IN13S CTTGTACAGGTTGGAAACTGAAC

1A2-IN14A CCACGGGCACCCTAAGAAGA

 

15 1A2-x14S CCGTGGGCTTCCTGGTGAGAG

1A2-IN15A TTGGTAAAGTGTCTGAAATGATGCTA

 

16 1A2-IN15S CACCCTGGATACCATGAATGTC

1A2-IN16A AACACAGTTCCAATCTTTCACATCACA

 

17 1A2-IN16S CAGTAGCCAAGATGGCAGAATC

1A2-x18A CCAGTAAGGCCGTTTGCTCCAG

 

18 1A2-IN17Snew GGGTCAAAGAAGAACCGAAA

1A2-IN18A AAAATGCAGTGTGGTCCATTAGG

 

19 1A2-IN18S TAATGTGTGCTGCCTCTACAGC

1A2-IN19A CATATAGCAGACGGGAGTGTAC

 

20 1A2-IN19S CTTGAGCTTCTCTTTACCTTGAC

1A2-IN20A GTTTAATGAAAAGAACAAAAGAAGAGGGA

 

21 1A2-x20S GGAGAGAGCGGTAACAAGGGT

1A2-IN21A ATAAGGCAGATGGAAAGCAGATG

 

Dijk_Opmaak.indd 157 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 79 T1 - Black CyanMagentaYellow

Page 159: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2158

22 1A2-IN21S* GGGTTGGGTGAAGTGTTTTGGCTTG

1A2-IN22A GAGGATGCTAAAGCTAATGACAC

 

23 1A2-x22S TAGTCCTGGTTCTCGTGGTCTTCCTG

1A2-IN23A* TAAAGAAGGAAGTAATGCCAGGTGTGA

 

24 1A2-IN23S AAAAAGTCGGGGGAAAAGGTGCCTT

1A2-IN24A TCTCCCCTGCTCTGCTTTCAGTCCT

 

25 1A2-IN24S TTTCATCCGTGGCAGCATCATAAGC

1A2-IN25A* CTGAGACTGGACTGATTCGCAG

 

26 1A2-IN25S* TGGAGCTGCATGGTGATGGATC

1A2-IN26A AGTGTGGTTCTTAGATGAATGCTATGTGA

 

27 1A2-IN26S CTTTGAGACATCTTAAACTACCTGCTT

1A2-IN27A

 

28 1A2-IN27S TGGCCATCTCCATTTTCAGTC

1A2-IN28A TGCTTCAGTCCTGAAATCATGT

 

29 1A2-IN28S GAGCTGTAAATCACCATACCGTAC

1A2-IN29A TGGCTCATTCTCTCCATCAGCAC

 

30 1A2-IN29S TGCACTCATGTAGATACTGCCAGGT

1A2-IN30A GACTTGTTGCAGGGTCATCAGTGGC

 

31 1A2-IN30S AAACCAGGGCTCGGAAGCTACACAA

1A2-IN31A GGTCCACTGGAATCGGATTGCTGTT

 

32 1A2-IN31S TCTCCCTCCTTTCAATAGCCCAGCC

1A2-IN32A GTGAAAACTTGGGCATCCTTGTGCA

 

33 1A2-IN32Snew2 TGCTTCAGGAGAGTGTACGGAA

1A2-IN33Anew* TTGACACTAGGAAGTTAGAAAATTCGGATA

 

33seq 1A2-IN32Sseq TTCAGGAGAGTGTACGGAAATTAGAAAAGAT

1A2-IN33Aseq TATTATTGTCATAGCCCAGGAGAGAAGGAA

 

Dijk_Opmaak.indd 158 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 79 T1 - Black CyanMagentaYellow

Page 160: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 159

34 1A2-IN33S GGAGTACCCTCCTTCTGAGAGTGGC

1A2-IN34A ATTGCTGGGGCTCTTTGGGACTAGG

 

35 1A2-IN34S ACTCTGTGAGATGTGCGTCAG

1A2-x36A GTTGGGGCCAGCAGGACCGAC

 

36 1A2-IN35S GAAGCCCTGTAAGTAAGAACCTG

1A2-IN36A GTTACAGCTCTGGTATTCCGAC

 

37 1A2-IN36S GATTTGCTGGTCCGGCTGTGAG

1A2-IN37A CTTCCGTTATTTTCCATCTTCTATC

 

38 1A2-IN37S TGCGGGAATGATCCACTTGAAGAAA

1A2-IN38A TCGGAATTGCTCTGAATAGAATGAA

 

39 1A2-IN38S GAAATTCCCATCTTACCCAAATTCTTG

1A2-IN39A GAAAAGCTGACTTCAGACCAGGAG

 

40 1A2-IN39S CATAAAGGAAGACAGGAGTTGC

1A2-IN40A CATCAACAAATAGATGCCACTTG

 

41 1A2-IN40S CTCACAATCTTCAAGCCAACCTGTG

1A2-IN41A TCTGTCACATTTGAAGTGGCAGCTT

 

42 1A2-IN41S GGAGGGGAAGGTTAGCATTCCATCG

1A2-IN42A AAAGCCCATTCTTTGGCCTAAGCAA

 

43 1A2-IN42S AGGGTTCGTTACTGAGCACTG

1A2-x44A CCACGGGGCCATGAGGACCAG

 

44 1A2-x43S ATGGTCAACCCGGACACAAG

1A2-IN44A CAACTTAGCTAGGCCCAAGATAC

 

45 1A2-IN44S GAGGAGTGGGGAGGGGTATCTT

1A2-IN45A CAGATGTTTTGGACTGATTCTCTCTAAAT

 

