Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Circ Res ; 134(9): 1160-1178, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38662861

RESUMEN

Heavy metals are harmful environmental pollutants that have attracted widespread attention due to their health hazards to human cardiovascular disease. Heavy metals, including lead, cadmium, mercury, arsenic, and chromium, are found in various sources such as air, water, soil, food, and industrial products. Recent research strongly suggests a connection between cardiovascular disease and exposure to toxic heavy metals. Epidemiological, basic, and clinical studies have revealed that heavy metals can promote the production of reactive oxygen species, which can then exacerbate reactive oxygen species generation and induce inflammation, resulting in endothelial dysfunction, lipid metabolism distribution, disruption of ion homeostasis, and epigenetic changes. Over time, heavy metal exposure eventually results in an increased risk of hypertension, arrhythmia, and atherosclerosis. Strengthening public health prevention and the application of chelation or antioxidants, such as vitamins and beta-carotene, along with minerals, such as selenium and zinc, can diminish the burden of cardiovascular disease attributable to metal exposure.


Asunto(s)
Enfermedades Cardiovasculares , Exposición a Riesgos Ambientales , Metales Pesados , Humanos , Metales Pesados/toxicidad , Metales Pesados/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/epidemiología , Exposición a Riesgos Ambientales/efectos adversos , Animales , Estrés Oxidativo/efectos de los fármacos , Contaminantes Ambientales/efectos adversos , Contaminantes Ambientales/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes
2.
Circ Res ; 124(1): 66-78, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30582453

RESUMEN

RATIONALE: Short QT syndrome (SQT) is a rare but arrhythmogenic disorder featured by shortened ventricular repolarization and a propensity toward life-threatening ventricular arrhythmias and sudden cardiac death. OBJECTIVE: This study aimed to investigate the single-cell mechanism of SQT using patient-specific and gene-corrected induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). METHODS AND RESULTS: One SQT patient carrying missense mutation T618I in potassium voltage-gated channel subfamily H member 2 ( KCNH2) was recruited as well as 2 healthy control subjects in this study. Control and SQT patient-specific iPSCs were generated from skin fibroblasts using nonintegrated Sendai virus. The KCNH2 T618I mutation was corrected by genome editing in SQT iPSC lines to generate isogenic controls. All iPSCs were differentiated into iPSC-CMs using monolayer-based differentiation protocol. SQT iPSC-CMs exhibited abnormal action potential phenotype featured by shortened action potential duration and increased beat-beat interval variability, when compared with control and gene-corrected iPSC-CMs. Furthermore, SQT iPSC-CMs showed KCNH2 gain-of-function with increased rapid delayed rectifying potassium current (IKr) density and enhanced membrane expression. Gene expression profiling of iPSC-CMs exhibited a differential cardiac ion-channel gene expression profile of SQT. Moreover, QTc of SQT patient and action potential durations of SQT iPSC-CMs were both normalized by quinidine, indicating that quinidine is beneficial to KCNH2 T618I of SQT. Importantly, shortened action potential duration phenotype observed in SQT iPSC-CMs was effectively rescued by a short-peptide scorpion toxin BmKKx2 with a mechanism of targeting KCNH2. CONCLUSIONS: We demonstrate that patient-specific and gene-corrected iPSC-CMs are able to recapitulate single-cell phenotype of SQT, which is caused by the gain-of-function mutation KCNH2 T618I. These findings will help elucidate the mechanisms underlying SQT and discover therapeutic drugs for treating the disease by using peptide toxins as lead compounds.


Asunto(s)
Potenciales de Acción/genética , Arritmias Cardíacas/genética , Canal de Potasio ERG1/genética , Mutación con Ganancia de Función , Edición Génica/métodos , Frecuencia Cardíaca/genética , Células Madre Pluripotentes Inducidas/metabolismo , Mutación Missense , Miocitos Cardíacos/metabolismo , Análisis de la Célula Individual/métodos , Adulto , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Sistemas CRISPR-Cas , Estudios de Casos y Controles , Línea Celular , Linaje de la Célula , Canal de Potasio ERG1/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Factores de Tiempo
3.
J Mol Cell Cardiol ; 142: 53-64, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32251671

