Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Int J Mol Sci ; 23(17)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36077200

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene and dilated cardiomyopathy (DCM) is a major cause of morbidity and mortality in DMD patients. We tested the hypothesis that DCM is caused by metabolic impairments by employing induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) generated from four DMD patients; an adult male, an adult female, a 7-year-old (7y) male and a 13-year-old (13y) male, all compared to two healthy volunteers. To test the hypothesis, we measured the bioenergetics, metabolomics, electrophysiology, mitochondrial morphology and mitochondrial activity of CMs, using respirometry, LC-MS, patch clamp, electron microscopy (EM) and confocal microscopy methods. We found that: (1) adult DMD CMs exhibited impaired energy metabolism and abnormal mitochondrial structure and function. (2) The 7y CMs demonstrated arrhythmia-free spontaneous firing along with "healthy-like" metabolic status, normal mitochondrial morphology and activity. In contrast, the 13y CMs were mildly arrhythmogenic and showed adult DMD-like bioenergetics deficiencies. (3) In DMD adult CMs, mitochondrial activities were attenuated by 45-48%, whereas the 7y CM activity was similar to that of healthy CMs. (4) In DMD CMs, but not in 7y CMs, there was a 75% decrease in the mitochondrial ATP production rate compared to healthy iPSC-CMs. In summary, DMD iPSC-CMs exhibit bioenergetic and metabolic impairments that are associated with rhythm disturbances corresponding to the patient's phenotype, thereby constituting novel targets for alleviating cardiomyopathy in DMD patients.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Cardiomyopathy, Dilated/metabolism , Cell Differentiation , Dystrophin/genetics , Energy Metabolism , Female , Humans , Male , Muscular Dystrophy, Duchenne/genetics , Myocytes, Cardiac/metabolism
2.
J Cell Mol Med ; 25(8): 3922-3934, 2021 04.
Article in English | MEDLINE | ID: mdl-33619882

ABSTRACT

Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is an X-linked disease affecting male and rarely adult heterozygous females, resulting in death by the late 20s to early 30s. Previous studies reported depressed left ventricular function in DMD patients which may result from deranged intracellular Ca2+ -handling. To decipher the mechanism(s) underlying the depressed LV function, we tested the hypothesis that iPSC-CMs generated from DMD patients feature blunted positive inotropic response to ß-adrenergic stimulation. To test the hypothesis, [Ca2+ ]i transients and contractions were recorded from healthy and DMD-CMs. While in healthy CMs (HC) isoproterenol caused a prominent positive inotropic effect, DMD-CMs displayed a blunted inotropic response. Next, we tested the functionality of the sarcoplasmic reticulum (SR) by measuring caffeine-induced Ca2+ release. In contrast to HC, DMD-CMs exhibited reduced caffeine-induced Ca2+ signal amplitude and recovery time. In support of the depleted SR Ca2+ stores hypothesis, in DMD-CMs the negative inotropic effects of ryanodine and cyclopiazonic acid were smaller than in HC. RNA-seq analyses demonstrated that in DMD CMs the RNA-expression levels of specific subunits of the L-type calcium channel, the ß1-adrenergic receptor (ADRß1) and adenylate cyclase were down-regulated by 3.5-, 2.8- and 3-fold, respectively, which collectively contribute to the depressed ß-adrenergic responsiveness.


Subject(s)
Adrenergic Agents/pharmacology , Calcium/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/pathology , Muscular Dystrophy, Duchenne/pathology , Myocardial Contraction , Myocytes, Cardiac/pathology , Adult , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Differentiation , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA-Seq , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology
3.
Int J Mol Sci ; 22(15)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34360639

ABSTRACT

LMNA-related dilated cardiomyopathy is an inherited heart disease caused by mutations in the LMNA gene encoding for lamin A/C. The disease is characterized by left ventricular enlargement and impaired systolic function associated with conduction defects and ventricular arrhythmias. We hypothesized that LMNA-mutated patients' induced Pluripotent Stem Cell-derived cardiomyocytes (iPSC-CMs) display electrophysiological abnormalities, thus constituting a suitable tool for deciphering the arrhythmogenic mechanisms of the disease, and possibly for developing novel therapeutic modalities. iPSC-CMs were generated from two related patients (father and son) carrying the same E342K mutation in the LMNA gene. Compared to control iPSC-CMs, LMNA-mutated iPSC-CMs exhibited the following electrophysiological abnormalities: (1) decreased spontaneous action potential beat rate and decreased pacemaker current (If) density; (2) prolonged action potential duration and increased L-type Ca2+ current (ICa,L) density; (3) delayed afterdepolarizations (DADs), arrhythmias and increased beat rate variability; (4) DADs, arrhythmias and cessation of spontaneous firing in response to ß-adrenergic stimulation and rapid pacing. Additionally, compared to healthy control, LMNA-mutated iPSC-CMs displayed nuclear morphological irregularities and gene expression alterations. Notably, KB-R7943, a selective inhibitor of the reverse-mode of the Na+/Ca2+ exchanger, blocked the DADs in LMNA-mutated iPSC-CMs. Our findings demonstrate cellular electrophysiological mechanisms underlying the arrhythmias in LMNA-related dilated cardiomyopathy.


Subject(s)
Arrhythmias, Cardiac/pathology , Calcium/metabolism , Cardiomyopathy, Dilated/pathology , Induced Pluripotent Stem Cells/pathology , Lamin Type A/genetics , Mutation , Myocytes, Cardiac/pathology , Action Potentials , Adult , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cell Differentiation , Electrophysiological Phenomena , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged , Myocytes, Cardiac/metabolism , Pedigree
4.
Int J Mol Sci ; 21(19)2020 Oct 03.
Article in English | MEDLINE | ID: mdl-33023024

ABSTRACT

: Over the years, numerous groups have employed human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) as a superb human-compatible model for investigating the function and dysfunction of cardiomyocytes, drug screening and toxicity, disease modeling and for the development of novel drugs for heart diseases. In this review, we discuss the broad use of iPSC-CMs for drug development and disease modeling, in two related themes. In the first theme-drug development, adverse drug reactions, mechanisms of cardiotoxicity and the need for efficient drug screening protocols-we discuss the critical need to screen old and new drugs, the process of drug development, marketing and Adverse Drug reactions (ADRs), drug-induced cardiotoxicity, safety screening during drug development, drug development and patient-specific effect and different mechanisms of ADRs. In the second theme-using iPSC-CMs for disease modeling and developing novel drugs for heart diseases-we discuss the rationale for using iPSC-CMs and modeling acquired and inherited heart diseases with iPSC-CMs.


Subject(s)
Cardiotoxicity/diagnosis , Heart Diseases/drug therapy , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Cardiotoxicity/drug therapy , Cell Differentiation/drug effects , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions , Heart Diseases/pathology , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology
5.
J Cell Mol Med ; 23(3): 2125-2135, 2019 03.
Article in English | MEDLINE | ID: mdl-30618214

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked progressive muscle degenerative disease, caused by mutations in the dystrophin gene and resulting in death because of respiratory or cardiac failure. To investigate the cardiac cellular manifestation of DMD, we generated induced pluripotent stem cells (iPSCs) and iPSC-derived cardiomyocytes (iPSC-CMs) from two DMD patients: a male and female manifesting heterozygous carrier. Dystrophin mRNA and protein expression were analysed by qRT-PCR, RNAseq, Western blot and immunofluorescence staining. For comprehensive electrophysiological analysis, current and voltage clamp were used to record transmembrane action potentials and ion currents, respectively. Microelectrode array was used to record extracellular electrograms. X-inactive specific transcript (XIST) and dystrophin expression analyses revealed that female iPSCs underwent X chromosome reactivation (XCR) or erosion of X chromosome inactivation, which was maintained in female iPSC-CMs displaying mixed X chromosome expression of wild type (WT) and mutated alleles. Both DMD female and male iPSC-CMs presented low spontaneous firing rate, arrhythmias and prolonged action potential duration. DMD female iPSC-CMs displayed increased beat rate variability (BRV). DMD male iPSC-CMs manifested decreased If density, and DMD female and male iPSC-CMs showed increased ICa,L density. Our findings demonstrate cellular mechanisms underlying electrophysiological abnormalities and cardiac arrhythmias in DMD.


Subject(s)
Heterozygote , Induced Pluripotent Stem Cells/physiology , Muscular Dystrophy, Duchenne/physiopathology , Myocytes, Cardiac/physiology , Action Potentials/genetics , Adult , Cell Differentiation/genetics , Dystrophin/genetics , Dystrophin/metabolism , Electrophysiological Phenomena , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Male , Microscopy, Electron, Transmission , Middle Aged , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure
6.
Methods Mol Biol ; 2454: 643-655, 2022.
Article in English | MEDLINE | ID: mdl-34128204

ABSTRACT

Induced pluripotent stem cells (iPSCs) were originally derived from adult somatic cells by ectopic expression of the stem cell transcription factors OCT3/4, SOX2, c-Myc, and KLF4. The characteristic features of iPSCs are similar to those of embryonic stem cells; they can be expanded indefinitely in vitro and differentiated into the three germ layers: endoderm, mesoderm, and ectoderm. The breakthrough discovery by Takahashi and Yamanaka that somatic cells can be "reprogrammed" to generate iPSCs has led to extensive use of iPSCs and their differentiated cells thereof, in diverse research areas, such as regenerative medicine, development, as well as establishment of disease-specific models, thus providing the platform for personalized patient-specific medicine.


Subject(s)
Induced Pluripotent Stem Cells , Adult , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/genetics , Embryonic Stem Cells , Endoderm , Humans , Induced Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL