Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 1441: 295-311, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884718

RESUMEN

Cardiac development is a fine-tuned process governed by complex transcriptional networks, in which transcription factors (TFs) interact with other regulatory layers. In this chapter, we introduce the core cardiac TFs including Gata, Hand, Nkx2, Mef2, Srf, and Tbx. These factors regulate each other's expression and can also act in a combinatorial manner on their downstream targets. Their disruption leads to various cardiac phenotypes in mice, and mutations in humans have been associated with congenital heart defects. In the second part of the chapter, we discuss different levels of regulation including cis-regulatory elements, chromatin structure, and microRNAs, which can interact with transcription factors, modulate their function, or are downstream targets. Finally, examples of disturbances of the cardiac regulatory network leading to congenital heart diseases in human are provided.


Asunto(s)
Redes Reguladoras de Genes , Cardiopatías Congénitas , Factores de Transcripción , Animales , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Corazón/fisiología , Miocardio/metabolismo
2.
Adv Exp Med Biol ; 1441: 629-644, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884738

RESUMEN

Tetralogy of Fallot (TOF) and double-outlet right ventricle (DORV) are conotruncal defects resulting from disturbances of the second heart field and the neural crest, which can occur as isolated malformations or as part of multiorgan syndromes. Their etiology is multifactorial and characterized by overlapping genetic causes. In this chapter, we present the different genetic alterations underlying the two diseases, which range from chromosomal abnormalities like aneuploidies and structural mutations to rare single nucleotide variations affecting distinct genes. For example, mutations in the cardiac transcription factors NKX2-5, GATA4, and HAND2 have been identified in isolated TOF cases, while mutations of TBX5 and 22q11 deletion, leading to haploinsufficiency of TBX1, cause Holt-Oram and DiGeorge syndrome, respectively. Moreover, genes involved in signaling pathways, laterality determination, and epigenetic mechanisms have also been found mutated in TOF and/or DORV patients. Finally, genome-wide association studies identified common single nucleotide polymorphisms associated with the risk for TOF.


Asunto(s)
Ventrículo Derecho con Doble Salida , Tetralogía de Fallot , Humanos , Tetralogía de Fallot/genética , Ventrículo Derecho con Doble Salida/genética , Mutación , Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple/genética , Predisposición Genética a la Enfermedad/genética , Factores de Transcripción/genética
3.
Adv Exp Med Biol ; 1441: 435-458, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884724

RESUMEN

Over the last few decades, the study of congenital heart disease (CHD) has benefited from various model systems and the development of molecular biological techniques enabling the analysis of single gene as well as global effects. In this chapter, we first describe different models including CHD patients and their families, animal models ranging from invertebrates to mammals, and various cell culture systems. Moreover, techniques to experimentally manipulate these models are discussed. Second, we introduce cardiac phenotyping technologies comprising the analysis of mouse and cell culture models, live imaging of cardiogenesis, and histological methods for fixed hearts. Finally, the most important and latest molecular biotechniques are described. These include genotyping technologies, different applications of next-generation sequencing, and the analysis of transcriptome, epigenome, proteome, and metabolome. In summary, the models and technologies presented in this chapter are essential to study the function and development of the heart and to understand the molecular pathways underlying CHD.


Asunto(s)
Cardiopatías Congénitas , Animales , Humanos , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Modelos Animales de Enfermedad , Ratones , Fenotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Técnicas de Cultivo de Célula/métodos
4.
Hum Mol Genet ; 23(12): 3115-28, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24459294

RESUMEN

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.


Asunto(s)
Predisposición Genética a la Enfermedad , Variación Genética , Estudio de Asociación del Genoma Completo/métodos , Miocardio/patología , Tetralogía de Fallot/genética , Tetralogía de Fallot/patología , Apoptosis , Secuencia de Bases , Proliferación Celular , Estudios de Cohortes , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Frecuencia de los Genes , Humanos , Datos de Secuencia Molecular , Herencia Multifactorial , Mutación , Miocardio/metabolismo , Análisis de Secuencia de ADN , Tetralogía de Fallot/sangre
5.
PLoS Genet ; 7(2): e1001313, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21379568

RESUMEN

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.


Asunto(s)
Redes Reguladoras de Genes , Histonas/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Procesamiento Proteico-Postraduccional/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Acetilación , Animales , Sitios de Unión , Factor de Transcripción GATA4/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción MEF2 , Ratones , Factores Reguladores Miogénicos/metabolismo , Unión Proteica , Reproducibilidad de los Resultados , Factor de Respuesta Sérica/metabolismo
6.
Bioinformatics ; 26(1): 123-4, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19880369

RESUMEN

MOTIVATION: Deep sequencing has become the method of choice for determining the small RNA content of a cell. Mapping the sequenced reads onto their reference genome serves as the basis for all further analyses, namely for identification and quantification. A method frequently used is Mega BLAST followed by several filtering steps, even though it is slow and inefficient for this task. Also, none of the currently available short read aligners has established itself for the particular task of small RNA mapping. RESULTS: We present MicroRazerS, a tool optimized for mapping small RNAs onto a reference genome. It is an order of magnitude faster than Mega BLAST and comparable in speed with other short read mapping tools. In addition, it is more sensitive and easy to handle and adjust. AVAILABILITY: MicroRazerS is part of the SeqAn C++ library and can be downloaded from http://www.seqan.de/projects/MicroRazerS.html.


Asunto(s)
Algoritmos , MicroARNs/genética , Alineación de Secuencia/métodos , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Secuencia de Bases , Datos de Secuencia Molecular
7.
J Cardiovasc Dev Dis ; 7(4)2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33276527

RESUMEN

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.

8.
Sci Rep ; 10(1): 10921, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32616843

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Miocitos Cardíacos/metabolismo , Tetralogía de Fallot/patología , Estudios de Casos y Controles , Diferenciación Celular , Células Clonales , Femenino , Regulación del Desarrollo de la Expresión Génica , Estudios de Asociación Genética , Mutación de Línea Germinal , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Piel/citología , Tetralogía de Fallot/genética , Transcripción Genética
9.
Dis Model Mech ; 13(12)2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33033063

RESUMEN

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.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Conectina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Estudios de Asociación Genética , Proteínas de la Membrana/genética , Tetralogía de Fallot/genética , Animales , Proteínas de Drosophila/metabolismo , Femenino , Humanos , Locomoción , Masculino , Proteínas de la Membrana/metabolismo , Músculos/metabolismo , Mutación/genética , Miocardio , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miofibrillas/metabolismo , Miofibrillas/patología , Especificidad de Órganos , Unión Proteica , Mapeo de Interacción de Proteínas
10.
Sci Rep ; 9(1): 19063, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31836860

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Tetralogía de Fallot/genética , Cromosomas Humanos Par 6/genética , Femenino , Perfilación de la Expresión Génica , Genoma Humano , Humanos , Masculino , MicroARNs/metabolismo , Anotación de Secuencia Molecular , Miocardio/metabolismo , Miocardio/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
PLoS One ; 12(6): e0179464, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28609469

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Histonas/genética , Proteína MioD/genética , Mioblastos/metabolismo , Animales , Línea Celular , Células Cultivadas , Análisis por Conglomerados , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Células HEK293 , Histonas/clasificación , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilación , Ratones , Desarrollo de Músculos/genética , Proteína MioD/metabolismo , Mioblastos/citología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Cardiovasc Res ; 112(1): 464-77, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27496870

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/genética , Metilación de ADN , Epigénesis Genética , Perfilación de la Expresión Génica/métodos , Defectos del Tabique Interventricular/genética , Proteínas Mitocondriales/genética , Tetralogía de Fallot/genética , Adaptación Fisiológica , Adulto , Estudios de Casos y Controles , Preescolar , Islas de CpG , Redes Reguladoras de Genes , Estudios de Asociación Genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Defectos del Tabique Interventricular/fisiopatología , Hemodinámica , Humanos , Lactante , Persona de Mediana Edad , Chaperonas Moleculares , Fenotipo , Análisis de Componente Principal , Regiones Promotoras Genéticas , Empalme del ARN , Reproducibilidad de los Resultados , Tetralogía de Fallot/fisiopatología , Adulto Joven
13.
Brief Funct Genomics ; 13(1): 51-65, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24095982

RESUMEN

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.


Asunto(s)
Variación Genética , Genómica/métodos , Cardiopatías Congénitas/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Bases de Datos Genéticas , Estudios de Asociación Genética , Humanos
14.
PLoS One ; 9(1): e85375, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24400131

RESUMEN

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.


Asunto(s)
Biología Computacional/métodos , Variaciones en el Número de Copia de ADN , Análisis de Secuencia de ADN/métodos , Tetralogía de Fallot/genética , Algoritmos , Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Cadenas de Markov
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA