Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(11)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37260361

RESUMEN

Human pluripotent stem cells (hPSCs), derived from individuals or genetically modified with disease-related mutations and variants, have revolutionised studies of human disease. Researchers are beginning to exploit the extraordinary potential of stem cell technology to screen for new drugs to treat intractable diseases, ideally without side-effects. However, a major problem is that the differentiated cell types on which these models are based are immature; they resemble fetal and not adult cells. Here, we discuss the nature and hurdles of hPSC maturation, using cardiomyocytes as an example. We review methods used to induce cardiomyocyte maturation in culture and consider remaining challenges for their integration into research on human disease and drug development pipelines.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Humanos , Miocitos Cardíacos/metabolismo , Diferenciación Celular
2.
Circ Res ; 132(7): 828-848, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36883446

RESUMEN

BACKGROUND: Signaling by cAMP is organized in multiple distinct subcellular nanodomains regulated by cAMP-hydrolyzing PDEs (phosphodiesterases). Cardiac ß-adrenergic signaling has served as the prototypical system to elucidate cAMP compartmentalization. Although studies in cardiac myocytes have provided an understanding of the location and properties of a handful of cAMP subcellular compartments, an overall view of the cellular landscape of cAMP nanodomains is missing. METHODS: Here, we combined an integrated phosphoproteomics approach that takes advantage of the unique role that individual PDEs play in the control of local cAMP, with network analysis to identify previously unrecognized cAMP nanodomains associated with ß-adrenergic stimulation. We then validated the composition and function of one of these nanodomains using biochemical, pharmacological, and genetic approaches and cardiac myocytes from both rodents and humans. RESULTS: We demonstrate the validity of the integrated phosphoproteomic strategy to pinpoint the location and provide critical cues to determine the function of previously unknown cAMP nanodomains. We characterize in detail one such compartment and demonstrate that the PDE3A2 isoform operates in a nuclear nanodomain that involves SMAD4 (SMAD family member 4) and HDAC-1 (histone deacetylase 1). Inhibition of PDE3 results in increased HDAC-1 phosphorylation, leading to inhibition of its deacetylase activity, derepression of gene transcription, and cardiac myocyte hypertrophic growth. CONCLUSIONS: We developed a strategy for detailed mapping of subcellular PDE-specific cAMP nanodomains. Our findings reveal a mechanism that explains the negative long-term clinical outcome observed in patients with heart failure treated with PDE3 inhibitors.


Asunto(s)
AMP Cíclico , Miocitos Cardíacos , Humanos , Proteómica , Hidrolasas Diéster Fosfóricas , Hipertrofia , Adrenérgicos
3.
Nat Rev Mol Cell Biol ; 13(11): 713-26, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23034453

RESUMEN

Worldwide increases in life expectancy have been paralleled by a greater prevalence of chronic and age-associated disorders, particularly of the cardiovascular, neural and metabolic systems. This has not been met by commensurate development of new drugs and therapies, which is in part owing to the difficulty in modelling human diseases in laboratory assays or experimental animals. Patient-specific induced pluripotent stem (iPS) cells are an emerging paradigm that may address this. Reprogrammed somatic cells from patients are already applied in disease modelling, drug testing and drug discovery, thus enabling researchers to undertake studies for treating diseases 'in a dish', which was previously inconceivable.


Asunto(s)
Células Madre Pluripotentes Inducidas , Modelos Biológicos , Envejecimiento , Animales , Bioingeniería , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/terapia , Diferenciación Celular , Células Cultivadas , Descubrimiento de Drogas , Humanos , Enfermedades Metabólicas/fisiopatología , Enfermedades Metabólicas/terapia , Miocitos Cardíacos/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Enfermedades Neurodegenerativas/terapia , Neuronas/fisiología
4.
Circ Res ; 128(6): 775-801, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33734815

RESUMEN

The ability of human pluripotent stem cells to form all cells of the body has provided many opportunities to study disease and produce cells that can be used for therapy in regenerative medicine. Even though beating cardiomyocytes were among the first cell types to be differentiated from human pluripotent stem cell, cardiac applications have advanced more slowly than those, for example, for the brain, eye, and pancreas. This is, in part, because simple 2-dimensional human pluripotent stem cell cardiomyocyte cultures appear to need crucial functional cues normally present in the 3-dimensional heart structure. Recent tissue engineering approaches combined with new insights into the dialogue between noncardiomyocytes and cardiomyocytes have addressed and provided solutions to issues such as cardiomyocyte immaturity and inability to recapitulate adult heart values for features like contraction force, electrophysiology, or metabolism. Three-dimensional bioengineered heart tissues are thus poised to contribute significantly to disease modeling, drug discovery, and safety pharmacology, as well as provide new modalities for heart repair. Here, we review the current status of 3-dimensional engineered heart tissues.


Asunto(s)
Corazón/fisiología , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Regeneración , Ingeniería de Tejidos/métodos , Animales , Técnicas de Reprogramación Celular/métodos , Humanos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(26): 15182-15192, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32554494

RESUMEN

The anthracycline doxorubicin (Doxo) and its analogs daunorubicin (Daun), epirubicin (Epi), and idarubicin (Ida) have been cornerstones of anticancer therapy for nearly five decades. However, their clinical application is limited by severe side effects, especially dose-dependent irreversible cardiotoxicity. Other detrimental side effects of anthracyclines include therapy-related malignancies and infertility. It is unclear whether these side effects are coupled to the chemotherapeutic efficacy. Doxo, Daun, Epi, and Ida execute two cellular activities: DNA damage, causing double-strand breaks (DSBs) following poisoning of topoisomerase II (Topo II), and chromatin damage, mediated through histone eviction at selected sites in the genome. Here we report that anthracycline-induced cardiotoxicity requires the combination of both cellular activities. Topo II poisons with either one of the activities fail to induce cardiotoxicity in mice and human cardiac microtissues, as observed for aclarubicin (Acla) and etoposide (Etop). Further, we show that Doxo can be detoxified by chemically separating these two activities. Anthracycline variants that induce chromatin damage without causing DSBs maintain similar anticancer potency in cell lines, mice, and human acute myeloid leukemia patients, implying that chromatin damage constitutes a major cytotoxic mechanism of anthracyclines. With these anthracyclines abstained from cardiotoxicity and therapy-related tumors, we thus uncoupled the side effects from anticancer efficacy. These results suggest that anthracycline variants acting primarily via chromatin damage may allow prolonged treatment of cancer patients and will improve the quality of life of cancer survivors.


Asunto(s)
Antineoplásicos/efectos adversos , Cromatina/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Doxorrubicina/efectos adversos , Animales , Línea Celular , Doxorrubicina/análogos & derivados , Doxorrubicina/síntesis química , Doxorrubicina/metabolismo , Doxorrubicina/uso terapéutico , Cardiopatías/inducido químicamente , Histonas , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones
6.
Nucleic Acids Res ; 48(2): 974-995, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31799604

RESUMEN

Genome editing typically involves recombination between donor nucleic acids and acceptor genomic sequences subjected to double-stranded DNA breaks (DSBs) made by programmable nucleases (e.g. CRISPR-Cas9). Yet, nucleases yield off-target mutations and, most pervasively, unpredictable target allele disruptions. Remarkably, to date, the untoward phenotypic consequences of disrupting allelic and non-allelic (e.g. pseudogene) sequences have received scant scrutiny and, crucially, remain to be addressed. Here, we demonstrate that gene-edited cells can lose fitness as a result of DSBs at allelic and non-allelic target sites and report that simultaneous single-stranded DNA break formation at donor and acceptor DNA by CRISPR-Cas9 nickases (in trans paired nicking) mostly overcomes such disruptive genotype-phenotype associations. Moreover, in trans paired nicking gene editing can efficiently and precisely add large DNA segments into essential and multiple-copy genomic sites. As shown herein by genotyping assays and high-throughput genome-wide sequencing of DNA translocations, this is achieved while circumventing most allelic and non-allelic mutations and chromosomal rearrangements characteristic of nuclease-dependent procedures. Our work demonstrates that in trans paired nicking retains target protein dosages in gene-edited cell populations and expands gene editing to chromosomal tracts previously not possible to modify seamlessly due to their recurrence in the genome or essentiality for cell function.


