Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 11296, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37438479

ABSTRACT

Familial hypertrophic cardiomyopathy (FHC) patients are advised to avoid strenuous exercise due to increased risk of arrhythmias. Mice expressing the human FHC-causing mutation R403Q in the myosin heavy chain gene (MYH6) recapitulate the human phenotype, including cytoskeletal disarray and increased arrhythmia susceptibility. Following in vivo administration of isoproterenol, mutant mice exhibited tachyarrhythmias, poor recovery and fatigue. Arrhythmias were attenuated with the ß-blocker atenolol and protein kinase A inhibitor PKI. Mutant cardiac myocytes had significantly prolonged action potentials and triggered automaticity due to reduced repolarization reserve and connexin 43 expression. Isoproterenol shortened cycle length, and escalated electrical instability. Surprisingly isoproterenol did not increase CaV1.2 current. We found alterations in CaV1.2-ß1 adrenergic receptor colocalization assessed using super-resolution nanoscopy, and increased CaV1.2 phosphorylation in mutant hearts. Our results reveal for the first time that altered ion channel expression, co-localization and ß-adrenergic receptor signaling associated with myocyte disarray contribute to electrical instability in the R403Q mutant heart.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial , Cardiomyopathy, Hypertrophic , Humans , Animals , Mice , Isoproterenol , Cardiomyopathy, Hypertrophic/genetics , Arrhythmias, Cardiac , Heart
2.
Stem Cell Res ; 71: 103153, 2023 09.
Article in English | MEDLINE | ID: mdl-37385135

ABSTRACT

Variants in the sodium voltage-gated channel alpha subunit 5 gene (SCN5A) produce variable cardiac phenotypes including Brugada syndrome, conduction disease and cardiomyopathy. These phenotypes can lead to life-threatening arrhythmias, heart failure, and sudden cardiac death. Novel variants in splice-site regions of SCN5A require functional studies to characterise their pathogenicity as they are poorly understood. The generation of an induced pluripotent stem cell line provides a valuable resource to investigate the functional effects of potential splice-disrupting variants in SCN5A.


Subject(s)
Induced Pluripotent Stem Cells , Ventricular Fibrillation , Humans , Ventricular Fibrillation/genetics , Induced Pluripotent Stem Cells/metabolism , Cardiac Conduction System Disease , Arrhythmias, Cardiac , NAV1.5 Voltage-Gated Sodium Channel/genetics , Sodium/metabolism , Mutation
3.
Circ Genom Precis Med ; 14(2): e003202, 2021 04.
Article in English | MEDLINE | ID: mdl-33657327

ABSTRACT

BACKGROUND: Transcriptome sequencing can improve genetic diagnosis of Mendelian diseases but requires access to tissue expressing disease-relevant transcripts. We explored genetic testing of hypertrophic cardiomyopathy using transcriptome sequencing of patient-specific human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs). We also explored whether antisense oligonucleotides (AOs) could inhibit aberrant mRNA splicing in hiPSC-CMs. METHODS: We derived hiPSC-CMs from patients with hypertrophic cardiomyopathy due to MYBPC3 splice-gain variants, or an unresolved genetic cause. We used transcriptome sequencing of hiPSC-CM RNA to identify pathogenic splicing and used AOs to inhibit this splicing. RESULTS: Transcriptome sequencing of hiPSC-CMs confirmed aberrant splicing in 2 people with previously identified MYBPC3 splice-gain variants (c.1090+453C>T and c.1224-52G>A). In a patient with an unresolved genetic cause of hypertrophic cardiomyopathy following genome sequencing, transcriptome sequencing of hiPSC-CMs revealed diverse cryptic exon splicing due to an MYBPC3 c.1928-569G>T variant, and this was confirmed in cardiac tissue from an affected sibling. Antisense oligonucleotide treatment demonstrated almost complete inhibition of cryptic exon splicing in one patient-specific hiPSC-CM line. CONCLUSIONS: Transcriptome sequencing of patient specific hiPSC-CMs solved a previously undiagnosed genetic cause of hypertrophic cardiomyopathy and may be a useful adjunct approach to genetic testing. Antisense oligonucleotide inhibition of cryptic exon splicing is a potential future personalized therapeutic option.


Subject(s)
Cardiomyopathy, Hypertrophic/pathology , Carrier Proteins/genetics , Transcriptome , Cardiomyopathy, Hypertrophic/genetics , Carrier Proteins/metabolism , Genotype , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/pharmacology , Polymorphism, Genetic , RNA Splicing/drug effects , Exome Sequencing
4.
Int J Mol Sci ; 21(23)2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33256189

ABSTRACT

Retinoblastoma binding protein 9 (RBBP9) is required for maintaining the expression of both pluripotency and cell cycle genes in human pluripotent stem cells (hPSCs). An siRNA-based study from our group showed it does so by influencing cell cycle progression through the RB/E2F pathway. In non-pluripotent cells, RBBP9 is also known to have serine hydrolase (SH) activity, acting on currently undefined target proteins. The role of RBBP9 SH activity in hPSCs, and during normal development, is currently unknown. To begin assessing whether RBBP9 SH activity might contribute to hPSC maintenance, hPSCs were treated with ML114-a selective chemical inhibitor of RBBP9 SH activity. Stem cells treated with ML114 showed significantly reduced population growth rate, colony size and progression through the cell cycle, with no observable change in cell morphology or decrease in pluripotency antigen expression-suggesting no initiation of hPSC differentiation. Consistent with this, hPSCs treated with ML114 retained the capacity for tri-lineage differentiation, as seen through teratoma formation. Subsequent microarray and Western blot analyses of ML114-treated hPSCs suggest the nuclear transcription factor Y subunit A (NFYA) may be a candidate effector of RBBP9 SH activity in hPSCs. These data support a role for RBBP9 in regulating hPSC proliferation independent of differentiation, whereby inhibition of RBBP9 SH activity de-couples decreased hPSC proliferation from initiation of differentiation.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Differentiation , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Pluripotent Stem Cells/cytology , Serine Proteinase Inhibitors/pharmacology , Biomarkers/metabolism , CCAAT-Binding Factor/metabolism , Cell Count , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colony-Forming Units Assay , Genomic Instability , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Karyotype , Neoplasm Proteins/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
5.
Stem Cell Res ; 37: 101450, 2019 05.
Article in English | MEDLINE | ID: mdl-31039485

ABSTRACT

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an arrhythmia syndrome characterized by adrenaline induced ventricular tachycardia. The primary genetic aetiologies underlying CPVT are either autosomal dominant or autosomal recessive inheritance, resulting from heterozygous mutations in cardiac ryanodine receptor (RYR2) and homozygous mutations in cardiac calsequestrin-2 (CASQ2), respectively. Recently, a large family with autosomal dominant CPVT due to a heterozygous mutation in CASQ2, p.Lys180Arg, was reported. This resource is the first induced pluripotent stem cell line generated from a patient with autosomal dominant CPVT due to a heterozygous mutation in CASQ2. Induced pluripotent stem cells were generated from the whole blood of a 40-year-old woman with severe CPVT who is heterozygous for the p.Lys180Arg CASQ2 mutation. Induced pluripotent stem cell (iPSC) characterization confirmed expression of pluripotency makers, trilineage differentiation potential, and the absence of exogenous pluripotency vector expression.


Subject(s)
Calsequestrin/genetics , Cell Differentiation , Cellular Reprogramming , Induced Pluripotent Stem Cells/pathology , Mutation , Myocytes, Cardiac/pathology , Tachycardia, Ventricular/genetics , Adult , Cells, Cultured , Female , Genes, Dominant , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Phenotype , Tachycardia, Ventricular/pathology
6.
Stem Cell Res ; 33: 269-273, 2018 12.
Article in English | MEDLINE | ID: mdl-30508693

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is an inherited cardiomyopathy characterized by left ventricular hypertrophy ≥15 mm in the absence of loading conditions. HCM has a prevalence of up to one in 200, and can result in significant adverse outcomes including heart failure and sudden cardiac death. An induced pluripotent stem cell (iPSC) line was generated from peripheral blood mononuclear cells obtained from the whole blood of a 38-year-old female patient with HCM in which genetic testing identified the well-known pathogenic p.Arg403Gln mutation in myosin heavy chain 7. iPSCs express pluripotency markers, demonstrate trilineage differentiation capacity, and display a normal 46,XX female karyotype. This resource will allow further assessment of the pathophysiological development of HCM.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Induced Pluripotent Stem Cells/metabolism , Myosin Heavy Chains/genetics , Adult , Cell Differentiation , Female , Humans , Mutation
7.
Stem Cell Res ; 33: 56-59, 2018 12.
Article in English | MEDLINE | ID: mdl-30316040

ABSTRACT

Hypertrophic cardiomyopathy is an inherited cardiomyopathy with a prevalence of up to 1 in 200, which can result in significant morbidity and mortality. An iPSC line was generated from peripheral blood mononuclear cells obtained from the whole blood of a 58-year-old male with hypertrophic cardiomyopathy who carries the heterozygous pathogenic myosin binding protein C mutation p.Arg502Trp. Induced pluripotent stem cells express pluripotency markers, demonstrate trilineage differentiation potential, and display a normal karyotype. This line is a useful resource for studying and modeling hypertrophic cardiomyopathy. Resource table.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Carrier Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , Cardiomyopathy, Hypertrophic/pathology , Humans , Male , Middle Aged , Mutation
8.
Development ; 145(1)2018 01 09.
Article in English | MEDLINE | ID: mdl-29217756

ABSTRACT

Cataracts cause vision loss and blindness by impairing the ability of the ocular lens to focus light onto the retina. Various cataract risk factors have been identified, including drug treatments, age, smoking and diabetes. However, the molecular events responsible for these different forms of cataract are ill-defined, and the advent of modern cataract surgery in the 1960s virtually eliminated access to human lenses for research. Here, we demonstrate large-scale production of light-focusing human micro-lenses from spheroidal masses of human lens epithelial cells purified from differentiating pluripotent stem cells. The purified lens cells and micro-lenses display similar morphology, cellular arrangement, mRNA expression and protein expression to human lens cells and lenses. Exposing the micro-lenses to the emergent cystic fibrosis drug Vx-770 reduces micro-lens transparency and focusing ability. These human micro-lenses provide a powerful and large-scale platform for defining molecular disease mechanisms caused by cataract risk factors, for anti-cataract drug screening and for clinically relevant toxicity assays.


Subject(s)
Aminophenols/adverse effects , Cataract/chemically induced , Cataract/metabolism , Lens, Crystalline/metabolism , Models, Biological , Pluripotent Stem Cells/metabolism , Quinolones/adverse effects , Aminophenols/pharmacology , Cataract/pathology , Humans , Lens, Crystalline/pathology , Pluripotent Stem Cells/pathology , Quinolones/pharmacology
9.
Anal Bioanal Chem ; 407(9): 2543-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25680633

ABSTRACT

Chitosan, being antimicrobial and biocompatible, is attractive as a cell growth substrate. To improve cell attachment, arginine-glycine-aspartic acid-serine (RGDS) peptides were covalently grafted to chitosan films, through the widely used coupling agents 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC-HCl) and N-hydroxysuccinimide (NHS), via the carboxylic acid function of the RGDS molecule. The grafting reaction was monitored, for the first time, in real time using free-solution capillary electrophoresis (CE). This enabled fast separation and determination of the peptide and all other reactants in one separation with no sample preparation. Covalent RGDS peptide grafting onto the chitosan film surface was demonstrated using solid-state NMR of swollen films. CE indicated that oligomers of RGDS, not simply RGDS, were grafted on the film, with a likely hyperbranched structure. To assess the functional properties of the grafted films, cell growth was compared on control and peptide-grafted chitosan films. Light microscopy and polymerase chain reaction (PCR) analysis demonstrated greatly improved cell attachment to RGDS-grafted chitosan films.


Subject(s)
Biocompatible Materials/chemical synthesis , Chitosan/chemistry , Electrophoresis, Capillary/methods , Peptides/chemistry , Biocompatible Materials/chemistry , Cell Adhesion , Cell Line , Cell Proliferation , Cell Survival , Cells/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...