Bookshelf » GeneReviews » Osteogenesis Imperfecta
 
gene
GeneReviews
PagonRoberta A
BirdThomas C
DolanCynthia R
SmithRichard JH
StephensKaren
University of Washington, Seattle2009
geneticspublic health

GeneTests Home Page About GeneTests Search GeneReviews on the GeneTests web site Laboratory Directory Clinic Directory Educational Materials Illustrated Glossary

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Osteogenesis Imperfecta
[Brittle Bone Disease, OI]

Robert D Steiner, MD
Oregon Health and Science University
Melanie G Pepin, MS, CGC
Department of Pathology
University of Washington Health Sciences Center
Peter H Byers, MD
Department of Pathology
University of Washington Health Sciences Center
28012005oi
Initial Posting: January 28, 2005.

*

*

*

Summary

Disease characteristics. Osteogenesis imperfecta (OI) is a group of disorders characterized by fractures with minimal or absent trauma, dentinogenesis imperfecta (DI), and, in adult years, hearing loss. The clinical features of OI represent a continuum ranging from perinatal lethality to individuals with severe skeletal deformities, mobility impairments, and very short stature to nearly asymptomatic individuals with a mild predisposition to fractures, normal stature, and normal lifespan. Fractures can occur in any bone, but are most common in the extremities. DI is characterized by grey or brown teeth that may appear translucent and wear down and break easily. Before the molecular basis of OI was understood, OI was classified into four types on the basis of mode of inheritance, clinical presentation, and radiographic findings. With detailed radiographic and bone morphologic studies and molecular genetic analyses, the classification has expanded to seven types and it is likely that more will emerge. This classification into types of OI is helpful in providing information about prognosis and management, but it should be remembered that many of the types of OI represent an artificial construct on a broad clinical entity.

Diagnosis/testing. The clinical diagnosis of OI is based on family history, a history of fractures, characteristic physical findings including scleral hue, and radiographic findings. Radiographic findings include fractures of varying ages and stages of healing, wormian bones, "codfish" vertebrae, and osteopenia. Analysis of bone biopsies is an adjunct to the diagnosis of OI type V and OI type VI. Biochemical testing (i.e., analysis of the structure and quantity of type I collagen synthesized in vitro by cultured dermal fibroblasts) detects abnormalities in 98% of individuals with OI type II, about 90% with OI type I, about 84% with OI type IV, and about 84% with OI type III. About 90% of individuals with OI types I, II, III, and IV (but none with OI types V, VI and VII) have an identifiable mutation in either COL1A1 or COL1A2. Such testing is clinically available.

Genetic counseling. Osteogenesis imperfecta types I-V are inherited in an autosomal dominant manner. OI type VII is inherited in an autosomal recessive manner, and the mode of inheritance of OI type VI is not yet certain. For types I-IV, the proportion of cases caused by a de novo mutation in either COL1A1 or COL1A2 varies by the severity of disease. Approximately 60% of individuals with mild OI have de novo mutations; virtually 100% of individuals with lethal (type II) OI and a smaller proportion of those with OI type II have a de novo mutation. Each child of an individual with a dominantly inherited form of OI has a 50% chance of inheriting the mutation and of developing some manifestations of OI. Prenatal testing in at-risk pregnancies can be performed by analysis of collagen synthesized by fetal cells obtained by chorionic villus sampling (CVS) at about 10-12 weeks' gestation if an abnormality of collagen has been identified in cultured cells from the proband. Biochemical analysis of collagen from amniocytes is not useful because amniocytes do not produce type I collagen. Prenatal testing in high-risk pregnancies can be performed by molecular genetic testing of COL1A1 and COL1A2 if the mutation has been identified in an affected relative. Prenatal ultrasound examination performed in a center with experience in diagnosing OI, and done at the appropriate gestational age, can be valuable in the prenatal diagnosis of the lethal form and most severe forms of OI prior to 20 weeks' gestation; fetuses affected with milder forms may be detected later in pregnancy when fractures or deformities occur.

Diagnosis

Clinical Diagnosis

The clinical diagnosis of OI depends on the presence of a number of features (see Table 1). No "diagnostic criteria," such as those for the types of Ehlers-Danlos syndrome, exist for types of OI. Features of OI:

  • Fractures with minimal or no trauma in the absence of other factors, such as abuse or other known disorders of bone

  • Short stature or stature shorter than predicted based on stature of unaffected family members, often with bone deformity

  • Blue sclerae

  • Dentinogenesis imperfecta (DI)

  • Progressive, post-pubertal hearing loss

  • Additional clinical features including ligamentous laxity and other signs of connective tissue abnormality

  • Family history of OI, usually consistent with autosomal dominant inheritance

Table 1. Clinical Features of OI by Type

TypeInheritanceSeverityFracturesBone DeformityStatureDIScleraeHearing Loss
IADMildFew to 100UncommonNormal or slightly short for familyRareBluePresent in about 50%
IIADPerinatal lethalMultiple fracture of ribs, minimal calvarial mineralization, platyspondyly, marked compression of long bonesSevereSeverely short stature+Dark blue
IIIAD; rare recessiveSevereThin ribs, platyspondyly, thin gracile bones with many fractures, "popcorn" epiphyses commonModerate to severeVery short+BlueFrequent
IVADModerate to mildMultipleMild to moderateVariably short stature+/-Normal to greySome
VADModerateMultiple with hypertrophic callusModerateVariableNoNormalNo
VIUncertainModerateMultipleRhizomelic shorteningMild short statureNoNormalNo
VIIARModerateMultipleYesMild short statureNoNormalNo

Radiographic features of OI (Table 2). The radiographic features of OI change with age. The major findings include the following:

  • Fractures of varying ages and stages of healing, often of the long bones but may also include ribs and skull. The metaphyseal chip fractures characteristic of abuse can be seen in a small number of children with OI.

  • "Codfish" vertebrae, which are the consequence of spinal compression fractures, seen primarily in the adult

  • Wormian bones, defined as "sutural bones which are 6 mm by 4 mm (in diameter) or larger, in excess of 10 in number, with a tendency to arrangement in a mosaic pattern" [Cremin et al 1982]. Wormian bones are suggestive of, but not pathognomic for, OI.

  • Protrusio acetabuli (The socket of the hip joint is too deep and the acetabulum bulges into the cavity of the pelvis causing intrapelvic protrusion of the acetabulum.)

  • Osteopenia detected by dual energy X-ray absorptiometry (DEXA). Of note, bone density can be normal, especially in OI type I, as DEXA measures mineral content rather than collagen content [Deodhar & Woolf 1994, Paterson & Mole 1994, Lund et al 1999]. In OI, changes with age of bone mineral density of the spine are primarily caused by increases in vertebral bone volume and secondarily caused by increased mineral density [Kurtz et al 1985, Moore et al 1998, Reinus et al 1998].

    Note: (1) A major determinant of bone density may be the patient's ability to ambulate. (2) Bone density standards for children under the age of two years have been determined after sampling very small populations, often fewer then ten, so that reliability is an issue. (3) Bone density standards for children are based on height; corrections for short stature of severely affected individuals may need to be made.

Table 2. Radiographic Findings of Osteogenesis Imperfecta by Type

TypeSeveritySkullBackExtremitiesOther
IMildWormian bonesCodfish vertebrae (adults)Thin corticesOsteopenia
IIPerinatal lethalUndermineralization; plaques of calcificationPlatyspondylySeverely deformed; broad, crumpled, bent femursSmall beaded ribs; findings are pathognomonic
IIISevereWormian bonesCodfish vertebrae; kyphoscoliosisFlared metaphyses ("popcorn"-like appearance [childhood]), bowing, thin corticesThin ribs, severe osteoporosis
IVIntermediate± Wormian bonesCodfish vertebraeThin corticesProtrusio acetabuli
VIntermediate?Wormian bones?Hypertrophic callus, usually of the femurs; mineralization of the interosseus membrane in the forearm
VIntermediate?Wormian bones?Similar to OI type IV
VIntermediate?Wormian bones?Similar to OI type IVRhizomelic shortening

Testing

Histomorphometric evaluation of iliac crest bone from individuals with types V, VI, and VII OI shows maintained lamellar structure, reduced cortical width and cancellous bone volume, and increased bone remodeling [Rauch et al 2000]. The distinction of OI type VI from OI types IV and V rests in part on a characteristic "fish scale" appearance under polarized light.

Analysis of type 1 collagen synthesized in vitro by culturing dermal fibroblasts obtained from a small skin biopsy shows the structure and quantity of the collagen. The sensitivity of biochemical testing is 87% in non-lethal forms of OI and about 98% in the lethal form. The sensitivity varies with clinical phenotype and reflects the small effect of some COL1A1 or COL1A2 mutations on type I collagen structure and quantity [Wenstrup et al 1990] as well as absence of COL1A1 or COL1A2 mutations in OI types V, VI, and VII, which were not recognized at the time of the survey completed by Wenstrup et al (1990).

Molecular Genetic Testing

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Genes. COL1A1 and COL1A2 are the two genes associated with OI types I, II, III, and IV. They encode the two chains pro α1(I) and pro α2(I), respectively, of type I procollagen.

Other loci

  • OI type VII maps to a 4.5-Mb region on the short arm of chromosome 3.

  • The loci for OI types V and VI have not been mapped.

Molecular genetic testing: Clinical uses

Molecular genetic testing: Clinical methods

Sequence analysis. Sequence analysis of COL1A1 and COL1A2 cDNA to detect mutations in the coding sequence and sequence analysis of COL1A1 and COL1A2 genomic DNA to detect mutations that alter either sequence or stability of mRNA identify close to 100% of mutations in the type I collagen genes. The mutations in most families are unique; only a few recurrent mutations (mostly CpG dinucleotides) are seen in more than one family. Mutation detection rate varies by OI type (Table 3).

The RNA for cDNA sequencing is generally derived from dermal fibroblasts. Although RNA from circulating leukocytes has been used, concerns about mono-allelic amplfication remain because of the very small amounts of illegitimate transcripts available.

Genomic DNA can be isolated from any tissue of the individual being tested.

Table 3 summarizes molecular genetic testing for this disorder.

Table 3. Molecular Genetic Testing Used in Osteogenesis Imperfecta

Test MethodMutations Detected 1 Mutation Detection RateTest
Availability
cDNA sequence analysisMissense mutations, small insertions or deletions, exon-skipping mutations of COL1A1 and COL1A2OI type I: ~100%
OI type II: 98%
OI type III: 60-70% 2
OI type IV: 70-80% 2
Clinical graphic element
Genomic DNA sequence analysisNonsense mutations, missense mutations, splice-site mutations of COL1A1 and COL1A2

1. Description of mutations in type I collagen genes follows the usual conventions with some exceptions (see Molecular Genetic Pathologenesis).
2. In OI type III, the frequency of identified mutations in type I collagen genes is lower than expected for dominant disorders; rare instances of recessive inheritance caused by mutations in type I collagen genes have been identified. Other genes no doubt also play a role.

Interpretation of test results

  • For issues to consider in interpretation of sequence analysis results, click here.

  • It is often important to have mRNA derived from cultured cells to interpret "sequence variants" that have not been previously recognized.

  • Analysis of genomic DNA may miss instances of whole gene deletion unless attention is paid to the state of common polymorphic nucleotides.

  • Sequence variants or small deletions that underlie the primers used for amplification of genomic DNA can lead to mono-allelic amplification; thus small deletions will be missed.

  • Although some features of the methods used may result in unsuccessful detection of alterations in collagen quantity or structure and/or mutation in COL1A1 or COL1A2, failure to identify such abnormalities substantially reduces the likelihood that the tested individual has OI types I, II, III, or IV.

  • In the case of some mutations — exon skipping or small deletions — it may be important to characterize the mutation itself at the genomic DNA level to be sure of the precise mutation.

  • It may be important to characterize the effects of some genomic mutations to be sure that they account for an abnormality at the mRNA or protein level.

Testing Strategy for a Proband

The sensitivity for indentifying OI by molecular genetic testing is similar to the sensitivity of analysis of the structure and quantity of type 1 collagen in cultured fibroblasts from a skin biopsy.

Clinical Description

Natural History

The severity of OI ranges from perinatal lethality to individuals with severe skeletal deformities, mobility impairments, and very short stature to nearly asymptomatic individuals with a mild predisposition to fractures, normal stature, and normal life span. Before the molecular basis of OI was understood, OI was classified into four types based on clinical presentation, radiographic features, family history, and natural history [Sillence et al 1979]. Recent classifications based on additional studies that include bone histomorphometry and molecular genetic characterization have identified additional types of OI (Table 1). OI types V, VI, and VII were identified among those with "OI type IV" because of unique clinical signs, abnormalities in bone, and the absence of COL1A1 and COL1A2 mutations; it is unclear if they will be grouped with OI in the future.

Although this classification into types of OI is helpful in providing information about prognosis and management, the features of different types of OI overlap and it is not always easy to classify individuals. It is helpful to remember that the severity of clinical and radiographic features lies on a continuum and that the "types" are defined using characteristics that appear to form clinical "nodes." Interfamilial variability is apparent among individuals with the same OI type and intrafamilial variability is apparent among individuals with the same mutation. Nonetheless, it is reasonable to continue to think of OI in terms of these types in order to provide information about the expected clinical course.

OI type I. OI type I is characterized by blue sclerae and normal stature. A small proportion of infants with OI type I have femoral bowing at birth. The first fractures may occur at birth or with diapering. More often, the first fractures occur when the infant begins to walk and, more importantly, to fall. Fractures generally occur at a rate of a few to several per year and then decrease in frequency after puberty. Fracture frequency often increases again late in adulthood, especially in postmenopausal women and men beyond the fifth decade [Paterson et al 1984]. Affected individuals may have anywhere from a few fractures to more than 100, but the fractures usually heal normally with no resulting deformity.

Most affected individuals have normal or near normal stature, but may be short compared to other members of their families.

Joint hypermobility may contribute to morbidity.

In their classification of OI, Sillence et al (1979) designated a subset of OI type I with DI (OI type IB). In DI, morbidity results not from dental decay, but rather from premature wearing down of the teeth, which interferes with chewing. DI can be a significant cosmetic issue. Dental eruption in OI type I can sometimes occur early.

Progressive hearing loss occurs in about 50% of adults with OI type I, beginning as a conductive hearing loss but becoming a sensorineural hearing loss in time.

OI type II. Abnormalities characteristic of OI type II are evident at birth. Weight and length are small for gestational age. The sclerae are dark blue and connective tissue is extremely fragile. The skull is large for the body size and soft to palpation. Callus formation on the ribs may be palpable. Extremities are short and bowed. Hips are usually flexed and abducted in a "frog-leg" position. Although some fetuses with OI type II die in utero or are spontaneously aborted, more typically infants die in the immediate perinatal period. More than 60% of affected infants die on the first day; 80% die within the first week; survival beyond one year is exceedingly rare and usually involves intensive support such as continuous assisted ventilation [Byers et al 1988]. Death usually results from pulmonary insufficiency related to the small thorax, rib fractures, or flail chest because of lack of stable ribs. Those who survive the first few days of life may not be able to take in sufficient calories because of respiratory distress.