Asunto(s)
Sistemas CRISPR-Cas/genética , ADN/genética , Desoxirribonucleasa I/química , Edición Génica/métodos , Secuencia de Bases , ADN/química , Roturas del ADN de Doble Cadena , Roturas del ADN de Cadena Simple , Desoxirribonucleasa I/genética , Endonucleasas/química , Marcación de Gen/métodos , Genoma/genética , Humanos , Mutación/genética , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/genética
7.
Biochem Biophys Res Commun ; 572: 118-124, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34364290

RESUMEN

BACKGROUND: Human induced pluripotent stem cells (hiPSCs) and their derivative cardiomyocytes (hiPSC-CMs) have been successfully used to study the electrical phenotype of cardiac ion channel diseases. However, strategies to mature CMs and more comprehensive systems recapitulating the heart complexity are required to advance our ability to capture adult phenotypes. METHODS: We differentiated wild-type (WT) and long QT syndrome type 1 (LQT1) hiPSCs into CMs, endothelial cells and cardiac fibroblasts. The three cell types were combined to form three-dimensional (3D) spheroids, termed "cardiac microtissues" (cMTs) and the electrophysiological properties were measured using 96-well multi-electrode arrays. RESULTS: LQT1 cMTs displayed prolonged field potential duration compared to WT controls, thus recapitulating the typical feature of LQTS. Isoprenaline caused a positive chronotropic effect on both LQT1 and WT cMTs. The 96-well multi-electrode array format proved suitable to detect electrical changes directly in the 3D tissues. CONCLUSIONS: 3D hiPSC cMTs are a scalable tool that can be used to identify LQT electrical hallmarks and drug responses. We anticipate this tool can be adopted by pharmaceutical companies to screen cardio-active compounds.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Síndrome de QT Prolongado/metabolismo , Miocitos Cardíacos/metabolismo , Células Cultivadas , Humanos , Miocitos Cardíacos/citología , Fenotipo
8.
Development ; 144(6): 1008-1017, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28279973

RESUMEN

Cardiomyocytes and endothelial cells in the heart are in close proximity and in constant dialogue. Endothelium regulates the size of the heart, supplies oxygen to the myocardium and secretes factors that support cardiomyocyte function. Robust and predictive cardiac disease models that faithfully recapitulate native human physiology in vitro would therefore ideally incorporate this cardiomyocyte-endothelium crosstalk. Here, we have generated and characterized human cardiac microtissues in vitro that integrate both cell types in complex 3D structures. We established conditions for simultaneous differentiation of cardiomyocytes and endothelial cells from human pluripotent stem cells following initial cardiac mesoderm induction. The endothelial cells expressed cardiac markers that were also present in primary cardiac microvasculature, suggesting cardiac endothelium identity. These cell populations were further enriched based on surface markers expression, then recombined allowing development of beating 3D structures termed cardiac microtissues. This in vitro model was robustly reproducible in both embryonic and induced pluripotent stem cells. It thus represents an advanced human stem cell-based platform for cardiovascular disease modelling and testing of relevant drugs.


Asunto(s)
Diferenciación Celular , Células Endoteliales/citología , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Ingeniería de Tejidos/métodos , Antígenos CD34/metabolismo , Separación Celular , Fenómenos Electrofisiológicos , Humanos , Mesodermo/citología , Células Madre Pluripotentes/metabolismo , Sarcómeros/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
9.
Circ Res ; 122(3): e5-e16, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29282212

RESUMEN

RATIONALE: There are several methods to measure cardiomyocyte and muscle contraction, but these require customized hardware, expensive apparatus, and advanced informatics or can only be used in single experimental models. Consequently, data and techniques have been difficult to reproduce across models and laboratories, analysis is time consuming, and only specialist researchers can quantify data. OBJECTIVE: Here, we describe and validate an automated, open-source software tool (MUSCLEMOTION) adaptable for use with standard laboratory and clinical imaging equipment that enables quantitative analysis of normal cardiac contraction, disease phenotypes, and pharmacological responses. METHODS AND RESULTS: MUSCLEMOTION allowed rapid and easy measurement of movement from high-speed movies in (1) 1-dimensional in vitro models, such as isolated adult and human pluripotent stem cell-derived cardiomyocytes; (2) 2-dimensional in vitro models, such as beating cardiomyocyte monolayers or small clusters of human pluripotent stem cell-derived cardiomyocytes; (3) 3-dimensional multicellular in vitro or in vivo contractile tissues, such as cardiac "organoids," engineered heart tissues, and zebrafish and human hearts. MUSCLEMOTION was effective under different recording conditions (bright-field microscopy with simultaneous patch-clamp recording, phase contrast microscopy, and traction force microscopy). Outcomes were virtually identical to the current gold standards for contraction measurement, such as optical flow, post deflection, edge-detection systems, or manual analyses. Finally, we used the algorithm to quantify contraction in in vitro and in vivo arrhythmia models and to measure pharmacological responses. CONCLUSIONS: Using a single open-source method for processing video recordings, we obtained reliable pharmacological data and measures of cardiac disease phenotype in experimental cell, animal, and human models.


Asunto(s)
Contracción Miocárdica , Miocitos Cardíacos/fisiología , Programas Informáticos , Algoritmos , Animales , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/fisiopatología , Fármacos Cardiovasculares/farmacología , Diferenciación Celular , Células Cultivadas , Subunidades beta de la Proteína de Unión al GTP/deficiencia , Subunidades beta de la Proteína de Unión al GTP/genética , Humanos , Síndrome de QT Prolongado/patología , Síndrome de QT Prolongado/fisiopatología , Masculino , Microscopía/métodos , Modelos Cardiovasculares , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp , Fenotipo , Células Madre Pluripotentes/citología , Conejos , Grabación en Video , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
10.
Int J Mol Sci ; 20(16)2019 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-31426283

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is an inherited heart disease characterized by sudden death in young people and featured by fibro-adipose myocardium replacement, malignant arrhythmias, and heart failure. To date, no etiological therapies are available. Mutations in desmosomal genes cause abnormal mechanical coupling, trigger pro-apoptotic signaling pathways, and induce fibro-adipose replacement. Here, we discuss the hypothesis that the ACM causative mechanism involves a defect in the expression and/or activity of the cardiac Ca2+ handling machinery, focusing on the available data supporting this hypothesis. The Ca2+ toolkit is heavily remodeled in cardiomyocytes derived from a mouse model of ACM defective of the desmosomal protein plakophilin-2. Furthermore, ACM-related mutations were found in genes encoding for proteins involved in excitation‒contraction coupling, e.g., type 2 ryanodine receptor and phospholamban. As a consequence, the sarcoplasmic reticulum becomes more eager to release Ca2+, thereby inducing delayed afterdepolarizations and impairing cardiac contractility. These data are supported by preliminary observations from patient induced pluripotent stem-cell-derived cardiomyocytes. Assessing the involvement of Ca2+ signaling in the pathogenesis of ACM could be beneficial in the treatment of this life-threatening disease.


Asunto(s)
Arritmias Cardíacas/patología , Calcio/metabolismo , Cardiomiopatías/patología , Desmosomas/patología , Miocitos Cardíacos/patología , Animales , Arritmias Cardíacas/metabolismo , Señalización del Calcio , Cardiomiopatías/metabolismo , Desmosomas/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Placofilinas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
11.
Int J Mol Sci ; 19(11)2018 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-30428582

RESUMEN

Loss-of-function long QT (LQT) mutations inducing LQT1 and LQT2 syndromes have been successfully translated to human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) used as disease-specific models. However, their in vitro investigation mainly relies on experiments using small numbers of cells. This is especially critical when working with cells as heterogeneous as hiPSC-CMs. We aim (i) to investigate in silico the ionic mechanisms underlying LQT1 and LQT2 hiPSC-CM phenotypic variability, and (ii) to enable massive in silico drug tests on mutant hiPSC-CMs. We combined (i) data of control and mutant slow and rapid delayed rectifying K⁺ currents, IKr and IKs respectively, (ii) a recent in silico hiPSC-CM model, and (iii) the population of models paradigm to generate control and mutant populations for LQT1 and LQT2 cardiomyocytes. Our four populations contain from 1008 to 3584 models. In line with the experimental in vitro data, mutant in silico hiPSC-CMs showed prolonged action potential (AP) duration (LQT1: +14%, LQT2: +39%) and large electrophysiological variability. Finally, the mutant populations were split into normal-like hiPSC-CMs (with action potential duration similar to control) and at risk hiPSC-CMs (with clearly prolonged action potential duration). At risk mutant hiPSC-CMs carried higher expression of L-type Ca2+, lower expression of IKr and increased sensitivity to quinidine as compared to mutant normal-like hiPSC-CMs, resulting in AP abnormalities. In conclusion, we were able to reproduce the two most common LQT syndromes with large-scale simulations, which enable investigating biophysical mechanisms difficult to assess in vitro, e.g., how variations of ion current expressions in a physiological range can impact on AP properties of mutant hiPSC-CMs.


Asunto(s)
Arritmias Cardíacas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de QT Prolongado/genética , Miocitos Cardíacos/metabolismo , Potenciales de Acción/fisiología , Arritmias Cardíacas/genética , Fenómenos Electrofisiológicos/genética , Fenómenos Electrofisiológicos/fisiología , Humanos , Mutación/genética , Técnicas de Placa-Clamp
12.
EMBO J ; 32(24): 3161-75, 2013 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-24213244

RESUMEN

Patient-specific induced pluripotent stem cells (iPSCs) will assist research on genetic cardiac maladies if the disease phenotype is recapitulated in vitro. However, genetic background variations may confound disease traits, especially for disorders with incomplete penetrance, such as long-QT syndromes (LQTS). To study the LQT2-associated c.A2987T (N996I) KCNH2 mutation under genetically defined conditions, we derived iPSCs from a patient carrying this mutation and corrected it. Furthermore, we introduced the same point mutation in human embryonic stem cells (hESCs), generating two genetically distinct isogenic pairs of LQTS and control lines. Correction of the mutation normalized the current (IKr) conducted by the HERG channel and the action potential (AP) duration in iPSC-derived cardiomyocytes (CMs). Introduction of the same mutation reduced IKr and prolonged the AP duration in hESC-derived CMs. Further characterization of N996I-HERG pathogenesis revealed a trafficking defect. Our results demonstrated that the c.A2987T KCNH2 mutation is the primary cause of the LQTS phenotype. Precise genetic modification of pluripotent stem cells provided a physiologically and functionally relevant human cellular context to reveal the pathogenic mechanism underlying this specific disease phenotype.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Síndrome de QT Prolongado/genética , Mutación , Células Madre Pluripotentes , Potenciales de Acción/genética , Adulto , Células Cultivadas , Canal de Potasio ERG1 , Células Madre Embrionarias/fisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Femenino , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Humanos , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Fenotipo , Células Madre Pluripotentes/fisiología , Transporte de Proteínas/genética , Factores de Transcripción/genética
13.
Proc Natl Acad Sci U S A ; 111(50): E5383-92, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25453094

RESUMEN

Jervell and Lange-Nielsen syndrome (JLNS) is one of the most severe life-threatening cardiac arrhythmias. Patients display delayed cardiac repolarization, associated high risk of sudden death due to ventricular tachycardia, and congenital bilateral deafness. In contrast to the autosomal dominant forms of long QT syndrome, JLNS is a recessive trait, resulting from homozygous (or compound heterozygous) mutations in KCNQ1 or KCNE1. These genes encode the α and ß subunits, respectively, of the ion channel conducting the slow component of the delayed rectifier K(+) current, IKs. We used complementary approaches, reprogramming patient cells and genetic engineering, to generate human induced pluripotent stem cell (hiPSC) models of JLNS, covering splice site (c.478-2A>T) and missense (c.1781G>A) mutations, the two major classes of JLNS-causing defects in KCNQ1. Electrophysiological comparison of hiPSC-derived cardiomyocytes (CMs) from homozygous JLNS, heterozygous, and wild-type lines recapitulated the typical and severe features of JLNS, including pronounced action and field potential prolongation and severe reduction or absence of IKs. We show that this phenotype had distinct underlying molecular mechanisms in the two sets of cell lines: the previously unidentified c.478-2A>T mutation was amorphic and gave rise to a strictly recessive phenotype in JLNS-CMs, whereas the missense c.1781G>A lesion caused a gene dosage-dependent channel reduction at the cell membrane. Moreover, adrenergic stimulation caused action potential prolongation specifically in JLNS-CMs. Furthermore, sensitivity to proarrhythmic drugs was strongly enhanced in JLNS-CMs but could be pharmacologically corrected. Our data provide mechanistic insight into distinct classes of JLNS-causing mutations and demonstrate the potential of hiPSC-CMs in drug evaluation.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Síndrome de Jervell-Lange Nielsen/tratamiento farmacológico , Síndrome de Jervell-Lange Nielsen/genética , Síndrome de Jervell-Lange Nielsen/fisiopatología , Canal de Potasio KCNQ1/genética , Modelos Biológicos , Fenotipo , Potenciales de Acción/fisiología , Análisis de Varianza , Secuencia de Bases , Línea Celular , Genes Recesivos/genética , Ingeniería Genética , Humanos , Técnicas In Vitro , Canal de Potasio KCNQ1/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense/genética , Miocitos Cardíacos/fisiología , Análisis de Secuencia de ADN
14.
Biochem Biophys Res Commun ; 467(4): 998-1005, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26456652

RESUMEN

One limitation in using human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) for disease modeling and cardiac safety pharmacology is their immature functional phenotype compared with adult cardiomyocytes. Here, we report that treatment of human embryonic stem cell derived cardiomyocytes (hESC-CMs) with dexamethasone, a synthetic glucocorticoid, activated glucocorticoid signaling which in turn improved their calcium handling properties and contractility. L-type calcium current and action potential properties were not affected by dexamethasone but significantly faster calcium decay, increased forces of contraction and sarcomeric lengths, were observed in hESC-CMs after dexamethasone exposure. Activating the glucocorticoid pathway can thus contribute to mediating hPSC-CMs maturation.


Asunto(s)
Calcio/metabolismo , Dexametasona/farmacología , Células Madre Embrionarias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Línea Celular , Dexametasona/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal
15.
Proc Natl Acad Sci U S A ; 109(29): 11866-71, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22745159

