Entry - *603381 - FILAMIN B; FLNB - OMIM
* 603381

FILAMIN B; FLNB


Alternative titles; symbols

FILAMIN, BETA
ACTIN-BINDING PROTEIN 276/278; ABP276/278


Other entities represented in this entry:

TRUNCATED ACTIN-BINDING PROTEIN, INCLUDED; TABP, INCLUDED
ACTIN-BINDING PROTEIN, TRUNCATED, INCLUDED
FILAMIN HOMOLOG 1, INCLUDED; FH1, INCLUDED

HGNC Approved Gene Symbol: FLNB

Cytogenetic location: 3p14.3     Genomic coordinates (GRCh38): 3:58,008,422-58,172,251 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
3p14.3 Atelosteogenesis, type I 108720 AD 3
Atelosteogenesis, type III 108721 AD 3
Boomerang dysplasia 112310 AD 3
Larsen syndrome 150250 AD 3
Spondylocarpotarsal synostosis syndrome 272460 AR 3

TEXT

Description

Filamins, such as FLNB, are actin-binding proteins that also interact with multiple receptors and intracellular proteins that regulate cytoskeleton-dependent cell proliferation, differentiation, and migration (Hu et al., 2014).


Cloning and Expression

The platelet GpIb complex (see 138720) mediates the adherence of platelets at the site of vascular injury through the binding of GpIb-alpha (231200) to subendothelial von Willebrand factor (VWF; 613160). In platelets, the GpIb complex is tightly bound to the actin cytoskeleton via an interaction of GpIb-alpha with ABP280 (filamin A; 300017). Using a yeast 2-hybrid screen with the cytoplasmic tail of GpIb-alpha as bait, Takafuta et al. (1998) isolated partial cDNAs encoding a novel filamin homolog that they designated beta-filamin. They used the partial cDNAs to screen a placenta library and recovered additional cDNAs corresponding to the entire beta-filamin coding region. Like ABP280, the predicted 2,602-amino acid protein contains an N-terminal actin-binding domain, a backbone of 24 tandem repeats, and 2 hinge regions. Excluding the unique first hinge region of beta-filamin, the sequences of beta-filamin and ABP280 are 70% identical. Antibodies against beta-filamin detected a 280-kD protein on Western blots of human umbilical vein endothelial cell (HUVEC) extracts and stained normal human endothelial cells in culture and in situ. Takafuta et al. (1998) determined that the GpIb-alpha-binding domain in beta-filamin is in repeats 17-20, a region that corresponds to the GpIb-alpha-binding domain in ABP280. Northern blot analysis revealed that beta-filamin is expressed as 2 approximately 9.5-kb mRNAs in many adult tissues. The 2 different transcripts appear to result from use of alternative polyadenylation signals. Takafuta et al. (1998) concluded that beta-filamin is a new member of the filamin family that may have significance for GpIb-alpha function in endothelial cells and platelets.

Independently, Xu et al. (1998) isolated cDNAs encoding beta-filamin, which they referred to as ABP278. These authors also identified alternatively spliced mRNAs encoding ABP276, a beta-filamin isoform missing the first hinge region. RT-PCR analysis indicated that the 2 isoforms were expressed at different relative levels in various human tissues.

The addition of thyroid-stimulating hormone (TSH; see 188540) to cultured thyroid follicular cells induces rapid and profound disruption of actin microfilaments. Using serum from a Graves disease (275000) patient, Leedman et al. (1993) identified a thyroid cDNA encoding TABP (truncated actin-binding protein), a predicted 195-amino acid protein with homology to the C terminus of ABP280. Both Xu et al. (1998) and Takafuta et al. (1998) considered TABP to be a truncated form of beta-filamin.


Mapping

By analysis of somatic cell hybrids, Zhang et al. (1998) mapped the FH1 gene to chromosome 3. Takafuta et al. (1998) refined the map position to 3p21.1-p14.3 based on inclusion of a previously mapped STS within the beta-filamin sequence. By FISH, Brocker et al. (1999) assigned the FLNB gene to 3p14.3. Chakarova et al. (2000) mapped FLNB to 3p14 by radiation hybrid analysis.


Gene Function

Using antigen-capture ELISA, Takafuta et al. (1998) found that beta-filamin associates with GpIb-alpha in both platelets and HUVEC extracts.

Mutations in the presenilin genes PS1 (104311) and PS2 (600759) account for approximately 50% of early-onset familial Alzheimer disease (AD; 104300). Zhang et al. (1998) identified beta-filamin as filamin homolog 1 (FH1), a filamin-related protein that interacts with the loop regions of PS1 and PS2. A monoclonal antibody recognizing both ABP280 and FH1 bound to blood vessels and astrocytes in the normal brain. In the brains of AD patients, Zhang et al. (1998) observed staining also in neurofibrillary tangles, neuropil threads, and dystrophic neurites within some senile plaques. The authors stated that detection of these presenilin-interacting proteins in these brain structures suggests that ABP280 and FH1 may be involved in the development of AD and that interactions between presenilins and ABP280/FH1 may be functionally significant. Takafuta et al. (1998) noted that the FH1 sequence is identical to the C-terminal 291 amino acids of beta-filamin except for 2 residues, making it very likely that FH1 represents the C-terminal region of beta-filamin.

Krakow et al. (2004) found that FLNB is expressed in human growth plate chondrocytes and in developing vertebral bodies in the mouse. The authors concluded that FLNB plays a role in vertebral segmentation, joint formation, and endochondral ossification.

Mutation in the X-linked gene filamin A (FLNA) can cause the neurologic disorder periventricular heterotopia (300049). Although periventricular heterotopia is characterized by a failure in neuronal migration into the cerebral cortex with consequent formation of nodules in the ventricular and subventricular zones, many neurons appear to migrate normally, even in males, suggesting compensatory mechanisms. Sheen et al. (2002) showed that, in mice, Flna mRNA was widely expressed in all brain cortical layers, whereas a homolog, Flnb, was most highly expressed in the ventricular and subventricular zones during development. In adulthood, widespread but reduced expression of Flna and Flnb persisted throughout the cerebral cortex. Flna and Flnb proteins were highly expressed in both the leading processes and somata of migratory neurons during corticogenesis. Postnatally, Flna immunoreactivity was largely localized to the cell body, whereas Flnb was localized to the soma and neuropil during neuronal differentiation. The putative Flnb homodimerization domain strongly interacted with itself or the corresponding homologous region of Flna, as shown by yeast 2-hybrid interaction. The 2 proteins colocalized within neuronal precursors by immunocytochemistry, and the existence of Flna-Flnb heterodimers could be detected by coimmunoprecipitation. Sheen et al. (2002) suggested that FLNA and FLNB may form both homodimers and heterodimers, and that their interaction could potentially compensate for the loss of FLNA function during cortical development within patients with periventricular heterotopia.

Using yeast 2-hybrid and immunoprecipitation analyses, Hu et al. (2014) found that mouse Flnb and Flna interacted directly with the actin nucleating-protein Fmn1 (136535). The filamins and Fmn1 colocalized in cytoplasm and, to a lesser extent, nucleus, and they were coexpressed in chondrocytes.


Molecular Genetics

Krakow et al. (2004) identified mutations in the FLNB gene in 4 human skeletal disorders: spondylocarpotarsal syndrome (SCT; 272460), Larsen syndrome (LRS; 150250), type I atelosteogenesis (AO1; 108720), and type III atelosteogenesis (AO3; 108721).

Biesecker (2004) commented that from the standpoint of the clinical geneticist, 4 distinct disorders result from mutation in the FLNB gene. In contrast, a basic scientist might view them as a single disorder with inconsequential phenotypic differences. The information concerning the multiple disorders caused by mutations in the FLNB gene followed closely on the heels of reports of mutations in the FLNA gene (300017) as the cause of 5 distinct disorders. This was of interest because there are overlapping phenotypic features in the disorders associated with FLNA and FLNB. Biesecker (2004) pointed out that the 'FLNB story' used information from the International Skeletal Dysplasia Registry (ISDR), which is maintained by a skilled group of clinical scientists and includes information on more than 12,000 cases of individuals with disorders that fall into 50 diagnostic groups. One reason for the success of the registry is that it combines clinical service with research archives. The motivation for a clinician to submit cases to the registry is that he or she can receive an expert opinion on the diagnosis (which is useful for medical care and estimating recurrence risks) and, as in the FLNB story, contribute to research.

In a 22-week male fetus previously studied by Krakow et al. (2004) and a 17-week male fetus previously described by Wessels et al. (2003), both diagnosed with boomerang dysplasia (BOOMD; 112310), Bicknell et al. (2005) identified heterozygosity for mutations in the FLNB gene, leu171 to arg (L171R; 603381.0009) and ser235 to pro (S235P; 603381.0010), respectively.

Farrington-Rock et al. (2006) found 14 novel missense mutations in FLNB in 15 unrelated patients with atelosteogenesis I and/or atelosteogenesis III. Most of the mutations resided in exons 2 and 3, which encode the CH2 domain of the actin-binding region of filamin B. The remaining mutations were found in exon 28 and exon 29, which encode repeats 14 and 15 of filamin B. Clinical and radiographic data were used to confirm the diagnosis of atelosteogenesis in all the patients. The diagnosis of type I was given to patients showing classic findings of absent, shortened, or distally tapered humeri and femora; absent, shortened, or bowed radii; shortened and bowed ulnae and tibiae; and absent fibulae. Other findings included vertebral hypoplasia with coronal clefts, 11 ribs, shortened wide distal phalanges, and unossified or partially ossified metacarpals and middle and proximal phalanges. In addition, most type I patients showed evidence of a hypoplastic pelvis, dislocations of the hips, elbows, and knees, and clubbed feet. With the exception of 1 patient where the pregnancy was terminated after 24 weeks' gestation, the patients given the diagnosis of type I either died in the neonatal period or were stillborn. The diagnosis of type III was given to patients when all bones were present in the extremities, when there was distal tapering of the humeri or femora, and where the small tubular bones of the hands and feet were shortened and broad. In 2 patients with type III, the fibulae were absent. Dislocation of the elbows, hips, and knees and clubbed feet were also present in the type III patients, as was vertebral hypoplasia with coronal clefting. Cartilage of these patients showed acellular areas within the growth plate and the presence of large multinuclear cells ('giant cells') within the resting zone as had been described previously.

Bicknell et al. (2007) identified heterozygous mutations in the FLNB gene (see, e.g., 603381.0011; 603381.0012) in 20 unrelated patients with Larsen syndrome. The distribution of mutations within the gene was nonrandom, with clusters of mutations in the actin-binding domains and filamin repeats 13 through 17 being the most common.

In a female infant with atelosteogenesis who died 3 hours after birth due to respiratory failure, Jeon et al. (2014) performed postmortem exome sequencing and identified heterozygosity for a de novo A173T mutation (603381.0015) in the FLNB gene that was not found in 50 healthy controls. The authors noted that most lethal FLNB-related disorders are caused by de novo mutations, and thus there is a low risk of recurrence in subsequent pregnancies.

In 7 families with SCT syndrome, Salian et al. (2018) identified 2 nonsense and 5 frameshift variants in the FLNB gene (see, e.g., 603381.0016), all in homozygous state.


Animal Model

Zhou et al. (2007) detected strong expression of the mouse Flnb gene in vascular endothelial cells and chondrocytes. In Flnb -/- mice, the authors observed a phenotype that resembled those of human skeletal disorders with mutations in the FLNB gene. Less than than 3% of Flnb -/- embryos reached term, indicating that the Flnb gene is important in embryonic development, whereas Flnb +/- mice were indistinguishable from their wildtype sibs. Flnb -/- embryos had impaired development of microvasculature and skeletal systems. The few that were born were very small and had scoliotic and kyphotic spines, lack of intervertebral discs, fusion of vertebral bodies, and reduced hyaline matrix in bones of the extremities, thorax, and vertebrae.

Farrington-Rock et al. (2008) generated Flnb -/- mice and observed a phenotype of short stature and skeletal abnormalities similar to those of individuals with spondylocarpotarsal synostosis syndrome (SCT; 272460). Newborn Flnb -/- mice had fusions between the neural arches of the vertebrae in the cervical and thoracic spine. At postnatal day 60, the vertebral fusions were more widespread and involved the vertebral bodies as well as the neural arches. In addition, fusions were seen in sternum and carpal bones. Analysis of the Flnb -/- mice phenotype showed that an absence of filamin B causes progressive vertebral fusions, in contrast to the previous hypothesis that SCT results from failure of normal spinal segmentation. Farrington-Rock et al. (2008) suggested that spinal segmentation can occur normally in the absence of filamin B, but that the protein is required for maintenance of intervertebral, carpal, and sternal joints, and the joint fusion process commences antenatally.

Hu et al. (2014) found that knockout (KO) of both Flnb and Fmn1 in mice resulted in a more severe reduction in body size, weight, and growth plate length than that observed in mice with KO of either gene alone. In Flnb/Fmn1 double-KO mice, shortening of long bones was associated with decreased chondrocyte proliferation and an overall delay in ossification. Comparison of Fmn1 KO mice with Flnb/Fmn1 double-KO mice revealed nonoverlapping functions for Fmn1 and Flnb in the prehypertrophic zone, with loss of Fmn1 resulting in a decrease in the width of the prehypertrophic zone, and loss of Flnb causing premature differentiation of the prehypertrophic zone.


ALLELIC VARIANTS ( 16 Selected Examples):

.0001 SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, SER2137TER
  
RCV000006767

In a consanguineous family with spondylocarpotarsal syndrome (SCT; 272460), Krakow et al. (2004) found that affected individuals were homozygous for a 6408delC mutation in exon 39 of the FLNB gene that predicted a translational frameshift and a stop codon 4 codons downstream.


.0002 SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, ARG818TER
  
RCV000006768...

In a nonconsanguineous family with spondylocarpotarsal syndrome (SCT; 272460), Krakow et al. (2004) found that the affected individual was a compound heterozygote for 2 mutations in the FLNB gene that predicted premature stop codons: arg818 to ter (R818X) and arg1607 to ter (R1607X; 603381.0003). The former mutation was a 2452C-T transition in exon 16; the latter, a 4819C-T transition in exon 28.


.0003 SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, ARG1607TER
  
RCV000006769

For discussion of the 4819C-T transition in exon 28 of the FLNB gene, resulting in an arg1607-to-ter (R1607X) substitution, that was found in compound heterozygous state in a patient with spondylocarpotarsal synostosis syndrome (SCT; 272460) by Krakow et al. (2004), see 603381.0002.


.0004 LARSEN SYNDROME

FLNB, PHE161CYS
  
RCV000030660...

In a family with Larsen syndrome (LRS; 150250), Krakow et al. (2004) found heterozygosity for a de novo missense mutation in the FLNB gene, 482T-G in exon 2, that predicted the substitution phe161-to-cys (F161C) in the second calponin homology domain (CHD2) of filamin B.


.0005 LARSEN SYNDROME

FLNB, GLY1586ARG
  
RCV000030661

In an individual with sporadically occurring Larsen syndrome (LRS; 150250), Krakow et al. (2004) found heterozygosity for a de novo mutation in the FLNB gene, 4756G-A in exon 29, that predicted the substitution gly1586-to-arg (G1586R) in repeat 14 of the protein.


.0006 ATELOSTEOGENESIS, TYPE I

FLNB, ALA173VAL
  
RCV000006770...

