324
Views
10
CrossRef citations to date
0
Altmetric
Review

Animal models of osteogenesis imperfecta: applications in clinical research

, , &
Pages 41-55 | Published online: 27 Sep 2016

Abstract

Osteogenesis imperfecta (OI), commonly known as brittle bone disease, is a genetic disease characterized by extreme bone fragility and consequent skeletal deformities. This connective tissue disorder is caused by mutations in the quality and quantity of the collagen that in turn affect the overall mechanical integrity of the bone, increasing its vulnerability to fracture. Animal models of the disease have played a critical role in the understanding of the pathology and causes of OI and in the investigation of a broad range of clinical therapies for the disease. Currently, at least 20 animal models have been officially recognized to represent the phenotype and biochemistry of the 17 different types of OI in humans. These include mice, dogs, and fish. Here, we describe each of the animal models and the type of OI they represent, and present their application in clinical research for treatments of OI, such as drug therapies (ie, bisphosphonates and sclerostin) and mechanical (ie, vibrational) loading. In the future, different dosages and lengths of treatment need to be further investigated on different animal models of OI using potentially promising treatments, such as cellular and chaperone therapies. A combination of therapies may also offer a viable treatment regime to improve bone quality and reduce fragility in animals before being introduced into clinical trials for OI patients.

Introduction

Osteogenesis imperfecta (OI), commonly known as brittle bone disease, is a genetic disorder characterized by extreme bone fragility, even if only exposed to mild trauma. The earliest known case of OI to occur in a human was determined by Peter Gray in 1969 in his analysis of bones of an approximately 3,000-year-old mummified infant.Citation1 However, it was only in 1788 that OI became recognized as a human disease, when it was given its first true pathological description.Citation1 By 1979, researchers had classified OI into four different types (I–IV), and identified OI I and IV to be caused by an autosomal-dominant gene and OI II and III to be caused by an autosomal-recessive gene, but the exact gene sequences responsible had not yet been determined.Citation2 Advances in the field of genetics have made it easier to characterize genetic sequences, and the genes linked to the development of OI have since been identified for all of the four originally defined types of OI, in addition to several of the more recently classified types.Citation3 This has been a major step in the advancement of clinical treatments for OI, as it is easier to identify and generate appropriate animal models for OI research by targeting specific genetic defects.

OI types I–IV

Characteristics in humans

When OI was first categorized into types I–IV, the description that was given to type I was “dominantly inherited OI with blue sclerae”, and included such characteristics as having a history of broken bones from a very young age, bowing of long bones in lower extremities, bruising easily, and hearing impairment.Citation2 Additional features observed in persons with OI I include being of normal height or slightly short in stature and not suffering from dentinogenesis imperfecta (DI).Citation4 OI II was labeled “lethal perinatal OI with radiographically crumpled femora and beaded ribs”, and was characterized as being a fatal condition either in utero or shortly after birth.Citation2 Features observed in individuals with OI II include multiple fractures at birth, obvious deformities, flattened long bones, and dark sclerae.Citation3 OI III, referred to as “progressively deforming OI with normal sclerae”, was characterized as having normal sclerae, distorted and fragile bones that progressively degenerated over time, ligament hypermobility, and a predisposition to DI.Citation2 Also, individuals with OI III tend to be very short in stature and have a triangular face and severe scoliosis.Citation3 Finally, OI IV was originally classified as “dominantly inherited OI with normal sclerae”, and had similar defining characteristics to OI I, but with normal sclerae, reduced likelihood of spontaneous bone fractures, and no hearing loss.Citation2

Therefore, in terms of severity, OI I–IV are ordered II>III>IV>I. Although OI I–IV are vastly different in how they affect humans, they all have a similar genetic basis.Citation4 Specifically, OI I is caused by a premature stop codon in COL1A1 and OI II–IV are caused by glycine substitutions in either the pro-α1(I) or pro-α2(I) collagen chains of COL1A1 or COL1A2.Citation3 It has been determined that the phenotypes for OI I–IV depend on the location of the mutation in the COL1A1 and COL1A2 genes; however, several different mutations occurring at different points in the sequence can lead to the same phenotype of OI.Citation5

Animal models

Mov-13 mouse: OI I and II

The Mov-13 mouse was first created by blocking transcription of the COL1A1 gene by integrating the Moloney leukemia virus at the 5′ end of the gene in what was referred to as the Mov-13 locus,Citation6 later shown to be the murine equivalent to human Col1a1.Citation7 The Mov-13 mutation in mice results in a failure to produce type I collagen.Citation8Citation10 The homozygous (Mov-13−/−) form of this mutation is lethal to mice.Citation6,Citation7,Citation9 Therefore, the Mov-13−/− mouse is considered a model for OI II,Citation11 but unfortunately it is unable to duplicate properly the development seen in humans.Citation12

The heterozygous Mov-13 mutation (Mov-13/+) is associated with a reduced amount of collagen in soft connective tissue, increased hearing loss over time, and a substantial reduction in both mechanical and material properties of long bones similar to that seen in humans affected by OI I.Citation13 A disorganized cortical lamellar structure in Mov-13/+ mice has been observed,Citation12 with a 22%–25% reduction in tissue-bending strength due to disrupted damage accumulation mechanisms in Mov-13/+ cortical bone.Citation14 While Mov-13/+ exhibits a 50% decrease in type I collagen production, the collagen that is produced has normal amounts of both pro-α1(I) and pro-α2(I) chains,Citation13,Citation15 making Mov-13/+ mice a proper model for OI I in humans.Citation16

oim mouse: OI I and III

One animal model of OI not created through the process of genetic engineering is the OI murine (oim) model. (The other five naturally occurring mutations are the G208A Col1a1 Golden Retriever, Col1a2 frameshift Beagle, SerpinH1 Dachshund, Chi zebrafish, and Frf−/− zebrafish.) It was produced in the Jackson Laboratory (Bar Harbor, ME, USA) as a result of breeding C3H/HeJ and C57BL/6JLe mice.Citation12 The mice present a spontaneous mutation in type I collagen structure with a deficiency of the pro-α2(I) chain, and exhibit phenotypic and biochemical features typical of the most debilitating, nonlethal forms of OI.Citation12 Characteristics observed in the homozygous oim mice (oim/oim) include susceptibility to fractures from a very young age, severe osteopenia, cortical thinning, bowing of long bones, bone deformities, joint laxity, small body size, and kyphosis.Citation12 Their bones have altered collagen cross-links,Citation17 reduced mineral size and heterogeneity,Citation18 reduced lamellar structure,Citation19 and increased vascular and osteocyte lacunar density.Citation20 oim/oim bones have inferior mechanical properties compared to wild-type (+/+) counterparts.Citation17,Citation20Citation22 Reduced fracture toughness, with a flat crack path and quick propagation, has been observed in these bones.Citation17 Similar characteristics have also been observed in OI III in humans,Citation23Citation26 for which the oim/oim mouse is a model.Citation12 Compared to oim/oim, the heterozygous oim mouse (oim/+) does not experience spontaneous fractures, but does have bone material properties that lie in between those found in oim/oim and wild-type mice, and is thus considered a model of OI I in humans.Citation27

Not only bones but also tendons are weaker in the oim mouse, as they are composed of collagen type I. Tensile testing performed on the tail tendons of oim/oim and oim/+ mice revealed a twofold reduction in the ultimate tensile strain compared to wild-type mice.Citation28

G610C mouse: OI I and IV

In 2004, the Cre/Lox strategy, a method for genetically engineering animals for biomedical research, was combined with embryonic stem cells to mutate a section of murine Col1a2 in the stem cells, resulting in G610C-positive stem cells that were then injected into the mouse strain C57BL/6J (B6) blastocysts.Citation29 Mice in the founder generation that retained the neomycin-targeting vector were termed Neo+ mice, and offspring from founder-generation males with Cre recombinase-expressing females were called Neo mice.Citation29 The founder line of G610C mice was homozygous, and is now commonly known as G610C/G610CNeo+, whereas heterozygous mice are known as +/G610CNeo+.Citation11 The common name for the Neo mouse (+/G610CNeo−) is the Amish mouse, because it models a large human Amish population consisting of 64 individuals exhibiting OI I/IV in both genetic code and phenotypic expression.Citation11,Citation29 Phenotypic characteristics of the mutant mice include decreased body weight, bone mineral density, and bone volume.Citation29 Additionally, a four-point bending test of femora indicated a decrease in mechanical properties with reduced stiffness, postyield ultimate displacement, failure load, and energy to failure compared to wild type.Citation29 Therefore, G610C/G610CNeo+, +/G610CNeo+, and +/G610CNeo− are all acceptable models of OI IV, and +/G610CNeo+ is also considered a model for OI I.Citation11

G859C Col1a1 mouse: OI II

In 1988, a site-directed mutagenesis approach was used to prepare transgenic mice expressing a mutated COL1A1 gene that resulted in perinatal death.Citation11,Citation30 The mutation was introduced to COL1A1 at residue 859 in the triple helical domain of the gene.Citation11,Citation30 Inspection of fetuses’ radiographs showed short and wavy ribs, poor bone mineralization, underdeveloped skeletons, and pliable limbs.Citation11,Citation30 There were also large cavities noted in the bones of the mice, indicating that a high level of resorption had occurred.Citation11,Citation31 Due to the disease characteristics expressed in the G859C mutagenic mice, they have been accepted as a model for OI II.Citation11

Aga2 mouse: OI II and III

A strain of mutagenic mice produced according to the standards of the German Mouse Clinic (http://www.mouseclinic.de), a system for the phenotyping of mutant mouse lines,Citation32 was created using the Munich N-ethyl-N-nitrosourea mutagenesis protocol.Citation11,Citation33 This new line was called the Aga2 (abnormal gait 2) mouse, as the Aga2/+ mice that were first produced exhibited an abnormal gait as a result of excessive hind-limb deformity.Citation33 The heterozygous (Aga2/+) mice expressed a range of phenotypic variation from skeletal deformities, fractures, fragility, disorganized trabecular bone and collagen structure, and osteopenia to perinatal death.Citation33 Aga2/+ mice that did not survive were classified as phenotypically similar to OI II.Citation33 Homozygous (Aga2−/−) mice that were produced from inter se breeding of Aga2/+ males and females all died in utero around embryonic stage E9.5.Citation33 Although the original research published on the Aga2 mice did not say which types of OI the Aga2/+ mice that survived to adulthood and the Aga2−/− mice were similar to, they have since been considered to be appropriate models for OI III and II, respectively.Citation11 However, it needs to be noted that although the mutation affects Col1a1, there has not yet been an equivalent genetic mutation identified in humans.Citation11

BrtlII and BrtlIV mice: OI II and IV

In 1999, two phenotypes of mice were created using the Cre/Lox system.Citation34 Initially, the researchers’ intent was to use a recombination strategy to substitute a cysteine for glycine at position 349 in type I collagen and create a mouse model of OI I, but the floxed stop-cassette placement instead led to the expression of an allele that was supposed to be suppressed.Citation34 Two chimeric males were obtained from this process, and after being mated with wild-type females, the resulting offspring that expressed the heterozygous mutation were described as having similar weight and size as healthy littermates, but they all died within hours of birth, and X-rays and skeletal staining revealed that they had multiple rib fractures, short vertebral bodies, and poor skull mineralization.Citation34 Due to the fact that the phenotype expressed in these mice was similar to OI II in humans, this initial strain of mice was named Brittle II (BrtlII).Citation34

To remove the stop cassette that led to the BrtlII phenotype, the two chimeric males were mated with EIIa-Cre mice, a strain of mice carrying a transgene mediated by the adenovirus EIIa promoter to target Cre-recombinase expression, and the resulting male founder-generation offspring that expressed the heterozygous mutation (F1-mosaic) were mated with wild-type CD1 and C3H/HeJ female mice to produce an F2 generation.Citation34 There was large variability in phenotype noted in the mutant F2 mice, ranging from perinatal death to long-term survival, and their bones exhibited deformities, fragility, osteopenia, and a disorganized trabecular structure.Citation34 These heterozygous mutant mice expressed the mutation that had been desired in the original recombination strategy, and were termed BrtlIV mice.Citation34 BrtlIV/+ mice have geometrically smaller bones with reduced cortical width and lower trabecular thickness and bone-volume density compared to wild-type mice, increasing their vulnerability to fracture.Citation35 Femoral elastic modulus, strength, and stiffness increase after puberty independently of the whole-bone geometry in BrtlIV/+ mice with values comparable with wild-type mice.Citation16 This strengthening is due to an increase in the material properties of the extracellular matrix without any typical corresponding structural adaptations, which means that BrtlIV/+ mouse femora still have a geometric organization that is less resistant to applied loads.Citation16 Mice also showed decreased molar volume and mineralized tissue volume in the teeth, although enamel properties were normal.Citation36 The BrtlIV mice are heterozygous mice (BrtlIV/+);Citation34 however, there is also a homozygous (BrtlIV/BrtlIV) type that exhibits a much milder form of OI than BrtlIV/+.Citation11,Citation37

Human COL1A1-minigene mouse: OI II–IV

A transgenic animal model of OI was produced by generating a minigene construct composed of sequences extracted from human COL1A1 that were missing a central region containing 41 exons,Citation38,Citation39 encompassing exons 6–46.Citation40 The construct resulted in shortened pro-α1(I) chains that ultimately prevented folding of the protein into a triple helix, leading to a lethal form of OI.Citation38,Citation39 Without proper folding, degradation of the three chains through procollagen suicide occurs, and leads to the depletion of normal pro-α(I) chains.Citation39,Citation41 Mice expressing high levels of the minigene exhibit a perinatal mortality rate up to 90%.Citation41 However, mice that only express a moderate minigene level tend to have a less severe phenotype, including low bone mineral density and collagen content, and a susceptibility to fractures.Citation38 Therefore, human COL1A1-minigene mice can be considered a model for OI II–IV.Citation11,Citation38

G208A Col1a1 Golden Retriever: OI III

Another naturally occurring animal model of OI is the G208A Col1a1 Golden Retriever, which was observed in a 12-week-old puppy that presented with DI and multiple bone fractures, including ribs and long bones.Citation11,Citation42,Citation43 Samples of collagen type I taken from the puppy’s skin showed that the collagen was not functioning normally.Citation42,Citation43 After it was determined that the puppy had naturally inherited a canine form of OI, samples of COL1A1 from both a healthy canine and the puppy with OI were sequenced and compared, and it was determined that the puppy’s OI was due to a substitution of alanine for glycine at amino acid position 208.Citation43 The location of the mutation is the namesake for this model, and because it is a naturally occurring mutation it is likely that the mutation is a heritable trait and could be reproduced in other dogs. However, the puppy used in these studies remains the only published case of this mutation having occurred in a canine.

Col1a2 frameshift Beagle: OI III

The second naturally occurring canine model of OI was identified with the discovery of a heterozygous COL1A2 frameshift mutation in the complementary DNA (cDNA) of a proband Beagle (CU3) exhibiting a history of multiple long-bone fractures and overhydroxylated type I collagen.Citation42,Citation44 Four nucleotides (3991–3994) were deleted and replaced, with nine in the COL1A2 region responsible for coding for the C-propeptide of the pro-α2(I) chain.Citation44 C-propeptides are essential for proper triple-helix formation, and mutations can lead to altered COL1A1 structure.Citation45 Also, the 30-amino acid code for human and canine pro-α2(I) sequences normally has 100% identity, which suggests the CU3 Beagle is a good model for human OI III.Citation46

Med−/− zebrafish: OI I–IV

In 1996 a homozygous mutant zebrafish was characterized as having curtain-like waves in its fin epithelium and was termed “microwaved” (Med−/−).Citation47 Fish with this mutation also exhibit delayed bone ossification, undulation of the larval fin, and severe reduction in bone density.Citation48 One of the constituents of zebrafish bone, actinotrichia, is partially formed by type I collagen, and Col1a1 has been identified in actinotrichia-forming cells.Citation49 Additionally, gene sequencing has been confirmed that Med−/− zebrafish exhibit an alanine-for-glycine substitution at position 888 in their cDNA, which is predicted to result in the substitution of a glutamic acid for a lysine.Citation48 This illustrates the validity of Med zebrafish as a model for OI I–IV.Citation48

Chi zebrafish: OI I–IV

Another zebrafish that serves as a model for OI is the Chihuahua (Chi) mutant. This mutation can occur either in heterozygous (Chi/+) or homozygous (Chi/Chi) form, but there has not been a phenotypic difference noted between the two.Citation50 It has, however, been determined that the mutation causes defective bone growth, with characteristics similar to those found in humans with OI.Citation3,Citation50 Radiographs show distorted bones with irregular radiodensity as early as 1 month of age.Citation50 The Chi mutation occurs at position 2207 in the cDNA, where the wild-type sequence is unaltered, but there is a base-pair change that causes a missense mutation and changes the encoded amino acid from glycine to aspartate.Citation50 The sequence of zebrafish COL1A1 is approximately 76% identical to the human gene, making it a good model for OI caused by COL1A1 mutations.Citation3,Citation50

OI types V–VIII

Characteristics in humans

In 2004, it was suggested that three new phenotypes be added to the classification of OI and referred to as OI V–VII.Citation4 A few years later, another phenotype was proposed and classified as OI VIII.Citation51 The features of the disorder classified as OI V include being slightly to moderately short in stature, with a dislocated head of the radius, a mineralized interosseous membrane, and normal sclerae.Citation4 Characteristics of OI VI include moderately short stature, presence of scoliosis, accumulation of osteoid in bone as well as a fish-scale bone-lamellation pattern, normal sclerae, and absence of DI.Citation4 Traits described for OI VII include slightly short stature with shortened humeri and femora, decreased angle between the head and shaft of the femora, normal sclerae, and no presence of DI.Citation4 Finally, the characteristics of the severe-to-fatal disease known as OI VIII include normal sclerae, bulbous metaphyses, and extremely short stature with very low bone mineralization.Citation51

Genetic causes of OI V–VII were unknown at the time it was proposed that they be added under the classification of OI, but have since been identified. In 2013, it was suggested that the expression of OI V is caused by a mutation in the IFITM5 gene,Citation52 which encodes a transmembrane protein important to osteoblast expression.Citation53 A mutation in the SER-PINF1 gene was identified as the cause of OI VI.Citation54 A genetic link between the decreased expression of CRTAP and the presence of OI VII had been discovered.Citation55 A genetic mutation caused by defects in the P3H1/LEPRE1 gene resulted in an osteopathic disease with OI-type characteristics,Citation51 later classified as OI VIII.Citation56

Animal models

Ifitm5 transgenic mouse: OIV

A mouse model with IFITM5 mutation was very recently created by first placing murine Ifitm5 cDNA under a Col1a1 2.3 kB promoter, and then using mutagenesis to achieve the correct mutation (c.-14C>T) that occurs in humans.Citation53 Mice that inherited mutant Ifitm5 had severe skeletal deformities, as well as perinatal death.Citation53 Additionally, the limbs of the mice lacked growth abnormalities, but they were facing downward at birth, and skeletal samples taken at E15.5 and E18.5 showed a consistent delay in mineralization. At E18.5 there was also the presence of forelimb and hind-limb fractures in utero, as well as extreme deformities of the rib cage.Citation53

Pedf−/− mouse: OI VI

Mice genetically modified to be deficient in Pedf were first created by replacing exons 3–6 of SERPINF1 with a cassette comprised of an internal ribosomal entry site, β-galactosidase genes, and neomycin-resistance genes.Citation57 After it was discovered that OI VI in humans is caused by mutations in SER-PINF1, this procedure was replicated to produce Pedf−/− mice for the purpose of studying the role of PEDF in bone and other tissues.Citation58 Bending tests performed on femora from wild-type and Pedf−/− mice indicated that while the ultimate force applied to both groups was about the same, the ultimate displacement in the Pedf−/− bone was approximately half of that in wild-type bone, with the energy of failure being 25% lower in the Pedf−/− mice.Citation58 Through this study, it was determined that Pedf−/− mice are a suitable model for OI VI.Citation58

Crtap−/− mouse: OI VII

Around the same time that the genetic cause of OI VII in humans was linked to a deficiency in CRTAP expression, a strain of homozygous Crtap-deficient (Crtap−/−) mice was produced using a homologous recombination approach.Citation59,Citation60 It has been found that Crtap−/− mice properly model OI VII in humans, as they have an abnormally high mineral content and increased mineral densities in their bones compared to wild-type mice.Citation60 Other characteristics noted in Crtap−/− mice include a progressive severe kyphoscoliosis, low bone density, and cartilage dysplasia.Citation11 However, Crtap−/− mice tend to have a less severe form of the disease than is found in most humans.Citation11,Citation56 Tensile testing done on the skin of Crtap−/− mice showed that the peak load and stiffness of the tissue were 60% lower than the skin of wild-type mice.Citation61

P3h1−/− mouse: OI VIII

In 2010, a method of homologous recombination was used to create a P3H1-knockout mouse by targeting exons 1–3 of P3H1.Citation62 The heterozygous (P3h1+) mice did not express an OI phenotype, but homozygous (P3h1−/−) mice exhibited significantly smaller body size, lower mineral density in the skull and long bones, progressive kyphoscoliosis, and shorter femora that had decreased stiffness and failure load compared to wild-type littermates.Citation62 P3h1−/− mice also demonstrate elevated auditory thresholds and altered morphology of the middle-ear bone joints, which make them an ideal model for investigating the mechanisms that cause hearing loss in OI patients.Citation63

OI types IX and X

Characteristics in humans

Recently, there have been several new genetic mutations identified that lead to a disorder similar to OI, and a few have been subsequently classified as OI types IX and X. PPIB was identified as the gene responsible for OI IX after patients initially diagnosed with OI II–IV were not found to have mutations present in COL1A1 or COL1A2.Citation64 In fact, multiple studies have shown that although the mutation occurs in PPIB, the phenotype expressed is similar to classical OI type II, III, or IV.Citation64Citation67 OI X, by comparison, is far less broad in its phenotype: the available data were obtained from a study of a single patient who exhibited small bone size, blue sclerae, multiple fractures, malformed bones, and DI.Citation68 The gene identified as causing the OI in this patient was HSP47/SERPINH1,Citation68 which is a molecular chaperone required for the maturation of collagen types I and IV in the endoplasmic reticulum (ER).Citation69,Citation70

Animal models

Ppib−/− mouse: OI IX

Between 2006 and 2007, several research articles were published indicating the roles of P3h1 and Crtap in the development of OI in humans and mice.Citation51,Citation55,Citation59,Citation71 There was also a relationship established between P3h1, Crtap, and Ppib,Citation59,Citation62,Citation71,Citation72 and it was found that they interact to form a complex in the rough ER that is believed to function as a collagen chaperone.Citation72 To investigate this relationship further, the Cre/Lox system was utilized to create a PPIB-knockout mouse by targeting exon 3 through homologous recombination.Citation11,Citation71 The heterozygous Ppib+/− mice that were produced using this technique did not exhibit any symptoms of OI, and were mated to produce Ppib−/− offspring,Citation71 which instead exhibit symptoms of OI, including decreased body size and weight, low bone mineral density and volume, progressive kyphosis, decreased skin stiffness, and increased laxity.Citation11,Citation71 Although the phenotype observed in the Ppib−/− mice is less severe than in humans, it is considered an appropriate model of OI IX.Citation11

Serpinh1 (Hsp47−/−) mouse: OI X

An HSP47-knockout mouse was created in 2000 as the result of a gene-targeting approach.Citation73 While first-generation heterozygous (Hsp47/+) mice displayed no apparent OI phenotype, second-generation homozygous (Hsp47−/−) embryos did not survive past E11.5, and displayed ruptured blood vessels and abnormally oriented epithelial tissues.Citation73 Mutant embryos were more translucent than wild-type mice, and appeared shortened.Citation73 Although Northern blot analysis showed that Hsp47 messenger RNA was lower in Hsp47/+, it was not expressed at all in Hsp47−/− mice.Citation73 Electron microscopy observations of collagen fibrils secreted by Hsp47−/− cells showed thin fibrils forming abnormal branches, thus indicating that the collagen type I secreted from the Hsp47−/− mouse cells could not form normal supramolecular fibrillar structures.Citation70 It is believed that Hsp47 plays an important role either in preventing the collagen triple helix from unfolding and providing stabilization during the progression of procollagen folding, or in preventing lateral association of procollagen triple helices in the ER.Citation73,Citation74

Serpinh1 (L326P) Dachshund: OI X

Dachshunds exhibiting multiple fractures and history of lameness were found to have different long bones, ribs, mandibles, and teeth from normal. Their long bone exhibited thin cortical bone lacking an organized Haversian system and secondary cancellous bone, and their teeth had a thin dentine layer.Citation75 A homozygosity-mapping approach determined the position of the mutation responsible for these abnormalities to be located at 24.66–29.4 Mb interval where the SERPINH1 gene maps.Citation76 The Serpinh1 Dachshund does not share an equivalent mutation noted in humans, but its phenotype makes it a suitable model for OI X.Citation11 Drögemüller et alCitation76 suggested the SERPINH1 canine mutation could be a valuable model for the identification of a fifth gene, in addition to COL1A1, COL1A2, CRTAP, and LEPRE1, that generates OI in humans.

OI types XI–XVII

Characteristics in humans

Since 2010, there have been an additional seven new OI types with similar phenotypes to but different genetic bases from the original four types of OI added to the OI-classification system. It was proposed that the syndrome resembling OI known as Bruck syndrome 1 be reclassified as OICitation77,Citation78 following research that identified a mutation in the FKBP10 gene as the cause of a recessive form of OI with characteristics similar to OI III.Citation79 This phenotype is now known as OI XI, and the suspected mechanism of the FKBP10 mutation is that it causes a decrease of collagen type I deposition in the extracellular matrix.Citation66

OI XII has been identified in a single proband with a phenotype similar to OI IV, but the cause of this OI type was linked to a frameshift mutation occurring in the SP7 gene.Citation80 OI XIII was identified as being caused by a homozygous missense mutation in the gene that encodes the BMP1/Toll-like proteinases, and results in a phenotype similar to OI III.Citation81 OI XIV was first described when a homozygous mutation in the TMEM38B gene was found to lead to a phenotype with characteristics in line with an autosomal-recessive type of OI, including osteopenia and fractures of varying severity, but normal teeth, sclerae, and hearing.Citation82

What is now known as OI XV was described in several publications when multiple mutations in the WNT1 gene were found in individuals displaying phenotypes characteristic of OI III–IV.Citation83Citation86 OI XVI has been documented in one proband family where two siblings were affected by severe OI characterized by frequent fractures starting in utero, varus deformities of the legs, soft skull bones with large fontanels, and beaded ribs.Citation87 Analysis of DNA from one sibling indicated a homozygous deletion in the CREB3L1 gene.Citation87 OI XVII was classified after two individuals diagnosed with OI IV were found to have a mutation in the SPARC gene.Citation88

Animal models

Fkbp10−/− mouse: OI XI

Mice containing the European conditional mutagenesis allele were created to study the function of Fkbp10.Citation89 In the first generation of heterozygotes (Fkbp10/+), no OI phenotype was noted.Citation89 However, homozygotes (Fkbp10−/−) obtained from breeding Fkbp10/+ mice exhibited delayed growth from E13.5 on, as well as downward-facing forelimbs, fragile tissue, flattened facial features, ER dilation caused by accumulation of abnormal procollagen chains in the ER, and perinatal death.Citation89 While humans with OI XI do not exhibit perinatal death, the effect the mutation has on the ER and collagen formation is phenotypically similar to Fkbp10−/− mice.Citation89

Osterix-null Sp7−/− mouse: OI XII

One initial method to create an Sp7-null mouse involved using a homologous recombination of embryonic stem cells to create nonphenotypic heterozygous (Sp7+/−) mice.Citation90 However, Sp7-null offspring obtained from Sp7+/− pairings all died within 15 minutes of birth following difficulty breathing, and exhibited severe limb deformities.Citation90 A second method used the Cre/Lox system to create conditional SP7-knockout mice with Col1a1-Cre (OsxFlox−;Col1a1-Cre), which were designed to inactivate SP7 without perinatal lethality.Citation91 OsxFlox−;Col1a1-Cre mice exhibited decreased trabecular bone mineralization, as well as osteopenia and cortical bone thinning.Citation91 Although both models existed before SP7 was linked to OI in humans, OsxFlox−;Col1a1-Cre may possibly be considered a model for OI XII, but further studies are needed in this regard.

Frf−/− zebrafish: OI XIII

It has been shown that mutations in BMP1 cause high bone mineral density and multiple fractures in human OI XIII.Citation48,Citation81 This mutation in zebrafish, termed frilly fins (Frf−/−), causes a ruffled larval fin, short body axis, malformed craniofacial bones and vertebrae, and reduced ossification and bone density.Citation48 Frf−/− was created by missense substitutions in BMP1, and led to osteoblasts exhibiting a more cuboidal structure than wild-type fish.Citation48 Bmp1 has been found to cleave and inactivate the Bmp2/4 inhibitor chordin, which aids in generating mature collagen type I, and thus it may disrupt osteogenesis when present at elevated levels.Citation92

Swaying (Wnt1Sw/Sw) mouse: OI XV

The Swaying (Sw) mouse was first described as having poor coordination, with a rotational movement pattern and cerebellar deficit.Citation93 The Sw mouse was later found to be phenotypically similar to mice with a targeted WNT1 mutation causing osteopenia and fragile bones,Citation94 and was subsequently determined to be caused by a frameshift mutation in WNT1 (Wnt1Sw), with homozygous (Wnt1Sw/Sw) mice expressing the Sw phenotype.Citation95 Following the classification of WNT1 mutations as a cause of documented OI in humans, Wnt1Sw/Sw was proposed to be a suitable model for OI XV.Citation96

Currently, there are no animal models for OI XIV, XVI, or XVII. However, the genes that were discovered to be the cause of OI XVI and XVII were selected for investigation based on genetic mutations in mice that cause similar phenotypes to what human cases presented with.Citation87,Citation88

Animal models in clinical research

Bone fracture and fragility are the primary symptoms of OI, and thus clinical research on treatments has been focused mainly on understanding the bone properties of the animal models for OI and on regaining mechanical integrity in these bones. Most of the research dedicated to finding treatments for OI has primarily utilized mouse models of classical OI ().

Table 1 Current animal models for human osteogenesis imperfecta (OI), with their genetic and phenotypic characteristics and clinical therapy applications

Pharmacological treatments

Many studies have tested drug treatment as a clinical therapy for OI. In particular, bisphosphonates, a class of antiresorptive drugs, have been widely used to treat OI in an effort to reduce fracture incidence and improve bone density in OI patients. Between the different bisphosphonates, alendronate and pamidronate have been the most used in clinical studies run on animals. Short-term therapy studies conducted on oim/oim and BrtlIV/+ mice for both alendronate and pamidronate injections showed a reduction in the fracture rate,Citation97,Citation98 with thicker and stiffer bones,Citation99Citation101 enhanced density,Citation97,Citation99Citation101 but no mineral maturity,Citation102 and variable biomechanical properties.Citation97Citation99,Citation101 In both models, bisphosphonate treatment altered the growth plate in mice by increasing their height, mainly at the proliferation and hypertrophic zones, reducing vascular invasionCitation103 and growth-plate cell turnover,Citation98 and with detrimental effects on osteoblasts and bone formation.Citation99 In trabecular oim/oim bone, alendronate increased trabecular number, decreasing thickness and separation and increasing bone volume density,Citation97,Citation104 and produced no improvement in strength or mechanical properties. This shows that the reduction in fracture rate is primarily attributable to an increase in bone mass.Citation104 High doses of alendronate were more effective in short-term therapy.Citation98

Long-term alendronate therapy in growing oim/oim mice also appeared to be effective in increasing metaphyseal density. The increased length and bowing of tibia metaphysis seen in short-term therapy was not observed in long-term treatment, suggesting a dissipation of these effects over time.Citation105 Sex was a factor contributing to the success of the treatment, as alendronate increased compositional heterogeneity in male mice, improving bone quality.Citation106

More recently, alendronate treatment was proved to affect the fracture-healing process of bone in BrtlIV+ mice positively.Citation107 Mice treated with alendronate before and after fracture exhibited increased callus volume, with decreased mineralization quality and quantity, and increased bone volume and torque at failure. However, no effect was observed if the drug treatment was used only before fracture.Citation107

RANKL inhibitors have shown promise in both humanCitation108,Citation109 and animalCitation97,Citation110Citation113 trials for the treatment of several different osteopathologies. When RANKL binds to RANK, it activates the pathway that leads to osteoclastogenesis,Citation109,Citation111Citation114 so the pharmacological application of RANKL inhibitors aims to prevent bone resorption by blocking the RANK pathway from being activated.Citation97,Citation108Citation114 The inhibitor denosumab, a fully human monoclonal antibody,Citation97,Citation111Citation114 has a higher specificity and affinity for RANKL than osteopro-tegerin,Citation112Citation114 a naturally occurring decoy RANKL receptor.Citation110Citation114 Unfortunately, denosumab is not recognized by murine RANKL,Citation97,Citation111Citation114 although preliminary studies done in primatesCitation97,Citation112 and knock-in mice expressing murine/human RANKCitation97,Citation113 have yielded positive results, including increased mineral content, density, and strength in primate bone,Citation97,Citation112 as well as suppressed resorption and increased density in human RANKL mouse boneCitation97,Citation113 following denosumab treatment.

The successful application of RANKL inhibitors in OI treatment was demonstrated in two children with OI VI who exhibited decreased bone resorption following administration of denosumab.Citation109 Animal research of RANKL inhibitors in the treatment of OI has so far been limited to the use of RANK-Fc,Citation97,Citation110,Citation111 a recombinant RANKL inhibitor with properties similar to osteoprotegerin.Citation97,Citation110,Citation114 The results of treating oim/oim mice with RANK-Fc show that it increases bone density,Citation97,Citation111 improves some mechanical and geometric properties of bone,Citation111 delays callus remodeling following fracture,Citation110 and reduces fracture incidence.Citation97 Inspired by the success of the teriparatide (human parathyroid hormone) as an anabolic drug for osteoporosis,Citation115Citation120 and as adjuvant for boneCitation121,Citation122 and allograftCitation123Citation125 healing, scientists have recently tested teriparatide in adults with OI I–IV.Citation126 Results showed an enhancement in vertebral bone mineral density and strength, and thus there is a need for animal research on the use of teriparatide for treatment of OI.

Finally, a study performed on BrtlIV/+ mice determined the effect of sclerostin antibody (SA), an anabolic bone therapeutic, on OI bone formation.Citation127 It was thought that SA could neutralize sclerostin, ultimately increasing bone formation by preventing the inhibition of anabolic canonical Wnt signaling.Citation127 While previous antiresorptive bisphosphonate-treatment studies using BrtlIV/+ and oim mice resulted in increased trabecular number but not thickness, the treatment study using SA instead found an increase in trabecular thickness but not number.Citation127 After 2 weeks of SA therapy, BrtlIV/+ mice had increased cortical and trabecular bone mass, reduced fragility in long bones, including improved stiffness, and the ability to withstand higher loads before breaking.Citation127 SA therapy increased the rate of bone formation, increasing both cortical and cancellous bone volume, and also improved the strength of cortical bone without affecting bone mineralization.Citation127 Although more research is needed, SA treatments could eventually prove to be an effective therapy for patients with OI IV in the future.

Stem cell transplantation

The oim/oim mouse has recently undergone stem cell therapies to alter developing OI bone pathology. Mesenchymal stem cell (MSC) therapy for tissue regeneration can improve damaged or diseased tissue when delivered to relevant locations.Citation128 MSCs injected into oim/oim mice from adult wild-type mice migrate to sites of new bone formation.Citation129 As an expansion on this study, human fetal blood MSCs, collected by cardiocentesis under ultrasound guidance, were considered as a therapy for oim/oim mice.Citation130,Citation131 Virgin oim/oim females were mated with oim/oim males and then underwent intrauterine transplantation at E13.5–E15 so that each fetus could be injected intraperitoneally with human fetal blood MSCs.Citation130 Offspring were analyzed at E18, week 1 after birth, and weeks 2, 4, 8, and 12 thereafter. At each point measured, findings showed a 70% reduction in the number of fractures with only 5% of cells ingrafted.Citation130 The fetal blood stem/stromal cells engrafted into oim/oim bone differentiated into osteoblasts, producing osteocalcin and Col1a2, which is absent in oim/oim mice.Citation132 As a result, these bones showed a clinically relevant increase in cortical bone strength, length, and thickness, and clustered cells around sites of bone formation and repair, suggesting osteogenic differentiation.Citation130,Citation132

Bone marrow stromal cells

Studies involving the transplant of bone marrow stromal cells (BMSCs) from wild-type mice into human COL1A1-minigene mice demonstrated that BMSCs could renew cells in multiple nonhematopoietic tissues, and may be a prospective treatment for OI.Citation133 Transplanted BMSCs have been shown to contribute to bone formation in vivo and to improve the mechanical properties of the recipient bone.Citation134 In a study conducted to determine the effect of transplanting BMSCs alone or suspended in a collagen matrix, BMSCs were harvested from the marrow of the femora and tibiae of wild-type mice syngeneic to oim mice, then cultured and injected into the femora of oim mice.Citation134 Mechanical testing of the bone conducted 6 weeks after implantation found that femora injected with BMSCs suspended in collagen matrix were stronger than those injected with BMSCs alone, and that in both BMSC test groups treated bone was stronger when compared to oim/oim.Citation134 Donor cells, tracked using green fluorescent proteins, differentiated into osteoblasts in vivo and actively participated in new bone formation.Citation134 Histological analysis of the femora showed that those injected with BMSCs suspended in collagen matrix had more bone deposition than those injected with BMSCs alone.Citation134 Comparison of the BMSC-treated samples with oim/oim samples indicated that all of the new bone deposition was a result of the combination of endogenous and exogenous cells.Citation134

BMSCs may not only be transplanted between adult mice but also in utero and in developing animals.Citation134,Citation135 In another study, BMSCs were transplanted from long bones of adult cytomegalovirus/eGFP CD1 transgenic mice into embryos of wild-type females mated with BrtlIV/+ males.Citation135 Of the pups that reached weaning age, 51.4% were wild-type and 48.5% BrtlIV/+ mice, demonstrating a nearly equal survival rate between the two genotypes.Citation135 Comparing these mice with a control group in which no treatments were administered, it was seen that BMSC treatment improved mechanical properties of bone in BrtlIV/+ mice,Citation135 with BMSC-treated BrtlIV/+ mice having significantly higher bone mineral density in both trabecular and cortical bone compared to untreated BrtlIV/+ mice.Citation135

Gene therapies

Several methods have been proposed for implementing gene therapies in the treatment of OI, including gene replacement, antisense approaches, gene supplementation, and gene delivery to bone.Citation136 One of the first studies in using animal models for gene replacement to treat OI was accomplished by transfecting a Col1a1-null allele taken from a Mov-13−/− embryo with human COL1A1.Citation137,Citation138 In this case, functional collagen type I was formed in the mouse through transgene expression.Citation137,Citation138 This collagen was composed of two human α1(I) and one mouse α2(I) chains, suggesting that this could be a means for using gene therapy in treating OI in humans.Citation137,Citation138

Another approach of gene therapy is gene supplementation, which in OI research could consist of the delivery of Col1a1 from healthy mice to a mouse with OI caused by a COL1A1 mutation. However, the challenge with this therapy is that most types of OI mutations are dominant-negative, meaning that the mutant allele disrupts the function of the normally functioning allele, so supplementation would not be effective without first blocking the expression of the mutant gene.Citation136 Delivery of the healthy gene is further complicated by the fact that COL1A1 and COL1A2 are large genes and difficult to manipulate.Citation138 The combination of gene supplementation with antisense gene therapy, a method of using antisense oligodeoxynucleotides, antisense RNA, or ribozymes to block transcription of certain gene sequences that are thought to cause the disease,Citation139 could make gene therapy more feasible.Citation136 One study involving linear oligonucleotide antisense therapy was done using human COL1A1-minigene mice.Citation41 Mating mice where both parents expressed antisense minigene produced offspring that inherited both genes, and the rate of occurrence of the lethal fragile-bone phenotype was reduced from 92% to 27%, demonstrating the effects of the therapy.Citation41,Citation140 One potential application for studying gene supplementation in an animal model is the use of the oim/oim mouse, as the deletion of the COL1A2 in these mice stops incorporation of pro-α2(I) chains into heterotrimers and leads to a buildup of pro-α1(I) heterotrimers in tissue.Citation136

One application of gene therapy that appears to hold a great deal of promise is gene delivery to bone. One way this can be done is through the use of noncoding RNA or a protein, which can be delivered using a vector, such as a virus.Citation141 However, the use of viral therapy is not ideal in the clinical setting, due to the risks involved with it.Citation142 Other studies have been performed to test the efficacy of gene delivery without a vector, as in one case using the delivery of naked plasmid DNA to the gastrocnemius of a wild-type ICR mouse.Citation143 Unfortunately, although this method did lead to bone formation, it was determined that this was not an efficient method of gene delivery.Citation142,Citation143 Other methods, such as RNA interfacing and using synthetic carriers for delivery, have also been attempted, with varying degrees of success,Citation142 and gene-delivery therapy still needs to be thoroughly tested before being safely implemented.

Mechanical therapies

Bone adapts both its mass and architecture to the mechanical loading applied upon it.Citation144 Therefore, mechanical therapies that do not result in fracture and do not involve drugs are very attractive for application in the OI population. One potential mechanical therapy, whole-body vibration (WBV), has been studied in young oim/oim mice, and resulted in the improvement of trabecular and cortical bone morphologies during hind-limb growth.Citation144 WBV involves placing mice into a container on a vertically oscillating plate, which effectively introduces a vibrational mechanical stimulus to normal activities.Citation145 Previous numerical studies have shown that increasing the frequency of the loading stimulus up to 30 Hz results in improved bone formation.Citation146 Vanleene and Shefelbine utilized high-frequency, low-amplitude WBV in order to determine its effect on the volume and mechanical properties of young oim/oim bone.Citation144 The study found that WBV caused an increase in oim/oim femoral and tibial cortical thickness and cross-sectional area, as well as an increase in trabecular bone volume in oim/oim tibiae.Citation144 WBV thus appears to be a potentially effective and safe treatment for increasing bone formation and possibly bone strength in OI. This therapy is particularly attractive for treating the growing skeletons of children with OI without the disadvantages of current long-term pharmacological therapies.

Discussion and conclusion

This review presented existing animal models for OI I–XVII and clinical therapies, some still emerging, that these models serve as a platform for OI research. Most of the animal models used for clinical trials were mice, representing moderate-to-severe conditions of the classic OI disease. In fact, mutagenesis studies that have led to the development of new models of OI have only been conducted using mice. The canine models of OI mentioned in this article all occurred spontaneously in animals that had not been bred for research purposes. It is probable that the bone structure of canines would more adequately model the disease physiology in humans than the bone structure of mice, but there are limiting factors that would make the development of canine OI models difficult.Citation11

Currently, treatments for human OI rely largely on bisphosphonates and exercise management, which can increase the amount of bone but not its quality.Citation37 More animal studies are needed to understand the actual efficacy and long-term effects of these treatments on bone, and also more research is envisaged to explore other therapies for the management and cure of OI. In this sense, animal models, and in particular mouse models of OI, offer an extremely valuable resource for the understanding of the disease and for the advancement of therapies for reducing fracture risk.

Mutagenic mouse models are relatively easy and quick to produce, and genetically assimilate mutations typical of human cases of OI. On the other hand, mouse bones are very small in size and do not allow for typical sample preparation as used in human bones (ie, milling out exact perfect beam samples from human bone for mechanical testing but not from a mouse bone, where the entire bone with its geometrical imperfections needs to be used). Also, the structure of mouse cortical bone is different from human cortical bone in that it is missing the Haversian system, as the whole mouse bone is about the size of a large osteon in human bone. This may have implications in terms of mechanics, because the lamellar osteonal structure is thought to provide toughness to human bone,Citation147 although recent research on oim/oim mouse bone has shown that possibly the lamellar structure can yield a similar toughness to mouse bone.Citation17 Apart from these two differences, mouse bone represents an indispensable resource to understand bone pathology and to anticipate the effects of potential therapies on humans, as mouse skeletal growth is accelerated, allowing for long-term investigation in a 6- to 12-month period.

In the future, there should be a much wider breadth of studies utilizing OI animal models for further understanding clinical applications and their translation to human therapies. It would be interesting to study methodological factors, such as drug dosage or mechanical loading, affecting treatment, and to investigate further long-term effects of treatment in each type of OI. Also, the combination of different therapies, such as using gene therapy with mechanical loading, may be a viable approach that needs further investigation. Therefore, more clinical studies should be conducted on the animal models presented here to determine which therapy holds the most promise for human OI. Finally, more studies allowing the direct translation of therapies from animal models to human cases should be put in place to facilitate implementation of new therapies.

Disclosure

The authors report no conflicts of interest in this work.

References

  • BaljetBAspects of the history of osteogenesis imperfecta (Vrolik’s syndrome)Ann Anat200218411711876477
  • SillenceDOSennADanksDMGenetic heterogeneity in osteogenesis imperfectaJ Med Genet1979162101116458828
  • Kamoun-GoldratASLe MerrerMFAnimal models of osteogenesis imperfecta and related syndromesJ Bone Miner Metab200725421121817593490
  • RauchFGlorieuxFHOsteogenesis imperfectaLancet200436394181377138515110498
  • BodianDLChanTFPoonAMutation and polymorphism spectrum in osteogenesis imperfecta type II: implications for genotype-phenotype relationshipsHum Mol Genet200918346347118996919
  • JaenischRHarbersKSchniekeAGermline integration of Moloney murine leukemia virus at the Mov13 locus leads to recessive lethal mutation and early embryonic deathCell19833212092166825169
  • HarbersKKuehnMDeliusHJaenischRInsertion of retrovirus into the first intron of α1(I) collagen gene to embryonic lethal mutation in miceProc Natl Acad Sci U S A1984815150415086324198
  • BarkerDDWuHHartungSBreindlMJaenischRRetrovirus-induced insertional mutagenesis: mechanism of collagen mutation in Mov13 miceMol Cell Biol19911110515451631922037
  • SmithROsteogenesis imperfecta: from phenotype to genotype and back againInt J Exp Pathol19947542332417947227
  • Iruela-ArispeMLVernonRBWuHJaenischRSageEHType I collagen-deficient Mov-13 mice do not retain SPARC in the extracellular matrix: implications for fibroblast functionDev Dyn199620721711838906420
  • GlorieuxFHSponsellerPDShapiroJRByersPHOsteogenesis Imperfecta: A Translational Approach to Brittle Bone DiseaseLondonAcademic Press2014
  • ChipmanSDSweetHOMcBrideDJDefective proα2(I) collagen synthesis in a recessive mutation in mice: a model of human osteogenesis imperfectaProc Natl Acad Sci U S A1993905170117058446583
  • BonadioJSaundersTLTsaiETransgenic mouse model of the mild dominant form of osteogenesis imperfectaProc Natl Acad Sci U S A19908718714571492402497
  • JepsenKJSchafflerMBKuhnJLGouletRWBonadioJGoldsteinSAType I collagen mutation alters the strength and fatigue behavior of Mov13 cortical tissueJ Biomech19973011–12114111479456382
  • KuznetsovaNVForlinoACabralWAMariniJCLeikinSStructure, stability and interactions of type I collagen with GLY349-CYS substitution in α1(I) chain in a murine osteogenesis imperfecta modelMatrix Biol200423210111215246109
  • KozloffKMCardenABergwitzCBrittle IV mouse model for osteogenesis imperfecta IV demonstrates postpubertal adaptations to improve whole bone strengthJ Bone Miner Res200419461462215005849
  • CarrieroAZimmermannEAPalusznyAHow tough is brittle bone? Investigating osteogenesis imperfecta in mouse boneJ Bone Miner Res20142961392140124420672
  • FratzlPParisOKlaushoferKLandisWBone mineralization in an osteogenesis imperfecta mouse model studied by small-angle X-ray scatteringJ Clin Invest19969723964028567960
  • NiyibiziCLiFPotential implications of cell therapy for osteogenesis imperfectaInt J Clin Rheumtol200941576620490372
  • CarrieroADoubeMVogtMAltered lacunar and vascular porosity in osteogenesis imperfecta mouse bone as revealed by synchrotron tomography contributes to bone fragilityBone20146111612424373921
  • McBrideDJrShapiroJDunnMBone geometry and strength measurements in aging mice with the oim mutationCalcif Tissue Int19986221721769437052
  • CamachoNPHouLToledanoTRThe material basis for reduced mechanical properties in oim mice bonesJ Bone Miner Res19991422642729933481
  • ColeWGThe Nicholas Andry Award – 1996: the molecular pathology of osteogenesis imperfectaClin Orthop Relat Res1997343235248
  • ColeWGAdvances in osteogenesis imperfectaClin Orthop Relat Res2002401616
  • BoydeATraversRGlorieuxFJonesSThe mineralization density of iliac crest bone from children with osteogenesis imperfectaCalcif Tissue Int199964318519010024373
  • NymanJSReyesMWangXEffect of ultrastructural changes on the toughness of boneMicron200536756658216169742
  • SabanJZussmanMHaveyRPatwardhanASchneiderGKingDHeterozygous oim mice exhibit a mild form of osteogenesis imperfectaBone19961965755798968022
  • MisofKLandisWKlaushoferKFratzlPCollagen from the osteogenesis imperfecta mouse model (oim) shows reduced resistance against tensile stressJ Clin Invest19971001409202055
  • DaleyEStreetenEASorkinJDVariable bone fragility associated with an Amish COL1A2 variant and a knock-in mouse modelJ Bone Miner Res201025224726119594296
  • StaceyABatemanJChoiTMascaraTColeWJaenischRPerinatal lethal osteogenesis imperfecta in transgenic mice bearing an engineered mutant pro-α1(I) collagen geneNature198833261601311362450280
  • BatemanJFMascaraTColeWGStaceyAJaenischRThe study of collagen structure and function by site-directed mutagenesis of collagen genesAnn N Y Acad Sci199058013243292337302
  • Gailus-DurnerVFuchsHBeckerLIntroducing the German Mouse Clinic: open access platform for standardized phenotypingNat Methods20052640340415908916
  • LisseTSThieleFFuchsHER stress-mediated apoptosis in a new mouse model of osteogenesis imperfectaPLoS Genet200842e718248096
  • ForlinoAPorterFDLeeEJWestphalHMariniJCUse of the Cre/lox recombination system to develop a non-lethal knock-in murine model for osteogenesis imperfecta with an α1(I) G349C substitution variability in phenotype in BrtlIV miceJ Biol Chem199927453379233793110608859
  • UvegesTECollin-OsdobyPCabralWACellular mechanism of decreased bone in Brtl mouse model of OI: imbalance of decreased osteoblast function and increased osteoclasts and their precursorsJ Bone Miner Res200823121983199418684089
  • BoskeyALVerdelisKSpevakLMineral and matrix changes in Brtl/+ teeth provide insights into mineralization mechanismsBiomed Res Int2013201329581223802117
  • ForlinoACabralWABarnesAMMariniJCNew perspectives on osteogenesis imperfectaNat Rev Endocrinol20117954055721670757
  • SokolovBPMaysPKKhillanJSProckopDJTissue- and development-specific expression in transgenic mice of a type I procollagen (COL1A1) minigene construct with 2.3 kB of the promoter region and 2 kB of the 3′-flanking region: specificity is independent of the putative regulatory sequences in the first intronBiochemistry19933235924292498369292
  • KhillanJSOlsenAKontusaariSSokolovBProckopDTransgenic mice that express a mini-gene version of the human gene for type I procollagen (COL1A1) develop a phenotype resembling a lethal form of osteogenesis imperfectaJ Biol Chem19912663423373233791744131
  • PereiraRKhillanJSHelminenHJHumeELProckopDJTransgenic mice expressing a partially deleted gene for type I procollagen (COL1A1): a breeding line with a phenotype of spontaneous fractures and decreased bone collagen and mineralJ Clin Invest19939127097168432871
  • KhillanJSLiSWProckopDJPartial rescue of a lethal phenotype of fragile bones in transgenic mice with a chimeric antisense gene directed against a mutated collagen geneProc Natl Acad Sci U S A19949114629863028022775
  • CampbellBWoottonJKrookLDeMarcoJMinorRClinical signs and diagnosis of osteogenesis imperfecta in three dogsJ Am Vet Med Assoc199721121831879227748
  • CampbellBGWoottonJAMacLeodJNMinorRRSequence of normal canine COL1A1 cDNA and identification of a heterozygous α1(I) collagen Gly208AIa mutation in a severe case of canine osteogenesis imperfectaArch Biochem Biophys20003841374611147834
  • CampbellBGWoottonJAMacleodJNMinorRRCanine COL1A2 mutation resulting in C-terminal truncation of pro-α2(I) and severe osteogenesis imperfectaJ Bone Miner Res20011661147115311393792
  • DeakSBVan Der RestMProckopDJAltered helical structure of a homotrimer of α1(I) chains synthesized by fibroblasts from a variant of osteogenesis imperfectaColl Relat Res1985543053134053561
  • CampbellBGWoottonJAMacLeodJNMinorRRSequence of canine COL1A2 cDNA: nucleotide substitutions affecting the cyanogen bromide peptide map of the α2(I) chainArch Biochem Biophys1998357167759721184
  • Van EedenFGranatoMSchachUGenetic analysis of fin formation in the zebrafish, Danio rerioDevelopment199612312552629007245
  • AsharaniPKeuppKSemlerOAttenuated BMP1 function compromises osteogenesis, leading to bone fragility in humans and zebrafishAm J Hum Genet201290466167422482805
  • DuránIMarí-BeffaMSantamaríaJBecerraJSantos-RuizLActinotrichia collagens and their role in fin formationDev Biol2011354116017221420398
  • FisherSJagadeeswaranPHalpernMERadiographic analysis of zebrafish skeletal defectsDev Biol20032641647614623232
  • CabralWAChangWBarnesAMProlyl 3-hydroxylase 1 deficiency causes a recessive metabolic bone disorder resembling lethal/severe osteogenesis imperfectaNat Genet200739335936517277775
  • ShapiroJRLietmanCGroverMPhenotypic variability of osteogenesis imperfecta type V caused by an IFITM5 mutationJ Bone Miner Res20132871523153023408678
  • LietmanCDMaromRMunivezEA transgenic mouse model of OI type V supports a neomorphic mechanism of the IFITM5 mutationJ Bone Miner Res201530348949825251575
  • HomanEPRauchFGrafeIMutations in SERPINF1 cause osteogenesis imperfecta type VIJ Bone Miner Res201126122798280321826736
  • BarnesAMChangWMorelloRDeficiency of cartilage-associated protein in recessive lethal osteogenesis imperfectaN Engl J Med2006355262757276417192541
  • MariniJCCabralWABarnesAMNull mutations in LEPRE1 and CRTAP cause severe recessive osteogenesis imperfectaCell Tissue Res20103391597019862557
  • DollJAStellmachVMBouckNPPigment epithelium-derived factor regulates the vasculature and mass of the prostate and pancreasNat Med20039677478012740569
  • BoganRRiddleRCLiZA mouse model for human osteogenesis imperfecta type VIJ Bone Miner Res20132871531153623413146
  • MorelloRBertinTKChenYCRTAP is required for prolyl 3-hydroxylation and mutations cause recessive osteogenesis imperfectaCell2006127229130417055431
  • Fratzl-ZelmanNMorelloRLeeBCRTAP deficiency leads to abnormally high bone matrix mineralization in a murine model and in children with osteogenesis imperfecta type VIIBone201046382082619895918
  • BaldridgeDLenningtonJWeisMGeneralized connective tissue disease in Crtap−/− mousePloS One201055e1056020485499
  • VrankaJAPokidyshevaEHayashiLProlyl 3-hydroxylase 1 null mice display abnormalities in fibrillar collagen-rich tissues such as tendons, skin, and bonesJ Biol Chem201028522172531726220363744
  • PokidyshevaETufaSBreseeCBrigandeJVBächingerHPProlyl 3-hydroxylase-1 null mice exhibit hearing impairment and abnormal morphology of the middle ear bone jointsMatrix Biol2013321394423186870
  • van DijkFSNesbittIMZwikstraEHPPIB mutations cause severe osteogenesis imperfectaAm J Hum Genet200985452152719781681
  • BarnesAMCarterEMCabralWALack of cyclophilin B in osteogenesis imperfecta with normal collagen foldingN Engl J Med2010362652152820089953
  • BarnesAMCabralWAWeisMAbsence of FKBP10 in recessive type XI osteogenesis imperfecta leads to diminished collagen cross-linking and reduced collagen deposition in extracellular matrixHum Mutat201233111589159822718341
  • PyottSMSchwarzeUChristiansenHEMutations in PPIB (cyclophilin B) delay type I procollagen chain association and result in perinatal lethal to moderate osteogenesis imperfecta phenotypesHum Mol Genet20112081595160921282188
  • ChristiansenHESchwarzeUPyottSMHomozygosity for a missense mutation in SERPINH1, which encodes the collagen chaperone protein HSP47, results in severe recessive osteogenesis imperfectaAm J Hum Genet201086338939820188343
  • NagataKHSP47 as a collagen-specific molecular chaperone: function and expression in normal mouse developmentSemin Cell Dev Biol200314527528214986857
  • IshidaYKubotaHYamamotoAKitamuraABächingerHPNagataKType I collagen in Hsp47-null cells is aggregated in endoplasmic reticulum and deficient in N-propeptide processing and fibrillogenesisMol Biol Cell20061752346235516525016
  • ChoiJWSutorSLLindquistLSevere osteogenesis imperfecta in cyclophilin B-deficient micePLoS Genet2009512e100075019997487
  • IshikawaYWirzJVrankaJANagataKBächingerHPBiochemical characterization of the prolyl 3-hydroxylase 1 cartilage-associated protein cyclophilin B complexJ Biol Chem200928426176411764719419969
  • NagaiNHosokawaMItoharaSEmbryonic lethality of molecular chaperone hsp47 knockout mice is associated with defects in collagen biosynthesisJ Cell Biol200015061499150610995453
  • TasabMBattenMRBulleidNJHsp47: a molecular chaperone that interacts with and stabilizes correctly-folded procollagenEMBO J200019102204221110811611
  • SeeligerFLeebTPetersMBrügmannMFehrMHewicker-TrautweinMOsteogenesis imperfecta in two litters of dachshundsVet Pathol200340553053912949410
  • DrögemüllerCBeckerDBrunnerAA missense mutation in the SERPINH1 gene in dachshunds with osteogenesis imperfectaPLoS Genet200957e100057919629171
  • AlanayYKrakowDResponse to Shaheen et alAm J Hum Genet2010872308
  • ShaheenRAl-OwainMSakatiNAlzayedZSAlkurayaFSFKBP10 and Bruck syndrome: phenotypic heterogeneity or call for reclassification?Am J Hum Genet201087230630720696291
  • AlanayYAvayganHCamachoNMutations in the gene encoding the RER protein FKBP65 cause autosomal-recessive osteogenesis imperfectaAm J Hum Genet201086455155920362275
  • LapunzinaPAglanMTemtamySIdentification of a frameshift mutation in osterix in a patient with recessive osteogenesis imperfectaAm J Hum Genet201087111011420579626
  • Martínez-GlezVValenciaMCaparrós-MartínJAIdentification of a mutation causing deficient BMP1/mTLD proteolytic activity in autosomal recessive osteogenesis imperfectaHum Mutat201233234335022052668
  • ShaheenRAlazamiAMAlshammariMJStudy of autosomal recessive osteogenesis imperfecta in Arabia reveals a novel locus defined by TMEM38B mutationJ Med Genet2012491063063523054245
  • KeuppKBeleggiaFKayseriliHMutations in WNT1 cause different forms of bone fragilityAm J Hum Genet201392456557423499309
  • PyottSMTranTTLeistritzDFWNT1 mutations in families affected by moderately severe and progressive recessive osteogenesis imperfectaAm J Hum Genet201392459059723499310
  • FahiminiyaSMajewskiJMortJMoffattPGlorieuxFHRauchFMutations in WNT1 are a cause of osteogenesis imperfectaJ Med Genet201350534534823434763
  • LaineCMJoengKSCampeauPMWNT1 mutations in early-onset osteoporosis and osteogenesis imperfectaN Engl J Med2013368191809181623656646
  • SymoensSMalfaitFD’hondtSDeficiency for the ER-stress transducer OASIS causes severe recessive osteogenesis imperfecta in humansOrphanet J Rare Dis2013815424079343
  • Mendoza-LondonoRFahiminiyaSMajewskiJRecessive osteogenesis imperfecta caused by missense mutations in SPARCAm J Hum Genet201596697998526027498
  • LietmanCDRajagopalAHomanEPConnective tissue alterations in Fkbp10−/− miceHum Mol Genet201423184822483124777781
  • NakashimaKZhouXKunkelGThe novel zinc finger- containing transcription factor osterix is required for osteoblast differentiation and bone formationCell20021081172911792318
  • BaekWYLeeMAJungJWPositive regulation of adult bone formation by osteoblast-specific transcription factor osterixJ Bone Miner Res20092461055106519113927
  • FisherSHalpernMEPatterning the zebrafish axial skeleton requires early chordin functionNat Genet199923444244610581032
  • LanePWSwayingMouse News Lett1967361
  • ThomasKRCapecchiMRTargeted disruption of the murine int-1 proto-oncogene resulting in severe abnormalities in midbrain and cerebellar developmentNature199034662878478502202907
  • ThomasKRMusciTSNeumannPECapecchiMRSwaying is a mutant allele of the proto-oncogene Wnt-1Cell19916759699761835670
  • JoengKSLeeYCJiangMMThe swaying mouse as a model of osteogenesis imperfecta caused by WNT1 mutationsHum Mol Genet201423154035404224634143
  • BargmanRPoshamRBoskeyADiCarloERaggioCPleshkoNComparable outcomes in fracture reduction and bone properties with RANKL inhibition and alendronate treatment in a mouse model of osteogenesis imperfectaOsteoporos Int20122331141115021901481
  • EvansKSheppardLRaoSMartinROberbauerAPamidronate alters the growth plate in the oim mouse model for osteogenesis imperfectaInt J Biomed Sci20095434535223675157
  • UvegesTEKozloffKMTyJMAlendronate treatment of the Brtl osteogenesis imperfecta mouse improves femoral geometry and load response before fracture but decreases predicted material properties and has detrimental effects on osteoblasts and bone formationJ Bone Miner Res200924584985919113917
  • McCarthyEARaggioCLHossackMDAlendronate treatment for infants with osteogenesis imperfecta: demonstration of efficacy in a mouse modelPediatr Res200252566067012409511
  • RaoSEvansKOberbauerAMartinRBisphosphonate treatment in the oim mouse model alters bone modeling during growthJ Biomech200841163371337619022450
  • CamachoNCarrollPRaggioCFourier transform infrared imaging spectroscopy (FT-IRIS) of mineralization in bisphosphonate-treated oim/oim miceCalcif Tissue Int200372560460912574874
  • EvansKLauSOberbauerAMartinRAlendronate affects long bone length and growth plate morphology in the oim mouse model for osteogenesis imperfectaBone200332326827412667554
  • MisofBMRoschgerPBaldiniTDifferential effects of alendronate treatment on bone from growing osteogenesis imperfecta and wild-type mouseBone200536115015815664013
  • QuenzerJBoachie-AdjeiKBoskeyALProckopDCamachoNPRaggioCLUse of alendronate in long-term treatment of transgenic OI micePoster presented at: 50th Annual Meeting of the Orthopaedic Research SocietyMarch 7–10, 2004San Francisco, CA
  • BoskeyALMarinoJSpevakLAre changes in composition in response to treatment of a mouse model of osteogenesis imperfecta sex-dependent?Clin Orthop Relat Res201547382587159825903941
  • MeganckJBegunDMcElderryJFracture healing with alendronate treatment in the Brtl/+ mouse model of osteogenesis imperfectaBone201356120421223774443
  • WangHDBoyceAMTsaiJYEffects of denosumab treatment and discontinuation on human growth platesJ Clin Endocrinol Metab201499389189724423331
  • SemlerONetzerCHoyer-KuhnHBeckerJEyselPSchoenauEFirst use of the RANKL antibody denosumab in osteogenesis imperfecta type VIJ Musculoskelet Neuronal Interact201212318318822947550
  • DelosDYangXRicciardiBFMyersERBostromMPCamachoNPThe effects of RANKL inhibition on fracture healing and bone strength in a mouse model of osteogenesis imperfectaJ Orthop Res200826215316417729310
  • BargmanRHuangABoskeyALRaggioCPleshkoNRANKL inhibition improves bone properties in a mouse model of osteogenesis imperfectaConnect Tissue Res201051212313120053133
  • KostenuikPJOsteoprotegerin and RANKL regulate bone resorption, density, geometry and strengthCurr Opin Pharmacol20055661862516188502
  • KostenuikPJNguyenHQMcCabeJDenosumab, a fully human monoclonal antibody to RANKL, inhibits bone resorption and increases BMD in knock-in mice that express chimeric (murine/human) RANKLJ Bone Miner Res200924218219519016581
  • AnastasilakisADToulisKAPolyzosSAAnastasilakisCDMakrasPLong-term treatment of osteoporosis: safety and efficacy appraisal of denosumabTher Clin Risk Manag2012829530622767993
  • NeerRMArnaudCDZanchettaJREffect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosisN Engl J Med2001344191434144111346808
  • JiangYZhaoJLiaoEYDaiRCWuXPGenantHKApplication of micro-CT assessment of 3-D bone microstructure in preclinical and clinical studiesJ Bone Miner Metab200523112213115984427
  • DempsterDWCosmanFKurlandESEffects of daily treatment with parathyroid hormone on bone microarchitecture and turnover in patients with osteoporosis: a paired biopsy studyJ Bone Miner Res200116101846185311585349
  • KurlandESCosmanFMcMahonDJRosenCJLindsayRBilezikianJPParathyroid hormone as a therapy for idiopathic osteoporosis in men: effects on bone mineral density and bone markersJ Clin Endocrinol Metab20008593069307610999788
  • RodanGAMartinTJTherapeutic approaches to bone diseasesScience200028954841508151410968781
  • BlackDMGreenspanSLEnsrudKEThe effects of parathyroid hormone and alendronate alone or in combination in postmenopausal osteoporosisN Engl J Med2003349131207121514500804
  • MognettiBMarinoSBarberisAExperimental stimulation of bone healing with teriparatide: histomorphometric and microhardness analysis in a mouse model of closed fractureCalcif Tissue Int201189216317121701938
  • KabackLASoungDYNaikATeriparatide (1-34 human PTH) regulation of osterix during fracture repairJ Cell Biochem2008105121922618494002
  • DhillonRSSchwarzEMTeriparatide therapy as an adjuvant for tissue engineering and integration of biomaterialsJ Mater Res2011461117113121857768
  • JacobsonJAYanoso-SchollLReynoldsDGTeriparatide therapy and beta-tricalcium phosphate enhance scaffold reconstruction of mouse femoral defectsTissue Eng Part A2010173–438939820807012
  • ReynoldsDGTakahataMLernerALO’KeefeRJSchwarzEMAwadHATeriparatide therapy enhances devitalized femoral allograft osseointegration and biomechanics in a murine modelBone201148356257020950720
  • OrwollESShapiroJVeithSEvaluation of teriparatide treatment in adults with osteogenesis imperfectaJ Clin Invest2014124249149824463451
  • SinderBPEddyMMOminskyMSCairdMSMariniJCKozl-offKMSclerostin antibody improves skeletal parameters in a Brtl/+ mouse model of osteogenesis imperfectaJ Bone Miner Res2013281738022836659
  • HorwitzEMAndreefMFrassoniFMesenchymal stromal cellsCurr Opin Hematol200613641942517053453
  • LiFWangXNiyibiziCDistribution of single-cell expanded marrow derived progenitors in a developing mouse model of osteogenesis imperfecta following systemic transplantationStem Cells200725123183319317823236
  • GuillotPVAbassOBassettJDIntrauterine transplantation of human fetal mesenchymal stem cells from first-trimester blood repairs bone and reduces fractures in osteogenesis imperfecta miceBlood200811131717172517967940
  • CampagnoliCRobertsIAKumarSBennettPRBellantuonoIFiskNMIdentification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrowBlood20019882396240211588036
  • VanleeneMSaldanhaZCloydKLTransplantation of human fetal blood stem cells in the osteogenesis imperfecta mouse leads to improvement in multiscale tissue propertiesBlood201111731053106021088133
  • PereiraRFO’HaraMDLaptevAVMarrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfectaProc Natl Acad Sci U S A1998953114211479448299
  • LiFWangXNiyibiziCBone marrow stromal cells contribute to bone formation following infusion into femoral cavities of a mouse model of osteogenesis imperfectaBone201047354655520570757
  • PanaroniCGioiaRLupiAIn utero transplantation of adult bone marrow decreases perinatal lethality and rescues the bone phenotype in the knockin murine model for classical, dominant osteogenesis imperfectaBlood2009114245946819414862
  • NiyibiziCWangSMiZRobbinsPGene therapy approaches for osteogenesis imperfectaGene Ther200411440841614724682
  • SchniekeADziadekMBatemanJIntroduction of the human pro-α1(I) collagen gene into pro-α1(I)-deficient Mov-13 mouse cells leads to formation of functional mouse-human hybrid type I collagenProc Natl Acad Sci U S A19878437647683468512
  • Millington-WardSMcMahonHPFarrarGJEmerging therapeutic approaches for osteogenesis imperfectaTrends Mol Med200511629930515949772
  • WeissBDavidkovaGZhouLWAntisense RNA gene therapy for studying and modulating biological processesCell Mol Life Sci199955333435810228554
  • ForlinoAMariniJCOsteogenesis imperfecta: prospects for molecular therapeuticsMol Genet Metab200071122523211001814
  • EvansCGene delivery to boneAdv Drug Deliv Rev201264121331134022480730
  • RoseLUludağHRealizing the potential of gene-based molecular therapies in bone repairJ Bone Miner Res201328112245226223553878
  • OsawaKOkuboYNakaoKKoyamaNBesshoKOsteoinduction by repeat plasmid injection of human bone morphogenetic protein-2J Gene Med2010121293794421069645
  • VanleeneMShefelbineSJTherapeutic impact of low amplitude high frequency whole body vibrations on the osteogenesis imperfecta mouse boneBone201353250751423352925
  • XieLRubinCJudexSEnhancement of the adolescent murine musculoskeletal system using low-level mechanical vibrationsJ Appl Physiol (1985)200810441056106218258802
  • MalachanneEDureisseixDJourdanFNumerical model of bone remodeling sensitive to loading frequency through a poroelastic behavior and internal fluid movementsJ Mech Behav Biomed Mater20114684985721616466
  • LauneyMEBuehlerMJRitchieROOn the mechanistic origins of toughness in boneAnnu Rev Mater Res2010402553