46 1A2-IN45S GCAGATTACCAGCAGAGGTGAGAGC

1A2-IN46A TGAAAATCCTTCTGAGCTGAAGGCC

 

Dijk_Opmaak.indd 159 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 80 T1 - Black CyanMagentaYellow

Page 161: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2160

47 1A2-IN46Snew GGAGAAAAGAGCCCCACTTTACA

1A2-IN47A* GACACCAGGTACATGTGAGCTG

 

48 1A2-IN47S CAAGAGAAGACAGTTCATCTCTG

1A2-IN48A TGGGGCTAACTTTAATGGGTTGTC

 

49 1A2-IN48S GAACATGCTTCCGTGTGAAGCTC

1A2-IN49A AGGGAAATGAGGTTGGGTGCTGGTT

 

50 1A2-IN49S TGTTACTTATGAGAGTCAGTATCTTTC

1A2-IN50A GTCCAATCAATCCATCTTCTAATGTG

 

51 1A2-IN50S CCCTTTTCCTAAGCTTGGATCTGAG

1A2-IN51A* TTAACCCCCCTTTAGACCCCCCTTG

 

52 1A2-IN51S TCAAAGGTTCAGATATTATCAGATTCAGA

1A2-IN52A* TTGCCCACAATTTAAGCAAGTAG

* Better seq. primer

figure 3 Type I (pro)collagen type I electrophoresis

P   C   P   C   P   C  

α1(III)3  

α1(I)  

α2(I)  

Procollagen   Collagen  medium   Collagen  cell  

A.

Proα1(III)

Proα1(I)

pCα1(III)

pCα1(I)

pNα1(I)

Proα2(I)

α2(I) pNα2(I)

pCα2(I) α1(I) - α1(III)

Figure 3A: (pro)collagen type I electrophoresis of a patient (P1 and P2) with a homozygous LEPRE1 mutation c.680C>T [p.(Arg210*)] compared with a healthy control (C).

Dijk_Opmaak.indd 160 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 80 T1 - Black CyanMagentaYellow

Page 162: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 161

α1(I)  

α2(I)  

α1(III)3  

α1(I)  -­‐  α1(III)  

Proα1(III)  

Proα1(I)  

pCα1(III)  

pCα1(I)  

Proα2(I)  

pNα1(I)  

pCα2(I)  

pNα2(I)  

α2(I)  

P   C   C   C  P   P  

B.

Procollagen Collagen medium Collagen cell

Figure 3B: (pro)collagen type I electrophoresis of a patient (P) with a heterozygous COL1A2 mutation c.1981G>C [p.(Gly661Arg)] compared with healthy controls.

α1(I)  α2(I)  

α1(III)3  

Collagen  medium   Collagen  cell  

P  C   C   C  P   C   P   C  

Collagen  cell  

C.

Figure 3C: (pro)collagen type I electrophoresis of a patient (P) with a heterozygous COL1A1 mutation c.2845G>A [p.(Gly949Ser)] compared with healthy controls (C).

α1(I)  α2(I)  

α1(III)3  

Collagen  medium   Collagen  cell   Procollagen  

C   C   C   C   C  P   P   P   P   P  

D.

Figure 3D: (pro)collagen type I electrophoresis of a patient (P) with a homozygous CRTAP mutation c.320_321del, [(p.Arg107Profs*53)] compared with healthy controls (C).

Dijk_Opmaak.indd 161 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 81 T1 - Black CyanMagentaYellow

Page 163: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter IV-2162

rEfErENCES

1. van Dijk FS, Pals G, van Rijn RR, Nikkels PGJ, Cobben JM: Classification of osteogenesis imperfecta revisited. Eur. J. Med. Genet. 2010; 53:1-5.

2. Byers PH, Krakow D, Nunes ME, Pepin M: Genetic evaluation of suspected osteogenesis imperfecta (OI). Genet. Med. 2006; 8:383-388.

3. Sillence DO, Senn A, Danks DM: Genetic heterogeneity in osteogenesis imperfecta. J. Med. Genet. 1979; 16:101-116.

4. Glorieux FH, Rauch F, Plotkin H et al: Type V osteogenesis imperfecta : a new form of brittle bone disease. J. Bone Miner. Res. 2000; 15:1650-1658.

5. Glorieux FH, Ward LM, Rauch F, Lalic L, Roughly PJ, Travers R. Osteogenesis Imperfecta Type VI: a form of brittle bone disease with mineralization defect. J. Bone Miner. Res. 2002; 17: 30-37.

6. Rauch F, Glorieux FH: Osteogenesis imperfecta. Lancet 2004; 363:1377-1385.

7. Breslau-Siderius EJ, Engelbert RH, Pals G, van der Sluijs JA. Bruck syndrome: a rare combination of bone fragility and multiple congenital joint contractures. J Pediatr Orthop B. 1998;7:35-38.

8. Ha-Vinh R, Alanay Y, Bank RA et al: Phenotypic and molecular characterization of Bruck syndrome (osteogenesis imperfecta with contractures of the large joints) caused by recessive mutation in PLOD2. Am. J. Med Genet. 2004; 131A:115-120.

9. Engel J, Prockop DJ: The zipper-like folding of collagen triple helices and the effects of mutations that might disrupt the zipper. Annu. Rev. Biophys. Biophys. Chem. 1991; 20:137-152.

10. Byers PH, Wallis GA, Willing MC: Osteogenesis imperfecta: translation of mutation to phenotype. J. Med. Genet. 1991; 28: 433-442.

11. Steiner RD, Pepin MG, Byers PH: Osteogenesis Imperfecta. Available at http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=gene&part=oi. Accessed May 3, 2011.

12. Willing MC, Pruchno CJ, Atkinson M, Byers PH: Osteogenesis imperfecta type I is commonly due to a COL1A1 null allele of type I collagen. Am. J. Hum. Genet. 1992; 51:508-515.

13. Willing MC, Deschenes SP, Scott DA et al: Osteogenesis Imperfecta type I: molecular heterogeneity for COL1A1 null alleles of type I collagen. Am. J. Hum. Genet. 1994; 55:638-647.

14. Nuytinck L, Sayli BS, Karen W, De Paepe A: Prenatal diagnosis of osteogenesis imperfecta type I by COL1A1 null-allele testing. Prenat. Diagn. 1999; 19:873-875.

15. van Dijk FS, Huizer M, Kariminejad A et al: Complete COL1A1 allele deletions in Osteogenesis Imperfecta. Genet. Med. 2010; 12:736-741.

16. Dalgleish R: The human type I collagen mutation database. Nucleic Acids Res. 1997; 25:181-187.

17. Dalgleish R: The human collagen mutation database 1998. Nucleic Acids Res. 1998; 26:253-255.

18. Ishikawa Y, Wirz J, Vranka JA, Nagata K, and Bächinger HP: Biochemical characterization of the prolyl 3-hydroxylase 1·cartilage-associated protein·cyclophilin B complex. J. Biol. Chem. 2009; 284: 17641-17647.

19. Barnes AM, Chang W, Morello R et al: Deficiency of cartilage-associated protein in recessive lethal osteogenesis imperfecta. N. Engl. J.Med. 2006; 355: 2757-2764.

20. Morello R, Bertin TK, Chen Y et al: CRTAP is required for prolyl 3- hydroxylation and mutations cause recessive osteogenesis imperfecta. Cell 2006; 127:291-304

21. Marini JC, Cabral WA, Barnes AM: Null mutations in LEPRE1 and CRTAP cause severe recessive osteogenesis imperfecta. Cell. Tissue Res. 2010; 339:59-70.

22. Cabral WA, Chang W, Barnes AM et al: Prolyl 3-hydroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfecta. Nat. Genet. 2007; 39: 359-365.

23. Willaert A, Malfait F, Symoens S et al: Recessive osteogenesis imperfecta caused by LEPRE1 mutations: clinical documentation and identification of the splice form responsible for prolyl 3-hydroxylation. J. Med. Genet. 2009; 46: 233-41.

24. van Dijk FS, Nesbitt IM, Zwikstra EH et al: PPIB mutations cause severe osteogenesis imperfecta. Am. J. Hum. Gen. 2009; 85: 521-527.

25. Barnes AM, Carter EM, Cabral WA et al: Lack of cyclophilin B in osteogenesis imperfecta with normal collagen folding. N. Engl. J.Med. 2010; 362:521-528.

26. Pyott SM, Schwarze U, Christiansen HE et al: Mutations in PPIB (cyclophilin B) delay type I procollagen chain association and result in perinatal lethal to moderate osteogenesis imperfecta phenotypes. Hum Mol Genet. 2011; 20:1595-1609.

Dijk_Opmaak.indd 162 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 81 T1 - Black CyanMagentaYellow

Page 164: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

EMQN Best Practice Guidelines for the Laboratory Diagnosis of Osteogenesis Imperfecta 163

27. Alanay Y, Avaygan H, Camacho N et al : Mutations in the gene encoding RER protein FKBP65 cause autosomal-recessive osteogenesis imperfecta. Am. J. Hum. Genet. 2010; 86, 551-559

28. Shaheen R, Al-Owain M, Sakati N, Alzayed ZS, Alkuraya FS: FKBP10 and Bruck syndrome: phenotypic heterogeneity or call for reclassification? Am J Hum Genet. 2010; 87:306-307

29. Kelley BP, Malfait F, Bonafe L et al: Mutations in FKBP10 cause recessive osteogenesis imperfecta and Bruck syndrome. J Bone Miner Res. 2011; 26: 666-672.

30. van der Slot AJ, Zuurmond AM, Bardoel AFJ et al: Identification of PLOD2 as telopeptide lysyl hydroxylase, an important enzyme in fibrosis. J. Biol. Chem 2003; 278: 40967-40972.

31. Christiansen HE, Schwarze U, Pyott SM et al: Homozygosity for a missense mutation in SERPINH1, which encodes the collagen chaperone protein HSP47, results in severe recessive osteogenesis imperfecta. Am. J. Hum. Genet. 2010; 86: 389-398.

32. Lapunzina P, Aglan M, Temtamy S et al: Identification of a frameshift mutation in Osterix in a patient with recessive osteogenesis imperfecta. Am. J. Hum. Genet. 2010; 87:110-114.

33. Becker J, Semler O, Gilissen C et al: Exome sequencing identifies truncating mutations in human SERPINF1 in autosomal-recessive osteogenesis imperfecta. Am J Hum Genet. 2011; 88:362-371

34. Marlowe A, Pepin MG, Byers PH: Testing for osteogenesis imperfecta in cases of suspected non-accidental injury. J. Med. Gen. 2002; 39:382-386.

35. Ablin DS, Greenspan A, Reinhart M, Grix A: Differentiation of child abuse from osteogenesis imperfecta. Am. J. Roentgenol. 1990; 154:1035-1046.

36. Steiner RD, Pepin M, Byers PH: Studies of collagen synthesis and structure in the differentiation of child abuse from osteogenesis imperfecta. J. Pediatr. 1996; 128: 542-547.

37. Bilo RAC, Robben, SGF, van Rijn RR. Forensic Aspects of Paediatric Fractures; Differentiating Accidental Trauma from Child Abuse. 1st edition. 2010. Heidelberg Dordrecht London New York, Springer.

38. Wenstrup RJ, Willing MC, Starman BJ, Byers PH: Distinct biochemical phenotypes predict clinical severity in nonlethal variants of osteogenesis imperfecta. Am. J. Hum. Genet 1990; 46: 975-982.

39. Körkkö J, Ala-Kokko L, De Paepe A, Nuytinck L, Earley J, Prockop DJ: Analysis of the COL1A1 and COL1A2 genes by PCR amplification and scanning by conformation-sensitive gel electrophoresis identifies only COL1A1 mutations in 15 patients with osteogenesis imperfecta type I:identification of common sequences of null-allele mutations. Am. J. Hum. Genet. 1998; 62: 98-110.

40. Sykes B, Ogilvie D, Wordsworth P et al: Consistent linkage of dominantly inherited osteogenesis imperfecta to the type I collagen loci: COL1A1 and COL1A2. Am. J. Hum. Genet. 1990; 46:293-307.

41. Steinmann B, Rao VH, Vogel A, Bruckner P, Gitzelmann R, Byers PH. Cysteine in the triple-helical domain of one allelic product of the α1(I) gene of type I collagen produces a lethal form of osteogenesis imperfecta. J Biol Chem. 1984; 259:11129-11138.

42. Bateman JF, Golub SB. Assessment of procollagen processing defects by fibroblasts cultured in the presence of dextran sulfate. Biochem. J. 1990; 267: 573-577

43. Pyott SM, Schwarze U, Christiansen HE et al: Recurrence of perinatal lethal osteogenesis imperfecta in sibships: parsing the risk between parental mosaicism for dominant mutations and autosomal recessive inheritance. Genet. Med. 2011; 13:125-130.

44. Dalgleish R, Flicek P, Cunningham F et al: Locus Reference Genomic sequences: an improved basis for describing human DNA variants. Genome Med. 2010; 2:24.

Dijk_Opmaak.indd 163 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 82 T1 - Black CyanMagentaYellow

Page 165: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 164 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 82 T1 - Black CyanMagentaYellow

Page 166: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter V

DiscussionSummary

Samenvatting voor de geïnteresseerde leek

Dijk_Opmaak.indd 165 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 83 T1 - Black CyanMagentaYellow

Page 167: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 166 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 83 T1 - Black CyanMagentaYellow

Page 168: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Discussion 167

DISCuSSION

OI is currently known to be clinically and genetically heterogeneous with an estimated 90% due to a

heterozygous causative variant in the COL1A1 or COL1A2 gene and approximately 10% due to causative

variants in recessive OI-related genes and other unknown genetic causes. Most of the newly discovered

genes for recessive OI have been found through candidate gene approach (CRTAP, LEPRE1, PPIB,

SERPINH1) or homozygosity mapping (FKBP10) and all appear to encode proteins that have a function

in collagen type I biosynthesis although the exact role of these proteins is still not known. Most recently

discovered genes (SP7 and SERPINF1) have been found with the use of (relatively) new techniques (SNP

array and whole exome sequencing, respectively). As laboratories increasingly will use next generation

sequencing, it will be more easy to find new but rare genetic causes of OI, provided the laboratory has

access to material of patients with OI due to an unknown genetic cause. Discovery of new genetic

causes in OI is of importance for determining the recurrence risk in families, and for prenatal and pre-

implantation genetic diagnosis (PGD) options. In the near future we expect that nearly all genetic

causes of OI will be elucidated. At that point and even now, the real challenge in OI research lies in

the development of treatment for both dominant and recessive OI. To develop possible therapies for

recessive OI, knowledge of the specific role of the proteins encoded by the newly discovered recessive

OI-related genes in the pathogenesis of OI is crucial.

Current treatment options (conservative and surgical treatment excluded) for OI are limited. For

dominant OI and probably also recessive OI, bisphosphonate therapy aimed at inhibiting bone turn-

over, is the main pharmacological therapy. However, it is uncertain whether the increase in bone mineral

density (BMD) that is observed, also results in fracture reduction and functional improvement [Philippi

et al., 2009]. Growth hormone therapy for short stature in OI type III and IV is under investigation [Marini

et al., 2010a].

It has been stated that the combination of stem cells with gene therapy offers the best approach for

treating OI [Niyibizi & Li, 2009]. Gene therapy for dominant OI is currently limited to in vitro studies and is

directed towards avoiding a dominant-negative effect by decreasing or silencing of the allele containing

the causative variant (antisense suppression therapy), which would transform a severe type of OI into

a mild OI type [Monti et al., 2010]. Clinical trials with bone marrow transplantation and mesenchymal

stem cell transplantation have been performed in a few individuals affected with severe dominant OI

[Horwitz et al., 1999; Horwitz et al., 2001; Horwitz et al., 2002]. Some improvement was observed but the

engraftment of donor cells in bone tissue appeared very low. Because of the inconclusive data of whole

marrow/mesenchymal stem cell transplation in clinical trials, it was decided to perform further research

in animal models [Niyibizi & Li, 2009].

In 2009 future perspectives for OI treatment were described by Niyibizi and Li consisting of (i)

development mechanisms of fibroblast reprogramming to generate embryonic-like stem cells (induced

Pluripotent Stem cells (iPS)) (ii) improve gene targeting to eliminate mutant alleles in stem cells, (iii)

improvements in designing novel methods to target stem cells to musculoskeletal system with high

efficiency [Niyibizi & Li., 2009].

Recently it has been discovered that a somatic cell can be reprogrammed to an alternative differentiated

Dijk_Opmaak.indd 167 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 84 T1 - Black CyanMagentaYellow

Page 169: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter V168

fate without first becoming a stem/progenitor cell [Masaki et al., 2010]. It appears now even possible

to dedifferentiate fibroblasts and redifferentiate them to osteoblasts (personal communication), which

means that prospects (i) and (iii) become more realistic possibilities. Various murine models (oim [Saban

et al., 1996], brtl IV [Kozloff et al., 2004], crtap-/- [Morello et al., 2006], Mov-13 [Bonadio et al., 1990],

OASIS-/- [Sekiya et al., 2010]) have been developed, which can be used for the purpose of investigating

gene therapy in OI (ii). However, the various antisense techniques (short oligonucleotides [Wang et al.,

1996], ribozymes [Grassi et al., 1997], silencing RNA [Millington-Ward et al., 2004]) all lack true specificity

against the mutant transcript. Another problem is stability of the antisense molecules apart from specific

problems which each technique. As such, antisense suppression therapy is currently limited to in vitro

studies [Monti et al., 2010].

In conclusion, for dominant and recessive OI, the currently most promising therapeutic approach would

be to transplant patient fibroblasts dedifferentiated and redifferentiated into osteoblast without the

genetic defect(s). In dominant OI, the main hurdle to overcome appears to be the elimination of mutant

alleles in the cells. Recessive OI may be more amenable to gene therapy because defective molecules

are not present in the matrix. However, no results from in vitro or in vivo studies are currently available.

More research is needed.

Dijk_Opmaak.indd 168 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 84 T1 - Black CyanMagentaYellow

Page 170: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Summary 169

SuMMAry

Chapter I-I describes the current knowledge concerning Osteogenesis Imperfecta (OI). OI is

characterized by bone fragility leading to fractures. Because of the clinical and supposed genetic

(based on pedigree findings) heterogeneity of OI, a classification in four types was proposed in 1979

by David Sillence. This classification consisted of OI type I (mild, autosomal dominant), type II (perinatal

lethal, autosomal recessive), type III (severe deforming, autosomal recessive) and type IV (moderately

deforming, autosomal dominant). After the discovery of heterozygous mutations in the COL1A1/2 genes

in all OI types, OI was considered to be an autosomal dominant disease and the Sillence classification

was used for clinical/radiological classification only. In 2004 and 2007 the Sillence classification was

expanded with OI types V-VIII because of distinct clinical features and/or different causative gene

mutations since in 2006 and 2007, recessive causative variants in CRTAP and LEPRE1 had been discovered

to cause OI as well. To date, 8 genes are known to cause recessive OI type II-B, III ,IV or Bruck syndrome (OI

with congentital contractures of the large joints) namely CRTAP, FKBP10, LEPRE1, PLOD2, PPIB, SERPINF1,

SERPINH1, SP7. Only 2 of these 8 genes (SP7 and SERPINF1) appear not to be involved in the collagen type

I biosynthesis. The increased knowledge concerning the genetic causes of OI changed in particular the

laboratory diagnosis of OI, which has now sequencing of the COL1A1/2 genes as a starting point. In the

past, the diagnostic flow started with collagen electrophoresis. With use of this technique collagen type

I production by dermal fibroblasts could be analyzed. In OI type I collagen type I production appeared

decreased and in the other OI types production of abnormal collagen type I occurred. Collagen

elctrophoresis still is important in certain selected cases but it is to be expected that it will be used less

frequently. The treatment of OI has not been influenced much by the discovery of genetic heterogeneity

in OI and has a multidiscipinary character. Further investigations of therapeutic approaches such as

bisphosphonates, growth hormone therapy, gene therapy and stem cell therapy are essential.

Chapter II-1 described for the first time in the literature, deletions of the complete COL1A1 allele in four

families with mild OI and no other phenotypic abnormalities. These findings underline the importance

of MLPA analysis of the COL1A1 gene in cases of suspected OI type I with no detectable mutation.

In Chapter III-I five families are described with novel causative variants in CRTAP encoding cartilage

associated protein (CRTAP). It was concluded that it appeared not possible to discriminate OI caused by

recessive variants in CRTAP from OI caused by dominant COL1A1/2 causative variants based on clinical/

radiological, histological or biochemical (collagen electrophoresis) characteristics.

Chapter III-2 is the first report in the literatur on PPIB mutations as a cause of OI type II-B/III as is seen with

causative variants in COL1A1/2, CRTAP and LEPRE1. Deficiency of CRTAP or prolyl 3-hydroxylase 1(P3H1)

had been reported in autosomal recessive lethal/severe OI. CRTAP, P3H1 and Cyclophylin B(CyPB) form

an intracellular collagen-modifying complex with probably multiple functions.

Chapter III-3 reports a large consanguineous Turkish family in which multiple individuals are affected

with autosomal recessive lethal or severe osteogenesis imperfecta (OI) due to a novel homozygous

LEPRE1 mutation. In one affected individual histological studies of bone tissue were performed, which

may indicate that the histology of LEPRE1 associated OI is indistinguishable from COL1A1/2, CRTAP and

PPIB related OI.

Dijk_Opmaak.indd 169 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 85 T1 - Black CyanMagentaYellow

Page 171: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Chapter V170

Chapter III-4 is a report of the first Indonesian patient with the very rare Bruck syndrome, due to a novel

homozygous causative FKBP10 variant .

In Chapter IV-1 a revised classification of OI is proposed with continued use of the Sillence criteria I,

II-A, II-B, II-C, III , IV, V and VI for clinical and radiological classification of OI with additional mentioning of

the causative mutated gene. Addition of a new type only because it has a different genetic cause (for

example OI type VII and VIII) is discouraged.

Chapter IV-2 summarizes the conclusions of the Best Practice meeting in Amsterdam concerning the

laboratory diagnosis of OI. Due to the genetic heterogeneity in OI, it was agreed to a new diagnostic

flow with DNA analysis (sequencing) of the COL1A1/2 genes as a starting point as opposed to protein

analysis.

Chapter V focuses on the future perspectives in OI research. Approximately 95-96% of genetic causes

of OI are discovered and with the next generation sequencing techniques it is to be expected that

other rare causes of OI will be discovered. This will shift the challenge in OI research to development

of therapy for OI. The combination of osteoblast transplantation (by de- and redifferentiating patient’s

own fibroblasts) and gene therapy may offer the best therapy for OI. More research into that particular

area is needed.

Dijk_Opmaak.indd 170 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 85 T1 - Black CyanMagentaYellow

Page 172: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Samenvatting voor de geïnteresseerde leek 171

SAMENVATTING VOOr DE GEINTErESSEErDE LEEK

De erfelijke ziekte Osteogenesis Imperfecta (OI) wordt gekenmerkt door botbreuken bij geen of geringe

aanleiding. De ernst van het ziektebeeld wisselt sterk. De Australische arts David Sillence stelde in 1979

daarom een onderverdeling van OI in vier typen voor t.w. OI type I (mild*), OI type II (letaal), OI type III

(ernstig deformerend) en OI type IV (matig deformerend). In hoofdstuk I is de huidige kennis over OI

samengevat en zijn voorbeelden van patiënten met type I, II, III of IV opgenomen.

Sillence was vanwege de verschillende overervingsvormen in de families die hij had onderzocht, van

mening dat er meerdere genetische oorzaken van OI waren. In 1983 werd echter duidelijk dat in vrijwel

alle OI typen sprake was van te weinig of afwijkend collageen type I door een dominante mutatie in het

COL1A1 of COL1A2 gen. Zo beschrijft hoofdstuk II een zeldzame mutatie namelijk het missen van een

volledig COL1A1 gen.

In enkele families met OI werd geen genetische oorzaak gevonden. De afgelopen vijf jaar zijn in zulke

families recessieve mutaties in 8 genen als oorzaak van OI ontdekt waarbij mutaties in 2 genen Bruck

syndroom veroorzaken (OI met aangeboren dwangstand van de grote gewrichten). De eiwitten

waarvoor deze genen coderen, hebben vrijwel allemaal een rol in het bewerkingsproces van collageen

type I. Hoofstuk III beschrijft families met mutaties in 4 van deze 8 genen (CRTAP (III-1), PPIB (III-2),

LEPRE1 (III-3), FKBP10 (III-4)) waarbij onze onderzoeksgroep als eerste in de wereld mutaties in het PPIB

gen ontdekte als oorzaak van OI (II-3).

Het feit dat OI wordt veroorzaakt door meerdere genetische oorzaken heeft invloed op het stellen van

de diagnose OI in de kliniek en in het laboratorium. In hoofdstuk IV-1 wordt voorgesteld de klinische/

radiologische classificatie van OI in 4 typen te handhaven (met ook de in 2004 toegevoegde zeer

zeldzame OI type V en VI vanwege onderscheidende kenmerken) en daarbij het oorzakelijke gendefect te

noemen dus bijvoorbeeld “OI type II, PPIB gerelateerd”. In hoofdstuk IV-2 worden richtlijnen beschreven

die door een groep internationale experts tijdens een bijeenkomst in Amsterdam zijn opgesteld. Een

belangrijke conclusie is dat voortaan gestart gaat worden met het analyseren van de COL1A1/2 genen

die in 90% van de mensen met OI, de oorzaak zijn.

Hoofdstuk V-1 stelt dat de komst van nieuwe laboratoriumtechnieken ertoe zal bijdragen dat de nog

onontdekte genetische oorzaken van OI snel bekend zullen worden. De uitdaging in OI onderzoek zal

dan waarschijnlijk verschuiven naar de behandeling van OI. De beste behandeling zou een transplantatie

zijn van botvormende cellen (osteoblasten) van de patiënt zonder genetisch(e) defect(en). Er is echter

nog veel onderzoek nodig om een dergelijke behandeling te kunnen gaan toepassen.

Dijk_Opmaak.indd 171 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 86 T1 - Black CyanMagentaYellow

Page 173: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 172 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 86 T1 - Black CyanMagentaYellow

Page 174: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

List of publications 173

LIST Of PubLICATIONS

2006

van Dijk fS, Knuistingh Neven A, Eekhof JAH. Snurken. Huisarts & wetenschap 2006; 49(2): 98-101

(written during internships)

2007

van Dijk fS & Knuistingh Neven A. Snurken in “Kleine kwalen in de huisartspraktijk” JAH Eekhof, A.

Knuistingh Neven, Th.J.M. Verheij. Maarssen: Elsevier gezondheidzorg, 2007, 479-482

van Dijk fS. Trommelvliesperforatie in “Kleine kwalen in de huisartspraktijk”JAH Eekhof, A. Knuistingh

Neven, Th. J.M. Verheij. Maarssen: Elsevier gezondheidszorg, 2007, 347-351

Te Veldhuis EC, Te Veldhuis AH, van Dijk fS, Kwee ML, van Hagen JM, Baart JA, van der Waal I. Rendu-

Osler-Weber disease: update of medical and dental considerations. Oral surg Oral Med. Oral Pathol. Oral

Radiol. Endod 2008; 105(2):38-41

Sapp JC, Turner JT, van de Kamp JM, van Dijk fS, Lowry RB, Biesecker LG. Newly delineated syndrome

of congenital lipomatous overgrowth, vascular malformations and epidermal nevi (CLOVE syndrome) in

seven patients. Am. J. Med. Genet. A. 2007; 143(24): 2944-58

2008

van Dijk fS, Meijers-Heijboer H, Pals G. Angiotensin II blockade Marfan’s syndrome. N Engl. J. Med. 2008;

