Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Eur J Med Genet ; 68: 104920, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38336121

ABSTRACT

T-Box Transcription Factor 5 (TBX5) variants are associated with Holt-Oram syndrome. Holt-Oram syndrome display phenotypic variability, regarding upper limb defects, congenital heart defects, and arrhythmias. To investigate the genotype-phenotype relationship between TBX5 variants and cardiac disease, we performed a systematic review of the literature. Through the systematic review we identified 108 variants in TBX5 associated with a cardiac phenotype in 277 patients. Arrhythmias were more frequent in patients with a missense variant (48% vs 30%, p = 0.009) and upper limb abnormalities were more frequent in patients with protein-truncating variants (85% vs 64%, p = 0.0008). We found clustering of missense variants in the T-box domain. Furthermore, we present a family with atrial septal defects. By whole exome sequencing, we identified a novel missense variant p.Phe232Leu in TBX5. The cardiac phenotype included atrial septal defect, arrhythmias, heart failure, and dilated cardiomyopathy. Clinical examination revealed subtle upper limb abnormalities. Thus, the family corresponds to the diagnostic criteria of Holt-Oram syndrome. We provide an overview of cardiac phenotypes associated with TBX5 variants and show an increased risk of arrhythmias associated to missense variants compared to protein-truncating variants. We report a novel missense variant in TBX5 in a family with an atypical Holt-Oram syndrome phenotype.


Subject(s)
Abnormalities, Multiple , Heart Defects, Congenital , Heart Septal Defects, Atrial , Lower Extremity Deformities, Congenital , Upper Extremity Deformities, Congenital , Humans , Heart Defects, Congenital/genetics , Heart Defects, Congenital/diagnosis , Heart Septal Defects, Atrial/genetics , Lower Extremity Deformities, Congenital/genetics , Phenotype , T-Box Domain Proteins/genetics , Upper Extremity Deformities, Congenital/genetics , Upper Extremity Deformities, Congenital/diagnosis
2.
Clin Transl Med ; 14(2): e1565, 2024 02.
Article in English | MEDLINE | ID: mdl-38328889

ABSTRACT

BACKGROUND: Heart failure due to myocardial infarction (MI) involves fibrosis driven by epicardium-derived cells (EPDCs) and cardiac fibroblasts, but strategies to inhibit and provide cardio-protection remains poor. The imprinted gene, non-canonical NOTCH ligand 1 (Dlk1), has previously been shown to mediate fibrosis in the skin, lung and liver, but very little is known on its effect in the heart. METHODS: Herein, human pericardial fluid/plasma and tissue biopsies were assessed for DLK1, whereas the spatiotemporal expression of Dlk1 was determined in mouse hearts. The Dlk1 heart phenotype in normal and MI hearts was assessed in transgenic mice either lacking or overexpressing Dlk1. Finally, in/ex vivo cell studies provided knowledge on the molecular mechanism. RESULTS: Dlk1 was demonstrated in non-myocytes of the developing human myocardium but exhibited a restricted pericardial expression in adulthood. Soluble DLK1 was twofold higher in pericardial fluid (median 45.7 [34.7 (IQR)) µg/L] from cardiovascular patients (n = 127) than in plasma (median 26.1 µg/L [11.1 (IQR)]. The spatial and temporal expression pattern of Dlk1 was recapitulated in mouse and rat hearts. Similar to humans lacking Dlk1, adult Dlk1-/- mice exhibited a relatively mild developmental, although consistent cardiac phenotype with some abnormalities in heart size, shape, thorax orientation and non-myocyte number, but were functionally normal. However, after MI, scar size was substantially reduced in Dlk1-/- hearts as compared with Dlk1+/+ littermates. In line, high levels of Dlk1 in transgenic mice Dlk1fl/fl xWT1GFPCre and Dlk1fl/fl xαMHCCre/+Tam increased scar size following MI. Further mechanistic and cellular insight demonstrated that pericardial Dlk1 mediates cardiac fibrosis through epithelial to mesenchymal transition (EMT) of the EPDC lineage by maintaining Integrin ß8 (Itgb8), a major activator of transforming growth factor ß and EMT. CONCLUSIONS: Our results suggest that pericardial Dlk1 embraces a, so far, unnoticed role in the heart augmenting cardiac fibrosis through EMT. Monitoring DLK1 levels as well as targeting pericardial DLK1 may thus offer new venues for cardio-protection.


Subject(s)
Epithelial-Mesenchymal Transition , Myocardial Infarction , Adult , Animals , Humans , Mice , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cicatrix/metabolism , Cicatrix/pathology , Epithelial-Mesenchymal Transition/genetics , Fibrosis , Ligands , Mice, Transgenic , Myocardial Infarction/genetics , Pericardium/metabolism , Thorax/pathology
3.
Genes (Basel) ; 13(4)2022 04 02.
Article in English | MEDLINE | ID: mdl-35456440

ABSTRACT

Microcephaly primary hereditary (MCPH) is a congenital disease characterized by nonsyndromic reduction in brain size due to impaired neurogenesis, often associated with a variable degree of intellectual disability (ID). The genetic etiology of MCPH is heterogeneous and comprises more than 20 loci, nearly all following a recessive inheritance pattern. The first causative gene identified, MCPH1 or Microcephalin, encodes a centrosomal protein that modulates chromosome condensation and cell cycle progression. It is also involved in DNA damage response and telomere maintenance in the nucleus. Despite numerous studies on MCPH1 function, MCPH1-affected individuals are rare and the available clinical reports are not sufficient to define the natural history of the disease. Here, we present a novel patient with congenital microcephaly, ID, language delay, short stature, and other minor features such as strabismus. magnetic resonance imaging revealed ventriculomegaly, simplified gyral pattern in the frontal lobes, and a neuronal migration defect. Genetic testing detected a homozygous deletion of exons 1-8 of MCPH1. We compare the patients' characteristics with a list of features from MCPH1 cases described in the literature, in an effort to provide additional clues for a comprehensive definition of disease presentation and evolution.


Subject(s)
Intellectual Disability , Microcephaly , Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Homozygote , Humans , Intellectual Disability/genetics , Microcephaly/genetics , Microcephaly/pathology , Sequence Deletion
4.
Am J Cardiol ; 173: 128-131, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35361477

ABSTRACT

Patients with atrial septal defects (ASDs) have increased mortality and morbidity. This can only partly be explained by hemodynamic changes caused by the ASD, suggesting additional underlying causes. Patients with an ASD have an increased burden of pathogenic gene variants in ASD-related genes, indicating genetics as an important factor in etiology. Inheritance of genetic variants with high impact can cause ASD in relatives (familial ASD). This study aimed to investigate whether lifelong outcomes were different in patients with familial ASD compared with patients with sporadic ASD. We used health registries and a nationwide cohort of 2,151 patients with ASD to compare the incidences of atrial fibrillation or flutter (together abbreviated as AF), heart failure, and mortality between patients with familial and sporadic ASD using Cox proportional hazard ratio and Fine and Gray analysis. Patients with familial ASD experienced AF and heart failure earlier in life than patients with sporadic ASD, with hazard ratios of 1.6 and 1.7, respectively. Subdistribution hazard ratios showed an increased risk of AF and heart failure in patients with familial ASD compared with patients with sporadic ASDs (2.3 and 3.1, respectively). Our results suggest that genetic variants with high impact may influence the outcomes of patients with ASD. In conclusion, patients with familial ASD have an increased risk and an earlier onset of AF and heart failure compared with patients with sporadic ASD, hence clinical awareness of arrhythmias and heart failure in patients with familial ASD may lead to timely treatment.


Subject(s)
Atrial Fibrillation , Heart Failure , Heart Septal Defects, Atrial , Atrial Fibrillation/epidemiology , Atrial Fibrillation/genetics , Cohort Studies , Heart Failure/complications , Heart Failure/epidemiology , Heart Failure/genetics , Heart Septal Defects, Atrial/complications , Heart Septal Defects, Atrial/epidemiology , Heart Septal Defects, Atrial/genetics , Humans , Registries
6.
PLoS Genet ; 17(7): e1009679, 2021 07.
Article in English | MEDLINE | ID: mdl-34324492

ABSTRACT

Numerous genetic studies have established a role for rare genomic variants in Congenital Heart Disease (CHD) at the copy number variation (CNV) and de novo variant (DNV) level. To identify novel haploinsufficient CHD disease genes, we performed an integrative analysis of CNVs and DNVs identified in probands with CHD including cases with sporadic thoracic aortic aneurysm. We assembled CNV data from 7,958 cases and 14,082 controls and performed a gene-wise analysis of the burden of rare genomic deletions in cases versus controls. In addition, we performed variation rate testing for DNVs identified in 2,489 parent-offspring trios. Our analysis revealed 21 genes which were significantly affected by rare CNVs and/or DNVs in probands. Fourteen of these genes have previously been associated with CHD while the remaining genes (FEZ1, MYO16, ARID1B, NALCN, WAC, KDM5B and WHSC1) have only been associated in small cases series or show new associations with CHD. In addition, a systems level analysis revealed affected protein-protein interaction networks involved in Notch signaling pathway, heart morphogenesis, DNA repair and cilia/centrosome function. Taken together, this approach highlights the importance of re-analyzing existing datasets to strengthen disease association and identify novel disease genes and pathways.


Subject(s)
DNA Copy Number Variations/genetics , Haploinsufficiency/genetics , Heart Defects, Congenital/genetics , Databases, Genetic , Gene Expression/genetics , Gene Expression Profiling/methods , Genetic Predisposition to Disease/genetics , Genomics/methods , Humans , Ion Channels/genetics , Membrane Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Transcriptome/genetics
7.
Nat Commun ; 11(1): 5816, 2020 11 16.
Article in English | MEDLINE | ID: mdl-33199730

ABSTRACT

Primary microcephaly (MCPH) is characterized by reduced brain size and intellectual disability. The exact pathophysiological mechanism underlying MCPH remains to be elucidated, but dysfunction of neuronal progenitors in the developing neocortex plays a major role. We identified a homozygous missense mutation (p.W155C) in Ribosomal RNA Processing 7 Homolog A, RRP7A, segregating with MCPH in a consanguineous family with 10 affected individuals. RRP7A is highly expressed in neural stem cells in developing human forebrain, and targeted mutation of Rrp7a leads to defects in neurogenesis and proliferation in a mouse stem cell model. RRP7A localizes to centrosomes, cilia and nucleoli, and patient-derived fibroblasts display defects in ribosomal RNA processing, primary cilia resorption, and cell cycle progression. Analysis of zebrafish embryos supported that the patient mutation in RRP7A causes reduced brain size, impaired neurogenesis and cell proliferation, and defective ribosomal RNA processing. These findings provide novel insight into human brain development and MCPH.


Subject(s)
Cilia/metabolism , Microcephaly/genetics , Neurogenesis , Organelle Biogenesis , RNA-Binding Proteins/genetics , Ribosomes/metabolism , Adult , Animals , Base Sequence , Brain/embryology , Brain/pathology , Cell Cycle , Cell Nucleolus/metabolism , Centrosome/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Mice , Mutation/genetics , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Pakistan , Pedigree , Protein Binding , RNA Processing, Post-Transcriptional , RNA, Ribosomal/genetics , RNA-Binding Proteins/metabolism , Zebrafish/embryology
8.
Biopreserv Biobank ; 16(4): 312-321, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30016130

ABSTRACT

Most university biobanks begin like other university research projects, that is, with an idea conceived by an individual researcher in pursuit of his/her own research interests, publications, funding, and career. Some biobanks, however, come to have scientific value that goes beyond the projects that were initially responsible for the collection of the samples and data they contain. Such value may derive from among other things the uniqueness of the samples in terms of their sheer volume, the quality of the samples, the ability to link the samples with information retrieved in disease registries, or the fact that the samples represent very rare diseases. This article focuses on biobanks of this kind, and the special obligations that publicly funded universities have to ensure the sustainability of biobanks with continued scientific value. We argue that universities should adopt policies to deal with the various, diverse issues which may arise during the lifecycle of a biobank. The policies should be flexible, accommodate the freedoms of individual researchers, and reflect the multifaceted nature of biobanks. Yet they should be specific enough to provide guidance and robust enough to safeguard legal norms and ethical values. The article sets out concrete recommendations which universities should consider and act upon.


Subject(s)
Biological Specimen Banks , Universities , Biomedical Research , Humans
9.
J Cell Biol ; 217(1): 151-161, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29237719

ABSTRACT

Primary cilia have pivotal roles as organizers of many different signaling pathways, including platelet-derived growth factor receptor α (PDGFRα) signaling, which, when aberrantly regulated, is associated with developmental disorders, tumorigenesis, and cancer. PDGFRα is up-regulated during ciliogenesis, and ciliary localization of the receptor is required for its appropriate ligand-mediated activation by PDGF-AA. However, the mechanisms regulating sorting of PDGFRα and feedback inhibition of PDGFRα signaling at the cilium are unknown. Here, we provide evidence that intraflagellar transport protein 20 (IFT20) interacts with E3 ubiquitin ligases c-Cbl and Cbl-b and is required for Cbl-mediated ubiquitination and internalization of PDGFRα for feedback inhibition of receptor signaling. In wild-type cells treated with PDGF-AA, c-Cbl becomes enriched in the cilium, and the receptor is subsequently ubiquitinated and internalized. In contrast, in IFT20-depleted cells, PDGFRα localizes aberrantly to the plasma membrane and is overactivated after ligand stimulation because of destabilization and degradation of c-Cbl and Cbl-b.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , 3T3 Cells , Animals , Cell Line , Cilia/metabolism , HEK293 Cells , Humans , Mice , Platelet-Derived Growth Factor/pharmacology , RNA Interference , Signal Transduction/genetics , Ubiquitination/physiology
10.
Eur J Med Genet ; 60(12): 627-630, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28778786

ABSTRACT

CDK5RAP2 gene encodes a centrosomal protein, highly expressed in fetal brain and essentially indispensable for its normal development, as biallelic mutations in it lead to primary microcephaly (MCPH). Despite being known as MCPH linked gene for more than a decade, the phenotypic spectrum of CDK5RAP2 mutations is still under explored as only eleven families have been reported worldwide. Here, we analyzed a consanguineous Pakistani MCPH family, characterized by moderate to severe intellectual disability, speech impairment, moderately short stature and sparse eyebrows. Whole exome sequencing of the proband identified a 2bp duplication in exon 34 of CDK5RAP2 that causes frame-shift, leading to a premature stop codon. The resultant transcript is resistant to nonsense mediated decay, suggesting that the mutation leads to a truncated protein lacking C-terminal domains; CDK5R1, and Cnn motif 2 (CM2), required for its localization to centrosome and Golgi Apparatus. Clinical variability observed in the family highlights the importance of further detailed clinical description of patients with CDK5RAP2 mutations.


Subject(s)
Eyebrows/abnormalities , Frameshift Mutation , Intellectual Disability/genetics , Intracellular Signaling Peptides and Proteins/genetics , Microcephaly/genetics , Nerve Tissue Proteins/genetics , Speech Disorders/genetics , Adult , Cell Cycle Proteins , Child , Codon, Terminator/genetics , Consanguinity , Female , Humans , Intellectual Disability/diagnosis , Intracellular Signaling Peptides and Proteins/metabolism , Male , Microcephaly/diagnosis , Nerve Tissue Proteins/metabolism , Nonsense Mediated mRNA Decay , Pedigree , Speech Disorders/diagnosis , Syndrome
11.
Cilia ; 6: 3, 2017.
Article in English | MEDLINE | ID: mdl-28344781

ABSTRACT

BACKGROUND: Three-dimensional explant spheroid formation is an ex vivo technique previously used in studies of airway epithelial ion and water transport. Explanted cells and sheets of nasal epithelium form fully differentiated spheroids enclosing a partly fluid-filled lumen with the ciliated apical surface facing the outside and accessible for analysis of ciliary function. METHODS: We performed a two-group comparison study of ciliary beat pattern and ciliary beat frequency in spheroids derived from nasal airway epithelium in patients with primary ciliary dyskinesia (PCD) and in healthy controls. Nasal ciliary cells and sheets were removed on day 1 by nasal brush biopsy and analyzed with regard to ciliary beat pattern-and frequency using high-speed video imaging for standard reference values. Three-dimensional explant spheroid formation was initiated in the same individual on the same day by incubation of cells and sheets from a separate brush biopsy. Harvested spheroids were analyzed earliest possible and values of spheroid ciliary beat pattern and frequency were compared to the corresponding reference values from day 1. RESULTS: Spheroids formed fast in serum-free culture medium. Formation was successful in 15 out of 18 (82%) sampled individuals. Thus, formation was successful in seven healthy controls and eight PCD patients, while unsuccessful in 3 with PCD due to infection. Median (range) number of days in culture before harvesting of spheroids was 4 (1-5) in healthy versus 2 (1-5) in PCD. Spheroid ciliary beat pattern and frequency were unchanged compared to their corresponding day 1 standard reference values. Spheroid ciliary beat frequency discriminated highly significant between healthy controls (9.3 Hz) and PCD patients (2.4 Hz) (P < 0.0001). Survival of spheroids was 16 days in a single healthy person. CONCLUSION: Patient-specific three-dimensional explant spheroid formation from a minimal invasive nasal brush biopsy is a feasible, fast and valid ex vivo method to assess ciliary function with potential of aiding the diagnosis of PCD. In addition, it may be a useful model in the investigation of pathophysiological aspects and drug effects in human nasal airway epithelium.

12.
World J Pediatr Congenit Heart Surg ; 7(2): 169-77, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26957399

ABSTRACT

BACKGROUND: Congenital heart disease (CHD) occurs in approximately 1% of all live births, and 3% to 8% of these have until now been considered familial cases, defined as the occurrence of two or more affected individuals in a family. The validity of CHD diagnoses in Danish administrative registry data has only been studied previously in highly selected patient populations. These studies identified high positive predictive values (PPVs) and recurrence risk ratios (RRRs-ratio between probabilities of CHD given family history of CHD and no family history). However, the RRR can be distorted if registry data are used indiscriminately. Here, we investigated the consequences of misclassifications for the RRR using validated diagnoses on Danish patients with familial CHD. METHODS: Danish citizens are assigned a civil registration number (CPR number) at birth or immigration, which acts as a unique identifier in the Danish registries, thus enabling connection of information from several registries. Utilizing the CPR number, we identified Danish patients with familial CHD and reviewed each patient's file. We compared diagnoses from the registries with those manually assigned, which enabled calculation of the PPVs of diagnoses in the Danish registries, and from this, we deduced the false discovery rate (FDR). To measure the consequences on the RRR, the FDR was applied to a simulated data set with true RRR values of 2 and 10. RESULTS: We validated diagnoses of 2,442 patients from 1,593 families. Of these, 874 patients were misclassified corresponding to an FDR of 36%. Applying this FDR on the simulated data sets, we found that the RRR decreased from 2 and 10 to 1.4 and 5.1, respectively. Lastly, we estimated that 11% of all cases with CHD were familial. CONCLUSION: We found that approximately one of nine of all cases with CHD are familial, and we also found that 36% of individuals with CHD in administrative medical registries are misclassified, which distort the RRR in simulated scenarios.


Subject(s)
Heart Defects, Congenital/genetics , Registries/standards , Data Accuracy , Denmark , Genetic Predisposition to Disease , Heart Defects, Congenital/diagnosis , Humans , Odds Ratio , Risk Assessment
14.
Congenit Heart Dis ; 11(3): 283-90, 2016 May.
Article in English | MEDLINE | ID: mdl-26679770

ABSTRACT

OBJECTIVE: Atrial septal defect (ASD) is the second most common congenital heart defect (CHD) and is observed in families as an autosomal dominant trait as well as in nonfamilial CHD. Mutations in the NKX2-5 gene, located on chromosome 5, are associated with ASD, often combined with conduction disturbances, cardiomyopathies, complex CHD, and sudden cardiac death as well. Here, we show that NKX2-5 mutations primarily occur in ASD patients with conduction disturbances and heritable ASD. Furthermore, these families are at increased risk of sudden cardiac death. RESULTS: We screened 39 probands with familial CHD for mutations in NKX2-5 and discovered a novel mutation in one family (2.5%) with ASD and atrioventricular block. A review of the literature revealed 59 different NKX2-5 mutations in 202 patients. Mutations were significantly more common in familial cases compared to nonfamilial cases (P = 7.1 × 10(-9) ). The majority of patients (74%) had ASD with conduction disturbance. Nineteen patients (15%) of 120 with familial ASD and conduction disturbance died from sudden cardiac death of which nine (8%) were confirmed mutation carriers, and 10 were possible carriers. CONCLUSIONS: NKX2-5 mutations mainly occur in familial CHD, the signature phenotype is ASD with conduction disturbances and mutation carriers are at increased risk of sudden cardiac death. We suggest that familial ASD patients should be screened for NKX2-5 mutations and, if they are mutation carriers, implantation of an implantable cardioverter-defibrillator should be considered in these patients.


Subject(s)
Death, Sudden, Cardiac/etiology , Heart Septal Defects, Atrial/genetics , Homeobox Protein Nkx-2.5/genetics , Mutation , Adult , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Child , Child, Preschool , Female , Genetic Association Studies , Genetic Predisposition to Disease , Heart Septal Defects, Atrial/diagnosis , Heart Septal Defects, Atrial/mortality , Heart Septal Defects, Atrial/physiopathology , Heredity , Heterozygote , Humans , Infant , Male , Pedigree , Phenotype , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Young Adult
15.
Cell Mol Life Sci ; 71(8): 1327-52, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23934094

ABSTRACT

Congenital heart disease (CHD) affects nearly 1 % of the population. It is a complex disease, which may be caused by multiple genetic and environmental factors. Studies in human genetics have led to the identification of more than 50 human genes, involved in isolated CHD or genetic syndromes, where CHD is part of the phenotype. Furthermore, mapping of genomic copy number variants and exome sequencing of CHD patients have led to the identification of a large number of candidate disease genes. Experiments in animal models, particularly in mice, have been used to verify human disease genes and to gain further insight into the molecular pathology behind CHD. The picture emerging from these studies suggest that genetic lesions associated with CHD affect a broad range of cellular signaling components, from ligands and receptors, across down-stream effector molecules to transcription factors and co-factors, including chromatin modifiers.


Subject(s)
Chromosome Aberrations , Genes/genetics , Genetic Predisposition to Disease/genetics , Heart Diseases/congenital , Heart Diseases/genetics , Molecular Biology/methods , Signal Transduction/genetics , Transcription Factors/genetics , Animals , DNA Copy Number Variations/genetics , Heterotaxy Syndrome/genetics , Humans , Mice , Models, Biological , Sarcomeres/genetics
16.
Organogenesis ; 10(1): 108-25, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24345806

ABSTRACT

Primary cilia are unique sensory organelles that coordinate a wide variety of different signaling pathways to control cellular processes during development and in tissue homeostasis. Defects in function or assembly of these antenna-like structures are therefore associated with a broad range of developmental disorders and diseases called ciliopathies. Recent studies have indicated a major role of different populations of cilia, including nodal and cardiac primary cilia, in coordinating heart development, and defects in these cilia are associated with congenital heart disease. Here, we present an overview of the role of nodal and cardiac primary cilia in heart development.


Subject(s)
Cilia , Heart/embryology , Organogenesis/physiology , Signal Transduction , Humans , Microscopy, Electron, Scanning , Transforming Growth Factor beta1/metabolism
17.
Cell Rep ; 3(6): 1806-14, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23746451

ABSTRACT

Transforming growth factor ß (TGF-ß) signaling is regulated by clathrin-dependent endocytosis (CDE) for the control of cellular processes during development and in tissue homeostasis. The primary cilium coordinates several signaling pathways, and the pocket surrounding the base and proximal part of the cilium is a site for CDE. We report here that TGF-ß receptors localize to the ciliary tip and endocytic vesicles at the ciliary base in fibroblasts and that TGF-ß stimulation increases receptor localization and activation of SMAD2/3 and ERK1/2 at the ciliary base. Inhibition of CDE reduced TGF-ß-mediated signaling at the cilium, and TGF-ß signaling and CDE activity are reduced at stunted primary cilia in Tg737orpk fibroblasts. Similarly, TGF-ß signaling during cardiomyogenesis correlated with accumulation of TGF-ß receptors and activation of SMAD2/3 at the ciliary base. Our results indicate that the primary cilium regulates TGF-ß signaling and that the ciliary pocket is a compartment for CDE-dependent regulation of signal transduction.


Subject(s)
Cilia/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/physiology , Endocytosis/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Signal Transduction , Up-Regulation
18.
Am J Med Genet A ; 158A(4): 720-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22383218

ABSTRACT

Recurrent copy number variants (CNVs) are found in a significant proportion of patients with congenital heart disease (CHD) and some of these CNVs are associated with other developmental defects. In some syndromic patients, CHD may be the first presenting symptom, thus screening of patients with CHD for CNVs in specific genomic regions may lead to early diagnosis and awareness of extracardiac symptoms. We designed a multiplex ligation-dependent probe amplification (MLPA) assay specifically for screening of CHD patients. The MLPA assay allows for simultaneous analysis of CNVs in 25 genomic regions previously associated with CHD. We screened blood samples from 402 CHD patients and identified 14 rare CNVs in 13 (3.2%) patients. Five CNVs were de novo and six where inherited from a healthy parent. The MLPA screen led to early syndrome diagnosis in two of these patients. We conclude that the MLPA assay detects clinically relevant CNVs and suggest that it could be used within pediatric cardiology as a first tier screen to detect clinically relevant CNVs and identify syndromic patients at an early stage.


Subject(s)
DNA Copy Number Variations/genetics , Gene Dosage/genetics , Heart Defects, Congenital/genetics , Adolescent , Aged , Child , Child, Preschool , Chromosome Aberrations , Female , Heart Diseases/genetics , Humans , Infant , Infant, Newborn , Male , Multiplex Polymerase Chain Reaction/methods , Nucleic Acid Amplification Techniques
19.
Ugeskr Laeger ; 173(3): 194-6, 2011 Jan 17.
Article in Danish | MEDLINE | ID: mdl-21241627

ABSTRACT

Congenital heart disease (CHD) affects 1% of the population. The aetiology of CHD is complex and largely unknown, comprising both environmental and genetic components. Recent progress in molecular cytogenetics has led to the identification of rare genomic copy number variants (CNVs) in a significant proportion of CHD patients. These novel results imply new diagnostic possibilities and may aid in the identification of novel disease genes.


Subject(s)
Heart Defects, Congenital/genetics , Chromosome Aberrations , Chromosome Deletion , DNA Copy Number Variations , Gene Dosage , Genetic Predisposition to Disease , Heart Defects, Congenital/diagnosis , Humans
20.
Am J Hum Genet ; 86(6): 839-49, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20493459

ABSTRACT

Congenital heart defects (CHDs) are the most common major developmental anomalies and the most frequent cause for perinatal mortality, but their etiology remains often obscure. We identified a locus for CHDs on 6q24-q25. Genotype-phenotype correlations in 12 patients carrying a chromosomal deletion on 6q delineated a critical 850 kb region on 6q25.1 harboring five genes. Bioinformatics prioritization of candidate genes in this locus for a role in CHDs identified the TGF-beta-activated kinase 1/MAP3K7 binding protein 2 gene (TAB2) as the top-ranking candidate gene. A role for this candidate gene in cardiac development was further supported by its conserved expression in the developing human and zebrafish heart. Moreover, a critical, dosage-sensitive role during development was demonstrated by the cardiac defects observed upon titrated knockdown of tab2 expression in zebrafish embryos. To definitively confirm the role of this candidate gene in CHDs, we performed mutation analysis of TAB2 in 402 patients with a CHD, which revealed two evolutionarily conserved missense mutations. Finally, a balanced translocation was identified, cosegregating with familial CHD. Mapping of the breakpoints demonstrated that this translocation disrupts TAB2. Taken together, these data clearly demonstrate a role for TAB2 in human cardiac development.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chromosome Deletion , Chromosomes, Human, Pair 6 , Heart Defects, Congenital/genetics , Amino Acid Sequence , Animals , Embryo, Mammalian , Female , Gene Knockdown Techniques , Genetic Association Studies , Heart/embryology , Humans , Male , Molecular Sequence Data , Mutation , Pedigree , Translocation, Genetic , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...