RESUMEN

Diabetes mellitus is a serious metabolic condition associated with a multitude of cardiovascular complications. Moreover, the prevalence of diabetes in heart failure populations is higher than that in control populations. However, the role of cardiomyocyte alterations in type 2 diabetes mellitus (T2DM) has not been well characterized and the underlying mechanisms remain elusive. In this study, two patients who were diagnosed as T2DM were recruited and patient-specific induced pluripotent stem cells (iPSCs) were generated from urine epithelial cells using nonintegrated Sendai virus. The iPSC lines derived from five healthy subjects were used as controls. All iPSCs were differentiated into cardiomyocytes (iPSC-CMs) using the monolayer-based differentiation protocol. T2DM iPSC-CMs exhibited various disease phenotypes, including cellular hypertrophy and lipid accumulation. Moreover, T2DM iPSC-CMs exhibited higher susceptibility to high-glucose/high-lipid challenge than control iPSC-CMs, manifesting an increase in apoptosis. RNA-Sequencing analysis revealed a differential transcriptome profile and abnormal activation of TGFß signaling pathway in T2DM iPSC-CMs. We went on to show that inhibition of TGFß significantly rescued the hypertrophic phenotype in T2DM iPSC-CMs. In conclusion, we demonstrate that the iPSC-CM model is able to recapitulate cellular phenotype of T2DM. Our results indicate that iPSC-CMs can therefore serve as a suitable model for investigating molecular mechanisms underlying diabetic cardiomyopathies and for screening therapeutic drugs.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Apoptosis/genética , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Biomarcadores , Estudios de Casos y Controles , Diferenciación Celular/genética , Células Cultivadas , Diabetes Mellitus Tipo 2/etiología , Células Epiteliales/metabolismo , Glucosa/metabolismo , Humanos , Inmunofenotipificación , Células Madre Pluripotentes Inducidas/citología , Metabolismo de los Lípidos , Miocitos Cardíacos/citología , Transcriptoma
4.
Plant Mol Biol ; 102(3): 271-285, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31838617

RESUMEN

KEY MESSAGE: H2 prolonged the vase life and improved the vase quality of cut roses through repressing endogenous ethylene production and alleviating ethylene signal transduction during the entire senescing period. Recently, the application of hydrogen gas (H2) was shown to improve postharvest quality and longevity in perishable horticultural products, but the specific regulation mechanism remains obscure. Here, endogenous ethylene production and the expression of genes in ethylene biosynthesis and signalling pathway were investigated to explore the crosstalk between H2 and ethylene during the senescence of cut roses. Our results revealed that addition of exogenous ethylene by ethephon accelerated the senescence of cut roses, in which 100 mg L-1 ethephon displayed the most obvious senescent phenotype. While the applied different concentrations (1%, 10%, 50% and 100%) of hydrogen-rich water (HRW) conducted different affects in alleviating the senescence of cut roses, and 1% HRW displayed the best ornamental quality and the longest vase life by reducing ethylene production, supported by the decrease of 1-aminocyclopropene-1-carboxylate (ACC) accumulation, ACC synthase (ACS) and ACC oxidase (ACO) activities, and Rh-ACS3 and Rh-ACO1 expressions in ethylene biosynthesis. In addition, HRW increased the transcripts of ethylene receptor genes Rh-ETR1 at blooming period from day 4 to day 6 and suppressed Rh-ETR3 at senescence phase at day 8 after harvest. Furthermore, the relevant affection of HRW on Rh-ETR1 and Rh-ETR3 expressions still existed when the ethylene production was compromised by adequate addition of exogenous ethylene in HRW-treated cut rose petals, and HRW directly repressed the protein level of Rh-ETR3 in a transient expression assay. Overall, the results suggested that H2 is involved in neutralizing ethylene-mediated postharvest in cut flowers.


Asunto(s)
Etilenos/antagonistas & inhibidores , Etilenos/biosíntesis , Flores/efectos de los fármacos , Hidrógeno/farmacología , Rosa/efectos de los fármacos , Rosa/metabolismo , Aminoácidos Cíclicos/metabolismo , Flores/enzimología , Flores/genética , Flores/crecimiento & desarrollo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Hidrógeno/metabolismo , Liasas/genética , Liasas/metabolismo , Compuestos Organofosforados , Fenotipo , Reguladores del Crecimiento de las Plantas , Proteínas de Plantas/genética , Receptores de Superficie Celular/genética , Rosa/enzimología , Rosa/genética , Transducción de Señal
5.
J Mol Cell Cardiol ; 126: 143-154, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30423318

RESUMEN

Cardiac hypertrophy is an adaptive response against increased workload featuring by an increase in left ventricular mass and a thickening left ventricle wall. Here, we showed the expression of transient receptor potential canonical 1 (TRPC1) is higher in hearts of patients with hypertrophic cardiomyopathy (HCM) or heart failure (HF) than that of normal hearts. To better understand the mechanisms of TRPC1 in regulating cellular hypertrophy of human-based cardiomyocytes, we generated human pluripotent stem cell lines of TRPC1 knockout by CRISPR/Cas9. We demonstrated that knockout of TRPC1 significantly attenuated cardiomyocyte hypertrophy phenotype induced by phorbol 12-myristate 13-acetate, which was associated with abnormal activation of NF-κB. In contrast, overexpression of TRPC1 induced cardiomyocyte hypertrophy, which can be reversed by inhibition of NF-κB. Taken together, we established a stable human-based cardiomyocyte hypertrophy model and highlighted molecular mechanisms underlying TRPC1-mediated hypertrophy, aiding the development of therapeutic drugs for HCM and HF by targeting TRPC1.


Asunto(s)
Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Células Madre Pluripotentes/metabolismo , Transducción de Señal , Canales Catiónicos TRPC/antagonistas & inhibidores , Secuencia de Bases , Cardiomegalia/genética , Cardiomegalia/patología , Humanos , Canales Catiónicos TRPC/metabolismo
6.
J Cell Mol Med ; 22(9): 4221-4235, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29993192

RESUMEN

Cadmium, a highly ubiquitous toxic heavy metal, has been widely recognized as an environmental and industrial pollutant, which confers serious threats to human health. The molecular mechanisms of the cadmium-induced cardiotoxicity (CIC) have not been studied in human cardiomyocytes at the cellular level. Here we showed that human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) can recapitulate the CIC at the cellular level. The cadmium-treated hPSC-CMs exhibited cellular phenotype including reduced cell viability, increased apoptosis, cardiac sarcomeric disorganization, elevated reactive oxygen species, altered action potential profile and cardiac arrhythmias. RNA-sequencing analysis revealed a differential transcriptome profile and activated MAPK signalling pathway in cadmium-treated hPSC-CMs, and suppression of P38 MAPK but not ERK MAPK or JNK MAPK rescued CIC phenotype. We further identified that suppression of PI3K/Akt signalling pathway is sufficient to reverse the CIC phenotype, which may play an important role in CIC. Taken together, our data indicate that hPSC-CMs can serve as a suitable model for the exploration of molecular mechanisms underlying CIC and for the discovery of CIC cardioprotective drugs.


Asunto(s)
Cloruro de Cadmio/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Cloruro de Cadmio/antagonistas & inhibidores , Cardiotoxicidad/genética , Cardiotoxicidad/metabolismo , Cardiotoxicidad/prevención & control , Diferenciación Celular/efectos de los fármacos , Línea Celular , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Imidas/farmacología , Insulina/farmacología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Morfolinas/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Quinolinas/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Circulation ; 127(16): 1677-91, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23519760