Histologic evaluation of bone from infants with OI type II shows marked reduction in collagen in secondary trabeculae and cortical bone [Horton et al 1980]. Cortical bone is hypercellular with large osteocytes. Trabeculae contain woven bone with large immature osteoblasts [Cole et al 1992, Cole & Dalgleish 1995].

OI type III. The diagnosis of OI type III is readily apparent at birth. Fractures in the newborn period, simply with handling of the infant, are common. In some affected infants, the number and severity of rib fractures leads to death from pulmonary failure in the first few weeks or months of life. Infants who survive this period generally fare well, although most do not walk without assistance and usually use a wheel chair or other assistance for mobility because of severe bone fragility and marked bone deformity. Affected individuals have as many as 200 fractures and progressive deformity even in the absence of fracture. OI type III is difficult to manage, even with intramedullary rod placement. Growth is extremely slow and adults with OI type III are among the shortest individuals known, with some having adult stature of less than one meter (three feet). Intellect is normal unless there have been intracerebral hemorrhages.

Even within OI type III, there is considerable heterogeneity at the clinical level. Some individuals have normal-appearing teeth and facial appearance while others have DI and a large head with enlarged ventricles that reflect the soft calvarium. Relative macrocephaly and barrel chest deformity are observed. Usually sclerae are blue in infancy but lighten with age. Hearing loss generally begins in the teenage years.

Basilar impression, an abnormality of the craniovertebral junction caused by descent of the skull on the cervical spine, is common. Basilar impression is characterized by invagination of the margins of the foramen magnum upward into the skull, resulting in protrusion of the odontoid process into the foramen magnum. Basilar impression may progress to brainstem compression, obstructive hydrocephalus, or syringomyelia because of direct mechanical blockage of normal CSF flow [Charnas & Marini 1993, Sillence 1994, Hayes et al 1999].

Symptoms become apparent with neck flexion. Findings include posterior skull pain, C2 sensory deficit, tingling in the 4th and 5th digits, and numbness in the medial forearm. When swimming, affected individuals may perceive that water temperature differs below and above the umbilicus. Lhermitte's sign (tingling on neck flexion) can be demonstrated at any stage. Basilar impression can cause headache with coughing, trigeminal neuralgia, loss of function of the extremities, or parasthesias. At its most severe levels of involvement, sleep apnea and death can occur.

OI type IV. OI type IV is characterized by mild short stature, DI, adult-onset hearing loss, and normal-to-grey sclerae. This is the most variable form of OI, ranging in severity from moderately severe to so mild that it may be difficult to make the diagnosis. Stature is variable and may vary markedly within the family. DI is common but may be mild. Sclerae are typically light blue or gray at birth but quickly lighten to near normal. Hearing loss occurs in some. Basilar impression/invagination can occur.

OI type V. This group was initially thought to have either OI type III or OI type IV because of short stature and fractures. Sclerae were generally white. Two features distinguish OI type V: striking hypertrophic callus formation, usually at the site of fractures and most often in the femoral shaft, and calcification of the interosseous membrane between the ulna and the radius that leads to inability to fully supinate and pronate the forearm.

OI type VI. The clinical features of OI type VI are similar to those of OI type IV. The defining features of OI type V are absent.

OI type VII. OI type VII is distinguished by rhizomelic shortening of all limbs.

Other considerations

Serum concentrations of vitamin D, calcium, phosphorous, and alkaline phosphatase, are typically normal; however, the latter is often elevated in response to fracture.

Facial features. Infants and children with OI are often described as having a triangular-shaped face. The skull is relatively large compared to body size.

Skin. Easy bruising can be a feature of OI.

Gastrointestinal. Although complaints of constipation are common in adults with OI who are mobile in wheelchairs, it is not clear if this is a complication of OI itself or of the mode of transport. Bowel obstruction can occur as a result of protrusio acetabuli [Lee et al 1995], but appears to be uncommon.

Cardiovascular. Mitral valve prolapse and aortic dilatation have been reported [McKusick 1972]. Aortic and mitral regurgitation have also been reported [Stein & Kloster 1977, Hortop et al 1986, Vetter et al 1989]. Both atrial and/or aortic rupture have been reported in OI; it is unclear if they occur more commonly in individuals with OI than in the general population. A minority of individuals with OI have a slightly larger than normal aortic root diameter, but the risk of progression or dissection is not increased [Hortop et al 1986].

Development. Delays in gross motor development are common in deforming types of OI. Development in other areas is usually normal, although significant joint hypermobility may delay acquisition of gross motor skills. Intellect is generally normal.

Life expectancy. Life expectancy for those with mild forms of OI is normal, whereas the most severely affected children with type II OI typically do not survive the neonatal period. The natural history of types of OI other than type II is not well understood or documented. Little has been published about adults with OI. Although mean life expectancy in the intermediate forms of OI (type III) may be shortened because of severe kyphoscoliosis and abnormal thoracic shape with attendant restrictive pulmonary disease and cardiac insufficiency, some individuals with these complications have a nearly normal life span [Paterson et al 1996].

Pregnancy. Fertility is normal in OI. Pregnancy in women with OI, especially those with OI type III, can be complicated because of a small pelvis, which may necessitate delivery by cesarian section. The mode of delivery of infants with OI has been examined to determine if the frequency of complications is higher with vaginal or cesarian section delivery. No difference in the frequency of complications was found, but a higher than expected frequency of cesarian sections as a result of non-vertex presentation was noted. The role of pregnancy in later fractures, loss of bone mineralization, progression of hearing loss, or any other physical consideration has not been examined in detail.

For most women who have OI, pregnancy is uncomplicated. The exception is in those women with OI who are very small and require pre-term cesarian section because of respiratory compromise. Joint laxity may increase, as it does with unaffected women, and reduce mobility in small moderately affected women. Bleeding is probably not more common than usual and complications of vaginal tearing during delivery are not common. It is uncertain whether post-partum pelvic relaxation is more common than usual.

Genotype-Phenotype Correlations

Genotype-phenotype correlation is not readily available and prediction of phenotype and natural history for unknown mutations has no precise rules.

OI type I almost always results from mutations in one COL1A1 allele that introduce premature termination codons and decrease the stability of mRNA. These mutations may occur by codon changes, by frame shifts, and by splice mutations that result in use of cryptic splice sites and frame shifts. The type I collagen molecule contains two pro α1(I) chains and a single α2(I) chain. If the number of available pro α1(I) chains decreases, the amount of the trimer manufactured is diminished because no more than one pro α2(I) chain can be accommodated per molecule.

Individuals with OI type IB (i.e., OI type I with DI) have mutations that alter the sequence of type I collagen chains, while those without DI have mutations that change the amount of type I collagen made.

OI types II, III, and IV all result from mutations that alter the structure of either pro α1(I) or pro α2(I) chains. The most common mutations result in substitution of another amino acid for glycine in the triple helical domain of either chain; serine, arginine, cysteine, and tryptophan result from substitutions in the first position of the glycine codon and alanine, valine, glutamic acid, and aspartic acid result from substitutions in the second position of the glycine codon. Substitutions in the pro α1(I) chain by arginine, valine, glutamic acid, aspartic acid, and tryptophan are almost always lethal if they occur in the carboxyl-terminal 70% of the triple helix and have a non-lethal, but still moderately severe phenotype, if they occur in the remainder of the chain. For the smaller side-chain residues (serine, alanine, and cysteine), the phenotypes are more variable and appear to reflect some characteristics of the stability profile of the triple helix that are not yet fully recognized. Much more variability occurs with mutations that affect glycine residues in the pro α2(I) chain, even with the large side-chain residues; therefore, it is more difficult to determine the genotype-phenotype relationship.

The other common mutations affect splice sites. Mutations that lead to exon skipping in the pro α1(I) chain beyond exon 14 and in the pro α2(I) chain beyond exon 25 are generally lethal. The phenotypes resulting from mutations in the upstream region are more variable and may lead to significant joint hypermobility.

A relatively small number of mutations that alter amino acid sequences in the carboxyl-terminal regions of both chains has been identified. These domains are used for chain association and mutations have the capacity to destroy this property or lead to abnormalities in chain association. The phenotypic effects of mutations that affect this domain appear to be milder when they result in exclusion rather than inclusion of the chain.

Mosaicism for dominant mutations has been recognized in OI types II, III, and IV. The phenotype of the mosaic individual can range from no identifiable characteristics of OI to one of the mild forms. Individuals mosaic for mutations that result in non-lethal forms of OI generally have no phenotypic features of OI, even when the mutation is present in a majority of somatic cells. Mosaicism for mutations that result in lethal OI can produce a mild OI phenotype if the mutation is present in the majority of somatic cells; otherwise, the mosaicism is generally asymptomatic.

Penetrance

The penetrance of the dominantly inherited forms of OI for which affected individuals are heterozygous for a COL1A1 or COL1A2 mutation is 100%, although expression may vary considerably, even in the same family.

Nomenclature

The classification scheme of "OI congenita" and "OI tarda" was discarded because fractures at birth can be noted in mild OI and infants with severe OI may not have fractures at birth.

In classifications of genetic conditions, OI may be considered a skeletal dysplasia, a connective tissue disorder, a disorder of collagen or extracellular matrix, or a disorder of bone fragility.

Prevalence

Considering all types, OI has a prevalence of approximately 6-7/100,000. The two mildest forms, OI type I and OI type IV, account for considerably more than half of all OI. OI is found in all racial and ethnic groups. A study in Edinburgh revealed an incidence of those types detectable at birth of one in 20,000 [Wynne-Davies & Gormley 1981]. The most recent estimates of prevalence come from Finland, where OI is thought to affect about 1/15,000 individuals [Kuurila et al 2002]. If the same prevalence is assumed for the US, then about 18,000 people in the US would be affected.

  • OI type I has a prevalence of approximately 3-4/100,000. Sillence (1979) reported an incidence in Australia of 3.5 per 100,000.

  • OI type II has an incidence of about 1-2/100,000. [Sillence (1979) reported an incidence in Australia of 1.6 per 100,000], translating to an approximate prevalence of 1/20,000 in those who survive early childhood mortality. OI type II is often not considered in prevalence data because of early lethality. Increasingly, OI type II is recognized in utero as a consequence of early screening by ultrasound examination, so that the birth frequency in developed countries is dropping.

  • OI type III has a prevalence of 1-2/100,000. Sillence (1979) reported an incidence in Australia of 1.6 per 100,000 for OI type III.

  • Sillence (1979) believed OI type IV to be uncommon. Since then it has been recognized that this is a relatively common form of OI, probably about as common as OI type I.

  • OI type V and OI type VI each account for about 5% of people with OI.

  • OI type VII has been found to date only in a Native Canadian population.

Differential Diagnosis

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

The differential diagnosis of OI depends largely on the age at which the individual is considered [Plotkin 2004]. Clinical features that help to differentiate OI from other conditions include characteristic triangular facies, blue sclerae, joint hypermobility, dental abnormalities, and, in adults, hearing loss.

In Utero

Early prenatal ultrasound examination or radiographic findings may lead to a consideration of hypophosphatasia, thanatophoric dysplasia, campomelic dysplasia, and achondrogenesis as well as OI type II. Experienced sonographers usually have little difficulty in distinguishing among these disorders. In some cases, either biochemical or molecular testing can be a useful adjunct.

Infancy and Childhood

  • Bruck syndrome (OMIM 259450) [Viljoen et al 1989, McPherson & Clemens 1997] is an autosomal recessive condition characterized by bone fragility, congenital joint contractures, clubfeet, normal or blue sclerae, and wormian bones. It results from defects in the lysyl hydroxylase that hydroxylates the amino-terminal lysyl residues involved in crosslink formation [Bank et al 1999].

  • Osteoporosis pseudoglioma syndrome (OMIM 259770) includes bone fragility and fractures, other skeletal deformities, pseudoglioma with blindness in infancy, and other anomalies. It is caused by mutations in the gene encoding the lipoprotein receptor-related protein 5 [Gong et al 1996, 2001].

  • Cole-Carpenter syndrome (OMIM 112240) is characterized by bone deformities, multiple fractures, ocular proptosis, shallow orbits, orbital craniosynostosis, frontal bossing, and hydrocephalus [Cole & Carpenter 1987].

  • Hadju-Cheney syndrome (OMIM 102500) is characterized by short stature, failure to thrive, conductive hearing loss, dysmorphic features, early tooth loss, genitourinary anomalies, osteopenia, pathologic fractures, wormian bones, failure of suture ossification, basilar impression, vertebral abnormalities, kyphoscoliosis, cervical instability, joint laxity, dislocation of the radial head, long bowed fibulae, pseudoclubbing, short distal digits, acroosteolysis, and hirsutism.

  • Gerodermia osteodysplastica (OMIM 231070) is characterized by dwarfism, lax skin, osteoporosis, wormian bones, fractures, vertebral compression, dysmorphic facies.

  • Idiopathic juvenile osteoporosis (IJO) typically presents in pre-adolescents with fractures and osteoporosis. The fracture susceptibility and osteoporosis usually resolve spontaneously with puberty. The etiology of IJO is unknown.

  • Dentiogenesis imperfecta. DI can occur separately from OI as an isolated familial condition as a result of mutations in the DSPP gene on chromosome 4 [Rajpar et al 2002].

  • Non-accidental trauma (child abuse). OI needs to be distinguished from child physical abuse/non-accidental trauma. The prevalence of physical abuse is much greater than the prevalence of OI. On rare occasion, both can occur in the same child. Patient history, family history, physical examination, radiographic imaging, and the clinical course all contribute to the distinction of OI from child abuse. The overlap in clinical features includes multiple or recurrent fractures, fractures that do not match the history of trauma, and the finding of fractures of varying ages and at different stages of healing [Carty 1988, Ablin et al 1990, Steiner et al 1996, Ablin & Sane 1997, Marlowe et al 2002]. The continued occurrence of fractures in a child who has been removed from a possibly abusive situation lends support to the possibility of OI. Metaphyseal and rib fractures, thought to be virtually pathognomonic for child abuse, can occur in OI.


    The presence or absence of blue sclerae is unreliable in distinguishing OI from child abuse because blue sclerae are often found in unaffected normal infants untilabout 18 months of age; children with OI type IV may not have blue sclerae.


    Family history is often unrevealing; families suspected of possible child abuse often provide an unverified family history of frequent fractures; conversely, the family history of individuals with OI often does not reveal any other affected individuals because of a de novo mutation in the proband or the presence of a mild phenotype in relatives.


    Laboratory testing (protein-based studies or molecular genetic testing of COL1A1 and COL1A2) often is not needed, and in some cases the time required to perform such testing can delay proper disposition of child abuse cases.

Management

Evaluations at Initial Diagnosis

  • Hearing evaluation

Treatment of Manifestations

Management focuses on supportive therapy to minimize fractures and maximize function, minimize disability, foster independence, and maintain overall health [Marini & Gerber 1997]. Ideally, OI is managed by a multidisciplinary team including specialists in medical therapy of OI, orthopedics, and rehabilitation medicine. Supportive therapy is individualized depending upon the severity, the degree of impairment, and the age of the patient. Considerable support is generally required by medical personnel to help parents feel comfortable caring for infants with OI type II.

Physical medicine treatment

  • Instruction of parents and other caregivers in safe handling techniques

  • Bracing of limbs with such devices as vacuum pants that are rigid

  • Orthotics to stabilize lax joints

  • Promotion of appropriate physical activity

  • Physical and occupational therapy for increased stability of bone, improved mobility, prevention of contractures, prevention of head and spinal deformity, aerobic fitness, and muscle strengthening

  • Endurance training [Marini 1988, Bleck 1981, Gerber et al 1990, Binder et al 1993, Marini & Gerber 1997]

  • Mobility devices such as scooters and chairs for children and modified automobiles for adults

  • Analgesics for pain

Orthopedic treatment

  • Fractures are treated as they would be in unaffected children and adults with attention to the following:

    • The period of immobility in children with OI should be shortened as much as is practical.

    • Casts should be small and lightweight.

    • Physical therapy should begin as soon as the cast is removed to promote mobility and enhance muscle strength and bone mass.

    • At this time, intramedullary rodding remains a mainstay of orthopedic care to provide anatomic positioning of limbs that permits more normal function. It is not yet clear if treatment with bisphosphonates will change the natural history of bone deformity and diminish the use of surgical intervention (see discussion of bisphosphonate treatment below).

  • Progressive spinal deformities are particularly difficult because of the poor quality of bone in severely affected children. Progressive scoliosis in severe OI does not respond to conservative management and response to surgical intervention may be limited.

Basilar impression. Basilar impression is best evaluated by CT or MRI scanning with views across the base of the skull. Criteria for surgical intervention are not well defined. If surgery is undertaken, it should be done in a center experienced in the procedures used.

Dental treatment. The goals are the maintenance of both primary and permanent dentition, a functional bite or occlusion, optimal gingival health, and overall appearance. Pediatric dentists are the most knowledgeable about DI in children. Some consensus exists that early dental restorative coverage of the primary molars, and if possible, aesthetic coverage of the upper anterior teeth is optimal. Plastic polymers are sometimes used to coat teeth. As anxiety can be an issue with children, pre-medication for anxiolysis, such as nitrous oxide analgesia or midazolam, can be used for treatment in a clinic setting.

If warranted, orthodontic treatment can be initiated, but care must be taken not to fracture teeth with orthodontic appliances because of the brittleness of the teeth.

Dental restorations in adults may best be done by a general dentist knowledgeable about OI or a specialist in prosthetic dentistry.

Treatment of hearing loss. Initial hearing loss in OI is usually conductive as a result of fractures of the bones of the middle ear or contracture and scarring of the incus. Surgical repair of the bones and creation of a prosthetic incus can improve unaided hearing.

Later hearing loss appears to have a significant sensorineural component that does not respond to middle ear surgery. Cochlear implantation has been used in a small number of individuals; outcome data are limited.

Prevention of Secondary Complications

General anesthesia. Special attention should be paid to anesthesia concerns including proper positioning on the operating room table, for which egg crate foam is recommended.

Surveillance

Children with OI should be seen by the dentist twice yearly, beginning in early childhood or even infancy.

Hearing evaluation should be performed at 3-5 year intervals after adolescence until hearing loss is identified.

Therapies Under Investigation

Bisphosphonates, analogs of pyrophosphate that decrease bone resorption, are being evaluated in both uncontrolled and controlled trials to assess the extent to which they can increase bone mass and bone strength and improve function in children with OI. These studies are still ongoing. Bisphosphonates have been used most extensively in severely affected children with OI; they may be useful in adults with OI as well [Adami et al 2003]. Because of the lack of published randomized controlled clinical trials, biosphosphonates should used with caution in the treatment of OI outside of research trials.

An open-label trial of cyclical intravenous Pamidronate (bisphosphonate) was reported by Glorieux and colleagues (1998) and the effects of relatively long term use in adults by Astrom and Soderhall (2002) and Zeitlin et al (2003). Falk et al (2003) replicated the study of Glorieux et al (1998) in children over 22 months of age, but did report one child with fracture non-union following treatment with Pamidronate. No randomized, placebo-controlled clinical trial of Pamidronate has been published. Bisphosphonate treatment has produced improvements in bone histomorphometry, increased bone mineral density (BMD), decreased some biochemical markers of bone resorption, and possibly reduced fracture risk. In addition, some investigators have reported decreased bone pain in young, but not older, children following treatment.

Pamidronate use is invasive and inconvenient, typically requiring intravenous infusions every three months four hours a day for three days and has real and potential complications. Recently, Pamidronate has been offered even to very young children with OI, but complications including transient asymptomatic hypocalcemia [Plotkin et al 2000] and symptomatic hypocalcemia [Chien et al 2002] have been noted. The long-term consequences of lowering bone turnover in children with OI are unknown, but may include delayed bone union after fracture or osteotomy.

A randomized, controlled clinical trial using the oral bisphosphonate, Alendronate, was recently completed and the results are being analyzed.

A newer bisphosphonate, Zoledronic acid, with a longer half life, greater potency, and more convenient dosing is being studied in children with OI.

Injection of human growth hormone has been used in clinical trials in children with OI and appears to improve linear growth rates and bone formation in some children [Antoniazzi et al 1996].

Bone marrow transplantation (BMT) to introduce normal mesenchymal stem cells that have the capacity to differentiate into normal osteoblasts is being evaluated [Horwitz et al 1999, 2001].

Search ClinicalTrials.gov for access to information on clinical studies for a wide range of diseases and conditions.

Other

Management of pregnancy and delivery. Pregnancy presents two issues: The risk to the mother who has OI and the risk to the child of having OI (See Genetic Counseling section). Delivery by cesarian section and by vagina have about the same rate of complications for infants with each type of OI. Delivery by cesarian section is more frequent than in the general population because a non-vertex presentation cannot be corrected by external manipulation. Women with significant skeletal deformity and short stature should be followed by high-risk prenatal care centers.

Management of lethal OI. It is appropriate to offer the parents the option of allowing the infant to expire without attempting interventions such as assisted ventilation.

Other therapies. Early trials of anabolic steroids, sodium fluoride, testosterone, vitamins C and D, flavinoids, and calcitonin showed minimal or no improvement in bone formation, or too small a sample size was utilized for meaningful conclusions [reviewed in Byers & Steiner 1992].

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Mode of Inheritance

Osteogenesis imperfecta types I, II, IV, and V are inherited in an autosomal dominant manner. Osteogenesis imperfecta type III is usually inherited in an autosomal dominant manner; rare instances of autosomal recessive inheritance have been observed. OI type VII is inherited in an autosomal recessive manner. The mode of inheritance of OI type VI is not yet certain.

Risk to Family Members - Autosomal Dominant

Parents of a proband

  • Many individuals diagnosed with the milder forms of OI (type I and some probands with type IV) have an affected parent.

  • The proportion of cases caused by de novo mutations varies by the severity of disease. Approximately 60% of individuals with mild OI have de novo mutations; virtually 100% of individuals with severe (type III) and lethal (type II) OI have de novo mutations.

Recommendations for the evaluation of parents of a proband with an apparent de novo mutation include clinical examination of the parents and molecular genetic testing if the mutation in the proband has been identified.

Sibs of a proband

Offspring of a proband. Each child of an individual with a dominantly inherited form of OI has a 50% chance of inheriting the mutation.

Other family members. The risk to other family members depends upon the status of the proband's parents. If a parent is found to be affected, his or her family members are at risk.

Risk to Family Members - Autosomal Recessive

Parents of a proband

  • The parents of an affected child are obligate heterozygotes and therefore carry one mutant allele.

  • Heterozygotes (carriers) are asymptomatic.

Sibs of a proband

  • At conception, the sibs of an affected individual have a 25% chance of being affected, a 50% chance of being asymptomatic carriers, and a 25% chance of being unaffected and not carriers.

  • Once an at-risk sib is known to be unaffected, the risk of his/her being a carrier is 2/3.

  • Heterozygotes (carriers) are asymptomatic.

Offspring of a proband. The offspring of an individual with OI type VII are obligate heterozygotes (carriers) for a disease-causing mutation.

Other family members of a proband. Sibs of the proband's parents are at 50 % risk of being carriers.

Carrier Detection

Carrier testing using molecular genetic techniques is not offered because it is not clinically available.

Related Genetic Counseling Issues

Considerations in families with an apparent de novo mutation. When neither parent of a proband with an autosomal dominant condition has clinical evidence of the disorder, it is likely that the proband has a de novo mutation or that one parent has germline mosaicism with or without somatic mosaicism. Other explanations, including alternate paternity or undisclosed adoption, could also be explored.

Family planning. The optimal time for determination of genetic risk and discussion of the availability of prenatal testing is before pregnancy.

DNA banking. DNA banking is the storage of DNA (typically extracted from white blood cells) for possible future use. Because it is likely that testing methodology and our understanding of genes, mutations, and diseases will improve in the future, consideration should be given to banking DNA of affected individuals. DNA banking is particularly relevant in situations in which the sensitivity of currently available testing is less than 100%. See DNA Banking for a list of laboratories offering this service.

Prenatal Testing

High-risk pregnancy

  • Molecular genetic testing. Analysis of DNA extracted from fetal cells obtained by amniocentesis usually performed at about 15-18 weeks' gestation* or chorionic villus sampling (CVS) at about 10-12 weeks' gestation is possible if the disease-causing allele of an affected family member has been identified.

  • Biochemical analysis of collagen from fetal cells obtained by CVS at about 10-12 weeks' gestation has been reported. An abnormality of collagen from cultured cells of the proband must be identified before this technique can be used for prenatal testing. Biochemical analysis of collagen from amniocytes is not useful because amniocytes do not produce type I collagen.

Note: OI type I cannot be identified prenatally even with CVS cells because the proportion of type I procollagen produced by the cells is reduced compared to control cells and resemble cells from individuals with OI. Thus false positive testing can be an issue.

  • Prenatal ultrasound examination performed in a center with experience in diagnosing OI, done at the appropriate gestational age, can be a valuable tool in the prenatal diagnosis of OI. Normally, ultrasound examination detects only the lethal and most severe forms of OI prior to 20 weeks' gestation; fetuses affected with milder forms may be detected later in pregnancy when fractures or deformity occur.

    • OI type II: The bony abnormalities can first be seen by ultrasound examination by about 13-14 weeks' gestation. By 16 weeks, femoral length is typically two or more weeks delayed, calvarial mineralization is essentially absent, and ribs generally have identified fractures.

    • OI type III: Limb length generally begins to fall off the growth curve at about 17-18 weeks' gestation; serial ultrasound examinations are required.

Low-risk pregnancy. Routine prenatal ultrasound examination may identify a fetus not known to be at risk for OI with findings suggestive of OI (Type II or III) including bowed, crumpled femurs, beaded ribs, evidence of fractures, and markedly diminished calvarial mineralization. As a part of the evaluation of such findings, molecular genetic testing for COL1A1 or COL1A2 mutations may be considered. Inability to identify a mutation, however, does not eliminate the diagnosis of OI in the fetus.

*Gestational age is expressed as menstrual weeks calculated either from the first day of the last normal menstrual period or by ultrasound measurements.

Molecular Genetics

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Table A. Molecular Genetics of Osteogenesis Imperfecta

Gene SymbolChromosomal LocusProtein Name
COL1A117q21.3-q22Collagen alpha 1(I) chain
COL1A27q22.1Collagen alpha 2(I) chain

Data are compiled from the following standard references: Gene symbol from HUGO; chromosomal locus, locus name, critical region, complementation group from OMIM; protein name from Swiss-Prot.

Table B. OMIM Entries for Osteogenesis Imperfecta

120150 COLLAGEN, TYPE I, ALPHA-1; COL1A1
120160 COLLAGEN, TYPE I, ALPHA-2; COL1A2
166200 OSTEOGENESIS IMPERFECTA, TYPE I
166210 OSTEOGENESIS IMPERFECTA CONGENITA; OIC
166220 OSTEOGENESIS IMPERFECTA, TYPE IV; OI4
259420 OSTEOGENESIS IMPERFECTA, PROGRESSIVELY DEFORMING, WITH NORMAL SCLERAE

Table C. Genomic Databases for Osteogenesis Imperfecta

Gene SymbolLocus SpecificEntrez GeneHGMD
COL1A1COL1A11277 (MIM No. 120150)COL1A1
COL1A2COL1A21278 (MIM No. 120160)COL1A2

For a description of the genomic databases listed, click here.

Molecular Genetic Pathogenesis

More than 90% of individuals with OI have mutations in one of the two genes, COL1A1 and COL1A2, that encode the chains of type I procollagen, the major protein in bone and most other connective tissues. The two general outcomes of these mutations are either a decrease in the amount of type I procollagen produced or the production of some abnormal type I procollagen molecules. OI type I is generally characterized by decreased production of type I procollagen. In the vast majority of instances, OI type I results from mutations in the COL1A1 gene that result in premature termination codons. The majority of these mutations are deletions or insertions of a small number of nucleotides, a number not divisible by three, in the coding sequences of exons throughout the gene. These mutations, single codon changes that introduce premature termination codons, and some splice-site mutations that lead to exclusive use of cryptic sites and generation of out-of-frame transcripts all lead to premature termination codons. The presence of a premature termination codon that is separated by one or more introns in the gene leads to marked instability of the mRNA derived from the mutant allele. As a consequence, the amount of COL1A1 mRNA is reduced to half the normal amount, with no compensation by the other allele. Type I procollagen is a trimer that must contain at least two pro α1(I) chains. With a reduction in the COL1A1 mRNA, an obligatory decrease in the production of type I procollagen occurs, although the protein produced is structurally normal. The diminished amount of type I collagen in bone appears to reduce the amount of bone that can be made and leads to brittle bones.

The majority of mutations that result in OI type II, III, and IV result in substitutions for glycine within the triple helical domain of the pro αchain. In the products of both genes, the triple helical domain contains 1014 amino acids in the repeating sequence of Gly-X-Y without interruption. The glycine residues are essential for the normal folding of the molecule and any substituting amino acid (eight possibilities result from single nucleotide substitutions in glycine codons) appears to result in OI. These mutations are designated by the position of glycine (1-1012) in the triple helical domain. There are 160 amino acids amino terminal to this position in the pro α1(I) chain and 90 in the pro α2(I) chain. In the major databases, both positions are generally reported.

The other types of OI result from synthesis of pro αchains that have altered sequences. The vast majority of these mutations result in substitutions of glycine residues in the triple helical portion of the chains of type I procollagen. The pro αchains consist of an amino-terminal propeptide, a triple helical segment of 1014 amino acids in which glycine is in every third position and prolines preceding glycine residues are generally hydroxylated as are some lysyl residues in the Y-position of the Gly-X-Y triplet. Glycine, the smallest amino acid, must be in the third position to allow proper chain folding to occur. When substituted, the propagation of the triple helix is delayed, additional post-translation modification occurs, and some of the assembled trimers are never secreted. The consequence of these mutations is that a diminished amount of type I procollagen is secreted and some of the protein in the matrix has an abnormal structure. Mutations in the two genes result in a different proportion of abnormal molecules, but all phenotypes can result from mutations in either gene. The clinical consequence in these circumstances appears to result from the position of the substituted glycine, the chain in which the substitution occurs, and the nature of the substituting amino acid.

Similar phenotypes (OI type II, III, or IV) can result from short deletions or duplications of single amino acids or Gly-X-Y triplets and from exon-skipping events. The relationship between genotype and phenotype is complex, although it appears that mutations closer to the 5' end of the coding sequence that affect the amino-terminal ends of the triple helical domains are likely to have milder clinical phenotypes. This is probably the reflection of chain association occurring at the carboxyl-terminal end of the chain so that a small part of the molecule is affected. Much is still to be learned about the final pathways of molecular pathogenesis.

Resources

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

Canadian Osteogenesis Imperfecta Society
128 Thornhill Crescent
Chatham, Ontario, Canada, N7L 4M3
Phone: 519 -436-0025

National Library of Medicine Genetics Home Reference
Osteogenesis Imperfecta

Osteogenesis Imperfecta Foundation
804 West Diamond Avenue, Suite 210
Gaithersburg, MD 20878
Phone: 800-981-2663; 301-947-0083
Fax: 301-947-0456
Email: bonelink@oif.org
http://www.oif.org

The Brittle Bone Society
30 Guthrie Street
Dundee, UK
DD1 5BS
Phone: (+44) 01382- 204446
Fax: (+44) 01382- 206771
Email: bbs@brittlebone.org
http://www.brittlebone.org/

Children's Brittle Bone Foundation
7701 95th Street
Pleasant Prairie, WI 53158
Phone: 866-694-2223
Fax: 262-947-0724
Email: info@cbbf.org
http://www.cbbf.org/

References

Medical Genetics Searches: A specialized PubMed search designed for clinicians that is located on the PubMed Clinical Queries page. graphic element

Literature Cited

Ablin DS, Greenspan A, Reinhart M, Grix A. Differentiation of child abuse from osteogenesis imperfecta. AJR Am J Roentgenol. 1990; 154: 103546. [PubMed]
Ablin DS, Sane SM. Non-accidental injury: confusion with temporary brittle bone disease and mild osteogenesis imperfecta. Pediatr Radiol. 1997; 27: 1113. [PubMed]
Adami S, Gatti D, Colapietro F, Fracassi E, Braga V, Rossini M, Tato L. Intravenous neridronate in adults with osteogenesis imperfecta. J Bone Miner Res. 2003; 18: 12630. [PubMed]
Antoniazzi F, Bertoldo F, Mottes M, Valli M, Sirpresi S, Zamboni G, Valentini R, Tato L. Growth hormone treatment in osteogenesis imperfecta with quantitative defect of type I collagen synthesis. J Pediatr. 1996; 129: 4329. [PubMed]
Astrom E, Soderhall S. Beneficial effect of long term intravenous bisphosphonate treatment of osteogenesis imperfecta. Arch Dis Child. 2002; 86: 35664. [PubMed]
Bank RA, Robins SP, Wijmenga C, Breslau-Siderius LJ, Bardoel AF, van der Sluijs HA, Pruijs HE, TeKoppele JM. 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. 1999; 96: 10548. [PubMed]
Binder H, Conway A, Hason S, Gerber LH, Marini J, Berry R, Weintrob J. Comprehensive rehabilitation of the child with osteogenesis imperfecta. Am J Med Genet. 1993; 45: 2659. [PubMed]
Bleck EE. Nonoperative treatment of osteogenesis imperfecta: orthotic and mobility management. Clin Orthop. 1981; 159: 11122. [PubMed]
Byers PH, Steiner RD. Osteogenesis imperfecta. Annu Rev Med. 1992; 43: 26982. [PubMed]
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: 23748. [PubMed]
Carty H. Brittle or battered. Arch Dis Child. 1988; 63: 3502. [PubMed]
Charnas LR, Marini JC. Communicating hydrocephalus, basilar invagination, and other neurologic features in osteogenesis imperfecta. Neurology. 1993; 43: 26038. [PubMed]
Chien YH, Chu SY, Hsu CC, Hwu WL. Pamidronate treatment of severe osteogenesis imperfecta in a newborn infant. J Inherit Metab Dis. 2002; 25: 5935.
Cole DE, Carpenter TO. Bone fragility, craniosynostosis, ocular proptosis, hydrocephalus, and distinctive facial features: a newly recognized type of osteogenesis imperfecta. J Pediatr. 1987; 110: 7680. [PubMed]
Cole WG, Dalgleish R. Perinatal lethal osteogenesis imperfecta. J Med Genet. 1995; 32: 2849. [PubMed]
Cole WG, Patterson E, Bonadio J, Campbell PE, Fortune DW. The clinicopathological features of three babies with osteogenesis imperfecta resulting from the substitution of glycine by valine in the pro alpha 1 (I) chain of type I procollagen. J Med Genet. 1992; 29: 1128. [PubMed]
Cremin B, Goodman H, Spranger J, Beighton P. Wormian bones in osteogenesis imperfecta and other disorders. Skeletal Radiol. 1982; 8: 358. [PubMed]
Deodhar AA, Woolf AD. Bone density measurement in osteogenesis imperfecta may well be important. Postgrad Med J. 1994; 70: 4634. [PubMed]
Falk MJ, Heeger S, Lynch KA, DeCaro KR, Bohach D, Gibson KS, Warman ML. Intravenous bisphosphonate therapy in children with osteogenesis imperfecta. Pediatrics. 2003; 111: 5738.
Gerber LH, Binder H, Weintrob J, Grange DK, Shapiro J, Fromherz W, Berry R, Conway A, Nason S, Marini J. Rehabilitation of children and infants with osteogenesis imperfecta. A program for ambulation. Clin Orthop. 1990; 251: 25462. [PubMed]
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. 1998; 339: 94752. [PubMed]
Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, Wang H, Cundy T, Glorieux FH, Lev D, Zacharin M, Oexle K, Marcelino J, Suwairi W, Heeger S, Sabatakos G, Apte S, Adkins WN, Allgrove J, Arslan-Kirchner M, Batch JA, Beighton P, Black GC, Boles RG, Boon LM, Borrone C, Brunner HG, Carle GF, Dallapiccola B, De Paepe A, Floege B, Halfhide ML, Hall B, Hennekam RC, Hirose T, Jans A, Juppner H, Kim CA, Keppler-Noreuil K, Kohlschuetter A, LaCombe D, Lambert M, Lemyre E, Letteboer T, Peltonen L, Ramesar RS, Romanengo M, Somer H, Steichen-Gersdorf E, Steinmann B, Sullivan B, Superti-Furga A, Swoboda W, van den Boogaard MJ, Van Hul W, Vikkula M, Votruba M, Zabel B, Garcia T, Baron R, Olsen BR, Warman ML. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001; 107: 51323. [PubMed]
Gong Y, Vikkula M, Boon L, Liu J, Beighton P, Ramesar R, Peltonen L, Somer H, Hirose T, Dallapiccola B, De Paepe A, Swoboda W, Zabel B, Superti-Furga A, Steinmann B, Brunner HG, Jans A, Boles RG, Adkins W, van den Boogaard MJ, Olsen BR, Warman ML. Osteoporosis-pseudoglioma syndrome, a disorder affecting skeletal strength and vision, is assigned to chromosome region 11q12-13. Am J Hum Genet. 1996; 59: 14651. [PubMed]
Grant SF, Reid DM, Blake G, Herd R, Fogelman I, Ralston SH. Reduced bone density and osteoporosis associated with a polymorphic Sp1 binding site in the collagen type I alpha 1 gene. Nat Genet. 1996; 14: 2035. [PubMed]
Hayes M, Parker G, Ell J, Sillence D. Basilar impression complicating osteogenesis imperfecta type IV: the clinical and neuroradiological findings in four cases. J Neurol Neurosurg Psychiatry. 1999; 66: 35764. [PubMed]
Horton WA, Dockery N, Sillence D, Rimoin DL. An embedding method for histochemical studies of undecalcified skeletal growth plate. Stain Technol. 1980; 55: 1929. [PubMed]
Hortop J, Tsipouras P, Hanley JA, Maron BJ, Shapiro JR. Cardiovascular involvement in osteogenesis imperfecta. Circulation. 1986; 73: 5461. [PubMed]
Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, Sussman M, Orchard P, Marx JC, Pyeritz RE, Brenner MK. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med. 1999; 5: 30913. [PubMed]
Horwitz EM, Prockop DJ, Gordon PL, Koo WW, Fitzpatrick LA, Neel MD, McCarville ME, Orchard PJ, Pyeritz RE, Brenner MK. Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta. Blood. 2001; 97: 122731. [PubMed]
Kurtz D, Morrish K, Shapiro J. Vertebral bone mineral content in osteogenesis imperfecta. Calcif Tissue Int. 1985; 37: 148. [PubMed]
Kuurila K, Kaitila I, Johansson R, Grenman R. Hearing loss in Finnish adults with osteogenesis imperfecta: a nationwide survey. Ann Otol Rhinol Laryngol. 2002; 111: 93946. [PubMed]
Lee JH, Gamble JG, Moore RE, Rinsky LA. Gastrointestinal problems in patients who have type-III osteogenesis imperfecta. J Bone Joint Surg Am. 1995; 77: 13526. [PubMed]
Lund AM, Molgaard C, Muller J, Skovby F. Bone mineral content and collagen defects in osteogenesis imperfecta. Acta Paediatr. 1999; 88: 10838.
Marini JC. Osteogenesis imperfecta: comprehensive management. Adv Pediatr. 1988; 35: 391426. [PubMed]
Marini JC, Gerber NL. Osteogenesis imperfecta. Rehabilitation and prospects for gene therapy. JAMA. 1997; 277: 74650. [PubMed]
Marlowe A, Pepin MG, Byers PH. Testing for osteogenesis imperfecta in cases of suspected non- accidental injury. J Med Genet. 2002; 39: 3826. [PubMed]
Mayer SA, Rubin BS, Starman BJ, Byers PH. Spontaneous multivessel cervical artery dissection in a patient with a substitution of alanine for glycine (G13A) in the alpha 1 (I) chain of type I collagen. Neurology. 1996; 47: 5526. [PubMed]
McKusick VA. Osteogenesis imperfecta. In: McKusick VA. Heritable Disorders of Connective Tissue, 4 ed. Mosby, St Louis. 1972
McPherson E, Clemens M. Bruck syndrome (osteogenesis imperfecta with congenital joint contractures): review and report on the first North American case. Am J Med Genet. 1997; 70: 2831. [PubMed]
Moore MS, Minch CM, Kruse RW, Harcke HT, Jacobson L, Taylor A. The role of dual energy x-ray absorptiometry in aiding the diagnosis of pediatric osteogenesis imperfecta. Am J Orthop. 1998; 27: 797801. [PubMed]
Nuytinck L, Freund M, Lagae L, Pierard GE, Hermanns-Le T, De Paepe A. Classical Ehlers-Danlos syndrome caused by a mutation in type I collagen. Am J Hum Genet. 2000; 66: 1398402. [PubMed]
Paterson CR, McAllion S, Stellman JL. Osteogenesis imperfecta after the menopause. N Engl J Med. 1984; 310: 16946. [PubMed]
Paterson CR, Mole PA. Bone density in osteogenesis imperfecta may well be normal. Postgrad Med J. 1994; 70: 1047. [PubMed]
Paterson CR, Ogston SA, Henry RM. Life expectancy in osteogenesis imperfecta. BMJ. 1996; 312: 351. [PubMed]
Plotkin H. Syndromes with congenital brittle bones. BMC Pediatr. 2004; 4: 16. [PubMed]
Plotkin H, Rauch F, Bishop NJ, Montpetit K, Ruck-Gibis J, Travers R, Glorieux FH. Pamidronate treatment of severe osteogenesis imperfecta in children under 3 years of age. J Clin Endocrinol Metab. 2000; 85: 184650. [PubMed]
Rajpar MH, Koch MJ, Davies RM, Mellody KT, Kielty CM, Dixon MJ. Mutation of the signal peptide region of the bicistronic gene DSPP affects translocation to the endoplasmic reticulum and results in defective dentine biomineralization. Hum Mol Genet. 2002; 11: 255965.
Rauch F, Travers R, Parfitt AM, Glorieux FH. Static and dynamic bone histomorphometry in children with osteogenesis imperfecta. Bone. 2000; 26: 5819. [PubMed]
Reinus WR, McAlister WH, Schranck F, Chines A, Whyte MP. Differing lumbar vertebral mineralization rates in ambulatory pediatric patients with osteogenesis imperfecta. Calcif Tissue Int. 1998; 62: 1720. [PubMed]
Sillence DO. Craniocervical abnormalities in osteogenesis imperfecta: genetic and molecular correlation. Pediatr Radiol. 1994; 24: 42730. [PubMed]
Sillence DO, Senn A, Danks DM. Genetic heterogeneity in osteogenesis imperfecta. J Med Genet. 1979; 16: 10116. [PubMed]
Stein D, Kloster FE. Valvular heart disease in osteogenesis imperfecta. Am Heart J. 1977; 94: 63741. [PubMed]
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: 5427. [PubMed]
Uitterlinden AG, Burger H, Huang Q, Yue F, McGuigan FE, Grant SF, Hofman A, van Leeuwen JP, Pols HA, Ralston SH. Relation of alleles of the collagen type Ialpha1 gene to bone density and the risk of osteoporotic fractures in postmenopausal women. N Engl J Med. 1998; 338: 101621. [PubMed]
Vetter U, Maierhofer B, Muller M, Lang D, Teller WM, Brenner R, Frohneberg D, Worsdorfer O. Osteogenesis imperfecta in childhood: cardiac and renal manifestations. Eur J Pediatr. 1989; 149: 1847. [PubMed]
Viljoen D, Versfeld G, Beighton P. Osteogenesis imperfecta with congenital joint contractures (Bruck syndrome). Clin Genet. 1989; 36: 1226. [PubMed]
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: 97582. [PubMed]
Wynne-Davies R, Gormley J. Clinical and genetic patterns in osteogenesis imperfecta. Clin Orthop. 1981; 159: 2635. [PubMed]
Zeitlin L, Rauch F, Plotkin H, Glorieux FH. Height and weight development during four years of therapy with cyclical intravenous pamidronate in children and adolescents with osteogenesis imperfecta types I, III, and IV. Pediatrics. 2003; 111: 10306.

Published Statements and Policies Regarding Genetic Testing

No specific guidelines regarding genetic testing for this disorder have been developed.

Next
GeneReviews2009
(navigation arrows) Go to previous chapter Go to next chapter Go to top of this page Go to bottom of this page Go to Table of Contents