In an individual with atelosteogenesis type I (AO1; 108720), Krakow et al. (2004) found heterozygosity for a point mutation, 518C-T, in exon 2 of the FNLB gene predicting an ala173-to-val (A173V) substitution in the second calponin homology domain (CHD2) of filamin B.


.0007 ATELOSTEOGENESIS, TYPE I

ATELOSTEOGENESIS, TYPE III, INCLUDED
FLNB, MET202VAL
  
RCV000006771...

In 1 individual with AO1 (108720) and in 1 with atelosteogenesis type III (AO3; 108721), Krakow et al. (2004) found heterozygosity for the same point mutation in exon 3 of the FLNB gene, 604A-G, predicting a met202-to-val (M202V) substitution in the second calponin homology domain (CHD2) of the protein.


.0008 ATELOSTEOGENESIS, TYPE III

FLNB, GLY751ARG
  
RCV000006773...

In an individual with atelosteogenesis type III (AO3; 108721), Krakow et al. (2004) found heterozygosity for a point mutation in exon 15 of the FLNB gene, 2251G-C, predicting a gly751-to-arg (G751R) substitution in repeat 6 of filamin B.


.0009 BOOMERANG DYSPLASIA

FLNBA, LEU171ARG
  
RCV000006774

In a 22-week male fetus with boomerang dysplasia (BOOMD; 112310), previously studied by Krakow et al. (2004), Bicknell et al. (2005) identified heterozygosity for a 512T-G transversion in the FLNB gene, predicted to cause a leu171-to-arg (L171R) substitution in the second calponin homology domain of filamin B. The authors noted that this residue is highly evolutionarily conserved among vertebrate filamins. The mutation was not found in the unaffected parents.


.0010 BOOMERANG DYSPLASIA

FLNB, SER235PRO
  
RCV000006775

In a 17-week male fetus with boomerang dysplasia (BOOMD; 112310), previously described by Wessels et al. (2003), Bicknell et al. (2005) identified heterozygosity for a 703T-C transition in exon 4 of the FLNB gene, predicted to cause a ser235-to-pro (S235P) substitution in the second calponin homology domain of filamin B. The authors noted that this residue is highly evolutionarily conserved among vertebrate filamins. The mutation was not found in 100 control chromosomes.


.0011 LARSEN SYNDROME

FLNB, GLU227LYS
  
RCV000030662...

In 13 affected individuals from a large family with Larsen syndrome (LRS; 150250), Bicknell et al. (2007) identified a heterozygous 679G-A transition in the FLNB gene, resulting in a glu227-to-lys (E227K) substitution. Clinical signs and symptoms of the disorder were variable in this family, although all had the characteristic facies and most had spatulate fingers and supernumerary carpal bones.


.0012 LARSEN SYNDROME

FLNB, GLY1691SER
  
RCV000030663...

In 6 of 20 unrelated patients with Larsen syndrome (LRS; 150250), Bicknell et al. (2007) identified a heterozygous 5071G-A transition in the FLNB gene, resulting in a gly1691-to-ser (G1691S) substitution.


.0013 SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, ARG2004TER
  
RCV000006776

In a 5-year-old boy with spondylocarpotarsal synostosis syndrome (SCT; 272460), Mitter et al. (2008) detected a homozygous 6010C-T transition in exon 36 of the FLNB gene, resulting in an arg2004-to-ter (R2004X) substitution. In addition to the typical findings of SCT, the boy demonstrated ossification delay of multiple epiphyses and bilateral proximal femoral epiphyseal dysplasia.


.0014 SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, GLY1850TER
  
RCV000006777

In an Italian girl with spondylocarpotarsal synostosis syndrome (SCT; 272460), born of consanguineous parents, Brunetti-Pierri et al. (2008) identified a homozygous 5548G-T transversion in the FLNB gene, resulting in a gly1850-to-ter (G1850X) substitution. She had short stature, scoliosis, short trunk, delayed bone age, vertebral fusions, and capitate-hamate fusion. She did not have facial dysmorphic features. Growth hormone (GH) deficiency was documented, but there was no response to GH administration. MRI scan did not show any abnormality of the hypothalamo-pituitary area, but there was platybasia and basilar impression, stenosis of the foramen magnum, but no signs of medullary compression at the cervicomedullary junction. A younger brother, who was heterozygous for the mutation, had short stature and transient GH deficiency.


.0015 ATELOSTEOGENESIS, TYPE I

FLNB, ALA173THR
  
RCV000114316

In a female infant with atelosteogenesis type I (AO1; 108720) who died 3 hours after birth due to respiratory failure, Jeon et al. (2014) identified heterozygosity for a de novo c.517G-A transition in exon 2 of the FLNB gene, resulting in an ala173-to-thr (A173T) substitution in the CH2 homology domain that was predicted to disrupt actin binding.


.0016 SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, 1-BP DUP, NT1592
  
RCV000782185

In 3 members of an Indian family (family VII) with spondylocarpotarsal synostosis syndrome (SCT; 272460), Salian et al. (2018) identified a homozygous duplication (c.1592dup, NM_001457.3) in the FLNB gene, resulting in a frameshift and a premature termination codon (His532ThrfsTer9). The mutation segregated with the disorder in the family. The patients were 2 boys and a girl, who ranged in age from 6.5 to 12 years. The severity of the phenotype was variable among the sibs.


REFERENCES

  1. Bicknell, L. S., Farrington-Rock, C., Shafeghati, Y., Rump, P., Alanay, Y., Alembik, Y., Al-Madani, N., Firth, H., Karimi-Nejad, M. H., Kim, C. A., Leask, K., Maisenbacher, M., and 14 others. A molecular and clinical study of Larsen syndrome caused by mutations in FLNB. J. Med. Genet. 44: 89-98, 2007. [PubMed: 16801345, images, related citations] [Full Text]

  2. Bicknell, L. S., Morgan, T., Bonafe, L., Wessels, M. W., Bialer, M. G., Willems, P. J., Cohn, D. H., Krakow, D., Robertson, S. P. Mutations in FLNB cause boomerang dysplasia. J. Med. Genet. 42: e43, 2005. Note: Electronic Article. [PubMed: 15994868, related citations] [Full Text]

  3. Biesecker, L. G. Phenotype matters. Nature Genet. 36: 323-324, 2004. [PubMed: 15054484, related citations] [Full Text]

  4. Brocker, F., Bardenheuer, W., Vieten, L., Julicher, K., Werner, N., Marquitan, G., Michael, D., Opalka, B., Schutte, J. Assignment of human filamin gene FLNB to human chromosome band 3p14.3 and identification of YACs containing the complete FLNB transcribed region. Cytogenet. Cell Genet. 85: 267-268, 1999. [PubMed: 10449914, related citations] [Full Text]

  5. Brunetti-Pierri, N., Esposito, B., De Brasi, D., Mattiacci, D. M., Krakow, D., Lee, B., Salerno, M. Spondylocarpotarsal synostosis: long-term follow-up of a case due to FLNB mutations. Am. J. Med. Genet. 146A: 1230-1233, 2008. [PubMed: 18386804, images, related citations] [Full Text]

  6. Chakarova, C., Wehnert, M. S., Uhl, K., Sakthivel, S., Vosberg, H.-P., van der Ven, P. F. M., Furst, D. O. Genomic structure and fine mapping of the two human filamin gene paralogues FLNB and FLNC and comparative analysis of the filamin gene family. Hum. Genet. 107: 597-611, 2000. [PubMed: 11153914, related citations] [Full Text]

  7. Farrington-Rock, C., Firestein, M. H., Bicknell, L. S., Superti-Furga, A., Bacino, C. A., Cormier-Daire, V., Le Merrer, M., Baumann, C., Roume, J., Rump, P., Verheij, J. B. G. M., Sweeney, E., Rimoin, D. L., Lachman, R. S., Robertson, S. P., Cohn, D. H., Krakow, D. Mutations in two regions of FLNB result in atelosteogenesis I and III. Hum. Mutat. 27: 705-710, 2006. [PubMed: 16752402, related citations] [Full Text]

  8. Farrington-Rock, C., Kirilova, V., Dillard-Telm, L., Borowsky, A. D., Chalk, S., Rock, M. J., Cohn, D. H., Krakow, D. Disruption of the Flnb gene in mice phenocopies the human disease spondylocarpotarsal synostosis syndrome. Hum. Molec. Genet. 17: 631-641, 2008. [PubMed: 17635842, images, related citations] [Full Text]

  9. Hu, J., Lu, J., Lian, G., Ferland, R. J., Dettenhofer, M., Sheen, V. L. Formin 1 and filamin B physically interact to coordinate chondrocyte proliferation and differentiation in the growth plate. Hum. Molec. Genet. 23: 4663-4673, 2014. [PubMed: 24760772, images, related citations] [Full Text]

  10. Jeon, G. W., Lee, M.-N., Jung, J. M., Hong, S. Y., Kim, Y. N., Sin, J. B., Ki, C.-S. Identification of a de novo heterozygous missense FLNB mutation in lethal atelosteogenesis type I by exome sequencing. Ann. Lab. Med. 34: 134-138, 2014. [PubMed: 24624349, images, related citations] [Full Text]

  11. Krakow, D., Robertson, S. P., King, L. M., Morgan, T., Sebald, E. T., Bertolotto, C., Wachsmann-Hogiu, S., Acuna, D., Shapiro, S. S., Takafuta, T., Aftimos, S., Kim, C. A., and 13 others. Mutations in the gene encoding filamin B disrupt vertebral segmentation, joint formation and skeletogenesis. Nature Genet. 36: 405-410, 2004. [PubMed: 14991055, related citations] [Full Text]

  12. Leedman, P. J., Faulkner-Jones, B., Cram, D. S., Harrison, P. J., West, J., O'Brien, E., Simpson, R., Coppel, R. L., Harrison, L. C. Cloning from the thyroid of a protein related to actin binding protein that is recognized by Graves disease immunoglobulins. Proc. Nat. Acad. Sci. 90: 5994-5998, 1993. [PubMed: 8327473, related citations] [Full Text]

  13. Mitter, D., Krakow, D., Farrington-Rock, C., Meinecke, P. Expanded clinical spectrum of spondylocarpotarsal synostosis syndrome and possible manifestation in a heterozygous father. Am. J. Med. Genet. 146A: 779-783, 2008. [PubMed: 18257094, related citations] [Full Text]

  14. Salian, S., Shukla, A., Shah, H., Bhat, S. N., Bhat, V. R., Nampoothiri, S., Shenoy, R., Phadke, S. R., Hariharan, S. V., Girisha, K. M. Seven additional families with spondylocarpotarsal synostosis syndrome with novel biallelic deleterious variants in FLNB. Clin. Genet. 94: 159-164, 2018. [PubMed: 29566257, related citations] [Full Text]

  15. Sheen, V. L., Feng, Y., Graham, D., Takafuta, T., Shapiro, S. S., Walsh, C. A. Filamin A and filamin B are co-expressed within neurons during periods of neuronal migration and can physically interact. Hum. Molec. Genet. 11: 2845-2854, 2002. [PubMed: 12393796, related citations] [Full Text]

  16. Takafuta, T., Wu, G., Murphy, G. F., Shapiro, S. S. Human beta-filamin is a new protein that interacts with the cytoplasmic tail of glycoprotein Ib-alpha. J. Biol. Chem. 273: 17531-17538, 1998. [PubMed: 9651345, related citations] [Full Text]

  17. Wessels, M. W., Den Hollander, N. S., De Krijger, R. R., Bonife, L., Superti-Furga, A., Nikkels, P. G., Willems, P. J. Prenatal diagnosis of boomerang dysplasia. Am. J. Med. Genet. 122A: 148-154, 2003. [PubMed: 12955767, related citations] [Full Text]

  18. Xu, W., Xie, Z., Chung, D. W., Davie, E. W. A novel human actin-binding protein homologue that binds to platelet glycoprotein Ib-alpha. Blood 92: 1268-1276, 1998. [PubMed: 9694715, related citations]

  19. Zhang, W., Han, S. W., McKeel, D. W., Goate, A., Wu, J. Y. Interaction of presenilins with the filamin family of actin-binding proteins. J. Neurosci. 18: 914-922, 1998. [PubMed: 9437013, images, related citations] [Full Text]

  20. Zhou, X., Tian, F., Sandzen, J., Cao, R., Flaberg, E., Szekely, L., Cao, Y., Ohlsson, C., Bergo, M. O., Boren, J., Akyurek, L. M. Filamin B deficiency in mice results in skeletal malformations and impaired microvascular development. Proc. Nat. Acad. Sci. 104: 3919-3924, 2007. [PubMed: 17360453, images, related citations] [Full Text]


Joanna S. Amberger - updated : 06/05/2019
Patricia A. Hartz - updated : 10/15/2014
Marla J. F. O'Neill - updated : 3/31/2014
Marla J. F. O'Neill - updated : 8/22/2011
Marla J. F. O'Neill - updated : 5/24/2010
Cassandra L. Kniffin - updated : 8/21/2008
Kelly A. Przylepa - updated : 4/11/2008
Cassandra L. Kniffin - updated : 3/23/2007
Cassandra L. Kniffin - updated : 2/26/2007
Victor A. McKusick - updated : 9/29/2006
Marla J. F. O'Neill - updated : 9/19/2005
Victor A. McKusick - updated : 4/5/2004
George E. Tiller - updated : 3/30/2004
Marla J. F. O'Neill - updated : 3/16/2004
Victor A. McKusick - updated : 12/18/2000
Carol A. Bocchini - updated : 10/1/1999
Creation Date:
Rebekah S. Rasooly : 12/23/1998
carol : 06/05/2019
mgross : 10/15/2014
carol : 3/31/2014
mcolton : 3/28/2014
carol : 8/22/2011
terry : 12/8/2010
carol : 10/4/2010
alopez : 5/24/2010
wwang : 10/16/2009
terry : 10/15/2009
wwang : 8/28/2008
ckniffin : 8/21/2008
carol : 4/11/2008
wwang : 4/11/2007
ckniffin : 3/23/2007
wwang : 3/2/2007
ckniffin : 2/26/2007
alopez : 10/13/2006
terry : 9/29/2006
wwang : 10/5/2005
terry : 9/19/2005
carol : 1/26/2005
alopez : 4/7/2004
terry : 4/5/2004
alopez : 4/2/2004
tkritzer : 3/31/2004
tkritzer : 3/30/2004
alopez : 3/23/2004
alopez : 3/23/2004
alopez : 3/22/2004
terry : 3/16/2004
mcapotos : 1/18/2001
terry : 12/18/2000
alopez : 9/5/2000
carol : 10/1/1999
alopez : 12/23/1998
alopez : 12/23/1998

* 603381

FILAMIN B; FLNB


Alternative titles; symbols

FILAMIN, BETA
ACTIN-BINDING PROTEIN 276/278; ABP276/278


Other entities represented in this entry:

TRUNCATED ACTIN-BINDING PROTEIN, INCLUDED; TABP, INCLUDED
ACTIN-BINDING PROTEIN, TRUNCATED, INCLUDED
FILAMIN HOMOLOG 1, INCLUDED; FH1, INCLUDED

HGNC Approved Gene Symbol: FLNB

SNOMEDCT: 254054000, 63387002, 702351004, 725141006, 725142004;  


Cytogenetic location: 3p14.3     Genomic coordinates (GRCh38): 3:58,008,422-58,172,251 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
3p14.3 Atelosteogenesis, type I 108720 Autosomal dominant 3
Atelosteogenesis, type III 108721 Autosomal dominant 3
Boomerang dysplasia 112310 Autosomal dominant 3
Larsen syndrome 150250 Autosomal dominant 3
Spondylocarpotarsal synostosis syndrome 272460 Autosomal recessive 3

TEXT

Description

Filamins, such as FLNB, are actin-binding proteins that also interact with multiple receptors and intracellular proteins that regulate cytoskeleton-dependent cell proliferation, differentiation, and migration (Hu et al., 2014).


Cloning and Expression

The platelet GpIb complex (see 138720) mediates the adherence of platelets at the site of vascular injury through the binding of GpIb-alpha (231200) to subendothelial von Willebrand factor (VWF; 613160). In platelets, the GpIb complex is tightly bound to the actin cytoskeleton via an interaction of GpIb-alpha with ABP280 (filamin A; 300017). Using a yeast 2-hybrid screen with the cytoplasmic tail of GpIb-alpha as bait, Takafuta et al. (1998) isolated partial cDNAs encoding a novel filamin homolog that they designated beta-filamin. They used the partial cDNAs to screen a placenta library and recovered additional cDNAs corresponding to the entire beta-filamin coding region. Like ABP280, the predicted 2,602-amino acid protein contains an N-terminal actin-binding domain, a backbone of 24 tandem repeats, and 2 hinge regions. Excluding the unique first hinge region of beta-filamin, the sequences of beta-filamin and ABP280 are 70% identical. Antibodies against beta-filamin detected a 280-kD protein on Western blots of human umbilical vein endothelial cell (HUVEC) extracts and stained normal human endothelial cells in culture and in situ. Takafuta et al. (1998) determined that the GpIb-alpha-binding domain in beta-filamin is in repeats 17-20, a region that corresponds to the GpIb-alpha-binding domain in ABP280. Northern blot analysis revealed that beta-filamin is expressed as 2 approximately 9.5-kb mRNAs in many adult tissues. The 2 different transcripts appear to result from use of alternative polyadenylation signals. Takafuta et al. (1998) concluded that beta-filamin is a new member of the filamin family that may have significance for GpIb-alpha function in endothelial cells and platelets.

Independently, Xu et al. (1998) isolated cDNAs encoding beta-filamin, which they referred to as ABP278. These authors also identified alternatively spliced mRNAs encoding ABP276, a beta-filamin isoform missing the first hinge region. RT-PCR analysis indicated that the 2 isoforms were expressed at different relative levels in various human tissues.

The addition of thyroid-stimulating hormone (TSH; see 188540) to cultured thyroid follicular cells induces rapid and profound disruption of actin microfilaments. Using serum from a Graves disease (275000) patient, Leedman et al. (1993) identified a thyroid cDNA encoding TABP (truncated actin-binding protein), a predicted 195-amino acid protein with homology to the C terminus of ABP280. Both Xu et al. (1998) and Takafuta et al. (1998) considered TABP to be a truncated form of beta-filamin.


Mapping

By analysis of somatic cell hybrids, Zhang et al. (1998) mapped the FH1 gene to chromosome 3. Takafuta et al. (1998) refined the map position to 3p21.1-p14.3 based on inclusion of a previously mapped STS within the beta-filamin sequence. By FISH, Brocker et al. (1999) assigned the FLNB gene to 3p14.3. Chakarova et al. (2000) mapped FLNB to 3p14 by radiation hybrid analysis.


Gene Function

Using antigen-capture ELISA, Takafuta et al. (1998) found that beta-filamin associates with GpIb-alpha in both platelets and HUVEC extracts.

Mutations in the presenilin genes PS1 (104311) and PS2 (600759) account for approximately 50% of early-onset familial Alzheimer disease (AD; 104300). Zhang et al. (1998) identified beta-filamin as filamin homolog 1 (FH1), a filamin-related protein that interacts with the loop regions of PS1 and PS2. A monoclonal antibody recognizing both ABP280 and FH1 bound to blood vessels and astrocytes in the normal brain. In the brains of AD patients, Zhang et al. (1998) observed staining also in neurofibrillary tangles, neuropil threads, and dystrophic neurites within some senile plaques. The authors stated that detection of these presenilin-interacting proteins in these brain structures suggests that ABP280 and FH1 may be involved in the development of AD and that interactions between presenilins and ABP280/FH1 may be functionally significant. Takafuta et al. (1998) noted that the FH1 sequence is identical to the C-terminal 291 amino acids of beta-filamin except for 2 residues, making it very likely that FH1 represents the C-terminal region of beta-filamin.

Krakow et al. (2004) found that FLNB is expressed in human growth plate chondrocytes and in developing vertebral bodies in the mouse. The authors concluded that FLNB plays a role in vertebral segmentation, joint formation, and endochondral ossification.

Mutation in the X-linked gene filamin A (FLNA) can cause the neurologic disorder periventricular heterotopia (300049). Although periventricular heterotopia is characterized by a failure in neuronal migration into the cerebral cortex with consequent formation of nodules in the ventricular and subventricular zones, many neurons appear to migrate normally, even in males, suggesting compensatory mechanisms. Sheen et al. (2002) showed that, in mice, Flna mRNA was widely expressed in all brain cortical layers, whereas a homolog, Flnb, was most highly expressed in the ventricular and subventricular zones during development. In adulthood, widespread but reduced expression of Flna and Flnb persisted throughout the cerebral cortex. Flna and Flnb proteins were highly expressed in both the leading processes and somata of migratory neurons during corticogenesis. Postnatally, Flna immunoreactivity was largely localized to the cell body, whereas Flnb was localized to the soma and neuropil during neuronal differentiation. The putative Flnb homodimerization domain strongly interacted with itself or the corresponding homologous region of Flna, as shown by yeast 2-hybrid interaction. The 2 proteins colocalized within neuronal precursors by immunocytochemistry, and the existence of Flna-Flnb heterodimers could be detected by coimmunoprecipitation. Sheen et al. (2002) suggested that FLNA and FLNB may form both homodimers and heterodimers, and that their interaction could potentially compensate for the loss of FLNA function during cortical development within patients with periventricular heterotopia.

Using yeast 2-hybrid and immunoprecipitation analyses, Hu et al. (2014) found that mouse Flnb and Flna interacted directly with the actin nucleating-protein Fmn1 (136535). The filamins and Fmn1 colocalized in cytoplasm and, to a lesser extent, nucleus, and they were coexpressed in chondrocytes.


Molecular Genetics

Krakow et al. (2004) identified mutations in the FLNB gene in 4 human skeletal disorders: spondylocarpotarsal syndrome (SCT; 272460), Larsen syndrome (LRS; 150250), type I atelosteogenesis (AO1; 108720), and type III atelosteogenesis (AO3; 108721).

Biesecker (2004) commented that from the standpoint of the clinical geneticist, 4 distinct disorders result from mutation in the FLNB gene. In contrast, a basic scientist might view them as a single disorder with inconsequential phenotypic differences. The information concerning the multiple disorders caused by mutations in the FLNB gene followed closely on the heels of reports of mutations in the FLNA gene (300017) as the cause of 5 distinct disorders. This was of interest because there are overlapping phenotypic features in the disorders associated with FLNA and FLNB. Biesecker (2004) pointed out that the 'FLNB story' used information from the International Skeletal Dysplasia Registry (ISDR), which is maintained by a skilled group of clinical scientists and includes information on more than 12,000 cases of individuals with disorders that fall into 50 diagnostic groups. One reason for the success of the registry is that it combines clinical service with research archives. The motivation for a clinician to submit cases to the registry is that he or she can receive an expert opinion on the diagnosis (which is useful for medical care and estimating recurrence risks) and, as in the FLNB story, contribute to research.

In a 22-week male fetus previously studied by Krakow et al. (2004) and a 17-week male fetus previously described by Wessels et al. (2003), both diagnosed with boomerang dysplasia (BOOMD; 112310), Bicknell et al. (2005) identified heterozygosity for mutations in the FLNB gene, leu171 to arg (L171R; 603381.0009) and ser235 to pro (S235P; 603381.0010), respectively.

Farrington-Rock et al. (2006) found 14 novel missense mutations in FLNB in 15 unrelated patients with atelosteogenesis I and/or atelosteogenesis III. Most of the mutations resided in exons 2 and 3, which encode the CH2 domain of the actin-binding region of filamin B. The remaining mutations were found in exon 28 and exon 29, which encode repeats 14 and 15 of filamin B. Clinical and radiographic data were used to confirm the diagnosis of atelosteogenesis in all the patients. The diagnosis of type I was given to patients showing classic findings of absent, shortened, or distally tapered humeri and femora; absent, shortened, or bowed radii; shortened and bowed ulnae and tibiae; and absent fibulae. Other findings included vertebral hypoplasia with coronal clefts, 11 ribs, shortened wide distal phalanges, and unossified or partially ossified metacarpals and middle and proximal phalanges. In addition, most type I patients showed evidence of a hypoplastic pelvis, dislocations of the hips, elbows, and knees, and clubbed feet. With the exception of 1 patient where the pregnancy was terminated after 24 weeks' gestation, the patients given the diagnosis of type I either died in the neonatal period or were stillborn. The diagnosis of type III was given to patients when all bones were present in the extremities, when there was distal tapering of the humeri or femora, and where the small tubular bones of the hands and feet were shortened and broad. In 2 patients with type III, the fibulae were absent. Dislocation of the elbows, hips, and knees and clubbed feet were also present in the type III patients, as was vertebral hypoplasia with coronal clefting. Cartilage of these patients showed acellular areas within the growth plate and the presence of large multinuclear cells ('giant cells') within the resting zone as had been described previously.

Bicknell et al. (2007) identified heterozygous mutations in the FLNB gene (see, e.g., 603381.0011; 603381.0012) in 20 unrelated patients with Larsen syndrome. The distribution of mutations within the gene was nonrandom, with clusters of mutations in the actin-binding domains and filamin repeats 13 through 17 being the most common.

In a female infant with atelosteogenesis who died 3 hours after birth due to respiratory failure, Jeon et al. (2014) performed postmortem exome sequencing and identified heterozygosity for a de novo A173T mutation (603381.0015) in the FLNB gene that was not found in 50 healthy controls. The authors noted that most lethal FLNB-related disorders are caused by de novo mutations, and thus there is a low risk of recurrence in subsequent pregnancies.

In 7 families with SCT syndrome, Salian et al. (2018) identified 2 nonsense and 5 frameshift variants in the FLNB gene (see, e.g., 603381.0016), all in homozygous state.


Animal Model

Zhou et al. (2007) detected strong expression of the mouse Flnb gene in vascular endothelial cells and chondrocytes. In Flnb -/- mice, the authors observed a phenotype that resembled those of human skeletal disorders with mutations in the FLNB gene. Less than than 3% of Flnb -/- embryos reached term, indicating that the Flnb gene is important in embryonic development, whereas Flnb +/- mice were indistinguishable from their wildtype sibs. Flnb -/- embryos had impaired development of microvasculature and skeletal systems. The few that were born were very small and had scoliotic and kyphotic spines, lack of intervertebral discs, fusion of vertebral bodies, and reduced hyaline matrix in bones of the extremities, thorax, and vertebrae.

Farrington-Rock et al. (2008) generated Flnb -/- mice and observed a phenotype of short stature and skeletal abnormalities similar to those of individuals with spondylocarpotarsal synostosis syndrome (SCT; 272460). Newborn Flnb -/- mice had fusions between the neural arches of the vertebrae in the cervical and thoracic spine. At postnatal day 60, the vertebral fusions were more widespread and involved the vertebral bodies as well as the neural arches. In addition, fusions were seen in sternum and carpal bones. Analysis of the Flnb -/- mice phenotype showed that an absence of filamin B causes progressive vertebral fusions, in contrast to the previous hypothesis that SCT results from failure of normal spinal segmentation. Farrington-Rock et al. (2008) suggested that spinal segmentation can occur normally in the absence of filamin B, but that the protein is required for maintenance of intervertebral, carpal, and sternal joints, and the joint fusion process commences antenatally.

Hu et al. (2014) found that knockout (KO) of both Flnb and Fmn1 in mice resulted in a more severe reduction in body size, weight, and growth plate length than that observed in mice with KO of either gene alone. In Flnb/Fmn1 double-KO mice, shortening of long bones was associated with decreased chondrocyte proliferation and an overall delay in ossification. Comparison of Fmn1 KO mice with Flnb/Fmn1 double-KO mice revealed nonoverlapping functions for Fmn1 and Flnb in the prehypertrophic zone, with loss of Fmn1 resulting in a decrease in the width of the prehypertrophic zone, and loss of Flnb causing premature differentiation of the prehypertrophic zone.


ALLELIC VARIANTS 16 Selected Examples):

.0001   SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, SER2137TER
SNP: rs80356521, gnomAD: rs80356521, ClinVar: RCV000006767

In a consanguineous family with spondylocarpotarsal syndrome (SCT; 272460), Krakow et al. (2004) found that affected individuals were homozygous for a 6408delC mutation in exon 39 of the FLNB gene that predicted a translational frameshift and a stop codon 4 codons downstream.


.0002   SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, ARG818TER
SNP: rs80356519, gnomAD: rs80356519, ClinVar: RCV000006768, RCV003555947

In a nonconsanguineous family with spondylocarpotarsal syndrome (SCT; 272460), Krakow et al. (2004) found that the affected individual was a compound heterozygote for 2 mutations in the FLNB gene that predicted premature stop codons: arg818 to ter (R818X) and arg1607 to ter (R1607X; 603381.0003). The former mutation was a 2452C-T transition in exon 16; the latter, a 4819C-T transition in exon 28.


.0003   SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, ARG1607TER
SNP: rs80356520, gnomAD: rs80356520, ClinVar: RCV000006769

For discussion of the 4819C-T transition in exon 28 of the FLNB gene, resulting in an arg1607-to-ter (R1607X) substitution, that was found in compound heterozygous state in a patient with spondylocarpotarsal synostosis syndrome (SCT; 272460) by Krakow et al. (2004), see 603381.0002.


.0004   LARSEN SYNDROME

FLNB, PHE161CYS
SNP: rs80356506, ClinVar: RCV000030660, RCV002276538

In a family with Larsen syndrome (LRS; 150250), Krakow et al. (2004) found heterozygosity for a de novo missense mutation in the FLNB gene, 482T-G in exon 2, that predicted the substitution phe161-to-cys (F161C) in the second calponin homology domain (CHD2) of filamin B.


.0005   LARSEN SYNDROME

FLNB, GLY1586ARG
SNP: rs80356513, ClinVar: RCV000030661

In an individual with sporadically occurring Larsen syndrome (LRS; 150250), Krakow et al. (2004) found heterozygosity for a de novo mutation in the FLNB gene, 4756G-A in exon 29, that predicted the substitution gly1586-to-arg (G1586R) in repeat 14 of the protein.


.0006   ATELOSTEOGENESIS, TYPE I

FLNB, ALA173VAL
SNP: rs121908894, ClinVar: RCV000006770, RCV003318540

In an individual with atelosteogenesis type I (AO1; 108720), Krakow et al. (2004) found heterozygosity for a point mutation, 518C-T, in exon 2 of the FNLB gene predicting an ala173-to-val (A173V) substitution in the second calponin homology domain (CHD2) of filamin B.


.0007   ATELOSTEOGENESIS, TYPE I

ATELOSTEOGENESIS, TYPE III, INCLUDED
FLNB, MET202VAL
SNP: rs121908895, ClinVar: RCV000006771, RCV000006772

In 1 individual with AO1 (108720) and in 1 with atelosteogenesis type III (AO3; 108721), Krakow et al. (2004) found heterozygosity for the same point mutation in exon 3 of the FLNB gene, 604A-G, predicting a met202-to-val (M202V) substitution in the second calponin homology domain (CHD2) of the protein.


.0008   ATELOSTEOGENESIS, TYPE III

FLNB, GLY751ARG
SNP: rs28937587, ClinVar: RCV000006773, RCV000508566

In an individual with atelosteogenesis type III (AO3; 108721), Krakow et al. (2004) found heterozygosity for a point mutation in exon 15 of the FLNB gene, 2251G-C, predicting a gly751-to-arg (G751R) substitution in repeat 6 of filamin B.


.0009   BOOMERANG DYSPLASIA

FLNBA, LEU171ARG
SNP: rs80356494, ClinVar: RCV000006774

In a 22-week male fetus with boomerang dysplasia (BOOMD; 112310), previously studied by Krakow et al. (2004), Bicknell et al. (2005) identified heterozygosity for a 512T-G transversion in the FLNB gene, predicted to cause a leu171-to-arg (L171R) substitution in the second calponin homology domain of filamin B. The authors noted that this residue is highly evolutionarily conserved among vertebrate filamins. The mutation was not found in the unaffected parents.


.0010   BOOMERANG DYSPLASIA

FLNB, SER235PRO
SNP: rs121908896, ClinVar: RCV000006775

In a 17-week male fetus with boomerang dysplasia (BOOMD; 112310), previously described by Wessels et al. (2003), Bicknell et al. (2005) identified heterozygosity for a 703T-C transition in exon 4 of the FLNB gene, predicted to cause a ser235-to-pro (S235P) substitution in the second calponin homology domain of filamin B. The authors noted that this residue is highly evolutionarily conserved among vertebrate filamins. The mutation was not found in 100 control chromosomes.


.0011   LARSEN SYNDROME

FLNB, GLU227LYS
SNP: rs80356508, ClinVar: RCV000030662, RCV001582470

In 13 affected individuals from a large family with Larsen syndrome (LRS; 150250), Bicknell et al. (2007) identified a heterozygous 679G-A transition in the FLNB gene, resulting in a glu227-to-lys (E227K) substitution. Clinical signs and symptoms of the disorder were variable in this family, although all had the characteristic facies and most had spatulate fingers and supernumerary carpal bones.


.0012   LARSEN SYNDROME

FLNB, GLY1691SER
SNP: rs80356503, ClinVar: RCV000030663, RCV001064985

In 6 of 20 unrelated patients with Larsen syndrome (LRS; 150250), Bicknell et al. (2007) identified a heterozygous 5071G-A transition in the FLNB gene, resulting in a gly1691-to-ser (G1691S) substitution.


.0013   SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, ARG2004TER
SNP: rs121908897, gnomAD: rs121908897, ClinVar: RCV000006776

In a 5-year-old boy with spondylocarpotarsal synostosis syndrome (SCT; 272460), Mitter et al. (2008) detected a homozygous 6010C-T transition in exon 36 of the FLNB gene, resulting in an arg2004-to-ter (R2004X) substitution. In addition to the typical findings of SCT, the boy demonstrated ossification delay of multiple epiphyses and bilateral proximal femoral epiphyseal dysplasia.


.0014   SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, GLY1850TER
SNP: rs121908898, gnomAD: rs121908898, ClinVar: RCV000006777

In an Italian girl with spondylocarpotarsal synostosis syndrome (SCT; 272460), born of consanguineous parents, Brunetti-Pierri et al. (2008) identified a homozygous 5548G-T transversion in the FLNB gene, resulting in a gly1850-to-ter (G1850X) substitution. She had short stature, scoliosis, short trunk, delayed bone age, vertebral fusions, and capitate-hamate fusion. She did not have facial dysmorphic features. Growth hormone (GH) deficiency was documented, but there was no response to GH administration. MRI scan did not show any abnormality of the hypothalamo-pituitary area, but there was platybasia and basilar impression, stenosis of the foramen magnum, but no signs of medullary compression at the cervicomedullary junction. A younger brother, who was heterozygous for the mutation, had short stature and transient GH deficiency.


.0015   ATELOSTEOGENESIS, TYPE I

FLNB, ALA173THR
SNP: rs587777259, ClinVar: RCV000114316

In a female infant with atelosteogenesis type I (AO1; 108720) who died 3 hours after birth due to respiratory failure, Jeon et al. (2014) identified heterozygosity for a de novo c.517G-A transition in exon 2 of the FLNB gene, resulting in an ala173-to-thr (A173T) substitution in the CH2 homology domain that was predicted to disrupt actin binding.


.0016   SPONDYLOCARPOTARSAL SYNOSTOSIS SYNDROME

FLNB, 1-BP DUP, NT1592
SNP: rs746105983, gnomAD: rs746105983, ClinVar: RCV000782185

In 3 members of an Indian family (family VII) with spondylocarpotarsal synostosis syndrome (SCT; 272460), Salian et al. (2018) identified a homozygous duplication (c.1592dup, NM_001457.3) in the FLNB gene, resulting in a frameshift and a premature termination codon (His532ThrfsTer9). The mutation segregated with the disorder in the family. The patients were 2 boys and a girl, who ranged in age from 6.5 to 12 years. The severity of the phenotype was variable among the sibs.


REFERENCES

  1. Bicknell, L. S., Farrington-Rock, C., Shafeghati, Y., Rump, P., Alanay, Y., Alembik, Y., Al-Madani, N., Firth, H., Karimi-Nejad, M. H., Kim, C. A., Leask, K., Maisenbacher, M., and 14 others. A molecular and clinical study of Larsen syndrome caused by mutations in FLNB. J. Med. Genet. 44: 89-98, 2007. [PubMed: 16801345] [Full Text: https://doi.org/10.1136/jmg.2006.043687]

  2. Bicknell, L. S., Morgan, T., Bonafe, L., Wessels, M. W., Bialer, M. G., Willems, P. J., Cohn, D. H., Krakow, D., Robertson, S. P. Mutations in FLNB cause boomerang dysplasia. J. Med. Genet. 42: e43, 2005. Note: Electronic Article. [PubMed: 15994868] [Full Text: https://doi.org/10.1136/jmg.2004.029967]

  3. Biesecker, L. G. Phenotype matters. Nature Genet. 36: 323-324, 2004. [PubMed: 15054484] [Full Text: https://doi.org/10.1038/ng0404-323]

  4. Brocker, F., Bardenheuer, W., Vieten, L., Julicher, K., Werner, N., Marquitan, G., Michael, D., Opalka, B., Schutte, J. Assignment of human filamin gene FLNB to human chromosome band 3p14.3 and identification of YACs containing the complete FLNB transcribed region. Cytogenet. Cell Genet. 85: 267-268, 1999. [PubMed: 10449914] [Full Text: https://doi.org/10.1159/000015309]

  5. Brunetti-Pierri, N., Esposito, B., De Brasi, D., Mattiacci, D. M., Krakow, D., Lee, B., Salerno, M. Spondylocarpotarsal synostosis: long-term follow-up of a case due to FLNB mutations. Am. J. Med. Genet. 146A: 1230-1233, 2008. [PubMed: 18386804] [Full Text: https://doi.org/10.1002/ajmg.a.32303]

  6. Chakarova, C., Wehnert, M. S., Uhl, K., Sakthivel, S., Vosberg, H.-P., van der Ven, P. F. M., Furst, D. O. Genomic structure and fine mapping of the two human filamin gene paralogues FLNB and FLNC and comparative analysis of the filamin gene family. Hum. Genet. 107: 597-611, 2000. [PubMed: 11153914] [Full Text: https://doi.org/10.1007/s004390000414]

  7. Farrington-Rock, C., Firestein, M. H., Bicknell, L. S., Superti-Furga, A., Bacino, C. A., Cormier-Daire, V., Le Merrer, M., Baumann, C., Roume, J., Rump, P., Verheij, J. B. G. M., Sweeney, E., Rimoin, D. L., Lachman, R. S., Robertson, S. P., Cohn, D. H., Krakow, D. Mutations in two regions of FLNB result in atelosteogenesis I and III. Hum. Mutat. 27: 705-710, 2006. [PubMed: 16752402] [Full Text: https://doi.org/10.1002/humu.20348]

  8. Farrington-Rock, C., Kirilova, V., Dillard-Telm, L., Borowsky, A. D., Chalk, S., Rock, M. J., Cohn, D. H., Krakow, D. Disruption of the Flnb gene in mice phenocopies the human disease spondylocarpotarsal synostosis syndrome. Hum. Molec. Genet. 17: 631-641, 2008. [PubMed: 17635842] [Full Text: https://doi.org/10.1093/hmg/ddm188]

  9. Hu, J., Lu, J., Lian, G., Ferland, R. J., Dettenhofer, M., Sheen, V. L. Formin 1 and filamin B physically interact to coordinate chondrocyte proliferation and differentiation in the growth plate. Hum. Molec. Genet. 23: 4663-4673, 2014. [PubMed: 24760772] [Full Text: https://doi.org/10.1093/hmg/ddu186]

  10. Jeon, G. W., Lee, M.-N., Jung, J. M., Hong, S. Y., Kim, Y. N., Sin, J. B., Ki, C.-S. Identification of a de novo heterozygous missense FLNB mutation in lethal atelosteogenesis type I by exome sequencing. Ann. Lab. Med. 34: 134-138, 2014. [PubMed: 24624349] [Full Text: https://doi.org/10.3343/alm.2014.34.2.134]

  11. Krakow, D., Robertson, S. P., King, L. M., Morgan, T., Sebald, E. T., Bertolotto, C., Wachsmann-Hogiu, S., Acuna, D., Shapiro, S. S., Takafuta, T., Aftimos, S., Kim, C. A., and 13 others. Mutations in the gene encoding filamin B disrupt vertebral segmentation, joint formation and skeletogenesis. Nature Genet. 36: 405-410, 2004. [PubMed: 14991055] [Full Text: https://doi.org/10.1038/ng1319]

  12. Leedman, P. J., Faulkner-Jones, B., Cram, D. S., Harrison, P. J., West, J., O'Brien, E., Simpson, R., Coppel, R. L., Harrison, L. C. Cloning from the thyroid of a protein related to actin binding protein that is recognized by Graves disease immunoglobulins. Proc. Nat. Acad. Sci. 90: 5994-5998, 1993. [PubMed: 8327473] [Full Text: https://doi.org/10.1073/pnas.90.13.5994]

  13. Mitter, D., Krakow, D., Farrington-Rock, C., Meinecke, P. Expanded clinical spectrum of spondylocarpotarsal synostosis syndrome and possible manifestation in a heterozygous father. Am. J. Med. Genet. 146A: 779-783, 2008. [PubMed: 18257094] [Full Text: https://doi.org/10.1002/ajmg.a.32230]

  14. Salian, S., Shukla, A., Shah, H., Bhat, S. N., Bhat, V. R., Nampoothiri, S., Shenoy, R., Phadke, S. R., Hariharan, S. V., Girisha, K. M. Seven additional families with spondylocarpotarsal synostosis syndrome with novel biallelic deleterious variants in FLNB. Clin. Genet. 94: 159-164, 2018. [PubMed: 29566257] [Full Text: https://doi.org/10.1111/cge.13252]

  15. Sheen, V. L., Feng, Y., Graham, D., Takafuta, T., Shapiro, S. S., Walsh, C. A. Filamin A and filamin B are co-expressed within neurons during periods of neuronal migration and can physically interact. Hum. Molec. Genet. 11: 2845-2854, 2002. [PubMed: 12393796] [Full Text: https://doi.org/10.1093/hmg/11.23.2845]

  16. Takafuta, T., Wu, G., Murphy, G. F., Shapiro, S. S. Human beta-filamin is a new protein that interacts with the cytoplasmic tail of glycoprotein Ib-alpha. J. Biol. Chem. 273: 17531-17538, 1998. [PubMed: 9651345] [Full Text: https://doi.org/10.1074/jbc.273.28.17531]

  17. Wessels, M. W., Den Hollander, N. S., De Krijger, R. R., Bonife, L., Superti-Furga, A., Nikkels, P. G., Willems, P. J. Prenatal diagnosis of boomerang dysplasia. Am. J. Med. Genet. 122A: 148-154, 2003. [PubMed: 12955767] [Full Text: https://doi.org/10.1002/ajmg.a.20239]

  18. Xu, W., Xie, Z., Chung, D. W., Davie, E. W. A novel human actin-binding protein homologue that binds to platelet glycoprotein Ib-alpha. Blood 92: 1268-1276, 1998. [PubMed: 9694715]

  19. Zhang, W., Han, S. W., McKeel, D. W., Goate, A., Wu, J. Y. Interaction of presenilins with the filamin family of actin-binding proteins. J. Neurosci. 18: 914-922, 1998. [PubMed: 9437013] [Full Text: https://doi.org/10.1523/JNEUROSCI.18-03-00914.1998]

  20. Zhou, X., Tian, F., Sandzen, J., Cao, R., Flaberg, E., Szekely, L., Cao, Y., Ohlsson, C., Bergo, M. O., Boren, J., Akyurek, L. M. Filamin B deficiency in mice results in skeletal malformations and impaired microvascular development. Proc. Nat. Acad. Sci. 104: 3919-3924, 2007. [PubMed: 17360453] [Full Text: https://doi.org/10.1073/pnas.0608360104]


Contributors:
Joanna S. Amberger - updated : 06/05/2019
Patricia A. Hartz - updated : 10/15/2014
Marla J. F. O'Neill - updated : 3/31/2014
Marla J. F. O'Neill - updated : 8/22/2011
Marla J. F. O'Neill - updated : 5/24/2010
Cassandra L. Kniffin - updated : 8/21/2008
Kelly A. Przylepa - updated : 4/11/2008
Cassandra L. Kniffin - updated : 3/23/2007
Cassandra L. Kniffin - updated : 2/26/2007
Victor A. McKusick - updated : 9/29/2006
Marla J. F. O'Neill - updated : 9/19/2005
Victor A. McKusick - updated : 4/5/2004
George E. Tiller - updated : 3/30/2004
Marla J. F. O'Neill - updated : 3/16/2004
Victor A. McKusick - updated : 12/18/2000
Carol A. Bocchini - updated : 10/1/1999

Creation Date:
Rebekah S. Rasooly : 12/23/1998

Edit History:
carol : 06/05/2019
mgross : 10/15/2014
carol : 3/31/2014
mcolton : 3/28/2014
carol : 8/22/2011
terry : 12/8/2010
carol : 10/4/2010
alopez : 5/24/2010
wwang : 10/16/2009
terry : 10/15/2009
wwang : 8/28/2008
ckniffin : 8/21/2008
carol : 4/11/2008
wwang : 4/11/2007
ckniffin : 3/23/2007
wwang : 3/2/2007
ckniffin : 2/26/2007
alopez : 10/13/2006
terry : 9/29/2006
wwang : 10/5/2005
terry : 9/19/2005
carol : 1/26/2005
alopez : 4/7/2004
terry : 4/5/2004
alopez : 4/2/2004
tkritzer : 3/31/2004
tkritzer : 3/30/2004
alopez : 3/23/2004
alopez : 3/23/2004
alopez : 3/22/2004
terry : 3/16/2004
mcapotos : 1/18/2001
terry : 12/18/2000
alopez : 9/5/2000
carol : 10/1/1999
alopez : 12/23/1998
alopez : 12/23/1998