359(16):1733

2009

van Dijk fS, Pals G , Van Rijn RR, Nikkels PGJ, Cobben JM. Classification of Osteogenesis Imperfecta

Revisited. A Mini-Review. European Journal of Medical Genetics. 2010 Jan-Feb;53(1):1-5

van Dijk fS , Nesbitt IM , Zwikstra EH, Nikkels PGJ, Piersma SR, Fratantoni SA, Jimenez CR , Huizer M,

Morsman AC, Cobben JM , van Roij MHH , Elting MW, Verbeke JIML, Wijnaendts LC , Shaw NJ , Högler

W, McKeown C , Sistermans EA, Dalton A , Meijers-Heijboer H , Pals G. PPIB mutations cause severe

Osteogenesis Imperfecta. American Journal of Human Genetics, 2009;85(4):521-7.

van Dijk fS, Nesbitt IM, Nikkels PGJ, Dalton A, Bongers EM, Kamp van de JM, Hilhorst-Hofstee Y, Den

Hollander NS, Lachmeijer AMA, Marcelis CL, Tan-Sindhunata MB, van Rijn RR, Meijers-Heijboer EJ, Cobben

JM, Pals G. CRTAP mutations in lethal and severe osteogenesis imperfecta: the importance of combining

biochemical and molecular genetic analysis. Eur J Hum Genet. 2009 Dec;17(12):1560-9.

van Dijk fS, Hamel BC, Mulder BJM, Timmermans J, Pals G, Cobben JM. Compound heterozygous

Marfan syndrome. Eur. J. Med. Genet. 2009 Jan-Febr; 52(1):1-5

Hornemann T, Penno K, Stephane R, Nicholson G, van Dijk fS, Rotthier A, Timmerman V, von Eckardstein

A. A critical comparison of mutations in hereditary sensory and autonomic neuropathy type I reveals

that the G387A mutation in the Serine-Palmitoyltransferase is not disease associated. Neurogenetics 2009;

10(2): 135-43

Dijk_Opmaak.indd 173 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 87 T1 - Black CyanMagentaYellow

Page 175: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

List of publications 174

2010

van Dijk fS, Huizer M, Kariminejad A, Marcelis CL, Plomp AS, Terhal PA , Meijers-Heijboer H, Weiss MM,

van Rijn RR , Cobben JM , Pals G. Complete COL1A1 allele deletions in Osteogenesis Imperfecta. Genet.

Med. 2010; 12:736-741.

van Dijk fS, Nikkels PG, den Hollander NS, Nesbitt IM, van Rijn RR, Cobben JM, Pals G. Lethal/ Severe

Osteogenesis Imperfecta in a Large Family: A Novel Homozygous LEPRE1 Mutation and Bone Histological

Findings. Pediatr Dev Pathol. 2011 May-Jun;14(3): 228-34.

van Dijk fS, Cobben JM, Pals G. Osteogenesis imperfecta, normal collagen folding, and lack of cyclophilin

B. N Engl J Med. 2010 May 20;362(20):1940-1

2011

van Dijk fS, van Thuijl HF, Wermeskerken A, van Rijn RR, Cobben JM. Solitary median maxillary central

incisor and congenital nasal pyriform aperture stenosis combined with asymmetric crying facies and

postaxial lower limb reduction defects: A unique combination of features. Eur J Med Genet. 2011;54: 284-6

van Dijk fS, Byers PH, Dalgleish R, Malfait F, Maugeri A, Rohrbach M, Symoens S, Sistermans EA, Pals G.

EMQN best practice guidelines for the laboratory diagnosis of Osteogenesis Imperfecta. Eur J Hum Genet.

2011 Aug 10. [Epub ahead of print]

van Dijk fS, Cobben JM, Kariminejad A, Maugeri A, Nikkels PGJ, van Rijn RR, Pals G. Osteogenesis

Imperfecta: a review with clinical examples. Accepted for publication in Molecular Syndromology.

Setijowati ED, van Dijk fS, Cobben JM, van Rijn RR, Sistermans EA, Faradz SMG, Kawiyana S, Pals G. A

novel homozygous 5 bp deletion in FKBP10 causes clinically Bruck syndrome in an Indonesian patient.

Accepted for publication in Eur J Med Genet.

Dijk_Opmaak.indd 174 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 87 T1 - Black CyanMagentaYellow

Page 176: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dankwoord / Acknowledgements 175

DANKWOOrD/ACKNOWLEDGEMENTS

Ik schrijf deze eerste zinnen van het dankwoord op een winderige avond op de laatste dag van maart

2011.Vandaag is mijn proefschrift in mijn hoofd af en daarom begin ik met datgene wat men normaliter

als eerste leest maar als laatste schrijft: het dankwoord.

Prof. Dr. E.J. Meijers-Heijboer (promotor), beste Hanne. Jij zag wat ik het meeste nodig had t.w. vrijheid

en je liet me de vrije teugel om “mijn ding te gaan doen”. Het resultaat is dit proefschrift maar weet wel:

ik ben nog niet klaar!

Dr. G. Pals (co-promotor), beste Gerard. Je hebt enorm veel ideeën en ervaring op het gebied van erfelijke

bindweefselaandoeningen. Samen hebben we een aantal van deze ideeën uitgewerkt en ik ben heel blij

dat jij me hebt begeleid. Ik hoop dat we nog lang kunnen blijven samenwerken!

Dr. P.J.G. Nikkels, beste Peter. In 2008 hadden wij voor het eerst contact, je hulp en inzichten als

kinderpatholoog gespecialiseerd in skeletaandoeningen zijn zeer waardevol, bedankt hiervoor!

