Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nat Commun ; 13(1): 4680, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35945219

ABSTRACT

DPF3, a component of the SWI/SNF chromatin remodeling complex, has been associated with clear cell renal cell carcinoma (ccRCC) in a genome-wide association study. However, the functional role of DPF3 in ccRCC development and progression remains unknown. In this study, we demonstrate that DPF3a, the short isoform of DPF3, promotes kidney cancer cell migration both in vitro and in vivo, consistent with the clinical observation that DPF3a is significantly upregulated in ccRCC patients with metastases. Mechanistically, DPF3a specifically interacts with SNIP1, via which it forms a complex with SMAD4 and p300 histone acetyltransferase (HAT), the major transcriptional regulators of TGF-ß signaling pathway. Moreover, the binding of DPF3a releases the repressive effect of SNIP1 on p300 HAT activity, leading to the increase in local histone acetylation and the activation of cell movement related genes. Overall, our findings reveal a metastasis-promoting function of DPF3, and further establish the link between SWI/SNF components and ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Signal Transduction , Carcinoma, Renal Cell/genetics , Chromatin , Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Genome-Wide Association Study , Humans , Kidney Neoplasms/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
2.
J Cardiovasc Dev Dis ; 7(4)2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33276527

ABSTRACT

Congenital heart defects (CHDs) are the most common birth defect in human with an incidence of almost 1% of all live births. Most cases have a multifactorial origin with both genetics and the environment playing a role in its development and progression. Adding an epigenetic component to this aspect is exemplified by monozygotic twins which share the same genetic background but have a different disease status. As a result, the interplay between the genetic, epigenetic and the environmental conditions might contribute to the etiology and phenotype. To date, the underlying causes of the majority of CHDs remain poorly understood. In this study, we performed genome-wide high-throughput sequencing to examine the genetic, structural genomic and epigenetic differences of two identical twin pairs discordant for Tetralogy of Fallot (TOF), representing the most common cyanotic form of CHDs. Our results show the almost identical genetic and structural genomic identity of the twins. In contrast, several epigenetic alterations could be observed given by DNA methylation changes in regulatory regions of known cardiac-relevant genes. Overall, this study provides first insights into the impact of genetic and especially epigenetic factors underlying monozygotic twins discordant for CHD like TOF.

3.
Dis Model Mech ; 13(12)2020 12 18.
Article in English | MEDLINE | ID: mdl-33033063

ABSTRACT

The causal genetic underpinnings of congenital heart diseases, which are often complex and multigenic, are still far from understood. Moreover, there are also predominantly monogenic heart defects, such as cardiomyopathies, with known disease genes for the majority of cases. In this study, we identified mutations in myomesin 2 (MYOM2) in patients with Tetralogy of Fallot (TOF), the most common cyanotic heart malformation, as well as in patients with hypertrophic cardiomyopathy (HCM), who do not exhibit any mutations in the known disease genes. MYOM2 is a major component of the myofibrillar M-band of the sarcomere, and a hub gene within interactions of sarcomere genes. We show that patient-derived cardiomyocytes exhibit myofibrillar disarray and reduced passive force with increasing sarcomere lengths. Moreover, our comprehensive functional analyses in the Drosophila animal model reveal that the so far uncharacterized fly gene CG14964 [herein referred to as Drosophila myomesin and myosin binding protein (dMnM)] may be an ortholog of MYOM2, as well as other myosin binding proteins. Its partial loss of function or moderate cardiac knockdown results in cardiac dilation, whereas more severely reduced function causes a constricted phenotype and an increase in sarcomere myosin protein. Moreover, compound heterozygous combinations of CG14964 and the sarcomere gene Mhc (MYH6/7) exhibited synergistic genetic interactions. In summary, our results suggest that MYOM2 not only plays a critical role in maintaining robust heart function but may also be a candidate gene for heart diseases such as HCM and TOF, as it is clearly involved in the development of the heart.This article has an associated First Person interview with Emilie Auxerre-Plantié and Tanja Nielsen, joint first authors of the paper.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Connectin/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Genetic Association Studies , Membrane Proteins/genetics , Tetralogy of Fallot/genetics , Animals , Drosophila Proteins/metabolism , Female , Humans , Locomotion , Male , Membrane Proteins/metabolism , Muscles/metabolism , Mutation/genetics , Myocardium , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/metabolism , Myofibrils/pathology , Organ Specificity , Protein Binding , Protein Interaction Mapping
4.
Sci Rep ; 10(1): 10921, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616843

ABSTRACT

Patient-specific induced pluripotent stem cells (ps-iPSCs) and their differentiated cell types are a powerful model system to gain insight into mechanisms driving early developmental and disease-associated regulatory networks. In this study, we use ps-iPSCs to gain insights into Tetralogy of Fallot (TOF), which represents the most common cyanotic heart defect in humans. iPSCs were generated and further differentiated to cardiomyocytes (CMs) using standard methods from two well-characterized TOF patients and their healthy relatives serving as controls. Patient-specific expression patterns and genetic variability were investigated using whole genome and transcriptome sequencing data. We first studied the clonal mutational burden of the derived iPSCs. In two out of three iPSC lines of patient TOF-01, we found a somatic mutation in the DNA-binding domain of tumor suppressor P53, which was not observed in the genomic DNA from blood. Further characterization of this mutation showed its functional impact. For patient TOF-02, potential disease-relevant differential gene expression between and across cardiac differentiation was shown. Here, clear differences at the later stages of differentiation could be observed between CMs of the patient and its controls. Overall, this study provides first insights into the complex molecular mechanisms underlying iPSC-derived cardiomyocyte differentiation and its transcriptional alterations in TOF.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Mutation , Myocytes, Cardiac/metabolism , Tetralogy of Fallot/pathology , Case-Control Studies , Cell Differentiation , Clone Cells , Female , Gene Expression Regulation, Developmental , Genetic Association Studies , Germ-Line Mutation , Humans , Male , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Skin/cytology , Tetralogy of Fallot/genetics , Transcription, Genetic
5.
Sci Rep ; 9(1): 19063, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836860

ABSTRACT

MicroRNAs (miRNAs) play an important role in guiding development and maintaining function of the human heart. Dysregulation of miRNAs has been linked to various congenital heart diseases including Tetralogy of Fallot (TOF), which represents the most common cyanotic heart malformation in humans. Several studies have identified dysregulated miRNAs in right ventricular (RV) tissues of TOF patients. In this study, we profiled genome-wide the whole transcriptome and analyzed the relationship of miRNAs and mRNAs of RV tissues of a homogeneous group of 22 non-syndromic TOF patients. Observed profiles were compared to profiles obtained from right and left ventricular tissue of normal hearts. To reduce the commonly observed large list of predicted target genes of dysregulated miRNAs, we applied a stringent target prediction pipeline integrating probabilities for miRNA-mRNA interaction. The final list of disease-related miRNA-mRNA pairs comprises novel as well as known miRNAs including miR-1 and miR-133, which are essential to cardiac development and function by regulating KCNJ2, FBN2, SLC38A3 and TNNI1. Overall, our study provides additional insights into post-transcriptional gene regulation of malformed hearts of TOF patients.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Tetralogy of Fallot/genetics , Chromosomes, Human, Pair 6/genetics , Female , Gene Expression Profiling , Genome, Human , Humans , Male , MicroRNAs/metabolism , Molecular Sequence Annotation , Myocardium/metabolism , Myocardium/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Science ; 359(6380): 1098-1099, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29590027

Subject(s)
Heart , Humans
7.
Heart Rhythm ; 15(2): 267-276, 2018 02.
Article in English | MEDLINE | ID: mdl-28917552

ABSTRACT

BACKGROUND: Mutations in the PRKAG2 gene encoding the γ-subunit of adenosine monophosphate kinase (AMPK) cause hypertrophic cardiomyopathy (HCM) and familial Wolff-Parkinson-White (WPW) syndrome. Patients carrying the R302Q mutation in PRKAG2 present with sinus bradycardia, escape rhythms, ventricular preexcitation, supraventricular tachycardia, and atrioventricular block. This mutation affects AMPK activity and increases glycogen storage in cardiomyocytes. The link between glycogen storage, WPW syndrome, HCM, and arrhythmias remains unknown. OBJECTIVE: The purpose of this study was to investigate the pathological changes caused by the PRKAG2 mutation. We tested the hypothesis that patient's induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) display clinical aspects of the disease. METHODS: Using clustered regularly interspaced short palindromic repeats (CRISPR) technology, we corrected the mutation and then generated isogenic iPSC-CMs. Action potentials were recorded from spontaneously firing and paced cardiomyocytes using the patch clamp technique. Using a microelectrode array setup, we recorded electrograms from iPSC-CMs clusters. Transmission electron microscopy was used to detect ultrastructural abnormalities in the mutated iPSC-CMs. RESULTS: PRKAG2-mutated iPSC-CMs exhibited abnormal firing patterns, delayed afterdepolarizations, triggered arrhythmias, and augmented beat rate variability. Importantly, CRISPR correction eliminated the electrophysiological abnormalities, the augmented glycogen, storage, and cardiomyocyte hypertrophy. CONCLUSION: PRKAG2-mutated iPSC-CMs displayed functional and structural abnormalities, which were abolished by correcting the mutation in the patient's iPSCs using CRISPR technology.


Subject(s)
AMP-Activated Protein Kinases/genetics , DNA/genetics , Induced Pluripotent Stem Cells/ultrastructure , Mutation , Myocytes, Cardiac/ultrastructure , Wolff-Parkinson-White Syndrome/genetics , AMP-Activated Protein Kinases/metabolism , Cardiac Electrophysiology , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Mutational Analysis , Electrophysiological Phenomena , Humans , Male , Microscopy, Electron, Transmission , Middle Aged , Myocytes, Cardiac/metabolism , Wolff-Parkinson-White Syndrome/metabolism , Wolff-Parkinson-White Syndrome/pathology
8.
PLoS One ; 12(6): e0179464, 2017.
Article in English | MEDLINE | ID: mdl-28609469

ABSTRACT

Post-translational modifications of histones play a key role in the regulation of gene expression during development and differentiation. Numerous studies have shown the dynamics of combinatorial regulation by transcription factors and histone modifications, in the sense that different combinations lead to distinct expression outcomes. Here, we investigated gene regulation by stable enrichment patterns of histone marks H3K4me2 and H3K4me3 in combination with the chromatin binding of the muscle tissue-specific transcription factor MyoD during myogenic differentiation of C2C12 cells. Using k-means clustering, we found that specific combinations of H3K4me2/3 profiles over and towards the gene body impact on gene expression and marks a subset of genes important for muscle development and differentiation. By further analysis, we found that the muscle key regulator MyoD was significantly enriched on this subset of genes and played a repressive role during myogenic differentiation. Among these genes, we identified the pluripotency gene Patz1, which is repressed during myogenic differentiation through direct binding of MyoD to promoter elements. These results point to the importance of integrating histone modifications and MyoD chromatin binding for coordinated gene activation and repression during myogenic differentiation.


Subject(s)
Cell Differentiation/genetics , Histones/genetics , MyoD Protein/genetics , Myoblasts/metabolism , Animals , Cell Line , Cells, Cultured , Cluster Analysis , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling/methods , Gene Ontology , HEK293 Cells , Histones/classification , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mice , Muscle Development/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Cardiovasc Res ; 112(1): 464-77, 2016 10.
Article in English | MEDLINE | ID: mdl-27496870

ABSTRACT

AIMS: For the majority of congenital heart diseases (CHDs), the full complexity of the causative molecular network, which is driven by genetic, epigenetic, and environmental factors, is yet to be elucidated. Epigenetic alterations are suggested to play a pivotal role in modulating the phenotypic expression of CHDs and their clinical course during life. Candidate approaches implied that DNA methylation might have a developmental role in CHD and contributes to the long-term progress of non-structural cardiac diseases. The aim of the present study is to define the postnatal epigenome of two common cardiac malformations, representing epigenetic memory, and adaption to hemodynamic alterations, which are jointly relevant for the disease course. METHODS AND RESULTS: We present the first analysis of genome-wide DNA methylation data obtained from myocardial biopsies of Tetralogy of Fallot (TOF) and ventricular septal defect patients. We defined stringent sets of differentially methylated regions between patients and controls, which are significantly enriched for genomic features like promoters, exons, and cardiac enhancers. For TOF, we linked DNA methylation with genome-wide expression data and found a significant overlap for hypermethylated promoters and down-regulated genes, and vice versa. We validated and replicated the methylation of selected CpGs and performed functional assays. We identified a hypermethylated novel developmental CpG island in the promoter of SCO2 and demonstrate its functional impact. Moreover, we discovered methylation changes co-localized with novel, differential splicing events among sarcomeric genes as well as transcription factor binding sites. Finally, we demonstrated the interaction of differentially methylated and expressed genes in TOF with mutated CHD genes in a molecular network. CONCLUSION: By interrogating DNA methylation and gene expression data, we identify two novel mechanism contributing to the phenotypic expression of CHDs: aberrant methylation of promoter CpG islands and methylation alterations leading to differential splicing.


Subject(s)
Carrier Proteins/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling/methods , Heart Septal Defects, Ventricular/genetics , Mitochondrial Proteins/genetics , Tetralogy of Fallot/genetics , Adaptation, Physiological , Adult , Case-Control Studies , Child, Preschool , CpG Islands , Gene Regulatory Networks , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , Heart Septal Defects, Ventricular/physiopathology , Hemodynamics , Humans , Infant , Middle Aged , Molecular Chaperones , Phenotype , Principal Component Analysis , Promoter Regions, Genetic , RNA Splicing , Reproducibility of Results , Tetralogy of Fallot/physiopathology , Young Adult
10.
Arch Med Sci ; 12(2): 263-78, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27186169

ABSTRACT

INTRODUCTION: Transgenic mice overexpressing mutated NEBL, encoding the cardiac-specific Z-disk protein nebulette, develop severe cardiac phenotypes. Since cardiomyopathies are commonly familial and because mutations in a single gene may result in variable phenotypes, we tested the hypothesis that NEBL mutations are associated with cardiomyopathy. MATERIAL AND METHODS: We analyzed 389 patients, including cohorts of patients with dilated cardiomyopathy (DCM), hypertrophic cardiomyopathy (HCM), and left ventricular non-compaction cardiomyopathy (LVNC). The 28 coding exons of the NEBL gene were sequenced. Further bioinformatic analysis was used to distinguish variants. RESULTS: In total, we identified six very rare heterozygous missense mutations in NEBL in 7 different patients (frequency 1.8%) in highly conserved codons. The mutations were not detectable in 320 Caucasian sex-matched unrelated individuals without cardiomyopathy and 192 Caucasian sex-matched blood donors without heart disease. Known cardiomyopathy genes were excluded in these patients. The mutations p.H171R and p.I652L were found in 2 HCM patients. Further, p.Q581R and p.S747L were detected in 2 DCM patients, while the mutation p.A175T was identified independently in two unrelated patients with DCM. One LVNC patient carried the mutation p.P916L. All HCM and DCM related mutations were located in the nebulin-like repeats, domains responsible for actin binding. Interestingly, the mutation associated with LVNC was located in the C-terminal serine-rich linker region. CONCLUSIONS: Our data suggest that NEBL mutations may cause various cardiomyopathies. We herein describe the first NEBL mutations in HCM and LVNC. Our findings underline the notion that the cardiomyopathies are true allelic diseases.

11.
Nucleic Acids Res ; 44(6): 2538-53, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26582913

ABSTRACT

DPF3 (BAF45c) is a member of the BAF chromatin remodeling complex. Two isoforms have been described, namely DPF3a and DPF3b. The latter binds to acetylated and methylated lysine residues of histones. Here, we elaborate on the role of DPF3a and describe a novel pathway of cardiac gene transcription leading to pathological cardiac hypertrophy. Upon hypertrophic stimuli, casein kinase 2 phosphorylates DPF3a at serine 348. This initiates the interaction of DPF3a with the transcriptional repressors HEY, followed by the release of HEY from the DNA. Moreover, BRG1 is bound by DPF3a, and is thus recruited to HEY genomic targets upon interaction of the two components. Consequently, the transcription of downstream targets such as NPPA and GATA4 is initiated and pathological cardiac hypertrophy is established. In human, DPF3a is significantly up-regulated in hypertrophic hearts of patients with hypertrophic cardiomyopathy or aortic stenosis. Taken together, we show that activation of DPF3a upon hypertrophic stimuli switches cardiac fetal gene expression from being silenced by HEY to being activated by BRG1. Thus, we present a novel pathway for pathological cardiac hypertrophy, whose inhibition is a long-term therapeutic goal for the treatment of the course of heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cardiomegaly/genetics , Chromatin Assembly and Disassembly , Chromatin/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Differentiation , Chromatin/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Myoblasts/cytology , Myoblasts/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Proteins/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
12.
PLoS One ; 9(1): e85375, 2014.
Article in English | MEDLINE | ID: mdl-24400131

ABSTRACT

Copy number variations (CNVs) are one of the main sources of variability in the human genome. Many CNVs are associated with various diseases including cardiovascular disease. In addition to hybridization-based methods, next-generation sequencing (NGS) technologies are increasingly used for CNV discovery. However, respective computational methods applicable to NGS data are still limited. We developed a novel CNV calling method based on outlier detection applicable to small cohorts, which is of particular interest for the discovery of individual CNVs within families, de novo CNVs in trios and/or small cohorts of specific phenotypes like rare diseases. Approximately 7,000 rare diseases are currently known, which collectively affect ∼6% of the population. For our method, we applied the Dixon's Q test to detect outliers and used a Hidden Markov Model for their assessment. The method can be used for data obtained by exome and targeted resequencing. We evaluated our outlier-based method in comparison to the CNV calling tool CoNIFER using eight HapMap exome samples and subsequently applied both methods to targeted resequencing data of patients with Tetralogy of Fallot (TOF), the most common cyanotic congenital heart disease. In both the HapMap samples and the TOF cases, our method is superior to CoNIFER, such that it identifies more true positive CNVs. Called CNVs in TOF cases were validated by qPCR and HapMap CNVs were confirmed with available array-CGH data. In the TOF patients, we found four copy number gains affecting three genes, of which two are important regulators of heart development (NOTCH1, ISL1) and one is located in a region associated with cardiac malformations (PRODH at 22q11). In summary, we present a novel CNV calling method based on outlier detection, which will be of particular interest for the analysis of de novo or individual CNVs in trios or cohorts up to 30 individuals, respectively.


Subject(s)
Computational Biology/methods , DNA Copy Number Variations , Sequence Analysis, DNA/methods , Tetralogy of Fallot/genetics , Algorithms , Exome , High-Throughput Nucleotide Sequencing , Humans , Markov Chains
13.
Hum Mol Genet ; 23(12): 3115-28, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24459294

ABSTRACT

Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease. Its genetic basis is demonstrated by an increased recurrence risk in siblings and familial cases. However, the majority of TOF are sporadic, isolated cases of undefined origin and it had been postulated that rare and private autosomal variations in concert define its genetic basis. To elucidate this hypothesis, we performed a multilevel study using targeted re-sequencing and whole-transcriptome profiling. We developed a novel concept based on a gene's mutation frequency to unravel the polygenic origin of TOF. We show that isolated TOF is caused by a combination of deleterious private and rare mutations in genes essential for apoptosis and cell growth, the assembly of the sarcomere as well as for the neural crest and secondary heart field, the cellular basis of the right ventricle and its outflow tract. Affected genes coincide in an interaction network with significant disturbances in expression shared by cases with a mutually affected TOF gene. The majority of genes show continuous expression during adulthood, which opens a new route to understand the diversity in the long-term clinical outcome of TOF cases. Our findings demonstrate that TOF has a polygenic origin and that understanding the genetic basis can lead to novel diagnostic and therapeutic routes. Moreover, the novel concept of the gene mutation frequency is a versatile measure and can be applied to other open genetic disorders.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study/methods , Myocardium/pathology , Tetralogy of Fallot/genetics , Tetralogy of Fallot/pathology , Apoptosis , Base Sequence , Cell Proliferation , Cohort Studies , Gene Expression Profiling , Gene Expression Regulation , Gene Frequency , Humans , Molecular Sequence Data , Multifactorial Inheritance , Mutation , Myocardium/metabolism , Sequence Analysis, DNA , Tetralogy of Fallot/blood
14.
Brief Funct Genomics ; 13(1): 51-65, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24095982

ABSTRACT

Congenital heart diseases (CHD) represent the most common birth defect in human. The majority of cases are caused by a combination of complex genetic alterations and environmental influences. In the past, many disease-causing mutations have been identified; however, there is still a large proportion of cardiac malformations with unknown precise origin. High-throughput sequencing technologies established during the last years offer novel opportunities to further study the genetic background underlying the disease. In this review, we provide a roadmap for designing and analyzing high-throughput sequencing studies focused on CHD, but also with general applicability to other complex diseases. The three main next-generation sequencing (NGS) platforms including their particular advantages and disadvantages are presented. To identify potentially disease-related genomic variations and genes, different filtering steps and gene prioritization strategies are discussed. In addition, available control datasets based on NGS are summarized. Finally, we provide an overview of current studies already using NGS technologies and showing that these techniques will help to further unravel the complex genetics underlying CHD.


Subject(s)
Genetic Variation , Genomics/methods , Heart Defects, Congenital/genetics , High-Throughput Nucleotide Sequencing/methods , Databases, Genetic , Genetic Association Studies , Humans
15.
Eur J Hum Genet ; 21(3): 294-300, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22892539

ABSTRACT

Recently, missense mutations in titin-associated proteins have been linked to the pathogenesis of dilated cardiomyopathy (DCM). The objective of this study was to search for novel disease-associated mutations in the two human titin-binding proteins myopalladin and its amino-terminal-interacting partner cardiac ankyrin-repeat protein (CARP). In a cohort of 255 cases with familial and sporadic DCM, we analyzed the coding regions and all corresponding intron flanks located in the MYPN and CARP-encoding ANKRD1 gene. Two heterozygous missense mutations were detected in the MYPN gene (p.R955W and p.P961L), but neither of these mutations was found in 300 healthy controls. Both mutations were located in the α-actinin-binding region of myopalladin. Endomyocardial biopsies from the p.R955W carrier showed normal subcellular localization of myopalladin and α-actinin in cardiac myocytes, while their regular sarcomeric staining pattern was significantly disrupted in the p.P961L carrier, indicating that disturbed myofibrillogenesis and altered sarcomere assembly are the cause of the disease. In the ANKRD1 gene, we identified synonymous base exchanges (c.108T>C and c.-79C>T, respectively), but no non-synonymous mutations. In summary, we have identified novel missense mutations in the third immunoglobulin-like domain of myopalladin, which have either no or profound effects on the molecular composition of the sarcomere. According to our epidemiological data, the prevalence of ANKRD1 mutations seems to be lower than that of its binding partner myopalladin, indicating the clinical significance of myopalladin for the functional integrity of the sarcomeric apparatus and the protection against DCM.


Subject(s)
Cardiomyopathy, Dilated/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Sarcomeres/metabolism , Adult , Cardiomyopathy, Dilated/pathology , Cohort Studies , Female , Humans , Male , Middle Aged , Mutation, Missense , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Repressor Proteins/genetics , Sarcomeres/genetics
16.
Mol Biosyst ; 8(2): 495-503, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22101280

ABSTRACT

The adaptation of the cellular network to functional changes, timing and patterning of gene expression is regulated by binding of transcription factors to gene regulatory elements, which in turn depends on co-occurring histone modifications. These two layers influence each other, enabling a further level of regulatory fine-tuning. We analyzed the interdependencies between histone 3 acetylation, histone 3 lysine 4 dimethylation, the transcription factor Srf and the histone acetyltransferase p300 in an in vivo model using chromatin immunoprecipitation in a time-series during cardiac maturation in mouse. We found a strong correlation between the presence of the two histone modifications and binding of Srf and p300. Using linear modeling techniques we could show that each factor contributes individually as well as conjointly to histone 3 acetylation and gene expression, probably aided by accompanying histone 3 lysine 4 dimethylation. We further demonstrate that changes in gene expression during cardiac maturation are attended by changes of the analyzed regulators while revealing a high variability of combinatorial regulation. Finally, we propose a model of Srf-driven gene expression in cardiomyocytes.


Subject(s)
E1A-Associated p300 Protein/metabolism , Heart/embryology , Histones/metabolism , Serum Response Factor/metabolism , Acetylation , Animals , Cell Line , Chromatin Immunoprecipitation/methods , DNA/metabolism , Gene Expression Regulation, Developmental , Methylation , Mice , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , Transcription, Genetic , Transcriptional Activation
17.
Cardiovasc Res ; 91(2): 269-78, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21527437

ABSTRACT

Even though the foundation of systems biology approaches to cardiac function was led more than fifty years ago, there has been slow progression over the last few decades. Systems biology studies were mainly focused on lower organisms, frequently on yeast. With the boost of high-throughput technologies, systems level analyses, building one backbone of systems biology, started to complement the single-gene focus in the fields of heart development and congenital heart disease. A challenge is to bring together the many uncovered molecular components driving heart development and eventually to establish computational models describing this complex developmental process. Congenital heart diseases represent overlapping phenotypes, reflecting the modularity of heart development. The aetiology of the majority of congenital heart disease is still unknown, and it is suggestive that understanding the biological network underlying heart development will enhance our understanding for its alteration. This review provides an overview of the framework for systems biology approaches focusing on the developing heart and its pathology. Recent methodological developments building the basis for future studies are highlighted and the knowledge gained is specified.


Subject(s)
Heart Defects, Congenital/embryology , Heart/embryology , Systems Biology , Animals , Computer Simulation , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Genomics , Heart/physiopathology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/physiopathology , Humans , Models, Cardiovascular , Morphogenesis , Phenotype , Proteomics , Systems Integration
18.
PLoS Genet ; 7(2): e1001313, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21379568

ABSTRACT

The transcriptome, as the pool of all transcribed elements in a given cell, is regulated by the interaction between different molecular levels, involving epigenetic, transcriptional, and post-transcriptional mechanisms. However, many previous studies investigated each of these levels individually, and little is known about their interdependency. We present a systems biology study integrating mRNA profiles with DNA-binding events of key cardiac transcription factors (Gata4, Mef2a, Nkx2.5, and Srf), activating histone modifications (H3ac, H4ac, H3K4me2, and H3K4me3), and microRNA profiles obtained in wild-type and RNAi-mediated knockdown. Finally, we confirmed conclusions primarily obtained in cardiomyocyte cell culture in a time-course of cardiac maturation in mouse around birth. We provide insights into the combinatorial regulation by cardiac transcription factors and show that they can partially compensate each other's function. Genes regulated by multiple transcription factors are less likely differentially expressed in RNAi knockdown of one respective factor. In addition to the analysis of the individual transcription factors, we found that histone 3 acetylation correlates with Srf- and Gata4-dependent gene expression and is complementarily reduced in cardiac Srf knockdown. Further, we found that altered microRNA expression in Srf knockdown potentially explains up to 45% of indirect mRNA targets. Considering all three levels of regulation, we present an Srf-centered transcription network providing on a single-gene level insights into the regulatory circuits establishing respective mRNA profiles. In summary, we show the combinatorial contribution of four DNA-binding transcription factors in regulating the cardiac transcriptome and provide evidence that histone modifications and microRNAs modulate their functional consequence. This opens a new perspective to understand heart development and the complexity cardiovascular disorders.


Subject(s)
Gene Regulatory Networks , Histones/metabolism , MicroRNAs/metabolism , Myocardium/metabolism , Protein Processing, Post-Translational/genetics , Transcription Factors/metabolism , Transcription, Genetic , Acetylation , Animals , Binding Sites , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , MEF2 Transcription Factors , Mice , Myogenic Regulatory Factors/metabolism , Protein Binding , Reproducibility of Results , Serum Response Factor/metabolism
19.
Circ Cardiovasc Genet ; 4(1): 43-50, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21127202

ABSTRACT

BACKGROUND: Ebstein anomaly is a rare congenital heart malformation characterized by adherence of the septal and posterior leaflets of the tricuspid valve to the underlying myocardium. An association between Ebstein anomaly with left ventricular noncompaction (LVNC) and mutations in MYH7 encoding ß-myosin heavy chain has been shown; in this report, we have screened for MYH7 mutations in a cohort of probands with Ebstein anomaly in a large population-based study. METHODS AND RESULTS: Mutational analysis in a cohort of 141 unrelated probands with Ebstein anomaly was performed by next-generation sequencing and direct DNA sequencing of MYH7. Heterozygous mutations were identified in 8 of 141 samples (6%). Seven distinct mutations were found; 5 were novel and 2 were known to cause hypertrophic cardiomyopathy. All mutations except for 1 3-bp deletion were missense mutations; 1 was a de novo change. Mutation-positive probands and family members showed various congenital heart malformations as well as LVNC. Among 8 mutation-positive probands, 6 had LVNC, whereas among 133 mutation-negative probands, none had LVNC. The frequency of MYH7 mutations was significantly different between probands with and without LVNC accompanying Ebstein anomaly (P<0.0001). LVNC segregated with the MYH7 mutation in the pedigrees of 3 of the probands, 1 of which also included another individual with Ebstein anomaly. CONCLUSIONS: Ebstein anomaly is a congenital heart malformation that is associated with mutations in MYH7. MYH7 mutations are predominantly found in Ebstein anomaly associated with LVNC and may warrant genetic testing and family evaluation in this subset of patients.


Subject(s)
Cardiac Myosins/genetics , Ebstein Anomaly/genetics , Mutation/genetics , Myosin Heavy Chains/genetics , Sarcomeres/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Cardiac Myosins/chemistry , Child , Child, Preschool , Cohort Studies , Ebstein Anomaly/diagnostic imaging , Family , Female , Humans , Infant , Male , Middle Aged , Molecular Sequence Data , Myosin Heavy Chains/chemistry , Pedigree , Ultrasonography , Young Adult
20.
Methods ; 50(4): S19-22, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20215016

ABSTRACT

Quantitative real-time PCR (qPCR) is a frequently used, sensitive and accurate method to study gene expression profiles. However, its throughput was so far limited for routine laboratories to 384 reactions per run based on the limitations of the available instruments. Recently, the LightCycler 1536 Instrument was launched providing a high-throughput solution for qPCR with the analysis of 1536 reactions in approximately 45 min. We assessed the accuracy and sensitivity of this novel technology for the analysis of gene expression profiles in combination with the Innovadyne Nanodrop Express pipetting robot. We compared expression profiles obtained for 42 genes in 71 samples between the Universal ProbeLibrary and the LightCycler 1536 Instrument and SYBR Green I and the ABI PRISM 7900HT system. We found that the results were highly reproducible between both systems. Beside the higher throughput, the advantage of the LightCycler 1536 Instrument was the reduced consumption of reagents and sample material.


Subject(s)
Gene Expression Profiling/instrumentation , Reverse Transcriptase Polymerase Chain Reaction/instrumentation , Gene Expression Profiling/methods , Heart Defects, Congenital/metabolism , Humans , Hydrolysis , Myocardium/chemistry , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...