RESUMEN

Long QT syndrome (LQTS) is a genetic disease characterized by a prolonged QT interval in an electrocardiogram (ECG), leading to higher risk of sudden cardiac death. Among the 12 identified genes causal to heritable LQTS, ∼90% of affected individuals harbor mutations in either KCNQ1 or human ether-a-go-go related genes (hERG), which encode two repolarizing potassium currents known as I(Ks) and I(Kr). The ability to quantitatively assess contributions of different current components is therefore important for investigating disease phenotypes and testing effectiveness of pharmacological modulation. Here we report a quantitative analysis by simulating cardiac action potentials of cultured human cardiomyocytes to match the experimental waveforms of both healthy control and LQT syndrome type 1 (LQT1) action potentials. The quantitative evaluation suggests that elevation of I(Kr) by reducing voltage sensitivity of inactivation, not via slowing of deactivation, could more effectively restore normal QT duration if I(Ks) is reduced. Using a unique specific chemical activator for I(Kr) that has a primary effect of causing a right shift of V(1/2) for inactivation, we then examined the duration changes of autonomous action potentials from differentiated human cardiomyocytes. Indeed, this activator causes dose-dependent shortening of the action potential durations and is able to normalize action potentials of cells of patients with LQT1. In contrast, an I(Kr) chemical activator of primary effects in slowing channel deactivation was not effective in modulating action potential durations. Our studies provide both the theoretical basis and experimental support for compensatory normalization of action potential duration by a pharmacological agent.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Activación del Canal Iónico/fisiología , Canales de Potasio KCNQ/metabolismo , Síndrome de QT Prolongado/genética , Modelos Biológicos , Acetamidas/farmacología , Animales , Células CHO , Células Cultivadas , Simulación por Computador , Cricetinae , Cricetulus , Cartilla de ADN/genética , Humanos , Canales de Potasio KCNQ/fisiología , Síndrome de QT Prolongado/patología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Piridinas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Bloqueadores de los Canales de Sodio/farmacología
16.
N Engl J Med ; 363(15): 1397-409, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20660394

RESUMEN

BACKGROUND: Long-QT syndromes are heritable diseases associated with prolongation of the QT interval on an electrocardiogram and a high risk of sudden cardiac death due to ventricular tachyarrhythmia. In long-QT syndrome type 1, mutations occur in the KCNQ1 gene, which encodes the repolarizing potassium channel mediating the delayed rectifier I(Ks) current. METHODS: We screened a family affected by long-QT syndrome type 1 and identified an autosomal dominant missense mutation (R190Q) in the KCNQ1 gene. We obtained dermal fibroblasts from two family members and two healthy controls and infected them with retroviral vectors encoding the human transcription factors OCT3/4, SOX2, KLF4, and c-MYC to generate pluripotent stem cells. With the use of a specific protocol, these cells were then directed to differentiate into cardiac myocytes. RESULTS: Induced pluripotent stem cells maintained the disease genotype of long-QT syndrome type 1 and generated functional myocytes. Individual cells showed a "ventricular," "atrial," or "nodal" phenotype, as evidenced by the expression of cell-type­specific markers and as seen in recordings of the action potentials in single cells. The duration of the action potential was markedly prolonged in "ventricular" and "atrial" cells derived from patients with long-QT syndrome type 1, as compared with cells from control subjects. Further characterization of the role of the R190Q­KCNQ1 mutation in the pathogenesis of long-QT syndrome type 1 revealed a dominant negative trafficking defect associated with a 70 to 80% reduction in I(Ks) current and altered channel activation and deactivation properties. Moreover, we showed that myocytes derived from patients with long-QT syndrome type 1 had an increased susceptibility to catecholamine-induced tachyarrhythmia and that beta-blockade attenuated this phenotype. CONCLUSIONS: We generated patient-specific pluripotent stem cells from members of a family affected by long-QT syndrome type 1 and induced them to differentiate into functional cardiac myocytes. The patient-derived cells recapitulated the electrophysiological features of the disorder. (Funded by the European Research Council and others.)


Asunto(s)
Potenciales de Acción , Células Madre Pluripotentes Inducidas/fisiología , Canal de Potasio KCNQ1/genética , Miocitos Cardíacos/citología , Síndrome de Romano-Ward/fisiopatología , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Adulto , Anciano , Cardiotónicos/farmacología , Niño , Femenino , Fibroblastos/citología , Expresión Génica , Humanos , Isoproterenol/farmacología , Factor 4 Similar a Kruppel , Masculino , Mutación Missense , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Linaje , Fenotipo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome de Romano-Ward/tratamiento farmacológico , Síndrome de Romano-Ward/genética
20.
Biomolecules ; 13(10)2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37892123

RESUMEN

Recent technical breakthroughs in genotyping and bioinformatics techniques have greatly facilitated the translation of genomics into clinical care [...].


Asunto(s)
Biología Computacional , Genómica , Humanos , Genómica/métodos , Enfermedades Raras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA