Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cardiovasc Res ; 117(3): 767-779, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-32173736

RESUMO

AIMS: In long QT syndrome (LQTS) patients, modifier genes modulate the arrhythmic risk associated with a disease-causing mutation. Their recognition can improve risk stratification and clinical management, but their discovery represents a challenge. We tested whether a cellular-driven approach could help to identify new modifier genes and especially their mechanism of action. METHODS AND RESULTS: We generated human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) from two patients carrying the same KCNQ1-Y111C mutation, but presenting opposite clinical phenotypes. We showed that the phenotype of the iPSC-CMs derived from the symptomatic patient is due to impaired trafficking and increased degradation of the mutant KCNQ1 and wild-type human ether-a-go-go-related gene. In the iPSC-CMs of the asymptomatic (AS) patient, the activity of an E3 ubiquitin-protein ligase (Nedd4L) involved in channel protein degradation was reduced and resulted in a decreased arrhythmogenic substrate. Two single-nucleotide variants (SNVs) on the Myotubularin-related protein 4 (MTMR4) gene, an interactor of Nedd4L, were identified by whole-exome sequencing as potential contributors to decreased Nedd4L activity. Correction of these SNVs by CRISPR/Cas9 unmasked the LQTS phenotype in AS cells. Importantly, the same MTMR4 variants were present in 77% of AS Y111C mutation carriers of a separate cohort. Thus, genetically mediated interference with Nedd4L activation seems associated with protective effects. CONCLUSION: Our finding represents the first demonstration of the cellular mechanism of action of a protective modifier gene in LQTS. It provides new clues for advanced risk stratification and paves the way for the design of new therapies targeting this specific molecular pathway.


Assuntos
Genes Modificadores , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/genética , Mutação , Polimorfismo de Nucleotídeo Único , Proteínas Tirosina Fosfatases não Receptoras/genética , Células Cultivadas , Predisposição Genética para Doença , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Canal de Potássio KCNQ1/metabolismo , Síndrome do QT Longo/metabolismo , Miócitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligases Nedd4/genética , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Fenótipo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Proteólise
2.
Int J Cardiol ; 298: 85-92, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31668660

RESUMO

AIMS: To recapitulate progressive human dilated cardiomyopathy (DCM) and heart block in the Lmna R225X mutant mice model and investigate the molecular basis of LMNA mutation induced cardiac conduction disorders (CD); To investigate the potential interventional impact of exercise endurance. METHODS AND RESULTS: A Lmna R225X knock-in mice model in either heterozygous or homozygous genotype was generated. Electrical remodeling was observed with higher occurrence of AV block from neonatal and aged mutant mice as measured by surface electrocardiogram and atrio-ventricular Wenckebach point detection. Histological and molecular profiles revealed an increase in apoptotic cells and activation of caspase-3 activities in heart tissue. Upon aging, extracellular cellular matrix (ECM) remodeling appeared with accumulation of collagen in Lmna R225X mutant hearts as visualized by Masson's trichrome stain. This could be explained by the upregulated ECM gene expression, such as Fibronectin: Fn1, collagen: Col12a1, intergrin: Itgb2 and 3, as detected by microarray gene chip. Also, endurance exercise for 3 month improved the ventricular ejection fraction, attenuated fibrosis and cardiomyocytes apoptosis in the aged mutant mice. CONCLUSIONS: The mechanism of LMNA nonsense mutation induced cardiac conduction defects through AV node fibrosis is due to upregulated ECM gene expression upon activation of cardiac apoptosis. Lmna R225X mutant mice hold the potential for serving as in vivo models to explore the mechanism and therapeutic methods for AV block or myopathy associated with the aging process.


Assuntos
Doença do Sistema de Condução Cardíaco/genética , Cardiomiopatia Dilatada/genética , Códon sem Sentido/genética , Lamina Tipo A/genética , Condicionamento Físico Animal/fisiologia , Animais , Animais Recém-Nascidos , Doença do Sistema de Condução Cardíaco/metabolismo , Doença do Sistema de Condução Cardíaco/terapia , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/terapia , Expressão Gênica , Técnicas de Introdução de Genes/métodos , Frequência Cardíaca/fisiologia , Lamina Tipo A/biossíntese , Camundongos , Condicionamento Físico Animal/métodos
3.
Stem Cell Res ; 37: 101431, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30974404

RESUMO

We generated human induced pluripotent stem cells (hiPSCs) from dermal fibroblasts of a male carrier of the heterozygous mutation c.1781 G > A p.R594Q on the KCNQ1 gene. hiPSCs, generated using four retroviruses each encoding for OCT4, SOX2, KLF4 and cMYC, display pluripotent stem cell characteristics, and can be differentiated into spontaneously beating cardiomyocytes (hiPSC-CMs).