Dr. E.A. Sistermans, beste Erik. Als hoofd van het DNA laboratorium van het VUmc heb jij me op belangrijke

momenten advies gegeven. Een belangrijk advies was om nu vaart te zetten achter het schrijven van

mijn proefschrift en die raad heb ik opgevolgd zoals je ziet!

Drs. A. Kariminejad. Dear Ariana, we met in 2009 in Teheran, Iran. As head of the clinical genetics

department in Teheran, you have sent DNA including clinical pictures and radiographs of interesting

patients to our laboratory. It is my sincere hope that we can maintain and extend our collaboration.

Dr. R.R. van Rijn, beste Rick. In 2008 ontmoette ik je op de afdeling kinderradiologie in het AMC. Je hebt

me erg geholpen met de radiologische beschrijvingen van de patiënten beschreven in onze publicaties,

bedankt!

Prof. Dr. P. Coucke, Prof. Dr. V. Everts, Prof. Dr. G. Mortier , Dr. P.G.J. Nikkels, Dr. E.A. Sistermans. Ik wil u allen

hartelijk bedanken voor het beoordelen van mijn proefschrift en de bereidheid om zitting te nemen in

de promotiecommissie.

Collega’s van de afdeling klinische genetica, polikliniek en laboratorium, ik voel me thuis bij jullie! Dank

voor jullie hulp bij mijn pogingen om onderzoek en patiëntenzorg zo goed mogelijk te combineren.

Jiddeke, vandaag ben je mijn paranimf. Behalve een hele goede vriendin ben je ook een supervisor

waarvan ik heb geleerd om me vast te bijten in een casus en alles tot op de bodem uit te zoeken. Dat

heeft me erg geholpen in mijn onderzoek.

Leila, jij bent vandaag ook mijn paranimf en je kent me al vanuit onze Leidse studententijd. Ik ben heel

blij dat jij vandaag bij me bent

Elsbeth, dank je wel voor je luisterend oor en je adviezen gedurende de afgelopen jaren, voor mij is dit

zeer waardevol.

Dijk_Opmaak.indd 175 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 88 T1 - Black CyanMagentaYellow

Page 177: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dankwoord / Acknowledgements 176

Sjanette, ook al heb ik je een jaar niet gesproken, we pakken altijd gewoon de draad weer op en op

belangrijke momenten ben je er, dank je wel.

Gea, je bent mijn kamergenoot en wonderlijk genoeg mijn klasgenootje van de basisschool in

Scherpenzeel en bewoner van ons oude huis in Scherpenzeel. In 2006 werd jij mijn collega en sindsdien

zitten we bij elkaar op de kamer. We steunen elkaar in barre tijden en heel belangrijk: jouw enorme

hoeveelheid proviand heeft me al van menig hypo gered!

Leden van Cordial Kallista en co-schap groepje 80, alhoewel jullie niet direct iets met dit proefschrift

te maken hebben, wil ik jullie toch noemen omdat ik met jullie fijne en ook leerzame momenten heb

beleefd.

Truus, om op mijn promotie te kunnen zijn, regelde je een vervroegde terugreis uit Australië, ik ben blij

dat je er bent!

Teije, je hebt al vaker gevraagd of ik eens een artikel wilde opsturen en ik heb het telkens beloofd maar

ik ben het elke keer toch vergeten. Je hebt even geduld moeten hebben maar nu is er mijn proefschrift

en daar staan de belangrijkste artikelen in!

Annemarie, mijn grote zus, mede dankzij jou is het gelukt om geneeskunde te gaan studeren, dat zal ik

nooit vergeten.

Lieve Papa en Mama, ik ben jullie derde en laatste kind maar niet altijd het makkelijkste….Dank jullie wel

voor alle support en liefde die ik krijg. Ik hou van jullie!

Jan Maarten, mijn grootste fan (en soms ook criticus) maar bovenal mijn lief. Wat een geluk dat ik jou ben

tegengekomen in mijn leven.

Mirle, kleine zonnestraal. Het is bijzonder hoeveel mensen jij blij kunt maken, simpelweg door er te zijn.

Ik hou van je!

Dijk_Opmaak.indd 176 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 88 T1 - Black CyanMagentaYellow

Page 178: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Curriculum Vitae 177

CurrICuLuM VITAE

Fleur Stephanie van Dijk was born on 8-7-1980, in Amersfoort, the Netherlands. From 1992-1998 she

attended “t Revius Lyceum” in Doorn. From 1998-2005 she studied Medicine at the University of Leiden.

In March 2005 she started a Volunteer internship in Clinical Genetics (keuze-coschap) at the department

of Clinical Genetics, Leiden University Medical Center. During this Volunteer internship her ambition to

become a clinical geneticist was raised. She commenced a residency in Clinical Genetics (AIOS) at the

VU medical center in Amsterdam under supervision of Prof. dr. Hanne Meijers-Heijboer combined with

research under supervision of Dr. Gerard Pals, specialized in laboratory diagnosis of various inherited

connective tissue disorders (July 2006-November 2012). This has resulted among others in the current

thesis. Together with Jan Maarten Cobben she has one daughter Mirle (2010).

Dijk_Opmaak.indd 177 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Front - 89 T1 - Black CyanMagentaYellow

Page 179: Osteogenesis Imperfecta; Clinical and Genetic Heterogeneity dissertation.pdf · Osteogenesis Imperfecta (OI) is characterized by susceptibility to bone fractures, with a severity

Dijk_Opmaak.indd 178 17-10-11 16:55

Dijk_PROEF_16x24 (all).ps Back - 89 T1 - Black CyanMagentaYellow