Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1441: 57-75, 2024.
Article in English | MEDLINE | ID: mdl-38884704

ABSTRACT

Congenital heart diseases (or congenital heart defects/disorders; CHDs) are structural abnormalities of the heart and/or great vessels that are present at birth. CHDs include an extensive range of defects that may be minor and require no intervention or may be life-limiting and require complex surgery shortly after birth. This chapter reviews the current knowledge on the genetic causes of CHD.


Subject(s)
Heart Defects, Congenital , Humans , Heart Defects, Congenital/genetics , Mutation
2.
J Anat ; 245(1): 70-83, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38419169

ABSTRACT

Congenital heart disease (CHD) is the most common congenital anomaly, with an overall incidence of approximately 1% in the United Kingdom. Exome sequencing in large CHD cohorts has been performed to provide insights into the genetic aetiology of CHD. This includes a study of 1891 probands by our group in collaboration with others, which identified three novel genes-CDK13, PRKD1, and CHD4, in patients with syndromic CHD. PRKD1 encodes a serine/threonine protein kinase, which is important in a variety of fundamental cellular functions. Individuals with a heterozygous mutation in PRKD1 may have facial dysmorphism, ectodermal dysplasia and may have CHDs such as pulmonary stenosis, atrioventricular septal defects, coarctation of the aorta and bicuspid aortic valve. To obtain a greater appreciation for the role that this essential protein kinase plays in cardiogenesis and CHD, we have analysed a Prkd1 transgenic mouse model (Prkd1em1) carrying deletion of exon 2, causing loss of function. High-resolution episcopic microscopy affords detailed morphological 3D analysis of the developing heart and provides evidence for an essential role of Prkd1 in both normal cardiac development and CHD. We show that homozygous deletion of Prkd1 is associated with complex forms of CHD such as atrioventricular septal defects, and bicuspid aortic and pulmonary valves, and is lethal. Even in heterozygotes, cardiac differences occur. However, given that 97% of Prkd1 heterozygous mice display normal heart development, it is likely that one normal allele is sufficient, with the defects seen most likely to represent sporadic events. Moreover, mRNA and protein expression levels were investigated by RT-qPCR and western immunoblotting, respectively. A significant reduction in Prkd1 mRNA levels was seen in homozygotes, but not heterozygotes, compared to WT littermates. While a trend towards lower PRKD1 protein expression was seen in the heterozygotes, the difference was only significant in the homozygotes. There was no compensation by the related Prkd2 and Prkd3 at transcript level, as evidenced by RT-qPCR. Overall, we demonstrate a vital role of Prkd1 in heart development and the aetiology of CHD.


Subject(s)
Heart Defects, Congenital , Heart , Animals , Mice , Heart Defects, Congenital/genetics , Heart/embryology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Mice, Transgenic
3.
J Anat ; 236(3): 549-563, 2020 03.
Article in English | MEDLINE | ID: mdl-31724174

ABSTRACT

Cardiogenesis is influenced by both environmental and genetic factors, with blood flow playing a critical role in cardiac remodelling. Perturbation of any of these factors could lead to abnormal heart development and hence the formation of congenital heart defects. Although abnormal blood flow has been associated with a number of heart defects, the effects of abnormal pressure load on the developing heart gene expression profile have to date not clearly been defined. To determine the heart transcriptional response to haemodynamic alteration during development, outflow tract (OFT) banding was employed in the chick embryo at Hamburger and Hamilton stage (HH) 21. Stereological and expression studies, including the use of global expression analysis by RNA sequencing with an optimised procedure for effective globin depletion, were subsequently performed on HH29 OFT-banded hearts and compared with sham control hearts, with further targeted expression investigations at HH35. The OFT-banded hearts were found to have an abnormal morphology with a rounded appearance and left-sided dilation in comparison with controls. Internal analysis showed they typically had a ventricular septal defect and reductions in the myocardial wall and trabeculae, with an increase in the lumen on the left side of the heart. There was also a significant reduction in apoptosis. The differentially expressed genes were found to be predominately involved in contraction, metabolism, apoptosis and neural development, suggesting a cardioprotective mechanism had been induced. Therefore, altered haemodynamics during development leads to left-sided dilation and differential expression of genes that may be associated with stress and maintaining cardiac output.


Subject(s)
Gene Expression Regulation , Heart Defects, Congenital/pathology , Hemodynamics/physiology , Animals , Chick Embryo , Disease Models, Animal , Heart Defects, Congenital/genetics , Sequence Analysis, RNA
4.
J Anat ; 234(6): 800-814, 2019 06.
Article in English | MEDLINE | ID: mdl-30882904

ABSTRACT

During embryo development, the heart is the first functioning organ. Although quiescent in the adult, the epicardium is essential during development to form a normal four-chambered heart. Epicardial-derived cells contribute to the heart as it develops with fibroblasts and vascular smooth muscle cells. Previous studies have shown that a heartbeat is required for epicardium formation, but no study to our knowledge has shown the effects of haemodynamic changes on the epicardium. Since the aetiologies of many congenital heart defects are unknown, we suggest that an alteration in the heart's haemodynamics might provide an explanatory basis for some of them. To change the heart's haemodynamics, outflow tract (OFT) banding using a double overhang knot was performed on HH21 chick embryos, with harvesting at different developmental stages. The epicardium of the heart was phenotypically and functionally characterised using a range of techniques. Upon alteration of haemodynamics, the epicardium exhibited abnormal morphology at HH29, even though migration of epicardial cells along the surface of the heart was found to be normal between HH24 and HH28. The abnormal epicardial phenotype was exacerbated at HH35 with severe changes in the structure of the extracellular matrix (ECM). A number of genes tied to ECM production were also differentially expressed in HH29 OFT-banded hearts, including DDR2 and collagen XII. At HH35, the differential expression of these genes was even greater with additional downregulation of collagen I and TCF21. In this study, the epicardium was found to be severely impacted by altered haemodynamics upon OFT banding. The increased volume of the epicardium at HH29, upon OFT-banding, and the expression changes of ECM markers were the first indicative signs of aberrations in epicardial architecture; by HH35, the phenotype had progressed. The decrease in epicardial thickness at HH35 suggests an increase in tension, with a force acting perpendicular to the surface of the epicardium. Although the developing epicardium and the blood flowing through the heart are separated by the endocardium and myocardium, the data presented here demonstrate that altering the blood flow affects the structure and molecular expression of the epicardial layer. Due to the intrinsic role the epicardium in cardiogenesis, defects in epicardial formation could have a role in the formation of a wide range of congenital heart defects.


Subject(s)
Hemodynamics/physiology , Pericardium/embryology , Animals , Chick Embryo , Organogenesis/physiology
5.
J Mol Cell Cardiol ; 114: 185-198, 2018 01.
Article in English | MEDLINE | ID: mdl-29174768

ABSTRACT

TBX5 plays a critical role in heart and forelimb development. Mutations in TBX5 cause Holt-Oram syndrome, an autosomal dominant condition that affects the formation of the heart and upper-limb. Several studies have provided significant insight into the role of TBX5 in cardiogenesis; however, how TBX5 activity is regulated by other factors is still unknown. Here we report that histone acetyltransferases KAT2A and KAT2B associate with TBX5 and acetylate it at Lys339. Acetylation potentiates its transcriptional activity and is required for nuclear retention. Morpholino-mediated knockdown of kat2a and kat2b transcripts in zebrafish severely perturb heart and limb development, mirroring the tbx5a knockdown phenotype. The phenotypes found in MO-injected embryos were also observed when we introduced mutations in the kat2a or kat2b genes using the CRISPR-Cas system. These studies highlight the importance of KAT2A and KAT2B modulation of TBX5 and their impact on heart and limb development.


Subject(s)
Extremities/embryology , Heart/embryology , Histone Acetyltransferases/metabolism , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Acetylation , Amino Acid Sequence , Animal Fins/embryology , Animals , CRISPR-Cas Systems/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Down-Regulation/drug effects , Embryo, Nonmammalian/metabolism , Embryonic Development/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Heart/drug effects , Histone Acetyltransferases/genetics , Morpholinos/pharmacology , Phenotype , T-Box Domain Proteins/chemistry , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
J Mol Cell Cardiol ; 108: 114-126, 2017 07.
Article in English | MEDLINE | ID: mdl-28576718

ABSTRACT

Intracardiac haemodynamics is crucial for normal cardiogenesis, with recent evidence showing valvulogenesis is haemodynamically dependent and inextricably linked with shear stress. Although valve anomalies have been associated with genetic mutations, often the cause is unknown. However, altered haemodynamics have been suggested as a pathogenic contributor to bicuspid aortic valve disease. Conversely, how abnormal haemodynamics impacts mitral valve development is still poorly understood. In order to analyse altered blood flow, the outflow tract of the chick heart was constricted using a ligature to increase cardiac pressure overload. Outflow tract-banding was performed at HH21, with harvesting at crucial valve development stages (HH26, HH29 and HH35). Although normal valve morphology was found in HH26 outflow tract banded hearts, smaller and dysmorphic mitral valve primordia were seen upon altered haemodynamics in histological and stereological analysis at HH29 and HH35. A decrease in apoptosis, and aberrant expression of a shear stress responsive gene and extracellular matrix markers in the endocardial cushions were seen in the chick HH29 outflow tract banded hearts. In addition, dysregulation of extracellular matrix (ECM) proteins fibrillin-2, type III collagen and tenascin were further demonstrated in more mature primordial mitral valve leaflets at HH35, with a concomitant decrease of ECM cross-linking enzyme, transglutaminase-2. These data provide compelling evidence that normal haemodynamics are a prerequisite for normal mitral valve morphogenesis, and abnormal blood flow could be a contributing factor in mitral valve defects, with differentiation as a possible underlying mechanism.


Subject(s)
Heart/embryology , Heart/physiology , Hemodynamics , Mitral Valve/embryology , Animals , Biomarkers , Chick Embryo , Extracellular Matrix , Gene Expression Profiling , Heart Defects, Congenital/etiology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Mitral Valve/abnormalities , Mitral Valve/metabolism , Organogenesis/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
7.
J Mol Cell Cardiol ; 106: 1-13, 2017 05.
Article in English | MEDLINE | ID: mdl-28359939

ABSTRACT

Tropomyosin 1 (TPM1) is an essential sarcomeric component, stabilising the thin filament and facilitating actin's interaction with myosin. A number of sarcomeric proteins, such as alpha myosin heavy chain, play crucial roles in cardiac development. Mutations in these genes have been linked to congenital heart defects (CHDs), occurring in approximately 1 in 145 live births. To date, TPM1 has not been associated with isolated CHDs. Analysis of 380 CHD cases revealed three novel mutations in the TPM1 gene; IVS1+2T>C, I130V, S229F and a polyadenylation signal site variant GATAAA/AATAAA. Analysis of IVS1+2T>C revealed aberrant pre-mRNA splicing. In addition, abnormal structural properties were found in hearts transfected with TPM1 carrying I130V and S229F mutations. Phenotypic analysis of TPM1 morpholino-treated embryos revealed roles for TPM1 in cardiac looping, atrial septation and ventricular trabeculae formation and increased apoptosis was seen within the heart. In addition, sarcomere assembly was affected and altered action potentials were exhibited. This study demonstrated that sarcomeric TPM1 plays vital roles in cardiogenesis and is a suitable candidate gene for screening individuals with isolated CHDs.


Subject(s)
Heart Defects, Congenital/genetics , Heart/growth & development , Myosin Heavy Chains/genetics , Tropomyosin/genetics , Actins/genetics , Female , Heart/physiopathology , Heart Defects, Congenital/pathology , Heart Ventricles/growth & development , Heart Ventricles/pathology , Humans , Male , Mutation/genetics , Phenotype , RNA Precursors/genetics , RNA Splicing/genetics , Sarcomeres/genetics
8.
J Cardiovasc Dev Dis ; 4(3)2017 Jul 07.
Article in English | MEDLINE | ID: mdl-29367539

ABSTRACT

Animals have frequently been used as models for human disorders and mutations. Following advances in genetic testing and treatment options, and the decreasing cost of these technologies in the clinic, mutations in both companion and commercial animals are now being investigated. A recent review highlighted the genes associated with both human and non-human dilated cardiomyopathy. Cardiac troponin T and dystrophin were observed to be associated with both human and turkey (troponin T) and canine (dystrophin) dilated cardiomyopathies. This review gives an overview of the work carried out in cardiac troponin T and dystrophin to date in both human and animal dilated cardiomyopathy.

9.
J Anat ; 229(3): 436-49, 2016 09.
Article in English | MEDLINE | ID: mdl-27194630

ABSTRACT

The heart is the first functioning organ to develop during embryogenesis. The formation of the heart is a tightly regulated and complex process, and alterations to its development can result in congenital heart defects. Mutations in sarcomeric proteins, such as alpha myosin heavy chain and cardiac alpha actin, have now been associated with congenital heart defects in humans, often with atrial septal defects. However, cardiac troponin T (cTNT encoded by gene TNNT2) has not. Using gene-specific antisense oligonucleotides, we have investigated the role of cTNT in chick cardiogenesis. TNNT2 is expressed throughout heart development and in the postnatal heart. TNNT2-morpholino treatment resulted in abnormal atrial septal growth and a reduction in the number of trabeculae in the developing primitive ventricular chamber. External analysis revealed the development of diverticula from the ventricular myocardial wall which showed no evidence of fibrosis and still retained a myocardial phenotype. Sarcomeric assembly appeared normal in these treated hearts. In humans, congenital ventricular diverticulum is a rare condition, which has not yet been genetically associated. However, abnormal haemodynamics is known to cause structural defects in the heart. Further, structural defects, including atrial septal defects and congenital diverticula, have previously been associated with conduction anomalies. Therefore, to provide mechanistic insights into the effect that cTNT knockdown has on the developing heart, quantitative PCR was performed to determine the expression of the shear stress responsive gene NOS3 and the conduction gene TBX3. Both genes were differentially expressed compared to controls. Therefore, a reduction in cTNT in the developing heart results in abnormal atrial septal formation and aberrant ventricular morphogenesis. We hypothesize that alterations to the haemodynamics, indicated by differential NOS3 expression, causes these abnormalities in growth in cTNT knockdown hearts. In addition, the muscular diverticula reported here suggest a novel role for mutations of structural sarcomeric proteins in the pathogenesis of congenital cardiac diverticula. From these studies, we suggest TNNT2 is a gene worthy of screening for those with a congenital heart defect, particularly atrial septal defects and ventricular diverticula.


Subject(s)
Heart Defects, Congenital , Heart/embryology , Troponin T/metabolism , Animals , Blotting, Western , Chick Embryo , Disease Models, Animal , Gene Knockdown Techniques , Immunohistochemistry , Real-Time Polymerase Chain Reaction
10.
Cell Mol Life Sci ; 70(7): 1221-39, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22955375

ABSTRACT

Myosin is an essential component of cardiac muscle, from the onset of cardiogenesis through to the adult heart. Although traditionally known for its role in energy transduction and force development, recent studies suggest that both myosin heavy-chain and myosin light-chain proteins are required for a correctly formed heart. Myosins are structural proteins that are not only expressed from early stages of heart development, but when mutated in humans they may give rise to congenital heart defects. This review will discuss the roles of myosin, specifically with regards to the developing heart. The expression of each myosin protein will be described, and the effects that altering expression has on the heart in embryogenesis in different animal models will be discussed. The human molecular genetics of the myosins will also be reviewed.


Subject(s)
Heart/embryology , Morphogenesis/genetics , Myosin Heavy Chains/physiology , Myosin Light Chains/physiology , Adult , Animals , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Humans , Models, Biological , Morphogenesis/physiology , Muscle Development/genetics , Muscle Development/physiology , Myocardium/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Light Chains/genetics , Myosin Light Chains/metabolism
11.
Development ; 138(18): 3955-66, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21862559

ABSTRACT

The expression and function of embryonic myosin heavy chain (eMYH) has not been investigated within the early developing heart. This is despite the knowledge that other structural proteins, such as alpha and beta myosin heavy chains and cardiac alpha actin, play crucial roles in atrial septal development and cardiac function. Most cases of atrial septal defects and cardiomyopathy are not associated with a known causative gene, suggesting that further analysis into candidate genes is required. Expression studies localised eMYH in the developing chick heart. eMYH knockdown was achieved using morpholinos in a temporal manner and functional studies were carried out using electrical and calcium signalling methodologies. Knockdown in the early embryo led to abnormal atrial septal development and heart enlargement. Intriguingly, action potentials of the eMYH knockdown hearts were abnormal in comparison with the alpha and beta myosin heavy chain knockdowns and controls. Although myofibrillogenesis appeared normal, in knockdown hearts the tissue integrity was affected owing to apparent focal points of myocyte loss and an increase in cell death. An expression profile of human skeletal myosin heavy chain genes suggests that human myosin heavy chain 3 is the functional homologue of the chick eMYH gene. These data provide compelling evidence that eMYH plays a crucial role in important processes in the early developing heart and, hence, is a candidate causative gene for atrial septal defects and cardiomyopathy.


Subject(s)
Heart/embryology , Heart/physiology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/physiology , Animals , Animals, Genetically Modified , Cardiomyopathy, Dilated/genetics , Chick Embryo , Embryo, Mammalian , Gene Knockdown Techniques , Heart/anatomy & histology , Heart Defects, Congenital/genetics , Humans , Mice , Morphogenesis/genetics , Myocardium/metabolism , Myosin Heavy Chains/metabolism , Organogenesis/genetics , Organogenesis/physiology , Survival , Validation Studies as Topic
12.
J Anat ; 214(6): 905-15, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19538634

ABSTRACT

Atrial septal defects are a common congenital heart defect in humans. Although mutations in different genes are now frequently being described, little is known about the processes and mechanisms behind the early stages of atrial septal development. By utilizing morpholino-induced knockdown in the chick we have analysed the role of alpha myosin heavy chain during early cardiogenesis in a temporal manner. Upon knockdown of alpha myosin heavy chain, three different phenotypes of the atrial septum were observed: (1) the atrial septum failed to initiate, (2) the septum was initiated but was growth restricted, or (3) incorrect specification occurred resulting in multiple septa forming. In addition, at a lower frequency, decreased alpha myosin heavy chain was found to give rise to an abnormally looped heart or an enlarged heart. Staining of the actin cytoskeleton indicated that many of the myofibrils in the knockdown hearts were not as mature as those observed in the controls, suggesting a mechanism for the defects seen. Therefore, these data suggest a role for alpha myosin heavy chain in modelling of the early heart and the range of defects to the atrial septum suggest roles in its initiation, specification and growth during development.


Subject(s)
Cytoskeleton/pathology , Heart/embryology , Ventricular Myosins/physiology , Actin Cytoskeleton/metabolism , Animals , Atrial Septum/embryology , Atrial Septum/metabolism , Atrial Septum/pathology , Cardiomegaly/embryology , Cardiomegaly/pathology , Chick Embryo , Embryonic Development , Gene Knockdown Techniques , Phenotype , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
13.
Methods Mol Biol ; 411: 1-11, 2007.
Article in English | MEDLINE | ID: mdl-18287634

ABSTRACT

This chapter provides information on beta-galactosidase staining of whole mouse embryos, organs, tissue sections, and cultured cells, as well as double staining with horseradish peroxidase and use as a tool for genotyping. Using these protocols, localization of beta-galactosidase can be visualized throughout development and in adult tissues. beta-Galactosidase staining may be used purely as a marker of gene expression and also as a tracer in cell lineage studies.


Subject(s)
Embryo, Mammalian/chemistry , Genes, Reporter , Staining and Labeling/methods , beta-Galactosidase/analysis , Animals , Coloring Agents/chemistry , Histocytochemistry , Immunoenzyme Techniques , Immunohistochemistry , Mice , beta-Galactosidase/genetics
14.
Nat Genet ; 37(4): 423-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15735645

ABSTRACT

Atrial septal defect is one of the most common forms of congenital heart malformation. We identified a new locus linked with atrial septal defect on chromosome 14q12 in a large family with dominantly inherited atrial septal defect. The underlying mutation is a missense substitution, I820N, in alpha-myosin heavy chain (MYH6), a structural protein expressed at high levels in the developing atria, which affects the binding of the heavy chain to its regulatory light chain. The cardiac transcription factor TBX5 strongly regulates expression of MYH6, but mutant forms of TBX5, which cause Holt-Oram syndrome, do not. Morpholino knock-down of expression of the chick MYH6 homolog eliminates the formation of the atrial septum without overtly affecting atrial chamber formation. These data provide evidence for a link between a transcription factor, a structural protein and congenital heart disease.


Subject(s)
Cardiac Myosins/genetics , Heart Septal Defects, Atrial/genetics , Mutation, Missense , Myosin Heavy Chains/genetics , T-Box Domain Proteins/genetics , Adult , Amino Acid Substitution , Animals , Cardiac Myosins/metabolism , Chick Embryo , Child , Child, Preschool , Female , Genetic Linkage , Heart Septal Defects, Atrial/embryology , Humans , Infant, Newborn , Male , Molecular Sequence Data , Myosin Heavy Chains/metabolism , Pedigree , T-Box Domain Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...