Skip to main content
Erschienen in: BMC Medical Genetics 1/2013

Open Access 01.12.2013 | Research article

Sequencing of NOTCH1, GATA5, TGFBR1 and TGFBR2genes in familial cases of bicuspid aortic valve

verfasst von: Ilenia Foffa, Lamia Ait Alì, Paola Panesi, Massimiliano Mariani, Pierluigi Festa, Nicoletta Botto, Cecilia Vecoli, Maria Grazia Andreassi

Erschienen in: BMC Medical Genetics | Ausgabe 1/2013

Abstract

Background

The purpose of our study was to investigate the potential contribution of germline mutations in NOTCH1, GATA5 and TGFBR1 and TGFBR2 genes in a cohort of Italian patients with familial Bicuspid Aortic Valve (BAV).

Methods

All the coding exons including adjacent intronic as well as 5 and 3 untranslated (UTR) sequences of NOTCH1, GATA5, TGFBR1 and TGFBR2 genes were screened by direct gene sequencing in 11 index patients (8 males; age = 42 ± 19 years) with familial BAV defined as two or more affected members.

Results

Two novel mutations, a missense and a nonsense mutation (Exon 5, p.P284L; Exon 26, p.Y1619X), were found in the NOTCH1 gene in two unrelated families. The mutations segregated with the disease in these families, and they were not found on 200 unrelated chromosomes from ethnically matched controls. No pathogenetic mutation was identified in GATA5, TGFBR1 and TGFBR2 genes.

Conclusions

Two novel NOTCH1 mutations were identified in two Italian families with BAV, highlighting the role of a NOTCH1 signaling pathway in BAV and its aortic complications. These findings are of relevance for genetic counseling and clinical care of families presenting with BAV. Future studies are needed in order to unravel the still largely unknown genetics of BAV.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2350-14-44) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

IF participated in the design of the study, carried out the molecular genetic studies, performed the statistical analysis and drafted the manuscript. MGA conceived the study, participated in its design and coordination and critically revised the manuscript. PP carried out the molecular genetic studies. MM, AAL and PF participated in design of the study and in the enrollment of the patients. NB and CV performed the genetic analysis and critically revised the manuscript. All authors read and approved the final manuscript.

Background

Bicuspid aortic valve (BAV) is the most common congenital cardiac malformation, with a prevalence estimated between 0.5% and 2% [1, 2]. Nearly all patients with BAV develop serious aortic complications, including significant valve regurgitation, severe aortic stenosis, aortic aneurysm and dissection [3, 4]. Therefore, the burden of BAV disease is more significant than any other congenital cardiac abnormalities [4]. Clinical studies have reported high heritability in families of patients with BAV [57], suggesting a Mendelian pattern of inheritance and the need for echocardiography screening in first-degree relatives of patients with BAV [8].
A family-based linkage analysis has identified genomic regions on chromosomes 18q, 5q and 13q responsible for BAV and associated cardiac malformations, but the precise genes within these regions were not defined [9].
To date, only mutations in the NOTCH1 gene, a signaling transmembrane receptor (2,556 amino acids) involved in multiple cell aspects of vascular development, have been associated with dominantly inherited BAV in a small number of families [10].
However, the rate of NOTCH1 gene mutations in familial forms of BAV and their association with different clinical complications remains incomplete [11, 12]. Interestingly, McBride et al. demonstrated that mutations in the gene NOTCH1 are found in cases of left ventricular outflow tract (LVOT) malformations including aortic valve stenosis, coarctation of the aorta and hypoplastic left heart syndrome [13]. In particular, a much higher rate of bicuspid aortic valve (BAV) has been found in families with LVOT malformations compared to the general population, suggesting that BAV may be a forme frusta of the more serious LVOT malformations [13].
Recently, a missense mutation in transforming growth factor-beta receptor 2 gene (TGFBR2) was found in a patient with BAV and aortic aneurysm [14], but earlier studies found no mutation in either TGFBR1 or TGFBR2 in patients with familial and sporadic BAV disease [15, 16].
Finally, GATA5 was reported to have an essential role in cardiac morphogenesis and in aortic valve development [17, 18], and targeted deletion of this gene in mice leads to partially penetrant BAV [19]. Accordingly, a very recent study investigated the relationship between GATA5 gene variations and BAV disease identifying rare non-synonymous variations in the functionally important GATA5 transcriptional activation domain [20]. Conversely from NOTCH1 gene, the involvement of GATA5 has not been described in other LVOT malformations.
However, further studies are needed to assess the involvement of NOTCH1, GATA5 and TGFBR1 and TGFBR2 mutations in patients with BAV and its associated aortic complications.
The purpose of our study was to investigate the potential contribution of germline mutations in NOTCH1, GATA5, TGFBR1 and TGFBR2 genes in a cohort of Italian patients with familial BAV.

Methods

Study population

A cohort of 11 unrelated Italian patients (8 males; age = 42 ± 19 years) with familial BAV were recruited for this study. Patients with sporadic BAV and/or with coexisting syndromes, such as Marfan syndrome, Turner syndrome and Di George syndrome or unknown syndromes were excluded from the study. Clinical evaluation consisted of medical history, physical examination, 2-dimensional echocardiography and a complete Magnetic Resonance Imaging (MRI) study. A detailed family history was obtained for each proband. Familial BAV was defined if two or more affected relatives had proven BAV diagnosed by echocardiography or cardiac magnetic resonance. Figure 1 shows the pedigree of BAV patients. Patient’s phenotype is reported in Table 1. Two of the probands (18%) had BAV with unknown morphology. Of the 9 probands with known BAV morphology, 7 (63%) had fusion of the right/left coronary commissure whereas 2 (18%) had a valve with anterior-posterior sinus without raphe.We also included a control cohort comprised of 100 ethnically unrelated individuals (60 male; age = 35 ± 16 years) without BAV as shown by echocardiography. This study was conducted with informed consent of every participant subject and was approved by the local Ethical Research Committee.
Table 1
Clinical and demographic characteristics of study population
Proband
Sex
Age
Diagnosis
Phenotype
Other cardiac malformation
A
M
40
AS and AAD
AP
/
B
M
25
AR and AAD
R-L
/
C
F
40
AR
unk
/
D
F
21
AR
R-L
CoA
E
M
37
BAV
R-L
/
F
M
74
AS and AAD
unk
/
G
M
17
AI and AAD
R-L
/
H
M
67
AR and AAD
R-L
/
I
M
38
AS
R-L
CoA
J
F
65
AS and AAD
AP
/
K
M
37
AR and AAD
R-L
/
AS: aortic stenosis; AAD: Ascending Aortic Dilation; AR: Aortic regurgitation; AI: aortic insufficiency; CoA: Aortic Coarctation; AP: anterior-posterior no raphe; R-L: right/left coronary commissure; unk: unknown.

Mutational sequencing analysis

A mutational screening was performed by direct gene sequencing of all coding exons including adjacent intronic as well as 5 and 3 untranslated sequences of NOTCH1, GATA5 and TGFBR1, TGFBR2 genes. Genomic DNA was extracted from peripheral blood cells using the QIAGEN BioRobot® EZ1 System. Exons and the flanking intronic sequences were amplified by polymerase chain reaction (PCR) using specific primers as described previously for NOTCH1 and TGFBR1 and TGFBR2[11, 21] and using Primer3 (v. 0.4.0) software (http://​frodo.​wi.​mit.​edu/​primer3/​) based on the cDNA sequences available in GenBank for GATA5 screening.
Approximately, PCR amplifications were performed with a volume of 50 microl mixture containing 1.5 mM MgCl2, 10 mMdNTP, 50 ng genomic DNA, 20 micromol of each primer and 2.5 U Taq DNA polymerase. PCR product was used for PCR sequencing reaction with the CEQ DTCS Quick Start Kit. The sequencing reaction products were purified by precipitation with ethanol, re-suspended in a sample loading solution and analyzed with a CEQ 8800 capillary sequence (Beckman Coulter, Germany) according to the manufacturer’s protocol. Resulting sequences were analyzed using CEQ 8800 software packages and aligned against a reference sequence obtained from GenBank.
For prediction of the functional consequences of mutations on protein sequence, we used software programs available on the internet, namely PolyPhen-2(http://​genetics.​bwh.​harvard.​edu/​pph/​) SIFT (http://​sift.​jcvi.​org/​) and Mutation Taster (http://​www.​mutationtaster.​org/​ ). PolyPhen-2 predicts the effect of an amino acid substitution on the structure and function of a protein, using sequence homology. The PolyPhen-2 score represents the probability that a substitution is damaging, so values nearer 1 are more confidently predicted to be deleterious (note that this is the opposite of SIFT).
SIFT uses sequence homology to predict whether an amino acid substitution will affect the protein function. SIFT score <0.05 indicates the amino acid substitution is damaging while scores ≥ 0.05 are predicted to be tolerant. Mutation Taster is a free, web-based application for rapid evaluation of the disease-causing potential of DNA sequence alterations. Mutation Taster integrates information from different biomedical databases and uses established analysis tools. Analyses comprise evolutionary conservation, splice-site changes, loss of protein features and changes that might affect the amount of mRNA.
Detected mutations in probands were confirmed by sequencing in the opposite direction from another two independent PCR products.
The presence of a novel mutation was assessed in 200 ethnically reference alleles derived from volunteers by PCR-based restriction fragment length polymorphism (PCR-RFLP) or by direct gene sequencing.

Results

Clinical and demographic characteristics of each patient are summarized in Table 1. The group comprised 73% males, and coarctation of the aorta (CoA) as coexistent cardiovascular anomalies occurred in 18% of the cohort.

Genetic analysis of NOTCH1gene

A mutational screening was performed by direct gene sequencing of all 34 coding exons including adjacent intronic as well as 5 and 3 untranslated sequences of NOTCH1 gene. Seventeen separate NOTCH1 sequence variants listed previously in public dbSNPs or in the literature were found in our patients, while five variants were novel. These included one synonymous variant (Exon 21, p.G1166G), two intronic (IVS11 + 63; IVS32-41) and two novel mutations (Exon 5,p.P284L; Exon 26, p.Y1619X). Details of the specific sequence variants and number of positive patients are summarized in Table 2.
Table 2
Genetic variants within the human NOTCH1 gene identified in familial BAV patients
Genomic position (NG_007458.1)
Position NOTCH1gene
cDNA position (NM_017617.3)
Protein position
Number of patients
rsSNP number (dbSNP)
g.26979 T > C
Exon 3
c.312 T > C
p.N104N
6
rs4489420
g.31330 C > T
Exon 5
c.851C > T
p.P284L
1
New
g.31331 G > A
Exon 5
c.852 G > A
p.P284P
4
rs2229975
g.34650 T > C
IVS 9-43
  
3
rs4880099
g.33525A > G
IVS 9 + 10
  
2
rs11145767
g.33601 C > A/T
IVS 9 + 86
  
2
rs113341997
g.33617C > T
IVS 9 + 102
  
1
rs10781498
g.33620C > T
IVS 9 + 105
  
1
rs11574887
g.34772C > T
Exon 10
c.1635 C > T
p.D545D
1
rs11574889
g.35062A > G
IVS 11-9
  
3
rs3124603
g.35367 G > A
IVS 11 + 63
  
1
New
g.35591C > T
IVS 12 + 94
  
1
rs62579232
g.36347G > A
IVS 13 + 70
  
2
rs3812609
g.37307 T > C
Exon 14
c.2265 T > C
p.N755N
4
rs2229971
g.39978G > A
IVS 16-4
  
3
rs3125001
g.40058C > T
Exon 17
c.2664 C > T
p.H888H
1
rs61751548
g.40085C > T
Exon 17
c.2691 C > T
p.A897A
1
rs11574895
g.42820 C > T
Exon 21
c.3498 C > T
p.G1166G
1
New
g.45953 C > G
Exon 26
c.4856 C > G
p.Y1619X
1
New
g.47532C > T
Exon 27
c.5094 C > T
p.D1698D
3
rs10521
g.48831 T > C
IVS 30-43
  
2
rs3124594
g.49928C > T
IVS 30-12
  
1
rs11574908
g.51487 C > T
IVS 32-41
  
1
New
The novel C > T heterozygous substitution in exon 5, resulting in the substitution of a Proline with a Leucine (P284L) at position 284, was found in the sequencing electropherograms of a 40-year-old male patient (Proband A; Figure 2). The identified missense mutation was absent in 200 ethnically reference alleles derived from volunteers. PolyPhen-2 analysis predicted the p.P284L mutation as probably damaging with a score of 0.993. This result was confirmed by SIFT (score: 0.04).
The missense mutation (p.P284L) identified in our proband, tested by Mutation Taster, induced the loss of an important residue with putative significance for calcium binding to epidermal growth factor (EGF)-like 7 domain.
The proband, at the age of 39 years, presented severe calcified aortic valve and normal diameter of ascending aorta (39 mm). One year later, the patient underwent elective aortic valve and ascending aorta repair (48 mm) replacement. Family history and echocardiography analysis revealed the presence of BAV in his 7-year-old son. Genetic screening of the proband’s child confirmed the presence of p.P284L mutation in exon 5 of NOTCH1 gene.
Moreover, we detected a novel heterozygous C-to-G transition, in exon 26, of nucleotide 4856 leading to a premature stop codon at position 1619 (p.Y1619X) instead of tyrosine in the sequencing electropherograms of a 38-year-old male patient (Proband I; Figure 2). The identified mutation was absent in 200 ethnically reference alleles derived from volunteers. At age 6 months the proband had surgery to correct the CoA, while at age 25 years he underwent elective aortic valve replacement. Family history and echocardiography analysis revealed the presence of BAV in his 5-year-old daughter. Genetic screening of the proband’s child confirmed the presence of p.Y1619X mutation in exon 26 of NOTCH1 gene.

Genetic analysis of GATA5gene

Sequencing of all 6 exons and splice signal sequences of GATA5 in our cohort revealed five synonymous, one non-synonymous and three intronic variants. Details of the specific sequence variants and number of positive patients are summarized in Table 3. We identified one novel intronic variant (C > T; IVS 5–15) in one patient, but it was also present in our control group.
Table 3
Genetic variants within the human GATA5 gene identified in familial BAV patients
Position GATA5gene
HGVS names
Protein position
Number of patients
rsSNP number (dbSNP)
Exon1
c.199 A > C
p.Thr67Pro
5
rs6142775
Exon 2
c.609 C > T
p.Asp203Asp
7
rs41305803
Exon 2
c.678 C > T
p.Leu226Leu
1
rs139428354
IVS 4-14
c.826-14 G > A
 
1
rs140408446
Exon 4
c.852 G > A
p.Lys284Lys
8
rs6587239
IVS 5-16
c.914-16 C > T
 
1
New
Exon 5
c.981 G > C
p.Ser327Ser
7
rs6061243
IVS 6-78
c.1039-78 C > G
 
5
rs113912772
Exon6
c.1128 A > G
p.Pro376Pro
11
rs6061550
NM_080473.4.

Genetic analysis of TGFBR1 and TGFBR2genes

TGFBR1 and TGFBR2 genes were analyzed by direct gene sequencing of all coding exons including adjacent intronic as well as 5 and 3 untranslated sequences. No pathogenetic mutation was identified; we found only known common genetic polymorphisms. Details of the specific sequence variants in TGFBR1 and TGFBR2 genes are summarized in Table 4.
Table 4
Genetic variants within the human TGFBR1 and TGFBR2 genes identified in familial BAV patients
Gene
Position TGFBR1 TGFBR2gene
HGVS names
Protein position
Number of patients
rsSNP number
TGFBR1
Exon 1
c.68_76delCGGCGGCG
p.Ala23Ala
4
rs11466445
 
IVS 7 + 24
c.1255 + 24 G > A
 
2
rs334354
TGFBR2
IVS 3 + 7
c.338 + 7 A > G
 
2
rs1155705
 
IVS 5-4
c.455-4 T > A
 
3
rs11466512
 
Exon 4
c.458delA
p.Lys153
8
rs79375991
TGFBR1: NM_004612.2 NG_007461.1.
TGFBR2: NM_001024847.2 NG_007490.1.

Discussion

In our cohort of 11 familial patients with BAV two novel mutations were found in the NOTCH1 gene in two unrelated families. The mutations segregated with the disease in these families, and they were not found in 200 chromosomes from ethnically matched controls.
On the contrary, analysis of GATA5, TGFBR1 and TGFBR2 genes identified only known genetic variants. Our findings confirm the important role of NOTCH1 as conserved intracellular regulator in the pathogenesis of congenital valve disease and in its complications. Indeed, the finding of two independent de novo mutations in this gene in a small number of affected subjects is compelling evidence of disease causation.
NOTCH1 encodes for a transmembrane protein that activates a signaling pathway with a major role in cardiac embryogenesis, including aortic and pulmonary valve development as well as the development and maintenance of the aorta and other great vessels [22]. Murine model studies indicate that Notch signaling promotes the epithelial-to mesenchyme transition that gives rise to the primordial cardiac valve, which later is sculpted into mature valves [23]. Moreover, a recent study showed that heterozygous Notch1-null (Notch1(+/)(−) mice had greater than fivefold more aortic valve calcification than age- and sex-matched wild type littermates [24].
Indeed, in vitro studies showed that Notch1 signaling repressed valvular Bmp2 expression, and de-repression of Bmp2 was involved in calcification induced by Notch1 inhibition. Thus, Notch1 signaling appears to prevent aortic valve calcification in part by repressing Bmp2 expression within the valve [24].
Defective NOTCH signaling might also contribute to aortic valve calcification by repressing the activity of the hairy-related family of transcription factors (Hrt), and thereby also repressing the Runx2/Cbfa1 pathway, a central transcriptional regulator of osteoblast cell fate [24, 25].
In addition, there is increasing evidence of interaction between the Notch1 pathway and transforming growth factor beta (TGFβ) [26]. This is particularly important in light of evidence showing that TGFβ plays an important role in vascular remodeling and alterations in TGFβ signaling activity might enhance matrix degradation [27, 28].
In 2005, the discovery of a nonsense (p.R1108X) and a frameshift mutation (p.H1505del) in two families represented the first demonstration of NOTCH1 germline mutations as a cause of BAV and severe valve calcification in non-syndromic autosomal-dominant human pedigrees [10].
Subsequently, two novel mutations (p.T596M and p.P1797H) located in highly conserved regions of the protein were identified in patients with sporadic BAV, providing evidence that mutations in NOTCH1 do not only play a role in familial BAV, but can also be observed in approximately 4% of sporadic cases [11]. Furthermore, a targeted mutational analysis of NOTCH1 demonstrated the presence of two novel mutations (p.A1343V and p.P1390T) among patients with BAV with sporadic and concomitant thoracic aortic aneurysms, providing evidence that mutations in NOTCH1 may also confer susceptibility to aortic aneurysm formation [12].
In a very recent paper, Kent et al. suggested a NOTCH1-dependent mechanism that produces stenotic, insufficient and/or calcified aortic valves with a rare aneurysm, and a NOTCH1-independent mechanism that produces highly penetrant ascending aortic aneurysm (AscAA) in the presence of a non-calcified and often normally functioning BAV [29]. Our data appear to agree with and fit well into the proposal made in this article by Kent et al., since we found two novel mutations in these patients with valve malformation, calcification, and dysfunction as predominant phenotype caused by altered NOTCH1 signaling.
Conversely, we did not identify any pathogenetic mutations in GATA5. The non-synonymous variant Thr67Pro located in the transcriptional activator domain encoded by exon 1, detected with a frequency of 45% in our population, is a common genetic polymorphism previously described [20, 30].
Regarding to TGFBR1 and TGFBR2 genes, in contrast with a recent paper showing a missense mutation in TGFBR2 gene in a patient with BAV and aortic aneurysm [14], our findings support the studies suggesting that mutations in these genes are rarely identified in patients with familial BAV [15, 16].

Conclusion

Although the small number of patients (11) might be an important study limitation, we have identified two novel NOTCH1 mutations in two Italian families with BAV. Our findings confirm the critical role of NOCTH1 in the development of aortic valve and aortic complications. Genetic screening for NOTCH1 mutation may be considered for diagnosis and family screening as part of the standard management of patients with family history of BAV. However, the etiology of this common valve disease is largely unknown. Future studies based on new genetic screening strategies, the so-called next-generation sequencing, are needed in order to unravel the still largely unknown genetics of BAV.
Abbreviations: BAV = Bicuspid Aortic Valve, TGFBR1 = transforming growth factor-beta receptorI gene; TGFBR2 = transforming growth factor-beta receptorII gene; PCR-RFLP = PCR-based restriction fragment length polymorphism; MAF = minor allele frequency; CoA = coarctation of the aorta; TGFβ = transforming growth factor beta; MRI = Magnetic Resonance Imaging; AscAA = ascending aortic aneurysm; LVOT = left ventricular outflow tract.

Acknowledgments

We are indebted with Ms. Alison Frank for her invaluable help in editing the manuscript.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

IF participated in the design of the study, carried out the molecular genetic studies, performed the statistical analysis and drafted the manuscript. MGA conceived the study, participated in its design and coordination and critically revised the manuscript. PP carried out the molecular genetic studies. MM, AAL and PF participated in design of the study and in the enrollment of the patients. NB and CV performed the genetic analysis and critically revised the manuscript. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
2.
Zurück zum Zitat Laforest B, Nemer M: Genetic insights into bicuspid aortic valve formation. Cardiol Res Pract. 2012, 2012: 180297-PubMedPubMedCentral Laforest B, Nemer M: Genetic insights into bicuspid aortic valve formation. Cardiol Res Pract. 2012, 2012: 180297-PubMedPubMedCentral
3.
Zurück zum Zitat Michelena HI, Khanna AD, Mahoney D, Margaryan E, Topilsky Y, Suri RM, Eidem B, Edwards WD, Sundt TM, Enriquez-Sarano M: Incidence of aortic complications in patients with bicuspid aortic valves. JAMA. 2011, 306: 1104-1112. 10.1001/jama.2011.1286.CrossRefPubMed Michelena HI, Khanna AD, Mahoney D, Margaryan E, Topilsky Y, Suri RM, Eidem B, Edwards WD, Sundt TM, Enriquez-Sarano M: Incidence of aortic complications in patients with bicuspid aortic valves. JAMA. 2011, 306: 1104-1112. 10.1001/jama.2011.1286.CrossRefPubMed
4.
Zurück zum Zitat Lewin MB, Otto CM: The bicuspid aortic valve: adverse outcomes from infancy to old age. Circulation. 2005, 111: 832-834. 10.1161/01.CIR.0000157137.59691.0B.CrossRefPubMed Lewin MB, Otto CM: The bicuspid aortic valve: adverse outcomes from infancy to old age. Circulation. 2005, 111: 832-834. 10.1161/01.CIR.0000157137.59691.0B.CrossRefPubMed
5.
Zurück zum Zitat Huntington K, Hunter AG, Chan KL: A prospective study to assess the frequency of familial clustering of congenital bicuspid aortic valve. J Am Coll Cardiol. 1997, 30: 1809-1812. 10.1016/S0735-1097(97)00372-0.CrossRefPubMed Huntington K, Hunter AG, Chan KL: A prospective study to assess the frequency of familial clustering of congenital bicuspid aortic valve. J Am Coll Cardiol. 1997, 30: 1809-1812. 10.1016/S0735-1097(97)00372-0.CrossRefPubMed
6.
Zurück zum Zitat Cripe L, Andelfinger G, Martin J, Shooner K, Benson DW: Bicuspid aortic valve is heritable. J Am Coll Cardiol. 2004, 44: 138-143. 10.1016/j.jacc.2004.03.050.CrossRefPubMed Cripe L, Andelfinger G, Martin J, Shooner K, Benson DW: Bicuspid aortic valve is heritable. J Am Coll Cardiol. 2004, 44: 138-143. 10.1016/j.jacc.2004.03.050.CrossRefPubMed
7.
Zurück zum Zitat Glick BN, Roberts WC: Congenitally bicuspid aortic valve in multiple family members. Am J Cardiol. 1994, 73: 400-404. 10.1016/0002-9149(94)90018-3.CrossRefPubMed Glick BN, Roberts WC: Congenitally bicuspid aortic valve in multiple family members. Am J Cardiol. 1994, 73: 400-404. 10.1016/0002-9149(94)90018-3.CrossRefPubMed
8.
Zurück zum Zitat Warnes CA, Williams RG, Bashore TM: ACC/AHA 2008 guidelines for the management of adults with congenital heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing committee to develop guidelines on the management of adults with congenital heart disease). J Am Coll Cardiol. 2008, 52: e1-e121. 10.1016/j.jacc.2008.05.007.CrossRef Warnes CA, Williams RG, Bashore TM: ACC/AHA 2008 guidelines for the management of adults with congenital heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing committee to develop guidelines on the management of adults with congenital heart disease). J Am Coll Cardiol. 2008, 52: e1-e121. 10.1016/j.jacc.2008.05.007.CrossRef
9.
Zurück zum Zitat Martin LJ, Ramachandran V, Cripe LH, Hinton RB, Andelfinger G, Tabangin M, Shooner K, Keddache M, Benson DW: Evidence in favor of linkage to human chromosomal regions 18q, 5q and 13q for bicuspid aortic valve and associated cardiovascular malformations. Hum Genet. 2007, 121: 275-284. 10.1007/s00439-006-0316-9.CrossRefPubMed Martin LJ, Ramachandran V, Cripe LH, Hinton RB, Andelfinger G, Tabangin M, Shooner K, Keddache M, Benson DW: Evidence in favor of linkage to human chromosomal regions 18q, 5q and 13q for bicuspid aortic valve and associated cardiovascular malformations. Hum Genet. 2007, 121: 275-284. 10.1007/s00439-006-0316-9.CrossRefPubMed
10.
Zurück zum Zitat Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King IN, Grossfeld PD, Srivastava D: Mutations in NOTCH1 cause aortic valve disease. Nature. 2005, 437: 270-274. 10.1038/nature03940.CrossRefPubMed Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King IN, Grossfeld PD, Srivastava D: Mutations in NOTCH1 cause aortic valve disease. Nature. 2005, 437: 270-274. 10.1038/nature03940.CrossRefPubMed
11.
Zurück zum Zitat Mohamed SA, Aherrahrou Z, Liptau H, Erasmi AW, Hagemann C, Wrobel S, Borzym K, Schunkert H, Sievers HH, Erdmann J: Novel missense mutations (p.T596M and p.P1797H) in NOTCH1 in patients with bicuspid aortic valve. Biochem Biophys Res Commun. 2006, 345: 1460-1465. 10.1016/j.bbrc.2006.05.046.CrossRefPubMed Mohamed SA, Aherrahrou Z, Liptau H, Erasmi AW, Hagemann C, Wrobel S, Borzym K, Schunkert H, Sievers HH, Erdmann J: Novel missense mutations (p.T596M and p.P1797H) in NOTCH1 in patients with bicuspid aortic valve. Biochem Biophys Res Commun. 2006, 345: 1460-1465. 10.1016/j.bbrc.2006.05.046.CrossRefPubMed
12.
Zurück zum Zitat McKellar SH, Tester DJ, Yagubyan M, Majumdar R, Ackerman MJ, Sundt TM: Novel NOTCH1 mutations in patients with bicuspid aortic valve disease and thoracic aortic aneurysms. J Thorac Cardiovasc Surg. 2007, 134: 290-296. 10.1016/j.jtcvs.2007.02.041.CrossRefPubMed McKellar SH, Tester DJ, Yagubyan M, Majumdar R, Ackerman MJ, Sundt TM: Novel NOTCH1 mutations in patients with bicuspid aortic valve disease and thoracic aortic aneurysms. J Thorac Cardiovasc Surg. 2007, 134: 290-296. 10.1016/j.jtcvs.2007.02.041.CrossRefPubMed
13.
Zurück zum Zitat McBride KL, Riley MF, Zender GA, Fitzgerald-Butt SM, Towbin JA, Belmont JW, Cole SE: NOTCH1 mutations in individuals with left ventricular outflow tract malformations reduce ligand-induced signaling. Hum Mol Genet. 2008, 17: 2886-2893. 10.1093/hmg/ddn187.CrossRefPubMedPubMedCentral McBride KL, Riley MF, Zender GA, Fitzgerald-Butt SM, Towbin JA, Belmont JW, Cole SE: NOTCH1 mutations in individuals with left ventricular outflow tract malformations reduce ligand-induced signaling. Hum Mol Genet. 2008, 17: 2886-2893. 10.1093/hmg/ddn187.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Girdauskas E, Schulz S, Borger MA, Mierzwa M, Kuntze T: Transforming growth factor-beta receptor Type II mutation in a patient with bicuspid aortic valve disease and intraoperative aortic dissection. Ann Thorac Surg. 2011, 91: e70-e71. 10.1016/j.athoracsur.2010.12.060.CrossRefPubMed Girdauskas E, Schulz S, Borger MA, Mierzwa M, Kuntze T: Transforming growth factor-beta receptor Type II mutation in a patient with bicuspid aortic valve disease and intraoperative aortic dissection. Ann Thorac Surg. 2011, 91: e70-e71. 10.1016/j.athoracsur.2010.12.060.CrossRefPubMed
15.
Zurück zum Zitat Loscalzo ML, Goh DL, Loeys B, Kent KC, Spevak PJ, Dietz HC: Familial thoracic aortic dilation and bicommissural aortic valve: a prospective analysis of natural history and inheritance. Am J Med Genet A. 2007, 143A: 1960-1967. 10.1002/ajmg.a.31872.CrossRefPubMed Loscalzo ML, Goh DL, Loeys B, Kent KC, Spevak PJ, Dietz HC: Familial thoracic aortic dilation and bicommissural aortic valve: a prospective analysis of natural history and inheritance. Am J Med Genet A. 2007, 143A: 1960-1967. 10.1002/ajmg.a.31872.CrossRefPubMed
16.
Zurück zum Zitat Arrington CB, Sower CT, Chuckwuk N, Stevens J, Leppert MF, Yetman AT, Bowles NE: Absence of TGFBR1 and TGFBR2 mutations in patients with bicuspid aortic valve and aortic dilation. Am J Cardiol. 2008, 102: 629-631. 10.1016/j.amjcard.2008.04.044.CrossRefPubMed Arrington CB, Sower CT, Chuckwuk N, Stevens J, Leppert MF, Yetman AT, Bowles NE: Absence of TGFBR1 and TGFBR2 mutations in patients with bicuspid aortic valve and aortic dilation. Am J Cardiol. 2008, 102: 629-631. 10.1016/j.amjcard.2008.04.044.CrossRefPubMed
17.
Zurück zum Zitat Laforest B, Nemer M: GATA5 interacts with GATA4 and GATA6 in outflow tract development. Dev Biol. 2011, 358: 368-378. 10.1016/j.ydbio.2011.07.037.CrossRefPubMed Laforest B, Nemer M: GATA5 interacts with GATA4 and GATA6 in outflow tract development. Dev Biol. 2011, 358: 368-378. 10.1016/j.ydbio.2011.07.037.CrossRefPubMed
18.
Zurück zum Zitat Charron F, Paradis P, Bronchain O, Nemre G, Nemer M: Cooperative interaction between GATA-4 and GATA-6 regulates myocardial gene expression. Moll Cell Biol. 1999, 19: 4355-CrossRef Charron F, Paradis P, Bronchain O, Nemre G, Nemer M: Cooperative interaction between GATA-4 and GATA-6 regulates myocardial gene expression. Moll Cell Biol. 1999, 19: 4355-CrossRef
19.
20.
Zurück zum Zitat Padang R, Bagnall RD, Richmond DR, Bannon P, Semsarian C: Rare non-synonymous variations in the transcriptional activation domains of GATA5 in bicuspid aortic valve disease. J Mol Cell Cardiol. 2012, 53: 277-281. 10.1016/j.yjmcc.2012.05.009.CrossRefPubMed Padang R, Bagnall RD, Richmond DR, Bannon P, Semsarian C: Rare non-synonymous variations in the transcriptional activation domains of GATA5 in bicuspid aortic valve disease. J Mol Cell Cardiol. 2012, 53: 277-281. 10.1016/j.yjmcc.2012.05.009.CrossRefPubMed
21.
Zurück zum Zitat Santiago-Sim T, Mathew-Joseph S, Pannu H, Milewicz DM, Seidman CE, Seidman JG, Kim DH: Sequencing of TGF-beta pathway genes in familial cases of intracranial aneurysm. Stroke. 2009, 40: 1604-1611. 10.1161/STROKEAHA.108.540245.CrossRefPubMedPubMedCentral Santiago-Sim T, Mathew-Joseph S, Pannu H, Milewicz DM, Seidman CE, Seidman JG, Kim DH: Sequencing of TGF-beta pathway genes in familial cases of intracranial aneurysm. Stroke. 2009, 40: 1604-1611. 10.1161/STROKEAHA.108.540245.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat O’Brien KD: Epidemiology and genetics of calcific aortic valve disease. J Investig Med. 2007, 55: 284-291. 10.2310/6650.2007.00010.CrossRefPubMed O’Brien KD: Epidemiology and genetics of calcific aortic valve disease. J Investig Med. 2007, 55: 284-291. 10.2310/6650.2007.00010.CrossRefPubMed
23.
Zurück zum Zitat Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC, de la Pompa JL: Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 2004, 18: 99-115. 10.1101/gad.276304.CrossRefPubMedPubMedCentral Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC, de la Pompa JL: Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 2004, 18: 99-115. 10.1101/gad.276304.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Nigam V, Srivastava D: Notch1 represses osteogenic pathways in aortic valve cells. J Mol Cell Cardiol. 2009, 47: 828-834. 10.1016/j.yjmcc.2009.08.008.CrossRefPubMedPubMedCentral Nigam V, Srivastava D: Notch1 represses osteogenic pathways in aortic valve cells. J Mol Cell Cardiol. 2009, 47: 828-834. 10.1016/j.yjmcc.2009.08.008.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat O’Brien KD: Pathogenesis of calcific aortic valve disease: a disease process comes of age (and a good deal more). Arterioscler Thromb Vasc Biol. 2006, 26: 1721-1728. 10.1161/01.ATV.0000227513.13697.ac.CrossRefPubMed O’Brien KD: Pathogenesis of calcific aortic valve disease: a disease process comes of age (and a good deal more). Arterioscler Thromb Vasc Biol. 2006, 26: 1721-1728. 10.1161/01.ATV.0000227513.13697.ac.CrossRefPubMed
26.
Zurück zum Zitat Klüppel M, Wrana JL: Turning it up a Notch: cross-talk between TGF beta and Notch signaling. Bioessays. 2005, 27: 115-118. 10.1002/bies.20187.CrossRefPubMed Klüppel M, Wrana JL: Turning it up a Notch: cross-talk between TGF beta and Notch signaling. Bioessays. 2005, 27: 115-118. 10.1002/bies.20187.CrossRefPubMed
27.
Zurück zum Zitat El-Hamamsy I, Yacoub MH: Cellular and molecular mechanisms of thoracic aortic aneurysms. Nat Rev Cardiol. 2009, 6: 771-786. 10.1038/nrcardio.2009.191.CrossRefPubMed El-Hamamsy I, Yacoub MH: Cellular and molecular mechanisms of thoracic aortic aneurysms. Nat Rev Cardiol. 2009, 6: 771-786. 10.1038/nrcardio.2009.191.CrossRefPubMed
28.
Zurück zum Zitat Yetman AT, Graham T: The dilated aorta in patients with congenital cardiac defects. J Am Coll Cardiol. 2009, 53: 461-467. 10.1016/j.jacc.2008.10.035.CrossRefPubMed Yetman AT, Graham T: The dilated aorta in patients with congenital cardiac defects. J Am Coll Cardiol. 2009, 53: 461-467. 10.1016/j.jacc.2008.10.035.CrossRefPubMed
29.
Zurück zum Zitat Kent KC, Crenshaw ML, Goh DL, Dietz HC: Genotype-phenotype correlation in patients with bicuspid aortic valve and aneurysm. J Thorac Cardiovasc Surg. 2012, pii: S0022-5223(12)01214-7 Kent KC, Crenshaw ML, Goh DL, Dietz HC: Genotype-phenotype correlation in patients with bicuspid aortic valve and aneurysm. J Thorac Cardiovasc Surg. 2012, pii: S0022-5223(12)01214-7
Metadaten
Titel
Sequencing of NOTCH1, GATA5, TGFBR1 and TGFBR2genes in familial cases of bicuspid aortic valve
verfasst von
Ilenia Foffa
Lamia Ait Alì
Paola Panesi
Massimiliano Mariani
Pierluigi Festa
Nicoletta Botto
Cecilia Vecoli
Maria Grazia Andreassi
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
BMC Medical Genetics / Ausgabe 1/2013
Elektronische ISSN: 1471-2350
DOI
https://doi.org/10.1186/1471-2350-14-44

Weitere Artikel der Ausgabe 1/2013

BMC Medical Genetics 1/2013 Zur Ausgabe