RESUMEN

BACKGROUND: Cardiotoxicity is a leading cause for drug attrition during pharmaceutical development and has resulted in numerous preventable patient deaths. Incidents of adverse cardiac drug reactions are more common in patients with preexisting heart disease than the general population. Here we generated a library of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from patients with various hereditary cardiac disorders to model differences in cardiac drug toxicity susceptibility for patients of different genetic backgrounds. METHODS AND RESULTS: Action potential duration and drug-induced arrhythmia were measured at the single cell level in hiPSC-CMs derived from healthy subjects and patients with hereditary long QT syndrome, familial hypertrophic cardiomyopathy, and familial dilated cardiomyopathy. Disease phenotypes were verified in long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy hiPSC-CMs by immunostaining and single cell patch clamp. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and the human ether-a-go-go-related gene expressing human embryonic kidney cells were used as controls. Single cell PCR confirmed expression of all cardiac ion channels in patient-specific hiPSC-CMs as well as hESC-CMs, but not in human embryonic kidney cells. Disease-specific hiPSC-CMs demonstrated increased susceptibility to known cardiotoxic drugs as measured by action potential duration and quantification of drug-induced arrhythmias such as early afterdepolarizations and delayed afterdepolarizations. CONCLUSIONS: We have recapitulated drug-induced cardiotoxicity profiles for healthy subjects, long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy patients at the single cell level for the first time. Our data indicate that healthy and diseased individuals exhibit different susceptibilities to cardiotoxic drugs and that use of disease-specific hiPSC-CMs may predict adverse drug responses more accurately than the standard human ether-a-go-go-related gene test or healthy control hiPSC-CM/hESC-CM screening assays.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica Familiar/genética , Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Predisposición Genética a la Enfermedad , Células Madre Pluripotentes Inducidas/citología , Síndrome de QT Prolongado/genética , Miocitos Cardíacos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica Familiar/patología , Diferenciación Celular , Línea Celular/efectos de los fármacos , Línea Celular/fisiología , Tamaño de la Célula , Cisaprida/toxicidad , Cuerpos Embrioides/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Perfilación de la Expresión Génica , Células HEK293/efectos de los fármacos , Células HEK293/fisiología , Humanos , Técnicas In Vitro , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Riñón/citología , Riñón/embriología , Síndrome de QT Prolongado/patología , Miocitos Cardíacos/fisiología , Miofibrillas/ultraestructura , Nicorandil/toxicidad , Técnicas de Placa-Clamp , Quinazolinas/toxicidad , Verapamilo/toxicidad
8.
Circulation ; 128(11 Suppl 1): S3-13, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24030418

RESUMEN

BACKGROUND: Drug-induced arrhythmia is one of the most common causes of drug development failure and withdrawal from market. This study tested whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with a low-impedance microelectrode array (MEA) system could improve on industry-standard preclinical cardiotoxicity screening methods, identify the effects of well-characterized drugs, and elucidate underlying risk factors for drug-induced arrhythmia. hiPSC-CMs may be advantageous over immortalized cell lines because they possess similar functional characteristics as primary human cardiomyocytes and can be generated in unlimited quantities. METHODS AND RESULTS: Pharmacological responses of beating embryoid bodies exposed to a comprehensive panel of drugs at 65 to 95 days postinduction were determined. Responses of hiPSC-CMs to drugs were qualitatively and quantitatively consistent with the reported drug effects in literature. Torsadogenic hERG blockers, such as sotalol and quinidine, produced statistically and physiologically significant effects, consistent with patch-clamp studies, on human embryonic stem cell-derived cardiomyocytes hESC-CMs. False-negative and false-positive hERG blockers were identified accurately. Consistent with published studies using animal models, early afterdepolarizations and ectopic beats were observed in 33% and 40% of embryoid bodies treated with sotalol and quinidine, respectively, compared with negligible early afterdepolarizations and ectopic beats in untreated controls. CONCLUSIONS: We found that drug-induced arrhythmias can be recapitulated in hiPSC-CMs and documented with low impedance MEA. Our data indicate that the MEA/hiPSC-CM assay is a sensitive, robust, and efficient platform for testing drug effectiveness and for arrhythmia screening. This system may hold great potential for reducing drug development costs and may provide significant advantages over current industry standard assays that use immortalized cell lines or animal models.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Fármacos Cardiovasculares/efectos adversos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Análisis de Matrices Tisulares/métodos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Adolescente , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Impedancia Eléctrica , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Microelectrodos , Miocitos Cardíacos/fisiología
9.
Nat Commun ; 15(1): 7000, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143095

RESUMEN

Mutations in the nuclear envelope (NE) protein lamin A/C (encoded by LMNA), cause a severe form of dilated cardiomyopathy (DCM) with early-onset life-threatening arrhythmias. However, molecular mechanisms underlying increased arrhythmogenesis in LMNA-related DCM (LMNA-DCM) remain largely unknown. Here we show that a frameshift mutation in LMNA causes abnormal Ca2+ handling, arrhythmias and disformed NE in LMNA-DCM patient-specific iPSC-derived cardiomyocytes (iPSC-CMs). Mechanistically, lamin A interacts with sirtuin 1 (SIRT1) where mutant lamin A/C accelerates degradation of SIRT1, leading to mitochondrial dysfunction and oxidative stress. Elevated reactive oxygen species (ROS) then activates the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-ryanodine receptor 2 (RYR2) pathway and aggravates the accumulation of SUN1 in mutant iPSC-CMs, contributing to arrhythmias and NE deformation, respectively. Taken together, the lamin A/C deficiency-mediated ROS disorder is revealed as central to LMNA-DCM development. Manipulation of impaired SIRT1 activity and excessive oxidative stress is a potential future therapeutic strategy for LMNA-DCM.


Asunto(s)
Cardiomiopatía Dilatada , Células Madre Pluripotentes Inducidas , Lamina Tipo A , Miocitos Cardíacos , Estrés Oxidativo , Especies Reactivas de Oxígeno , Sirtuina 1 , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Lamina Tipo A/metabolismo , Lamina Tipo A/genética , Células Madre Pluripotentes Inducidas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Sirtuina 1/metabolismo , Sirtuina 1/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fenotipo , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Mutación del Sistema de Lectura , Calcio/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Membrana Nuclear/metabolismo , Mitocondrias/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética
10.
Stem Cell Res ; 72: 103202, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37708613

RESUMEN

The transient receptor potential vanilloid subfamily 1 (TRPV1) is a polymodal nociceptor that is highly expressed in sensory nerves. Activation of TRPV1 receptors excites primary afferent nociceptors by opening cation channels, allowing the influx of Na+ and Ca2+ ions into the cytoplasm. Here, a TRPV1 knockout human embryonic stem cell line was generated using the CRISPR/Cas9 genome-editing technology to further study the function of TRPV1. The cell line confirmed with normal pluripotency and karyotype.


Asunto(s)
Células Madre Embrionarias Humanas , Células Madre Pluripotentes , Humanos , Sistemas CRISPR-Cas/genética , Nociceptores/metabolismo , Línea Celular , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Embrionarias Humanas/metabolismo
11.
Stem Cell Res Ther ; 14(1): 241, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37679791

RESUMEN

BACKGROUND: Mutations in the cardiac sodium channel gene SCN5A cause Brugada syndrome (BrS), an arrhythmic disorder that is a leading cause of sudden death and lacks effective treatment. An association between SCN5A and Wnt/ß-catenin signaling has been recently established. However, the role of Wnt/ß-catenin signaling in BrS and underlying mechanisms remains unknown. METHODS: Three healthy control subjects and one BrS patient carrying a novel frameshift mutation (T1788fs) in the SCN5A gene were recruited in this study. Control and BrS patient-specific induced pluripotent stem cells (iPSCs) were generated from skin fibroblasts using nonintegrated Sendai virus. All iPSCs were differentiated into cardiomyocytes using monolayer-based differentiation protocol. Action potentials and sodium currents were recorded from control and BrS iPSC-derived cardiomyocytes (iPSC-CMs) by single-cell patch clamp. RESULTS: BrS iPSC-CMs exhibited increased burden of arrhythmias and abnormal action potential profile featured by slower depolarization, decreased action potential amplitude, and increased beating interval variation. Moreover, BrS iPSC-CMs showed cardiac sodium channel (Nav1.5) loss-of-function as compared to control iPSC-CMs. Interestingly, the electrophysiological abnormalities and Nav1.5 loss-of-function observed in BrS iPSC-CMs were accompanied by aberrant activation of Wnt/ß-catenin signaling. Notably, inhibition of Wnt/ß-catenin significantly rescued Nav1.5 defects and arrhythmic phenotype in BrS iPSC-CMs. Mechanistically, SCN5A-encoded Nav1.5 interacts with ß-catenin, and reduced expression of Nav1.5 leads to re-localization of ß-catenin in BrS iPSC-CMs, which aberrantly activates Wnt/ß-catenin signaling to suppress SCN5A transcription. CONCLUSIONS: Our findings suggest that aberrant activation of Wnt/ß-catenin signaling contributes to the pathogenesis of SCN5A-related BrS and point to Wnt/ß-catenin as a potential therapeutic target.


Asunto(s)
Síndrome de Brugada , Células Madre Pluripotentes Inducidas , Humanos , Síndrome de Brugada/genética , Miocitos Cardíacos , beta Catenina/genética
12.
EBioMedicine ; 95: 104741, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37544203

RESUMEN

BACKGROUND: Brugada syndrome (BrS) is a cardiac channelopathy that can result in sudden cardiac death (SCD). SCN5A is the most frequent gene linked to BrS, but the genotype-phenotype correlations are not completely matched. Clinical phenotypes of a particular SCN5A variant may range from asymptomatic to SCD. Here, we used comparison of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) derived from a SCN5A mutation-positive (D356Y) BrS family with severely affected proband, asymptomatic mutation carriers (AMCs) and healthy controls to investigate this variation. METHODS: 26 iPSC lines were generated from skin fibroblasts using nonintegrated Sendai virus. The generated iPSCs were differentiated into cardiomyocytes using a monolayer-based differentiation protocol. FINDINGS: D356Y iPSC-CMs exhibited increased beat interval variability, slower depolarization, cardiac arrhythmias, defects of Na+ channel function and irregular Ca2+ signaling, when compared to controls. Importantly, the phenotype severity observed in AMC iPSC-CMs was milder than that of proband iPSC-CMs, an observation exacerbated by flecainide. Interestingly, the iPSC-CMs of the proband exhibited markedly decreased Ca2+ currents in comparison with control and AMC iPSC-CMs. CRISPR/Cas9-mediated genome editing to correct D356Y in proband iPSC-CMs effectively rescued the arrhythmic phenotype and restored Na+ and Ca2+ currents. Moreover, drug screening using established BrS iPSC-CM models demonstrated that quinidine and sotalol possessed antiarrhythmic effects in an individual-dependent manner. Clinically, venous and oral administration of calcium partially reduced the malignant arrhythmic events of the proband in mid-term follow-up. INTERPRETATION: Patient-specific and genome-edited iPSC-CMs can recapitulate the varying phenotypic severity of BrS. Our findings suggest that preservation of the Ca2+ currents might be a compensatory mechanism to resist arrhythmogenesis in BrS AMCs. FUNDING: National Key R&D Program of China (2017YFA0103700), National Natural Science Foundation of China (81922006, 81870175), Natural Science Foundation of Zhejiang Province (LD21H020001, LR15H020001), National Natural Science Foundation of China (81970269), Key Research and Development Program of Zhejiang Province (2019C03022) and Natural Science Foundation of Zhejiang Province (LY16H020002).


Asunto(s)
Síndrome de Brugada , Células Madre Pluripotentes Inducidas , Humanos , Síndrome de Brugada/genética , Síndrome de Brugada/patología , Miocitos Cardíacos , Arritmias Cardíacas/patología , Mutación , Muerte Súbita Cardíaca/patología
13.
Stem Cell Res Ther ; 14(1): 92, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37061738

RESUMEN

BACKGROUND: Although human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) are a promising cell resource for cardiovascular research, these cells exhibit an immature phenotype that hampers their potential applications. The inwardly rectifying potassium channel Kir2.1, encoded by the KCNJ2 gene, has been thought as an important target for promoting electrical maturation of iPSC-CMs. However, a comprehensive characterization of morphological and functional changes in iPSC-CMs overexpressing KCNJ2 (KCNJ2 OE) is still lacking. METHODS: iPSC-CMs were generated using a 2D in vitro monolayer differentiation protocol. Human KCNJ2 construct with green fluorescent protein (GFP) tag was created and overexpressed in iPSC-CMs via lentiviral transduction. The mixture of iPSC-CMs and mesenchymal cells was cocultured with decellularized natural heart matrix for generation of 3D human engineered heart tissues (EHTs). RESULTS: We showed that mRNA expression level of KCNJ2 in iPSC-CMs was dramatically lower than that in human left ventricular tissues. KCNJ2 OE iPSC-CMs yielded significantly increased protein expression of Kir2.1 and current density of Kir2.1-encoded IK1. The larger IK1 linked to a quiescent phenotype that required pacing to elicit action potentials in KCNJ2 OE iPSC-CMs, which can be reversed by IK1 blocker BaCl2. KCNJ2 OE also led to significantly hyperpolarized maximal diastolic potential (MDP), shortened action potential duration (APD) and increased maximal upstroke velocity. The enhanced electrophysiological maturation in KCNJ2 OE iPSC-CMs was accompanied by improvements in Ca2+ signaling, mitochondrial energy metabolism and transcriptomic profile. Notably, KCNJ2 OE iPSC-CMs exhibited enlarged cell size and more elongated and stretched shape, indicating a morphological phenotype toward structural maturation. Drug testing using hERG blocker E-4031 revealed that a more stable MDP in KCNJ2 OE iPSC-CMs allowed for obtaining significant drug response of APD prolongation in a concentration-dependent manner. Moreover, KCNJ2 OE iPSC-CMs formed more mature human EHTs with better tissue structure and cell junction. CONCLUSIONS: Overexpression of KCNJ2 can robustly enhance maturation of iPSC-CMs in electrophysiology, Ca2+ signaling, metabolism, transcriptomic profile, cardiomyocyte structure and tissue engineering, thus providing more accurate cellular model for elucidating cellular and molecular mechanisms of cardiovascular diseases, screening drug-induced cardiotoxicity, and developing personalized and precision cardiovascular medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas , Canales de Potasio de Rectificación Interna , Humanos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Diferenciación Celular/genética , Técnicas de Cocultivo , Cardiotoxicidad , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo
14.
Stem Cell Res ; 64: 102898, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36041398

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an inherited cardiovascular disease characterized by left ventricular hypertrophy and a high risk of sudden death. In this study, a skin biopsy was obtained from a HCM patient harboring a heterozygous missense mutation (c.3764C>A; p.A1225D) in the myosin binding protein C3 (MYBPC3) gene. The isolated fibroblasts were reprogrammed using non-integrated Sendai viral method to establish the patient-specific induced pluripotent stem cell (iPSC) line. The established iPSC line displayed normal morphology and karyotype, expressed pluripotency markers, and can differentiate into three germ layers in vivo.


Asunto(s)
Cardiomiopatía Hipertrófica , Células Madre Pluripotentes Inducidas , Humanos , Cardiomiopatía Hipertrófica/patología , Heterocigoto , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Miosinas/metabolismo
15.
Stem Cell Res ; 64: 102883, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35944310

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an autosomal dominant inherited cardiovascular disease characterized by left ventricular hypertrophy and cardiomyocyte disarray. In this study, a skin biopsy was obtained from a HCM patient, who carried a missense mutation (c.4384G > A; p.E1462K) in the myosin heavy chain 7 (MYH7) gene. The skin fibroblasts were subsequently reprogrammed with a non-integrated Sendai viral method to generate a patient-specific induced pluripotent stem cell (iPSC) line. The generated iPSC line showed typical morphology and normal karyotype, expressed pluripotency markers, and was capable to differentiate into three germ layers.


Asunto(s)
Cardiomiopatía Hipertrófica , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cadenas Pesadas de Miosina/genética , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Mutación/genética , Mutación Missense , Miosinas Cardíacas/genética
16.
Stem Cell Res ; 62: 102813, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35569348

RESUMEN

Long QT syndrome (LQT) is an inherited primary arrhythmic disorder characterized by prolonged QT interval on the surface electrocardiogram and life-threatening arrhythmia. In this study, a skin biopsy was obtained from an LQT type 2 (LQT2) patient, who carried a nonsense mutation (c.1956C > A; p.Y652X) in the potassium voltage-gated channel subfamily H member 2 (KCNH2) gene. The skin fibroblasts were reprogrammed by non-integrated Sendai viral method to generate a patient-specific induced pluripotent stem cell (iPSC) line. The generated iPSC line showed typical embryonic stem cell-like morphology, exhibited normal karyotype, expressed pluripotency markers, and was capable to differentiate into three germ layers.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome de QT Prolongado , Arritmias Cardíacas/metabolismo , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de QT Prolongado/metabolismo , Mutación/genética
17.
J Inflamm Res ; 14: 3489-3500, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326657

RESUMEN

PURPOSE: Peptidyl arginine deiminase, type VI (PADI6), a member of the subcortical maternal complex, plays an important role in oocyte growth and the development of fertilized oocytes. Human patients with PADI6 mutations can suffer from multiple reproductive deficiencies including hydatidiform moles and miscarriages. Recent studies have demonstrated that the Hippo signaling pathway plays a central role in the specification of the first cell fates and the maintenance of the human placental trophoblast epithelium. The present study aimed to verify the hypothesis that PADI6 regulates the biological functions of trophoblast cells by targeting YAP1 and to explore the mechanism by which PADI6 accomplishes this in trophoblast cells. METHODS: Villi from HMs and human trophoblast cell lines were used to identify the localization of PADI6 and YAP1 by immunohistochemistry and immunocytochemistry. PADI6 overexpression and knockdown were induced in human trophoblast cells. Co-immunoprecipitation was used to explore the interaction between PADI6 and YAP1. Wound healing, Transwell and EdU staining assays were used to detect migration, invasion and proliferation. Flow cytometric analysis was used to analyze the cell cycle and apoptosis. ß-Tubulin and F-actin levels were determined by Western blot, quantitative real-time PCR and phalloidin staining. RESULTS: The results showed that PADI6 and YAP1 had the same expression pattern in villi and colocalized in the cytotrophoblast. An interaction between PADI6 and YAP1 was also confirmed in human trophoblast cell lines. We found that PADI6 positively regulated the expression of YAP1. Functionally, overexpression of PADI6 promoted cell cycle progression and enhanced migration, invasion, proliferation and apoptosis, whereas downregulation of PADI6 showed the opposite effects. CONCLUSION: This study demonstrates that YAP1 is a novel target of PADI6 that serves as an important regulator of trophoblast dysfunction. The crosstalk between the Hippo/YAP1 pathway and the SCMC might be a new topic to explore to uncover the pathological mechanisms of HMs.

18.
Stem Cell Res ; 53: 102361, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34087990

RESUMEN

Recurrent hydatidiform mole (RHM) is characterized by the occurrence of at least twice hydatidiform mole. Unlike sporadic complete hydatidiform moles (CHMs), which are androgenetic with 2 paternal chromosomes, CHMs associated with familial recurrence are genetically biparental with a maternal and a paternal chromosome. NLRP7 mutations have been reported in 55% of RHM cases. Here, we generated induced pluripotent stem cells (iPSCs) from a patent with NLRP7 gene mutation c.1261C > T by reprogramming peripheral blood mononuclear cells by non-integrated method. The resulting iPSCs carrying NLRP7 mutation, had normal karyotype, expressed pluripotency markers, and could differentiate into three germ layersin vivo.


Asunto(s)
Mola Hidatiforme , Células Madre Pluripotentes Inducidas , Proteínas Adaptadoras Transductoras de Señales/genética , Femenino , Humanos , Mola Hidatiforme/genética , Leucocitos Mononucleares , Mutación/genética , Recurrencia Local de Neoplasia , Embarazo
19.
Stem Cell Reports ; 16(9): 2305-2319, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34388364

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a rare disease associated with high morbidity and mortality. Existing evidence suggests that the central pathogenesis to aHUS might be endothelial cell damage. Nevertheless, the role of endothelial cell alterations in aHUS has not been well characterized and the underlying mechanisms remain unclear. Utilizing an induced pluripotent stem cell-derived endothelial cell (iPSC-EC) model, we showed that anti-complement factor H autoantibody-associated aHUS patient-specific iPSC-ECs exhibited an intrinsic defect in endothelial functions. Stimulation using aHUS serums exacerbated endothelial dysfunctions, leading to cell apoptosis in iPSC-ECs. Importantly, we identified p38 as a novel signaling pathway contributing to endothelial dysfunctions in aHUS. These results illustrate that iPSC-ECs can be a reliable model to recapitulate EC pathological features, thus providing a unique platform for gaining mechanistic insights into EC injury in aHUS. Our findings highlight that the p38 MAPK signaling pathway can be a therapeutic target for treatment of aHUS.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/etiología , Síndrome Hemolítico Urémico Atípico/metabolismo , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Apoptosis , Síndrome Hemolítico Urémico Atípico/diagnóstico , Autoanticuerpos/inmunología , Autoinmunidad , Biomarcadores , Factor H de Complemento/inmunología , Susceptibilidad a Enfermedades , Células Endoteliales/citología , Endotelio/metabolismo , Endotelio/fisiopatología , Humanos , Fenotipo
20.
Stem Cell Res ; 48: 101965, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32916635

RESUMEN

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited disease in which the right ventricular myocardium is replaced by progressive fibrous adipose tissue. ARVC is clinically characterized by right ventricular enlargement, ventricular arrhythmia, and sudden cardiac death. It eventually leads to heart failure, and thus has a significant impact on the patient's health. In this study, human dermal fibroblasts were obtained from a patient with ARVC, which were subsequently reprogrammed with a non-integrated Sendai virus to generate a patient-specific induced pluripotent stem cell (iPSC) line. The iPSC line exhibited normal karyotype and morphology, expressed pluripotency markers, and was capable of differentiating into three germ layers.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Células Madre Pluripotentes Inducidas , Displasia Ventricular Derecha Arritmogénica/genética , Línea Celular , Fibroblastos , Humanos , Mutación , Placofilinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA