Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
1.
Circ Arrhythm Electrophysiol ; 13(10): e008712, 2020 10.
Article in English | MEDLINE | ID: mdl-32755394

ABSTRACT

BACKGROUND: Mutations in the nuclear envelope genes encoding LMNA and EMD are responsible for Emery-Dreifuss muscular dystrophy. However, LMNA mutations often manifest dilated cardiomyopathy with conduction disturbance without obvious skeletal myopathic complications. On the contrary, the phenotypic spectrums of EMD mutations are less clear. Our aims were to determine the prevalence of nonsyndromic forms of emerinopathy, which may underlie genetically undefined isolated cardiac conduction disturbance, and the etiology of thromboembolic complications associated with EMD mutations. METHODS: Targeted exon sequencing was performed in 87 probands with familial sick sinus syndrome (n=36) and a progressive cardiac conduction defect (n=51). RESULTS: We identified 3 X-linked recessive EMD mutations (start-loss, splicing, missense) in families with cardiac conduction disease. All 3 probands shared a common clinical phenotype of progressive atrial arrhythmias that ultimately resulted in atrial standstill associated with left ventricular noncompaction (LVNC), but they lacked early contractures and progressive muscle wasting and weakness characteristic of Emery-Dreifuss muscular dystrophy. Because the association of LVNC with EMD has never been reported, we further genetically screened 102 LVNC patients and found a frameshift EMD mutation in a boy with progressive atrial standstill and LVNC without complications of muscular dystrophy. All 6 male EMD mutation carriers of 4 families underwent pacemaker or defibrillator implantation, whereas 2 female carriers were asymptomatic. Notably, a strong family history of stroke observed in these families was probably due to the increased risk of thromboembolism attributable to both atrial standstill and LVNC. CONCLUSIONS: Cardiac emerinopathy is a novel nonsyndromic X-linked progressive atrial standstill associated with LVNC and increased risk of thromboembolism.


Subject(s)
Cardiomyopathies/genetics , Genetic Diseases, Inborn/genetics , Heart Atria/abnormalities , Heart Block/genetics , Isolated Noncompaction of the Ventricular Myocardium/genetics , Membrane Proteins/genetics , Mutation , Nuclear Proteins/genetics , Stroke/etiology , Thromboembolism/etiology , X-Linked Emery-Dreifuss Muscular Dystrophy/genetics , Adolescent , Adult , Aged , Cardiac Conduction System Disease/complications , Cardiac Conduction System Disease/diagnosis , Cardiac Conduction System Disease/genetics , Cardiomyopathies/complications , Cardiomyopathies/diagnosis , Child , Female , Genetic Diseases, Inborn/complications , Genetic Diseases, Inborn/diagnosis , Genetic Predisposition to Disease , Heart Block/complications , Heart Block/diagnosis , Humans , Isolated Noncompaction of the Ventricular Myocardium/complications , Isolated Noncompaction of the Ventricular Myocardium/diagnosis , Male , Middle Aged , Phenotype , Sick Sinus Syndrome/complications , Sick Sinus Syndrome/genetics , Stroke/diagnostic imaging , Thromboembolism/diagnostic imaging , X-Linked Emery-Dreifuss Muscular Dystrophy/complications , X-Linked Emery-Dreifuss Muscular Dystrophy/diagnosis , Young Adult
2.
Cell Physiol Biochem ; 54(4): 696-706, 2020 Jul 25.
Article in English | MEDLINE | ID: mdl-32706220

ABSTRACT

BACKGROUND/AIMS: Mutations of desmosomal genes are known to cause arrhythmogenic cardiomyopathy characterized by arrhythmias and sudden cardiac death. Previously, we described a novel genetic variant H1684R in desmoplakin gene (DSP), associated with a progressive cardiac conduction disease (PCCD). In the present study, we aimed to investigate an effect of the DSP-H1684R genetic variant on the activity of ion channels. METHODS: We used cardiomyocytes derived from induced pluripotent stem cells (iPSC cardiomyocytes) from a patient with DSP-H1684R genetic variant and from two healthy donors. Immunofluorescent staining and western blot analyses were used to characterize patient-specific cardiomyocytes. By the whole-cell voltage-clamp technique we estimated the activity of voltage-gated sodium, calcium, and potassium channels that are responsible for action potential generation and its shape. Action potentials' parameters were measured using whole-cell current-clamp technique. RESULTS: In patient-specific cardiomyocytes we observed both lower amplitudes of currents through sodium Nav1.5 channels and L-type calcium channels, but higher amplitude of current through transient-outward potassium channels in comparison to donor cardiomyocytes. Current-clamp measurements revealed shortening of action-potential in DSP-H1684R-carrying iPSC cardiomyocytes. Therefore, observed alterations in the channels activity might have a great impact on the properties of action potential and development of PCCD. CONCLUSION: Our results show that desmoplakin genetic variants, besides conduction slowing caused by structural heart remodeling, could affect multiple ion channel activity aggravating arrhythmia manifestation in PCCD.


Subject(s)
Cardiac Conduction System Disease/genetics , Desmoplakins/genetics , Heart Block/genetics , Induced Pluripotent Stem Cells/drug effects , Ion Channels/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Action Potentials/physiology , Calcium Channels/physiology , Cardiac Conduction System Disease/metabolism , Desmoplakins/metabolism , Fluorescent Antibody Technique , Heart Block/metabolism , Humans , Ion Channels/metabolism , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/physiology , Voltage-Gated Sodium Channels/physiology
3.
PLoS One ; 15(6): e0233007, 2020.
Article in English | MEDLINE | ID: mdl-32492036

ABSTRACT

BACKGROUND: In humans, stillbirth describes the death of a fetus before birth after 28 weeks gestation, and accounts for approximately 2.6 million deaths worldwide annually. In high-income countries, up to half of stillbirths have an unknown cause and are described as "unexplained stillbirths"; this lack of understanding impairs efforts to prevent stillbirth. There are also few animal models of stillbirth, but those that have been described usually have significant placental abnormalities. This study describes a novel mutant murine model of fetal death with atrial conduction block due to an ErbB2 missense mutation which is not associated with abnormal placental morphology. METHODS: Phenotypic characterisation and histological analysis of the mutant mouse model was conducted. The mRNA distribution of the early cardiomyocyte marker Nkx2-5 was assessed via in situ hybridisation. Cardiac structure was quantified and cellular morphology evaluated by electron microscopy. Immunostaining was employed to quantify placental structure and cell characteristics on matched heterozygous and homozygous mutant placental samples. RESULTS: There were no structural abnormalities observed in hearts of mutant embryos. Comparable Nkx2-5 expression was observed in hearts of mutants and controls, suggesting normal cardiac specification. Additionally, there was no significant difference in the weight, placenta dimensions, giant cell characteristics, labyrinth tissue composition, levels of apoptosis, proliferation or vascularisation between placentas of homozygous mutant mice and controls. CONCLUSION: Embryonic lethality in the ErbB2 homozygous mutant mouse cannot be attributed to placental pathology. As such, we conclude the ErbB2M802R mutant is a model of stillbirth with a non-placental cause of death. The mechanism of the atrial block resulting from ErbB2 mutation and its role in embryonic death is still unclear. Studying this mutant mouse model could identify candidate genes involved in stillbirth associated with structural or functional cardiac defects.


Subject(s)
Heart Defects, Congenital/genetics , Mutation, Missense , Receptor, ErbB-2/genetics , Stillbirth/genetics , Animals , Disease Models, Animal , Female , Heart Block/congenital , Heart Block/genetics , Heart Block/metabolism , Heart Block/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heterozygote , Homeobox Protein Nkx-2.5/genetics , Homozygote , Humans , Mice , Mice, Mutant Strains , Myocardium/metabolism , Myocardium/pathology , Placenta/abnormalities , Placenta/pathology , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Pediatrics ; 146(1)2020 07.
Article in English | MEDLINE | ID: mdl-32561613

ABSTRACT

Flecainide acetate is a Vaughan-Williams class IC antiarrhythmic drug prescribed for the treatment of supraventricular arrhythmias. It has a narrow therapeutic index and proarrhythmic effects even at therapeutic doses. Flecainide is metabolized by a CYP2D6 enzyme that exhibits polymorphism. In this case report, we present, to our best knowledge, the first case of toxicity contributed by genetic polymorphism in an infant. Our patient with recurrent supraventricular tachycardia was treated with a therapeutic dose of flecainide but developed heart block requiring extracorporeal membrane oxygenation support and subsequent treatment with lipid emulsion therapy. He was found to have supratherapeutic serum flecainide concentration, and gene testing revealed the patient to be an intermediate metabolizer. With this case report, we reinforce the importance of evaluating the CYP2D6 genotype before drug initiation in the neonatal population and recommend regular monitoring of serum flecainide levels and electrocardiograms in these patients.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Electrocardiography , Flecainide/adverse effects , Heart Block/chemically induced , Polymorphism, Genetic , Tachycardia, Supraventricular/drug therapy , Cytochrome P-450 CYP2D6/metabolism , Genotype , Heart Block/genetics , Heart Block/metabolism , Humans , Infant, Newborn , Tachycardia, Supraventricular/physiopathology , Voltage-Gated Sodium Channel Blockers/adverse effects
5.
Mol Med Rep ; 21(6): 2459-2465, 2020 06.
Article in English | MEDLINE | ID: mdl-32323820

ABSTRACT

Progressive cardiac conduction defect (PCCD) is an inherited autosomal dominant cardiac disorder characterized by an age­dependent cardiac electrical conduction block. Several genes have been associated with the genetic pathogenesis of PCCD. The present study aimed to identify the causal mutation of PCCD and to investigate the association between genotype and phenotype in a Chinese family with PCCD. A total of 39 family members were included in the present study. All subjects participated in physical, biochemical, electrocardiography and echocardiography examinations. Whole­exome sequencing was performed for four individuals from the same generation, including three patients with PCCD and one normal control with no cardiovascular disease. Sanger sequencing and in silico analysis were used to identify the causal mutation. Whole­exome sequencing and variant identification revealed a candidate nonsense mutation (c.1443C>A, p.Tyr481*) in lamin A/C (LMNA). The mutation was identified in seven patients (including the proband) and two asymptomatic mutation carriers, but it was not detected in 100 control subjects of matched ancestry. Clinical examinations identified typical symptoms in patients with PCCD, including bradycardia and various types of conduction defect, and excluded other phenotypes related to the LMNA mutation. The genotype and phenotype were co­associated among all participants. In the present study, the c.1443C>A mutation in the LMNA gene was identified as a potential cause of PCCD. In silico analysis predicted that the identified mutation was damaging through its effect on the lamin tail domain of LMNA. From the present study, it could be suggested that genetic screening and family counseling, early pacemaker implantation or a sudden death in the family may be essential for risk stratification and treatment of patients with PCCD.


Subject(s)
Cardiac Conduction System Disease/diagnosis , Heart Block/diagnosis , Lamin Type A/genetics , Adolescent , Adult , Aged , Base Sequence , Cardiac Conduction System Disease/genetics , Codon, Nonsense , Electrocardiography , Female , Genotype , Heart Block/genetics , Humans , Lamin Type A/chemistry , Middle Aged , Pedigree , Protein Structure, Tertiary , Exome Sequencing , Young Adult
6.
Stem Cell Res ; 43: 101720, 2020 03.
Article in English | MEDLINE | ID: mdl-32062131

ABSTRACT

Human iPSC cell lines (FAMRCi004-A and FAMRCi004-B) were generated from patient with progressive cardiac conduction disease and sick sinus syndrome carrying DSP p.His1684Arg genetic variant. Patient-specific adipose tissue-derived mesenchymal multipotent stromal cells were reprogrammed using non-integrative Sendai viruses. Established iPSC lines showed normal karyotype, expressed pluripotent markers and were able to differentiate toward three germ layers in vitro. The reported iPSC lines could be useful tool for in vitro modeling of progressive cardiac conduction disease associated with mutations in desmosomal genes.


Subject(s)
Cardiac Conduction System Disease/genetics , Desmoplakins/genetics , Heart Block/genetics , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation , Humans
7.
Cardiovasc Res ; 116(8): 1446-1457, 2020 07 01.
Article in English | MEDLINE | ID: mdl-31589297

ABSTRACT

AIMS: Investigating human heart development and applying this to deviations resulting in disease is incomplete without molecular characterization of the cell types required for normal functioning. We investigated foetal human heart single-cell transcriptomes from mid-gestational healthy and anti-SSA/Ro associated congenital heart block (CHB) samples. METHODS AND RESULTS: Three healthy foetal human hearts (19th to 22nd week of gestation) and one foetal heart affected by autoimmune-associated CHB (21st week of gestation) were subjected to enzymatic dissociation using the Langendorff preparation to obtain single-cell suspensions followed by 10× Genomics- and Illumina-based single-cell RNA-sequencing (scRNA-seq). In addition to the myocytes, fibroblasts, immune cells, and other minor cell types, previously uncharacterized diverse sub-populations of endothelial cells were identified in the human heart. Differential gene expression analysis revealed increased and heterogeneous interferon responses in varied cell types of the CHB heart compared with the healthy controls. In addition, we also identified matrisome transcripts enriched in CHB stromal cells that potentially contribute to extracellular matrix deposition and subsequent fibrosis. CONCLUSION: These data provide an information-rich resource to further our understanding of human heart development, which, as illustrated by comparison to a heart exposed to a maternal autoimmune environment, can be leveraged to provide insight into the pathogenesis of disease.


Subject(s)
Antibodies, Antinuclear/immunology , Autoimmunity , Fetal Heart/immunology , Fetal Heart/pathology , Heart Block/congenital , Transcriptome , Case-Control Studies , Gene Expression Profiling , Gestational Age , Heart Block/embryology , Heart Block/genetics , Heart Block/immunology , Heart Block/pathology , Humans , RNA-Seq , Single-Cell Analysis
9.
Gene ; 714: 143990, 2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31326550

ABSTRACT

BACKGROUND: Progressive cardiac conduction defect (PCCD), also known as Lenegre-Lev disease, is one of the most common heart conduction abnormalities. Previous studies have screened for known mutation sites that cause heart block in a 68-person family with a history of PCCD, revealed no mutations. OBJECTIVE: To screen pathogenic genes of the PCCD family and to study the function of the gene mutations related to heart block diseases. METHODS: Whole exome sequencing (WES) was performed on two PCCD patients and one non-PCCD family member to find the related pathogenic gene. After family co-segregation and preliminary functional analysis, we identified the mutant gene CLCA2. To study the function of this gene, we constructed mutant-gene mice using CRISPR-Cas9 technology, and electrocardiogram monitoring was performed after genotype verification. RESULTS: The CLCA2 c.G1725T mutation was identified and co-segregated with the phenotype. The analysis showed that the CLCA2 c.G1725T mutation is harmful and mainly affects protein glycosylation. Immunofluorescence staining revealed that CLCA2 was highly expressed in the sinoatrial node (SAN) tissues. Electrocardiogram monitoring of the mice revealed that CLCA2 point mutations induced mild conduction block and ectopic pacemakers. CONCLUSION: Our findings indicate that a novel heterozygous missense mutation c.G1725T of the CLCA2 gene may be associated with heart block disease and the mutation in this gene may lead to sinus node lesions and conduction blocking.


Subject(s)
Chloride Channels/genetics , Heart Block/genetics , Mutation, Missense/genetics , Amino Acid Sequence , Animals , Electrocardiography/methods , Female , Heterozygote , Humans , Male , Mice , Pedigree , Phenotype , Point Mutation/genetics , Sinoatrial Node/pathology
10.
Cardiol Rev ; 27(1): 23-33, 2019.
Article in English | MEDLINE | ID: mdl-29528859

ABSTRACT

Inherited primary arrhythmia syndromes are genetically determined disorders of cardiac ion channels or ion channel macromolecular complexes usually associated with a higher risk of sudden cardiac death. These conditions have a very broad spectrum of clinical manifestations, ranging from an asymptomatic course to syncope, atrial and ventricular arrhythmias, and conduction disturbances, but may produce sudden infant death syndrome and unexplained sudden cardiac death in apparently healthy individuals. During the last 20 years, the evolving knowledge on the genetic basis of inherited arrhythmia syndromes has dramatically reshaped our understanding of these conditions and, consequently, had a great impact on patient care. Based on the knowledge of the genetic substrates, specific risk factors for individual genotypes have been identified, and various investigations have been launched with the intention of developing a gene- and even mutation-specific therapy. Preliminary results from animal studies suggest that gene therapy rescues the normal ion channel function and thereby prevents cardiac events in some primary arrhythmia syndromes, which suggests that upon appropriate validation in a clinical setting, it may become available for affected patients. The purpose of this review is to provide clinicians with a contemporary insight into the role of genetic testing in the diagnosis, therapy, and prognosis of patients with primary arrhythmia syndromes, and the clinical implications of screening family members who are at risk of sudden cardiac death.


Subject(s)
Arrhythmias, Cardiac/diagnosis , Cardiac Conduction System Disease/diagnosis , Genetic Testing , Heart Block/diagnosis , Long QT Syndrome/diagnosis , Tachycardia, Ventricular/diagnosis , Arrhythmias, Cardiac/genetics , Cardiac Conduction System Disease/genetics , Heart Block/genetics , Humans , Long QT Syndrome/genetics , Tachycardia, Ventricular/genetics
12.
Europace ; 20(10): 1692-1698, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29579189

ABSTRACT

Aims: Cardiac atrial arrhythmias are the most common type of heart rhythm disorders. Its genetic elucidation remains challenging with poor understanding of cellular and molecular processes. These arrhythmias usually affect elderly population but in rare cases, young children may also suffer from such electrical diseases. Severe complications, including stroke, are commonly age related. This study aims to identify a genetic link between electro-mechanic atrial dysfunction and stroke in children. Methods and results: In two unrelated boys of 11 and 14 years with both stroke and atrial arrhythmias, the clinical phenotype was determined through a complete physical examination, electrocardiogram (ECG), Holter ECG, and computed tomography. The genetic testing was performed on a large 95 genes panel implicated in myocardial electrical imbalance, using the next generation sequencing method. The panel also includes the genes usually associated with the development of cardiomyopathies. In one child, a left atrial dilation was observed. The 2nd boy suffered from atrial standstill. Both suffered from atrial bradycardia, flutter, and fibrillation. The complete genetic testing revealed the SCN5A c.3823G>A (p.D1275N) mutation in the first family, c.1141-2A>G and c.3157G>A (p.E1053K) mutations in the second family. Conclusion: Our results strengthen the association between Nav1.5 mutations and the occurrence of stroke in young patients. It emphasizes the need to look for atrial myopathy in the decision process for anticoagulation in young patients with atrial arrhythmic events.


Subject(s)
Atrial Fibrillation/complications , Atrial Flutter/complications , Atrial Function, Left , Bradycardia/complications , Cardiomyopathies/complications , Genetic Diseases, Inborn/complications , Heart Atria/abnormalities , Heart Block/complications , Stroke/etiology , Adolescent , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Flutter/genetics , Atrial Flutter/physiopathology , Bradycardia/genetics , Bradycardia/physiopathology , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Child , Electrocardiography , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/physiopathology , Heart Atria/physiopathology , Heart Block/genetics , Heart Block/physiopathology , Humans , Male , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Phenotype
14.
Arthritis Rheumatol ; 69(11): 2170-2174, 2017 11.
Article in English | MEDLINE | ID: mdl-29045069

ABSTRACT

OBJECTIVE: Fetal exposure to maternal anti-SSA/Ro antibodies is necessary but not sufficient for the development of autoimmune congenital heart block (CHB), suggesting that other factors, such as fetal genetic predisposition, are important. Given the previously described association between major histocompatibility complex alleles and CHB risk, we undertook the present study to test the hypothesis that a variant form of HLA-C Asn80Lys, which binds with high affinity to an inhibitory killer cell immunoglobulin-like receptor (KIR) and thus renders natural killer (NK) cells incapable of restricting inflammation, contributes to the development of CHB. METHODS: Members of 192 pedigrees in the US and Europe (194 cases of CHB, 91 unaffected siblings, 152 fathers, 167 mothers) and 1,073 out-of-study controls were genotyped on the Immunochip single-nucleotide polymorphism microarray. Imputation was used to identify associations at HLA-C Asn80Lys (Asn, C1; Lys, C2) and KIR. Tests for association were performed using logistic regression. McNemar's test and the pedigree disequilibrium test (PDT) were used for matched analyses between affected and unaffected children. RESULTS: Compared with out-of-study controls of the same sex, the C2 allele was less frequent in the mothers (odds ratio [OR] 0.63, P = 0.0014) and more frequent in the fathers (OR 1.40, P = 0.0123), yielding a significant sex-by-C2 interaction (P = 0.0002). The C2 allele was more frequent in affected siblings than in unaffected siblings (OR 3.67, P = 0.0025), which was consistent with the PDT results (P = 0.016); these results were observed in both sexes and across the US and European cohorts. There was no difference in the frequency of the inhibitory KIR genotype (KIR AA) between affected and unaffected children (P = 0.55). CONCLUSION: These data establish C2 as a novel genetic risk factor associated with CHB. This observation supports a model in which fetuses with C2 ligand expression and maternal anti-SSA/Ro positivity may have impaired NK cell surveillance, resulting in unchecked cardiac inflammation and scarring.


Subject(s)
HLA-C Antigens/genetics , Heart Block/congenital , Antibodies, Antinuclear/immunology , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Europe , Fathers , Female , Genotype , Heart Block/genetics , Heart Block/immunology , Humans , Infant, Newborn , Logistic Models , Male , Mothers , Odds Ratio , Pedigree , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Sex Factors , Siblings , United States
15.
Biochem Biophys Res Commun ; 494(1-2): 346-351, 2017 12 09.
Article in English | MEDLINE | ID: mdl-28989025

ABSTRACT

Malfunction of nodal pacemaker (Pm) cardiomyocytes (CMs) due to diseases or aging leads to rhythm generation disorders, necessitating electronic Pm implantation. We functionally reprogrammed human pluripotent stem cell (hPSC) derived-ventricular (V) CMs into -PmCMs via recombinant adeno-associated virus serotype 9 (rAAV9)-mediated overexpression of engineered HCN1 channel (HCN1ΔΔΔ) whose S3-S4 linker has been strategically deleted by design to promote cardiac pacemaking. rAAV9-HCN1ΔΔΔ-reprogrammed hPSC-PmCMs converted from -VCMs showed automaticity and action potential parameters typical of native nodal PmCMs. Implantation of rAAV9-HCN1ΔΔΔ-based BPm in a preclinical porcine model of complete heart block significantly reduced the dependence on device-supported pacing and generated spontaneous heart rhythms from the BPm. Collectively, these results have further laid the groundwork on BPm for future translation.


Subject(s)
Dependovirus/metabolism , Heart Block/therapy , Heart Ventricles/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Potassium Channels/metabolism , Action Potentials/physiology , Animals , Cell Differentiation , Cellular Reprogramming , Dependovirus/genetics , Disease Models, Animal , Gene Expression , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Block/genetics , Heart Block/metabolism , Heart Block/physiopathology , Heart Rate/physiology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Myocytes, Cardiac/cytology , Pacemaker, Artificial , Pluripotent Stem Cells/cytology , Potassium Channels/genetics , Swine
16.
Am J Physiol Heart Circ Physiol ; 313(3): H631-H640, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28626076

ABSTRACT

The signature lesion of SSA/Ro autoantibody-associated congenital heart block (CHB) is fibrosis and a macrophage infiltrate, supporting an experimental focus on cues influencing the fibroblast component. The transcriptomes of human fetal cardiac fibroblasts were analyzed using two complementary approaches. Cardiac injury conditions were simulated in vitro by incubating human fetal cardiac fibroblasts with supernatants from macrophages transfected with the SSA/Ro-associated noncoding Y ssRNA. The top 10 upregulated transcripts in the stimulated fibroblasts reflected a type I interferon (IFN) response [e.g., IFN-induced protein 44-like (IFI44L), of MX dynamin-like GTPase (MX)1, MX2, and radical S-adenosyl methionine domain containing 2 (Rsad2)]. Within the fibrotic pathway, transcript levels of endothelin-1 (EDN1), phosphodiesterase (PDE)4D, chemokine (C-X-C motif) ligand (CXCL)2, and CXCL3 were upregulated, while others, including adenomedullin, RAP guanine nucleotide exchange factor 3 (RAPGEF3), tissue inhibitor of metalloproteinase (TIMP)1, TIMP3, and dual specificity phosphatase 1, were downregulated. Agnostic Database for Annotation, Visualization and Integrated Discovery analysis revealed a significant increase in inflammatory genes, including complement C3A receptor 1 (C3AR1), F2R-like thrombin/trypsin receptor 3, and neutrophil cytosolic factor 2. In addition, stimulated fibroblasts expressed high levels of phospho-MADS box transcription enhancer factor 2 [a substrate of MAPK5 (ERK5)], which was inhibited by BIX-02189, a specific inhibitor of ERK5. Translation to human disease leveraged an unprecedented opportunity to interrogate the transcriptome of fibroblasts freshly isolated and cell sorted without stimulation from a fetal heart with CHB and a matched healthy heart. Consistent with the in vitro data, five IFN response genes were among the top 10 most highly expressed transcripts in CHB fibroblasts. In addition, the expression of matrix-related genes reflected fibrosis. These data support the novel finding that cardiac injury in CHB may occur secondary to abnormal remodeling due in part to upregulation of type 1 IFN response genes.NEW & NOTEWORTHY Congenital heart block is a rare disease of the fetal heart associated with maternal anti-Ro autoantibodies which can result in death and for survivors, lifelong pacing. This study provides in vivo and in vitro transcriptome-support that injury may be mediated by an effect of Type I Interferon on fetal fibroblasts.


Subject(s)
Antibodies, Antinuclear/metabolism , Fetal Heart/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Heart Block/congenital , Inflammation Mediators/metabolism , Interferon Type I/metabolism , Transcriptome , Adult , Antibodies, Antinuclear/genetics , Antibodies, Antinuclear/immunology , Cells, Cultured , Culture Media, Conditioned/metabolism , Female , Fetal Heart/immunology , Fetal Heart/pathology , Fibroblasts/pathology , Fibrosis , Gene Expression Profiling/methods , Gene Expression Regulation , Heart Block/genetics , Heart Block/immunology , Heart Block/metabolism , Heart Block/pathology , Humans , Inflammation Mediators/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon Type I/immunology , Macrophages/immunology , Macrophages/metabolism , Myocardium , Paracrine Communication , Pregnancy , Transfection
17.
Horm Res Paediatr ; 87(5): 350-353, 2017.
Article in English | MEDLINE | ID: mdl-27631398

ABSTRACT

BACKGROUND: Myxedema coma (MC), a medical emergency defined as severe hypothyroidism leading to altered mental status, is more common in older women with hypothyroidism. METHODS/RESULTS: A 7-year-old Caucasian male with chromosome 1q deletion presented with altered mental status preceded by milestone regression. His presenting labs results were: thyroid-stimulating hormone (TSH) 0.501 µIU/ml and free thyroxine (T4) <0.5 ng/dl. His morning cortisol level was 8.1 µg/dl with repeat testing, while TSH was 1.119 µIU/ml and free T4 was 0.5 ng/dl. Low-dose cosyntropin test showed baseline and peak cortisol levels of 1.9 and 16 µg/dl, respectively. Aside from altered mental status, heart block was present in addition to hypothermia and hypercarbia. Diffuse cerebral cortical and corpus callosum atrophy were seen on MRI. An intravenous (i.v.) stress dose of hydrocortisone was administered for 24 h prior to an i.v. loading dose of levothyroxine. His activity level subsequently returned to baseline within 48 h after treatment had been initiated. CONCLUSION: Though MC is rare, occurring mainly with noncompliance in primary hypothyroidism, it may occur at the diagnosis of secondary hypothyroidism. Based on features like hypothermia, hypoventilation, and cardiovascular instability occurring in the setting of central hypothyroidism, it should be suspected and managed urgently in order to avert the associated high mortality resulting from treatment delays.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Congenital Hypothyroidism , Mental Disorders , Agenesis of Corpus Callosum/blood , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/pathology , Cerebral Cortex/abnormalities , Child , Congenital Hypothyroidism/blood , Congenital Hypothyroidism/genetics , Congenital Hypothyroidism/pathology , Corpus Callosum/pathology , Cortisone/administration & dosage , Cortisone/blood , Heart Block/blood , Heart Block/genetics , Heart Block/pathology , Humans , Hypothermia/blood , Hypothermia/genetics , Hypothermia/pathology , Male , Mental Disorders/blood , Mental Disorders/genetics , Mental Disorders/pathology , Thyrotropin/blood , Thyroxine/blood
18.
Dis Markers ; 2016: 3684965, 2016.
Article in English | MEDLINE | ID: mdl-28018021

ABSTRACT

Background. Congenital heart block is characterized by blockage of electrical impulses from the atrioventricular node (AV node) to the ventricles. This blockage can be caused by ion channel impairment that is the result of genetic variation. This study aimed to investigate the possible causative variants in a Thai family with complete heart block by using whole exome sequencing. Methods. Genomic DNA was collected from a family consisting of five family members in three generations in which one of three children in generation III had complete heart block. Whole exome sequencing was performed on one complete heart block affected child and one unaffected sibling. Bioinformatics was used to identify annotated and filtered variants. Candidate variants were validated and the segregation analysis of other family members was performed. Results. This study identified compound heterozygous variants, c.101G>A and c.3832G>A, in the SCN5A gene and c.28730C>T in the TTN gene. Conclusions. Compound heterozygous variants in the SCN5A gene were found in the complete heart block affected child but these two variants were found only in the this affected sibling and were not found in other unaffected family members. Hence, these variants in the SCN5A gene were the most possible disease-causing variants in this family.


Subject(s)
Heart Block/congenital , Heterozygote , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Adult , Child , Connectin/genetics , Exome , Female , Heart Block/genetics , Humans , Male , Middle Aged , Pedigree
19.
Chin Med J (Engl) ; 129(12): 1419-24, 2016 Jun 20.
Article in English | MEDLINE | ID: mdl-27270536

ABSTRACT

BACKGROUND: Kearns-Sayre syndrome (KSS) is a mitochondrial DNA (mtDNA) deletion disorder characterized by a triad of onset before 20 years of age, ophthalmoplegia, and pigmentary retinopathy. The heart and central nervous system are commonly involved. We summarized clinical and brain magnetic resonance imaging (MRI) features of a cohort of Chinese KSS patients. METHODS: Nineteen patients confirmed by muscle biopsy and mtDNA analysis were enrolled. We examined clinical profiles, mainly focusing on changes in electrocardiogram (ECG) and brain MRI. The correlation between genotype and phenotype was statistically analyzed. RESULTS: The mean age of onset was 9.6 ± 4.3 years, with all developing the classic triad at the time of diagnosis. Heart conduction block was detected in 63.2%, with four initially presenting as bundle branch block and developing into complete atrioventricular block over 3-72 months. Brain MRI showed symmetric high-T2 signals in 100% of cerebral and cerebellar white matter, as well as brainstem, 46.7% of basal ganglia, and 53.3% of thalamus. There were two patterns of cerebral white matter involvements, one with selective subcortical U-fibers and the other with periventricular white matter. The size of mtDNA deletion did not significantly correlate with age of onset or percentage of ragged blue fibers on muscle pathology. CONCLUSIONS: The clinical features of KSS evolve dynamically, affecting the cardiac conduction system predominantly, highlighting the significance of ECG monitoring. Brain MRI showed changes involving both the white matter and deep gray nuclei. Clinical presentation or severity of muscle pathological changes is not related to the size of mtDNA deletions.


Subject(s)
Brain/pathology , Kearns-Sayre Syndrome/diagnosis , Magnetic Resonance Imaging/methods , Adolescent , Brain/physiology , Child , Child, Preschool , DNA, Mitochondrial/genetics , Female , Genotype , Heart Block/diagnosis , Heart Block/genetics , Heart Block/physiopathology , Humans , Kearns-Sayre Syndrome/genetics , Kearns-Sayre Syndrome/physiopathology , Male
20.
Zhonghua Xin Xue Guan Bing Za Zhi ; 44(5): 411-5, 2016 May 24.
Article in Chinese | MEDLINE | ID: mdl-27220576

ABSTRACT

OBJECTIVE: To define the potential causative gene mutation in a Chinese pedigree with progressive cardiac conduction defect (PCCD). METHODS: Sanger sequencing was performed to define potential causative gene mutation in a four-generation family with 68 members including seven PCCD patients (5 male) from 2010 to 2015.No causative gene was detected by screening known candidate genes related to PCCD including SCN5A, NKX2.5 and LMNA.High-throughput sequencing technology on exon-enriched DNA was then used to search the causative genes in 2 patients and one normal family member. RESULTS: Eight new non-synonymous single nucleotide variants including AQP7 gene (exon5: c.T343C: p.Y115H), CACNA1B gene (NM_001243812: exon19: c.A2986G: p.T996A), CATSPERB gene (exon27: c.C3254G: p.P1085R), CLCA2 gene (exon11: c.G1725T: p.W575C), CLCA3P gene (ncRNA_intronic), MYLK-AS1 gene (ncRNA_intronic), TTN gene (ncRNA_UTR3), LMNA gene (LMNA: NM_170708: exon5: c.C922T: p.Q308X) were identified by comparing and filtering the results with known public databases.Then, more detailed biological analysis on these 8 genes was conducted.Traditional Sanger sequencing validated the exome sequencing results, and found that the mutation c. 1725Gï¹¥T in gene CLCA2 segregated with the phenotype of this PCCD pedigree.The mutation c. 1725Gï¹¥T in gene CLCA2 was thus be considered as the causative PCCD gene in this pedigree from the perspective of genetics and genomics. CONCLUSION: The heterozygote mutation c. 1725Gï¹¥T in gene CLCA2 might be causative gene in this PCCD pedigree.This finding adds new gene mutation variant responsible for PCCD.


Subject(s)
Chloride Channels/genetics , Heart Block/genetics , Asian People , Cardiac Conduction System Disease , China , DNA Mutational Analysis , Exons , Female , Humans , Male , Mutation , Pedigree , Phenotype , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...