Open Access

A novel FBN1 mutation causes autosomal dominant Marfan syndrome

  • Authors:
    • Ying Xiao
    • Xiaoqi Liu
    • Xiaoxin Guo
    • Liping Liu
    • Linxin Jiang
    • Qi Wang
    • Bo Gong
  • View Affiliations

  • Published online on: September 20, 2017     https://doi.org/10.3892/mmr.2017.7544
  • Pages: 7321-7328
  • Copyright: © Xiao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Marfan syndrome (MFS) is an inherited and systemic disorder. It has been reported that mutations in the fibrillin‑1 gene (FBN1) account for ~90% of autosomal dominant cases of MFS. This study was conducted to screen mutations of FBN1 in a Chinese family with autosomal dominant MFS; four individuals including two patients with MFS were recruited. The family members underwent complete physical, cardiovascular and ophthalmologic examinations. Genomic DNA samples were collected from the family along with 383 unrelated healthy subjects. FBN1 coding regions were amplified by polymerase chain reaction and analyzed by direct sequencing. SIFT and PolyPhen‑2 were used to predict the possible structural and functional alterations of the protein. A novel heterozygous mutation c.1708 T>G (p.C570G) in exon 14 was identified, which led to a substitution of cysteine by glycine at codon 570 (p.C570G). The mutation was identified as being associated with the MFS phenotype in the affected members of this family. However, the unaffected family members and the 383 normal controls lacked the mutation. Multiple sequence alignment of the human FBN1 protein revealed that this novel mutation occurred within a highly conserved region of the FBN1 protein across different species and may induce structural alterations in this functional domain. The spectrum of MFS‑associated mutations in the FBN1 gene has been enriched from this study; this may improve understanding of the molecular pathogenesis and clinical diagnosis of MFS.

Introduction

Marfan syndrome (MFS) is an autosomal dominant hereditary disease comprising a disorder of fibrous connective tissue involving the ocular, skeletal and cardiovascular systems (1). According to the Ghent criteria, patients with malfunctions of at least two organ systems could be diagnosed with MFS (2). Aortic root dilatation/dissection and lens dislocation were two cardinal manifestations to establish an unequivocal diagnosis of MFS in patients with positive family history. Due to the large clinical variability of MFS, and several other connective tissue disorders with comparable clinical features, distinguishing MFS from those similar syndromes is still challenging.

Increasing evidence indicates that heredity holds a key role in the development of MFS. It has been reported that MFS generally results from mutations in the human fibrillin-1 (FBN1) gene (3,4). At present, >3,000 mutations have been identified in relation to MFS. Most mutations are specific to a family with MFS, whereas ~10% of FBN1 mutations are shared by different families (5). Located at chromosome 15q-21.1 with 65 exons, the FBN1 gene encodes a secreted 350 kDa glycoprotein (6). Human FBN1 protein shares conserved sequences with other species. FBN1 protein constitutes extracellular microfibrils and controls the stability, as well as the microfibril assembly. Mutations within the FBN1 gene may disrupt microfibril formation, leading to abnormalities of fibrillin and eventually weakening the connective tissue (7).

In the present study, the entire coding region of FBN1 was analyzed, and a novel mutation in exon 14 of FBN1 was identified in all affected members. The newly identified FBN1 mutation in a Chinese family with MFS further emphasizes the important role of FBN1 in the mechanism of MFS development. The present study not only expanded the mutation spectrum of FBN1 resulting in MFS development in a Chinese family, but is also likely to aid understanding of the molecular pathogenesis and clinical diagnosis of FBN1-associated MFS.

Materials and methods

Subjects

A family with MFS was recruited from the Shandong Provincial Hospital Affiliated to Shandong University (Jinan, China) (Fig. 1). This study was conducted in accordance to the tenets of The Declaration of Helsinki and was approved by the Institutional Review Boards of the Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital (Chengdu, China), and the Shandong Provincial Hospital Affiliated to Shandong University. A total of 383 ethnically matched, unrelated and normal healthy individuals were recruited from the Hospital of University of Electronic Science and Technology of China & Sichuan Provincial People's Hospital (255 males and 128 females; mean age at recruitment 55.26±8.78 years). These control individuals had no medical history associated with any related diseases. Written informed consent was obtained from all participants prior to the study.

Clinical diagnosis

Two of the family members were diagnosed with MFS according to the revised Ghent criteria (2). Non-consanguineous marriages were found in the family; clinical information of the affected family members is summarized in Table I. All members of this family underwent complete physical, cardiovascular and ophthalmologic examinations. Unrelated healthy individuals also underwent the same examinations.

Table I.

Clinical details of the patients with Marfan syndrome in the family.

Table I.

Clinical details of the patients with Marfan syndrome in the family.

CharacteristicProband (I:1)Proband's daughter (II:2)
Age (years)448
GenderMF
Ectopialentis++
Myopia++
Strabismus+, exotropia+, exotropia
Glaucoma
Retinal detachment+
Height (cm)184134
Arm span (cm)186137
AS/H1.011.02
Overgrowth of the long bones++
Arachnodactyly++
Scoliosis
Pectus excavatum
Pectus carinatum+
Flatfeet++
Mitral valve prolapse
Aortic aneurysm+ (ruptured 5 years ago then formed aortic dissection; Bentall surgery was performed at that time)
Aortic root dimension (mm)25.0 (artificial vessel diameter)29.1

[i] M, male; F, female; AS, arm span; H, Height.

Mutation screening

Genomic DNA samples were extracted from peripheral blood using a Blood DNA extraction kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The whole coding region of FBN1 (NM_000138.4) was amplified by polymerase chain reaction (PCR) with 35 cycles (30 sec at 95°C for initial denaturation, 30 sec for annealing at different temperatures as shown in Table II, and 30 sec at 72°C for extension), using a GeneAmp® PCR system 9700 (Applied Biosystems; Thermo Scientific Inc.). Sequencing primers of all the exons were designed using Primer 5.0 (Premier Biosoft International, Palo Alto, CA, USA; Table II). Amplified PCR products were purified and sequenced directly (BigDye Terminators Sequencing kit) with an Automated Genetic Analysis system 3130 (both from Applied Biosystems; Thermo Fisher Scientific, Inc.). Comparative amino acid sequence analysis of the human FBN1 protein was performed across different species using HomoloGene (https://www.ncbi.nlm.nih.gov/homologene/?term=FBN1). The potentially damaging effects of the mutation on the structure and function of FBN1 was predicted using SIFT (http://sift.jcvi.org) and PolyPhen-2 (http://genetics.bwh.harvard.edu/pph2/).

Table II.

Primers used for mutation screening of the FBN1 gene.

Table II.

Primers used for mutation screening of the FBN1 gene.

Primer namePrimer sequence (5′-3′)Product size (bp)Annealing temperature (°C)
FBN11&2F TCGGGGATTTGTCTCTGTGT434  59
FBN11&2R GCCCGTTGTTCTGGATCTTG
FBN13F ACCAACCCAGCATTGAGTCT308  60
FBN13R TTCTAAGGCTCCCCATGCAA
FBN14F TTGTGAGGGACCTGAGAACC296  59
FBN14R TTGCAGGAAAGAGGAAAGCC
FBN15F CAACTCCTGTGAGCTGTTGC278  60
FBN15R AAACATGCTGTGTCCCAGGT
FBN16F GTCCTTCCAGAGGACCACAA228  60
FBN16R CAGCTTTAGGTACCAGCATGTC
FBN17F GCATGATGGTTCCTGCTTTT380  60
FBN17R GCAGTCAGCGAAATTGTGAA
FBN18F TTCCAAATATTGTGATGGACAAA448  60
FBN18R ACAGGGTTTTTCTGGTCCAA
FBN19F GCTGTTTCCAGGGACATGAT441  60
FBN19R TTTATGGGAGGCAAAACGTC
FBN110F AGCCCCAGTGTGAAGTATGG396  60
FBN110R TTCCCTGGACGTCATCTCTT
FBN111F TGACTTCTGTGGGCCTATGA300  59
FBN111R TTAACTTGAACAATGCAAGAAAAA
FBN112F TTGTCACCAGACGACCTTTG383  60
FBN112R CCACCAAGTTTGGGGTAAGTT
FBN113F AAAAGGAACCCAGAAAGTCTTAGAA295  60
FBN113R CTTCCGGCATGGGTTATTTA
FBN114F GGAGGGAGGGGGAAATAAA244  60
FBN114R ACTGCAATGGAAGGAGAGGA
FBN115F GATCTTATTTGGATGAAAGTTAGCC400  59
FBN115R AGTCAGGTTTCCCAAACCAA
FBN116F TTCCCCATTTTCAAGGGTTA294  61
FBN116R CGTTTGTTACCATTGGGCTTT
FBN117F GGGGGTTCTCATCTGTTTGA242  60
FBN117R CAGTACGAGGGCATCTCCAT
FBN118F ACCAAGGGCAGGATCTACCT188  60
FBN118R ACCCACAAGAAAGCCTGATG
FBN119F CCTGTAGCTCCTAAGGTCATTACA300  60
FBN119R CTCCCAGCAATGAAAGAAGG
FBN120F CAAAGTTTGGGCCCTTTTTA226  59
FBN120R TGGCATTCCAAAAGATAGCA
FBN121F GGCCCAAGACTAGATTTTAGCA243  60
FBN121R TTTTGCAGGAAAAGCTGACA
FBN122F AATGTCAGCTTTTCCTGCAA368  59
FBN122R TGAAATACTAGGCTTCCCCTTT
FBN123F TGTCAGAACTGCAAAGTCTGG204  60
FBN123R GACAGCTTTATCCAGTCCGAGT
FBN124F TGCTATTCAGGCACCCTAGA400  59
FBN124R TGGAGTGTGTGTCTGTACCTGA
FBN125F AACAGAGTGTTGGCAGTTTGG373  60
FBN125R CTGAGATCATGAAAATGCATCC
FBN126&27F GACCTCCTGACTGCTTGCTC494  60
FBN126&27R CAAAGCTTCATGGAATCCTTCT
FBN128&29F GAGTGCTTGGTCTGGTGGAG564  61
FBN128&29R AGCGATGAAAACAAAACTCAGA
FBN130F GGGACAGACATCCAAACCAT249  62
FBN130R CAAAGCCTGGGCCCTAAAC
FBN131F CTCACTGAACAGTGGAACCAA280  59
FBN131R GCTCTCTTTGGAATGCTGGT280  59
FBN132F GAATCTTTCTATCACTGACCCAAAC
FBN132R TCGAGGGGAAAGTACTCAATG325  59
FBN133&34F CATTTGTGCTGAGCCTTTTTC495  60
FBN133&34R GAATGCCTGGCTTCTCTGAC
FBN135F TGCTGCACTGGAAAGTTGAT231  60
FBN135R AGTGGCTTCCCCATCAGTTA
FBN136F TGCCCAGATTGGTGTTAGAT400  59
FBN136R CAGGTCTGAGAAAAGGTATCTGTG
FBN137&38F AGATTGGGCCCTGTTCTTTT819  60
FBN137&38R TTGGGAATAAGGTCCCCTCT
FBN139&40F TCAGACGGGCAGAGTAACAA496  59
FBN139&40R CCATATTCTGGTTTTGCAGGT
FBN141F AGGCCATTCCAAAATGTGAA249  60
FBN141R TTGTGAGCTCTCTTCCTCTTTGT
FBN142F ATTTCCCACATGGCATCAC300  60
FBN142R TGCTTCCTTCGCTAAGACTGA
FBN143F CTATCCTCCCATCCCACCTT273  60
FBN143R CAGGGTGTTTGCACAGTTTG
FBN144F CACAGGGATCATGTGCTGTC315  60
FBN144R TCCACACCATGCCCTTTACT
FBN145F GGCTTTGTTGACTGGACACC218  62
FBN145R GTAGGCATGTCCAGCCTGTG
FBN146F GAGCTAGGATTACTCCTGAGAATGA398  59
FBN146R TCATGTTCAGATTGCCAAAGA
FBN147F GGCCTGGTGAACCCTAAAAT247  60
FBN147R TTCCTTTGCTGATGCACAAT
FBN148F TGCTGGGATTATGACATCTTTG292  60
FBN148R TTTTCCTCCAGGTTTCCAGA
FBN149F CCAGTGGGAACCTCTTCCTT205  60
FBN149R GACACCCGACACTCCTCATT
FBN150F TGATGTCTCCATCGTGTTTTG208  61
FBN150R ATTGAAAGCCCAAAGCCTTC
FBN151F GGAAAGCAACTGAAGGGTGT263590
FBN151R GCCTACAGTCTTACTTACATCATGG
FBN152&53F GGAGAAGCTTGTAATGAATTGCT594  60
FBN152&53R AACTTATTTCAGTGCCATCTTGG
FBN154F TTTGGACACATTCCTGGTTTC207  60
FBN154R CAACCAATTGTTCCCAGGAT
FBN155F CCTTTTGTTGCTGTCCATGAT249  60
FBN155R AGGGAAGCTTTGAGGGACAT
FBN156F TCATACTCAACAGAGCAGAAGGA363  59
FBN156R CAAGAACTCAGAGCCCAGGT
FBN157F AAGGAACAAAGGGAGGGAAG392  60
FBN157R CAGTCATTACGGCATCTCCA
FBN158F CTGACATCCCCTTTGCCATA277  61
FBN158R TCCCTGCAAGTATTTTTGGAC
FBN159&60F CACTGAAGTGACCCCCTACA600  60
FBN159&60R TGAGGGGCAATGGTCAAT
FBN161&62F TGTTGGCTTGACTCAAATGC600  61
FBN161&62R CCTCCACAAGGATTCACCAG
FBN163F TGGTGGCTCTGCTTCTTTTT178  60
FBN163R GCCATGCATCTTGAGAGTGA
FBN164F AAGTGGCCAGATCCAATGTC334  60
FBN164R ACCATGACCAGGAAGAGCAC
FBN165F CATCTATGCTCCCCTTCTGC243  60
FBN165R TTCCACCACAGGAGACATCA
FBN166F GCAGCATAAGGCAGAAAATTG583  60
FBN166R TGATTCTGATTGGGGGAAAA

[i] FBN1, fibrillin-1; F, forward; R, reverse; bp, base pair.

Results

Clinical findings

The parents and two daughters of a family from Shandong, China, were included in the present study (Fig. 1). Other relatives of this family were not willing to be tested and so additional clinical details were unattainable. Two affected patients (I:1 and II:2) exhibited similar clinical symptoms, including ectopialentis, myopia and strabismus (Fig. 2 and Table I). The left eye of the proband (I:1) underwent refractive lensectomy and vitrectomy combined with silicone oil tamponade after retinal detachment 2 years prior to the current study; following retinal re-attachment, silicone oil was removed 3 months later. The two patients both had the same facial and skeletal features, including arachnodactyly, flat feet and dilation of the aortic root (Fig. 3 and Table I). The proband had pectus carinatum and aortic aneurysm. The patient received Bentall surgery and underwent aortic arch replacement 5 years prior to the current study, as their aortic aneurysm ruptured and formed aortic dissection (Fig. 4). The other two members of the family had no features of MFS.

Mutation screening of FBN1

Direct sequencing of the whole coding region of FBN1 detected a novel missense mutation c.1708 T>G (p.C570G), situated at nucleotide 570 in exon 14 of the coding region (Fig. 5A). This heterozygous mutation was detected in the two affected patients (I:1 and II:2) but was not found in the unaffected mother and daughter (I:2 and II:1) of the family and in the 383 ethnically matched healthy subjects. Therefore, c.1708 T>G (p.C570G) cosegregated to the patients with MFS in this family. Multiple sequencing alignment of human FBN1 protein with various species revealed that the novel mutation occurred within a highly conserved region of the calcium binding epidermal growth factor-like (cbEGF) domain (Fig. 5C). This mutation is a T>G transition, converting cysteine to glycine at amino acid 570 (p.C570G). This amino acid substitution in the FBN1 protein was predicted to be damaging by SIFT and PolyPhen-2.

Discussion

It has been reported that MFS is mainly caused by mutations in the FBN1 gene, which was the first gene identified to cause MFS (8). Of all the identified mutations in the FBN1 gene, 38.6% result in a truncated FBN1 protein and 60.3% represent missense mutations across different ethnic groups (9). FBN1 mutations may cause abnormalities in the formation of microfibrils and fibrillin. As a result, connective tissues weaken (10). A novel FBN1 heterozygous missense mutation, c.1708 T>G (p.C570G) was identified within a Chinese family associated with MFS in the present study.

FBN1 is an important component of microfibrils and is expressed in many human tissues, including in zonules, the cardiovascular system, cartilage, tendon and cornea. The protein serves a role in the formation of zonules and is secreted from ciliary bodies of non-pigmented cells (11). FBN1 protein is composed of repeated modules, including cbEGF and transforming growth factor-1 binding protein-like domains, and is responsible for maintaining microfibers in an ordered arrangement (12,13). The majority of identified missense mutations in FBN1 are localized in cbEGF (14). The mutated monomer of FBN1 could interfere with the polymerization of fibrillin and microfiber aggregation (15). FBN1 mutations within cbEGF modules may disrupt the stability of elastic fibers and render FBN1 susceptible to proteolysis. As a result, the transforming growth factor-β signaling activity that affects extracellular matrix formation may malfunction (4,16).

In the present study, a novel c.1708 T>G (p.C570G) heterozygous missense mutation of the FBN1 gene was reported in a Chinese family with MFS. Three similar missense mutations: c.1709G>A (p.C570Y) (17), c.1709G>C (p.C570S) (18) and c.1709G>C (p.C570R) (19) have been reported in sporadic cases; however, clinical data in these studies were not obtained. In this pedigree, c.1708 T>G (p.C570G) in FBN1 was detected in the two patients with MFS (I:1 and II:2). The proband (I:1) initially came to Shandong Provincial Hospital to see an ophthalmologist and was found to suffer from ectopialentis, myopia and strabismus in both eyes. The proband and the affected daughter (II:2) had similar facial and skeletal features of MFS, including arachnodactyly, flat feet and dilation of aortic root. In addition, pectus carinatum, aortic dissection and retinal detachment were also detected in the proband. These findings suggested that the clinical manifestations of the patient with MFS became more evident with age. This mutation was not included in the Exome Aggregation Consortium dataset; c.1708 T>G (p.C570G) of FBN1 was not detected in the mother (I:2) and another daughter (II:1) of this family, or in the 383 unrelated normal controls during the mutation screening in the present study. This indicated that c.1708 T>G (p.C570G) of FBN1 cosegregated with affected MFS patients and may serve an important role in the pathogenesis of MFS development in this pedigree.

The p.C570G mutation of FBN1 identified in this family with MFS resulted in a substitution of a highly conserved cysteine residue for glycine in a cbEGF domain of FBN1. This mutation is predicted to abolish one disulfide bond and thus affect the sixth conserved cysteine (C6) of the cbEGF domain; disulfide bonds are essential for the correct EGF-like domain structure. SIFT and PolyPhen-2 predictions indicated that this mutation is critical to protein function, supporting a possible pathogenic effect of this mutation. Evidence has revealed that most FBN1 mutations are clustered in exons 24–32, a hot spot region associated with classic and severe forms of MFS (17,20); mutations in exons 12–15 encoding cbEGF-like domains (C3-C6) cause a mild phenotype of MFS with possible late cardiovascular involvement (21). Evidence from the present study consistently indicated that the identified heterozygous mutation, c.1708T>G, is located at exon 14 and that this cysteine substitution detected in the proband resulted in pectus carinatum and aortic dissection. These two factors correlated with increasing age. However, evident symptoms were not detected in the young affected daughter (II:2), even though significant dilation of the aortic root was identified. Nevertheless, further functional analyses are required to confirm the role of FBN1 and its underlying mechanisms in MFS.

In conclusion, a novel heterozygous mutation, c.1708 T>G (p.C570G), in the FBN1 gene was identified in a Chinese family associated with MFS. The results from the present study enrich the spectrum of MFS-associated mutations of FBN1 and may aid presymptomatic molecular diagnosis of undetermined cases of MFS.

Acknowledgements

The present study was supported by grants from the Natural Science Foundation of China [grant nos. 81670853 (B.G) and 81371048 (B.G)], the Department of Science and Technology of Sichuan Province [grant no. 2015HH0031 (B.G.)] and the Health and Family Planning Commission of Sichuan Province of China [grant no. 16ZD028 (B.G.)].

References

1 

Judge DP and Dietz HC: Marfan's syndrome. Lancet. 366:1965–1976. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Radonic T, de Witte P, Groenink M, de Bruin-Bon RA, Timmermans J, Scholte AJ, van den Berg MP, Baars MJ, van Tintelen JP, Kempers M, et al: Critical appraisal of the revised Ghent criteria for diagnosis of Marfan syndrome. Clin Genet. 80:346–353. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Zhao F, Pan X, Zhao K and Zhao C: Novel mutations of fibrillin-1 gene correlate with different phenotypes of Marfan syndrome in Chinese families. Mol Vis. 19:751–758. 2013.PubMed/NCBI

4 

Boileau C, Jondeau G, Mizuguchi T and Matsumoto N: Molecular genetics of Marfan syndrome. Curr Opin Cardiol. 20:194–200. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Baetens M, Van Laer L, De Leeneer K, Hellemans J, De Schrijver J, Van De Voorde H, Renard M, Dietz H, Lacro RV, Menten B, et al: Applying massive parallel sequencing to molecular diagnosis of Marfan and Loeys-Dietz syndromes. Hum Mutat. 32:1053–1062. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Faivre L, Collod-Beroud G, Child A, Callewaert B, Loeys BL, Binquet C, Gautier E, Arbustini E, Mayer K, Arslan-Kirchner M, et al: Contribution of molecular analyses in diagnosing Marfan syndrome and type I fibrillinopathies: An international study of 1009 probands. J Med Genet. 45:384–390. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Vollbrandt T, Tiedemann K, El-Hallous E, Lin G, Brinckmann J, John H, Bätge B, Notbohm H and Reinhardt DP: Consequences of cysteine mutations in calcium-binding epidermal growth factor modules of fibrillin-1. J Biol Chem. 279:32924–32931. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Dietz HC, Cutting GR, Pyeritz RE, Maslen CL, Sakai LY, Corson GM, Puffenberger EG, Hamosh A, Nanthakumar EJ, Curristin SM, et al: Marfan syndrome caused by a recurrent de novo missense mutation in the fibrillin gene. Nature. 352:337–339. 1991. View Article : Google Scholar : PubMed/NCBI

9 

Faivre L, Collod-Beroud G, Loeys BL, Child A, Binquet C, Gautier E, Callewaert B, Arbustini E, Mayer K, Arslan-Kirchner M, et al: Effect of mutation type and location on clinical outcome in 1,013 probands with Marfan syndrome or related phenotypes and FBN1 mutations: An international study. Am J Hum Genet. 81:454–466. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Cañadas V, Vilacosta I, Bruna I and Fuster V: Marfan syndrome. Part 1: Pathophysiology and diagnosis. Nat Rev Cardiol. 7:256–265. 2010.PubMed/NCBI

11 

Dureau P: Pathophysiology of zonular diseases. Curr Opin Ophthalmol. 19:27–30. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Whiteman P and Handford PA: Defective secretion of recombinant fragments of fibrillin-1: Implications of protein misfolding for the pathogenesis of Marfan syndrome and related disorders. Hum Mol Genet. 12:727–737. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Werner JM, Knott V, Handford PA, Campbell ID and Downing AK: Backbone dynamics of a cbEGF domain pair in the presence of calcium. J Mol Biol. 296:1065–1078. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Dietz HC, Saraiva JM, Pyeritz RE, Cutting GR and Francomano CA: Clustering of fibrillin (FBN1) missense mutations in Marfan syndrome patients at cysteine residues in EGF-like domains. Hum Mutat. 1:366–374. 1992. View Article : Google Scholar : PubMed/NCBI

15 

Dietz HC, McIntosh I, Sakai LY, Corson GM, Chalberg SC, Pyeritz RE and Francomano CA: Four novel FBN1 mutations: Significance for mutant transcript level and EGF-like domain calcium binding in the pathogenesis of Marfan syndrome. Genomics. 17:468–475. 1993. View Article : Google Scholar : PubMed/NCBI

16 

Mizuguchi T, Collod-Beroud G, Akiyama T, Abifadel M, Harada N, Morisaki T, Allard D, Varret M, Claustres M, Morisaki H, et al: Heterozygous TGFBR2 mutations in Marfan syndrome. Nat Genet. 36:855–860. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Loeys B, Nuytinck L, Delvaux I, De Bie S and De Paepe A: Genotype and phenotype analysis of 171 patients referred for molecular study of the fibrillin-1 gene FBN1 because of suspected Marfan syndrome. Arch Intern Med. 161:2447–2454. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Ogawa N, Imai Y, Takahashi Y, Nawata K, Hara K, Nishimura H, Kato M, Takeda N, Kohro T, Morita H, et al: Evaluating Japanese patients with the Marfan syndrome using high-throughput microarray-based mutational analysis of fibrillin-1 gene. Am J Cardiol. 108:1801–1807. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Schrijver I, Liu W, Brenn T, Furthmayr H and Francke U: Cysteine substitutions in epidermal growth factor-like domains of fibrillin-1: Distinct effects on biochemical and clinical phenotypes. Am J Hum Genet. 65:1007–1020. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Tiecke F, Katzke S, Booms P, Robinson PN, Neumann L, Godfrey M, Mathews KR, Scheuner M, Hinkel GK, Brenner RE, et al: Classic, atypically severe and neonatal Marfan syndrome: Twelve mutations and genotype-phenotype correlations in FBN1 exons 24–40. Eur J Hum Genet. 9:13–21. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Pepe G, Lapini I, Evangelisti L, Attanasio M, Giusti B, Lucarini L, Fattori R, Pellicanò G, Scrivanti M, Porciani MC, et al: Is ectopia lentis in some cases a mild phenotypic expression of Marfan syndrome? Need for a long-term follow-up. Mol Vis. 13:2242–2247. 2007.PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xiao Y, Liu X, Guo X, Liu L, Jiang L, Wang Q and Gong B: A novel FBN1 mutation causes autosomal dominant Marfan syndrome. Mol Med Rep 16: 7321-7328, 2017
APA
Xiao, Y., Liu, X., Guo, X., Liu, L., Jiang, L., Wang, Q., & Gong, B. (2017). A novel FBN1 mutation causes autosomal dominant Marfan syndrome. Molecular Medicine Reports, 16, 7321-7328. https://doi.org/10.3892/mmr.2017.7544
MLA
Xiao, Y., Liu, X., Guo, X., Liu, L., Jiang, L., Wang, Q., Gong, B."A novel FBN1 mutation causes autosomal dominant Marfan syndrome". Molecular Medicine Reports 16.5 (2017): 7321-7328.
Chicago
Xiao, Y., Liu, X., Guo, X., Liu, L., Jiang, L., Wang, Q., Gong, B."A novel FBN1 mutation causes autosomal dominant Marfan syndrome". Molecular Medicine Reports 16, no. 5 (2017): 7321-7328. https://doi.org/10.3892/mmr.2017.7544