Assuntos
Diferenciação Celular , Fibroblastos/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Canal de Potássio KCNQ1/genética , Mutação , Miócitos Cardíacos/patologia , Síndrome de Romano-Ward/genética , Adulto , Células Cultivadas , Reprogramação Celular , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Masculino , Miócitos Cardíacos/metabolismo , Fenótipo , Síndrome de Romano-Ward/patologia
4.
Stem Cell Res ; 37: 101437, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31009818

RESUMO

We generated human induced pluripotent stem cells (hiPSCs) from dermal fibroblasts of a woman carrier of the heterozygous mutation c.568C > T p.R190W on the KCNQ1 gene. hiPSCs, obtained using four retroviruses enconding the reprogramming factors OCT4, SOX2, cMYC and KLF4, display pluripotent stem cell characteristics, and can be differentiated into spontaneously beating cardiomyocytes (hiPSC-CMs).


Assuntos
Diferenciação Celular , Reprogramação Celular , Fibroblastos/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Canal de Potássio KCNQ1/genética , Mutação , Síndrome de Romano-Ward/genética , Adulto , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Heterozigoto , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Síndrome de Romano-Ward/patologia
5.
Sci Rep ; 8(1): 14872, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30291295

RESUMO

Empagliflozin, a sodium-glucose co-transporter (SGLT) inhibitor, reduces heart failure and sudden cardiac death but the underlying mechanisms remain elusive. In cardiomyocytes, SGLT1 and SGLT2 expression is upregulated in diabetes mellitus, heart failure, and myocardial infarction. We hypothesise that empagliflozin exerts direct effects on cardiomyocytes that attenuate diabetic cardiomyopathy. To test this hypothesis, cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) were used to test the potential effects of empagliflozin on neutralization of cardiac dysfunction induced by diabetic-like cultures. Our results indicated that insulin-free high glucose culture significantly increased the size of and NPPB, SGLT1 and SGLT2 expression of hiPSC-derived cardiomyocytes. In addition, high glucose-treated hiPSC-derived cardiomyocytes exhibited reduced contractility regardless of the increased calcium transient capacity. Interestingly, application of empagliflozin before or after high glucose treatment effectively reduced the high glucose-induced cardiac abnormalities. Since application of empagliflozin did not significantly alter viability or glycolytic capacity of the hiPSC-derived cardiomyocytes, it is plausible that empagliflozin exerts its effects via the down-regulation of SGLT1, SGLT2 and GLUT1 expression. These observations provide supportive evidence that may help explain its unexpected benefit observed in the EMPA-REG trial.


Assuntos
Compostos Benzidrílicos/farmacologia , Glucose/metabolismo , Glucosídeos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Linhagem Celular , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Miócitos Cardíacos/metabolismo
6.
Sci Rep ; 8(1): 10846, 2018 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-30022097

RESUMO

Targeted next generation sequencing of gene panels has become a popular tool for the genetic diagnosis of hypertrophic (HCM) and dilated cardiomyopathy (DCM). However, it is uncertain whether the use of Whole Exome Sequencing (WES) represents a more effective approach for diagnosis of cases with HCM and DCM. In this study, we performed indirect comparisons of the coverage and diagnostic yield of WES on genes and variants related to HCM and DCM versus 4 different commercial gene panels using 40 HCM and DCM patients, assuming perfect coverage in those panels. We identified 6 pathogenic or likely pathogenic among 14 HCM patients (diagnostic yield 43%). 3 pathogenic or likely pathogenic were found among the 26 DCM patients (diagnostic yield 12%). The coverage was similar to that of four existing commercial gene panels due to the clustering of mutation within MYH7, MYBPC3, TPM1, TNT2, and TTN. Moreover, the coverage of WES appeared inadequate for TNNI3 and PLN. We conclude that most of the pathogenic variants for HCM and DCM can be found within a small number of genes which were covered by all the commercial gene panels, and the application of WES did not increase diagnostic yield.


Assuntos
Cardiomiopatia Dilatada/diagnóstico , Cardiomiopatia Hipertrófica/diagnóstico , Sequenciamento do Exoma/métodos , Marcadores Genéticos , Mutação , Análise de Sequência de DNA/métodos , Cardiomiopatia Dilatada/genética , Cardiomiopatia Hipertrófica/genética , Feminino , Testes Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade
7.
Stem Cell Res ; 29: 157-161, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29677589

RESUMO

We report the generation of human induced pluripotent stem cells (hiPSCs) from dermal fibroblasts of a female patient carrier of the two compound heterozygous mutations c.568 C>T p.R190W (maternal allele), and c.1781 G>A p.R594Q (paternal allele) on the KCNQ1 gene, causing Jervell and Lange-Nielsen Syndrome (JLNS). To obtain hiPSCs, we used the classical approach of the four retroviruses each encoding for a reprogramming factor OCT4, SOX2, KLF4, cMYC. The obtained hiPSC clones display pluripotent stem cell characteristics, and differentiate into spontaneously beating cardiomyocytes (hiPSC-CMs).


Assuntos
Heterozigoto , Células-Tronco Pluripotentes Induzidas , Síndrome de Jervell-Lange Nielsen , Canal de Potássio KCNQ1/genética , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Linhagem Celular , Criança , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome de Jervell-Lange Nielsen/genética , Síndrome de Jervell-Lange Nielsen/metabolismo , Síndrome de Jervell-Lange Nielsen/patologia , Fator 4 Semelhante a Kruppel
8.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2964-2972, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28754452

RESUMO

Dilated cardiomyopathy (DCM) is cardiac disease characterized by increased left ventricular chamber volume and decreased systolic function. DCM patient-specific human induced-pluripotent stem cells-derived cardiomyocytes (DCM-hiPSC-CMs) were generated. We found that uniaxial stretch elicited a cytosolic [Ca2+]i rise in hiPSC-CMs. Compared to control-hiPSC-CMs, DCM-hiPSC-CMs displayed a greater magnitude of [Ca2+]i responses to the cell stretch of 10-15% elongation in length. This stretch-induced [Ca2+]i rise was abolished by removal of extracellular Ca2+ and markedly attenuated by TRPV4 inhibitors HC-067047 and RN-1734. Application of nifedipine and tranilast also reduced the [Ca2+]i response but to a lesser degree. Moreover, the augmented [Ca2+]i was decreased by cytochalasin D treatment. Taken together, our study for the first time demonstrated an abnormal TRPV4-related mechanosensitive Ca2+ signaling in DCM-hiPSC-CMs.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Cardiomiopatia Dilatada/metabolismo , Citosol/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mecanotransdução Celular , Miócitos Cardíacos/metabolismo , Canais de Cátion TRPV/metabolismo , Cardiomiopatia Dilatada/patologia , Citosol/patologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Miócitos Cardíacos/patologia
9.
J Am Heart Assoc ; 6(8)2017 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-28754655

RESUMO

BACKGROUND: Precision medicine is an emerging approach to disease treatment and prevention that takes into account individual variability in the environment, lifestyle, and genetic makeup of patients. Patient-specific human induced pluripotent stem cells hold promise to transform precision medicine into real-life clinical practice. Lamin A/C (LMNA)-related cardiomyopathy is the most common inherited cardiomyopathy in which a substantial proportion of mutations in the LMNA gene are of nonsense mutation. PTC124 induces translational read-through over the premature stop codon and restores production of the full-length proteins from the affected genes. In this study we generated human induced pluripotent stem cells-derived cardiomyocytes from patients who harbored different LMNA mutations (nonsense and frameshift) to evaluate the potential therapeutic effects of PTC124 in LMNA-related cardiomyopathy. METHODS AND RESULTS: We generated human induced pluripotent stem cells lines from 3 patients who carried distinctive mutations (R225X, Q354X, and T518fs) in the LMNA gene. The cardiomyocytes derived from these human induced pluripotent stem cells lines reproduced the pathophysiological hallmarks of LMNA-related cardiomyopathy. Interestingly, PTC124 treatment increased the production of full-length LMNA proteins in only the R225X mutant, not in other mutations. Functional evaluation experiments on the R225X mutant further demonstrated that PTC124 treatment not only reduced nuclear blebbing and electrical stress-induced apoptosis but also improved the excitation-contraction coupling of the affected cardiomyocytes. CONCLUSIONS: Using cardiomyocytes derived from human induced pluripotent stem cells carrying different LMNA mutations, we demonstrated that the effect of PTC124 is codon selective. A premature stop codon UGA appeared to be most responsive to PTC124 treatment.


Assuntos
Cardiomiopatia Dilatada/tratamento farmacológico , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Lamina Tipo A/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Oxidiazóis/farmacologia , Adulto , Apoptose/efeitos dos fármacos , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/fisiopatologia , Códon sem Sentido , Estimulação Elétrica , Acoplamento Excitação-Contração/efeitos dos fármacos , Mutação da Fase de Leitura , Regulação da Expressão Gênica/efeitos dos fármacos , Predisposição Genética para Doença , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Lamina Tipo A/genética , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo
10.
Circulation ; 134(18): 1373-1389, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27678261

RESUMO

BACKGROUND: Danon disease is an X-linked disorder that leads to fatal cardiomyopathy caused by a deficiency in lysosome-associated membrane protein-2 (LAMP2). In female patients, a later onset and less severe clinical phenotype have been attributed to the random inactivation of the X chromosome carrying the mutant diseased allele. We generated a patient-specific induced pluripotent stem cell (iPSCs)-based model of Danon disease to evaluate the therapeutic potential of Xi-chromosome reactivation using a DNA methylation inhibitor. METHODS: Using whole-exome sequencing, we identified a nonsense mutation (c.520C>T, exon 4) of the LAMP2 gene in a family with Danon disease. We generated iPSC lines from somatic cells derived from the affected mother and her 2 sons, and we then differentiated them into cardiomyocytes (iPSC-CMs) for modeling the histological and functional signatures, including autophagy failure of Danon disease. RESULTS: Our iPSC-CM platform provides evidence that random inactivation of the wild-type and mutant LAMP2 alleles on the X chromosome is responsible for the unusual phenotype in female patients with Danon disease. In vitro, iPSC-CMs from these patients reproduced the histological features and autophagy failure of Danon disease. Administration of the DNA demethylating agent 5-aza-2'-deoxycytidine reactivated the silent LAMP2 allele in iPSCs and iPSC-CMs in female patients with Danon disease and ameliorated their autophagy failure, supporting the application of a patient-specific iPSC platform for disease modeling and drug screening. CONCLUSIONS: Our iPSC-CM platform provides novel mechanistic and therapeutic insights into the contribution of random X chromosome inactivation to disease phenotype in X-linked Danon disease.


Assuntos
Autofagia , Azacitidina/farmacologia , Cromossomos Humanos X/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo , Adulto , Alelos , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular , Feminino , Doença de Depósito de Glicogênio Tipo IIb/genética , Doença de Depósito de Glicogênio Tipo IIb/metabolismo , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/biossíntese , Proteína 2 de Membrana Associada ao Lisossomo/genética , Masculino
11.
Stem Cell Res Ther ; 7(1): 139, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27649756

RESUMO

Laminopathy is a disease closely related to deficiency of the nuclear matrix protein lamin A/C or failure in prelamin A processing, and leads to accumulation of the misfold protein causing progeria. The resultant disrupted lamin function is highly associated with abnormal nuclear architecture, cell senescence, apoptosis, and unstable genome integrity. To date, the effects of loss in nuclear integrity on the susceptible organ, striated muscle, have been commonly associated with muscular dystrophy, dilated cardiac myopathy (DCM), and conduction defeats, but have not been studied intensively. In this review, we aim to summarize recent breakthroughs in an in vivo laminopathy model and in vitro study using patient-specific human induced pluripotent stem cells (iPSCs) that reproduce the pathophysiological phenotype for further drug screening. We describe several in-vivo transgenic mouse models to elucidate the effects of Lmna H222P, N195K mutations, and LMNA knockout on cardiac function, in terms of hemodynamic and electrical signal propagation; certain strategies targeted on stress-related MAPK are mentioned. We will also discuss human iPSC cardiomyocytes serving as a platform to reveal the underlying mechanisms, such as the altered mechanical sensation in electrical coupling of the heart conduction system and ion channel alternation in relation to altered nuclear architecture, and furthermore to enable screening of drugs that can attenuate this cardiac premature aging phenotype by inhibition of prelamin misfolding and oxidative stress, and also enhancement of autophagy protein clearance and cardiac-protective microRNA.


Assuntos
Cardiomiopatia Dilatada/genética , Lamina Tipo A/genética , Modelos Cardiovasculares , Mutação , Progéria/genética , Deficiências na Proteostase/genética , Animais , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiotônicos/farmacologia , Expressão Gênica , Instabilidade Genômica , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Lamina Tipo A/deficiência , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Progéria/tratamento farmacológico , Progéria/metabolismo , Progéria/patologia , Dobramento de Proteína , Deficiências na Proteostase/tratamento farmacológico , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia
12.
Stem Cells ; 34(11): 2693-2706, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27333945

RESUMO

Recent pre-clinical and clinical studies have suggested that endogenous cardiospheres (eCS) are potentially safe and effective for cardiac regeneration following myocardial infarction (MI). Nevertheless the preparation of autologous eCS requires invasive myocardial biopsy with limited yield. We describe a novel approach to generate induced cardiospheres (iCS) from adult skin fibroblasts via somatic reprogramming. After infection with Sox2, Klf4, and Oct4, iCS were generated from mouse adult skin fibroblasts treated with Gsk3ß inhibitor-(2'Z,3'E)- 6-Bromoindirubin-3'-oxime and Oncostatin M. They resembled eCS, but contained a higher percentage of cells expressing Mesp1, Isl1, and Nkx2.5. They were differentiated into functional cardiomyocytes in vitro with similar electrophysiological properties, calcium transient and contractile function to eCS and mouse embryonic stem cell-derived cardiomyocytes. Transplantation of iCS (1 × 106 cells) into mouse myocardium following MI had similar effects to transplantation of eCS but significantly better than saline or fibroblast in improving left ventricular ejection fraction, increasing anterior/septal ventricular wall thickness and capillary density in the infarcted region 4 weeks after transplantation. No tumor formation was observed. iCS generated from adult skin fibroblasts by somatic reprogramming and a cocktail of Gsk3ß inhibitor-6-Bromoindirubin-3'-oxime and Oncostatin M may represent a novel source for cell therapy in MI. Stem Cells 2016;34:2693-2706.


Assuntos
Reprogramação Celular , Fibroblastos/metabolismo , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Regeneração/fisiologia , Esferoides Celulares/transplante , Potenciais de Ação , Animais , Cálcio/metabolismo , Diferenciação Celular , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Expressão Gênica , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Indóis/farmacologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Oncostatina M/farmacologia , Oximas/farmacologia , Cultura Primária de Células , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Pele/citologia , Pele/efeitos dos fármacos , Pele/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Transdução Genética , Função Ventricular Esquerda/fisiologia
13.
Int J Cardiol ; 203: 964-71, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26625322

RESUMO

BACKGROUND: Friedreich's ataxia (FRDA), a recessive neurodegenerative disorder commonly associated with hypertrophic cardiomyopathy, is caused by silencing of the frataxin (FXN) gene encoding the mitochondrial protein involved in iron-sulfur cluster biosynthesis. METHODS: Application of our previously established FRDA human induced pluripotent stem cell (hiPSC) derived cardiomyocytes model as a platform to assess the efficacy of treatment with either the antioxidant coenzyme Q10 analog, idebenone (IDE) or the iron chelator, deferiprone (DFP), which are both under clinical trial. RESULTS: DFP was able to more significantly suppress synthesis of reactive oxygen species (ROS) than IDE at the dosages of 25 µM and 10nM respectively which agreed with the reduced rate of intracellular accumulation of iron by DFP treatment from 25 to 50 µM. With regard to cardiac electrical-contraction (EC) coupling function, decay velocity of calcium handling kinetics in FRDA-hiPSC-cardiomyocytes was significantly improved by DFP treatment but not by IDE. Further mechanistic studies revealed that DFP also modulated iron induced mitochondrial stress as reflected by mitochondria network disorganization and decline level of respiratory chain protein, succinate dehydrogenase (CxII) and cytochrome c oxidase (COXIV). In addition, iron-response protein (IRP-1) regulatory loop was overridden by DFP as reflected by resumed level of ferritin (FTH) back to basal level and the attenuated transferrin receptor (TSFR) mRNA level suppression thereby reducing further iron uptake. CONCLUSIONS: DFP modulated iron homeostasis in FRDA-hiPSC-cardiomyocytes and effectively relieved stress-stimulation related to cardiomyopathy. The resuming of redox condition led to the significantly improved cardiac prime events, cardiac electrical-coupling during contraction.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Ataxia de Friedreich/terapia , Células-Tronco Pluripotentes Induzidas , Ferro/metabolismo , Miócitos Cardíacos/metabolismo , Piridonas/farmacologia , Ubiquinona/análogos & derivados , Antioxidantes/farmacologia , Deferiprona , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Regulação da Expressão Gênica , Homeostase , Humanos , Quelantes de Ferro/farmacologia , Proteínas de Ligação ao Ferro/biossíntese , Proteínas de Ligação ao Ferro/genética , Miócitos Cardíacos/patologia , Estresse Oxidativo , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquinona/farmacologia , Frataxina
14.
Methods Mol Biol ; 1353: 191-213, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26126449

RESUMO

Advances in differentiation of cardiomyocytes from human induced pluripotent stem cell (hiPSC) were emerged as a tool for modeling of cardiovascular disease that recapitulates the phenotype for the purpose of drug screening, biomarker discovery, and testing of single-nucleotide polymorphism (SNP) as a modifier for disease stratification. Here, we describe the (1) retroviral reprogramming strategies in the generation of human iPSC, (2) methodology in characterization of iPSC in order to identify the stem cell clones with the best quality, and (3) protocol of cardiac differentiation by modulation of Wnt signaling and ß-catenin pathway.


Assuntos
Técnicas de Cultura de Células/métodos , Reprogramação Celular , Corpos Embrioides/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Amidas/farmacologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Colágeno/química , Combinação de Medicamentos , Embrião de Mamíferos , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Inibidores Enzimáticos/farmacologia , Células Alimentadoras/citologia , Fibroblastos/citologia , Expressão Gênica , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Laminina/química , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Cultura Primária de Células , Proteoglicanas/química , Piridinas/farmacologia , Pirimidinas/farmacologia , Retroviridae/genética , Teratoma/genética , Teratoma/metabolismo , Teratoma/patologia , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo
15.
Biochem Biophys Rep ; 5: 335-345, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28955840

RESUMO

Patients with Danon disease may suffer from severe cardiomyopathy, skeletal muscle dysfunction as well as varying degrees of mental retardation, in which the primary deficiency of lysosomal membrane-associated protein-2 (LAMP2) is considerably associated. Owing to the scarcity of human neurons, the pathological role of LAMP2 deficiency in neural injury of humans remains largely elusive. However, the application of induced pluripotent stem cells (iPSCs) may shed light on overcoming such scarcity. In this study, we obtained iPSCs derived from a patient carrying a mutated LAMP2 gene that is associated with Danon disease. By differentiating such LAMP2-deficient iPSCs into cerebral cortical neurons and with the aid of various biochemical assays, we demonstrated that the LAMP2-deficient neurons are more susceptible to mild oxidative stress-induced injury. The data from MTT assay and apoptotic analysis demonstrated that there was no notable difference in cellular viability between the normal and LAMP2-deficient neurons under non-stressed condition. When exposed to mild oxidative stress (10 µM H2O2), the LAMP2-deficient neurons exhibited a significant increase in apoptosis. Surprisingly, we did not observe any aberrant accumulation of autophagic materials in the LAMP2-deficient neurons under such stress condition. Our results from cellular fractionation and inhibitor blockade experiments further revealed that oxidative stress-induced apoptosis in the LAMP2-deficient cortical neurons was caused by increased abundance of cytosolic cathepsin L. These results suggest the involvement of lysosomal membrane permeabilization in the LAMP2 deficiency associated neural injury.

16.
Curr Stem Cell Res Ther ; 9(5): 401-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24947903

RESUMO

Cardiovascular disease is the leading cause of death worldwide. Despite significant progress in understanding of the disease mechanisms, most therapies remain at best palliative. Few therapeutic approaches offer direct tissue repair and regeneration. Cell-based therapy offers a promising approach that involves transplantation of healthy and functional cells to replenish damaged cells and repair injured tissue. Endothelial dysfunction is one of the most important mechanisms of cardiovascular disease, thus endothelial progenitor cells (EPC) and their derivatives have been investigated as a potential source for cell therapy. In pre-clinical and pilot clinical studies, treatment with EPCs or their derivatives as well as their co-transplantation with other cell types has shown some initial promising results. In this review, we will first describe the importance of endothelial cells and EPC homeostasis in the pathophysiology of cardiovascular disease. The potential sources of EPCs, including their isolation and purification, differentiation from pluripotent stem cells and adult stem cells, and trans-differentiation from somatic cells will then be summarized. Lastly, the application of target genome editing tools, such as Zinc Finger Nuclease (ZFN), Transcription Activator Like Effector Nucleases (TALEN) and RNA Guided Endo Nuclease (RGEN) to modify EPCs and their derivatives will be described. These technologies promise to further improve the therapeutic potential of EPCs and their derivatives to treat cardiovascular disease.


Assuntos
Doenças Cardiovasculares/terapia , Células Progenitoras Endoteliais/transplante , Animais , Doenças Cardiovasculares/patologia , Diferenciação Celular , Células Endoteliais/fisiologia , Células Progenitoras Endoteliais/fisiologia , Técnicas de Transferência de Genes , Terapia Genética , Humanos
17.
Pflugers Arch ; 466(9): 1831-44, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24327207

RESUMO

Friedreich ataxia (FRDA), a recessive neurodegenerative disorder commonly associated with hypertrophic cardiomyopathy, is due to GAA repeat expansions within the first intron of the frataxin (FXN) gene encoding the mitochondrial protein involved in iron-sulfur cluster biosynthesis. The triplet codon repeats lead to heterochromatin-mediated gene silencing and loss of frataxin. Nevertheless, inadequacy of existing FRDA-cardiac cellular models limited cardiomyopathy studies. We tested the hypothesis that iron homeostasis deregulation accelerates reduction in energy synthesis dynamics which contributes to impaired cardiac calcium homeostasis and contractile force. Silencing of FXN expressions occurred both in somatic FRDA-skin fibroblasts and two of the induced pluripotent stem cells (iPSC) clones; a sign of stress condition was shown in FRDA-iPSC cardiomyocytes with disorganized mitochondrial network and mitochondrial DNA (mtDNA) depletion; hypertrophic cardiac stress responses were observed by an increase in α-actinin-positive cell sizes revealed by FACS analysis as well as elevation in brain natriuretic peptide (BNP) gene expression; the intracellular iron accumulated in FRDA cardiomyocytes might be due to attenuated negative feedback response of transferring receptor (TSFR) expression and positive feedback response of ferritin (FTH1); energy synthesis dynamics, in terms of ATP production rate, was impaired in FRDA-iPSC cardiomyocytes, which were prone to iron overload condition. Energetic insufficiency determined slower Ca(2+) transients by retarding calcium reuptake to sarcoplasmic reticulum (SR) and impaired the positive inotropic and chronotropic responses to adrenergic stimulation. Our data showed for the first time that FRDA-iPSCs cardiac derivatives represent promising models to study cardiac stress response due to impaired iron homeostasis condition and mitochondrial damages. The cardiomyopathy phenotype was accelerated in an iron-overloaded condition early in calcium homeostasis aspect.


Assuntos
Cardiomiopatias , Ataxia de Friedreich/complicações , Técnicas In Vitro , Células-Tronco Pluripotentes , Adulto , Cardiomiopatias/etiologia , Feminino , Ataxia de Friedreich/genética , Humanos , Sobrecarga de Ferro/complicações , Proteínas de Ligação ao Ferro/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Frataxina
18.
J Biomed Nanotechnol ; 10(10): 2562-85, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25992409

RESUMO

Human inherited cardiomyopathies are one of the major etiologies for heart failure which are associated with significant mortality and morbidity. Unfortunately, there are lack of effective specific therapies for human cardiomyopathies due to the limited understanding on their pathophysiology. Currently, most of the mechanistic studies of human cardiomyopathy are based on transgenic mouse models and invasive collection of limited amount of myocardial biopsy specimen. Disease-specific stem-cells are already available for studying single-gene mutation related diseases, such as cystic fibrosis and fragile X syndrome. The possibility of obtaining stem-cells using induced pluripotent stem cell (iPSC) technology provides the opportunity to generate stem cells carrying an inherited disease phenotype that will then serve as an invaluable model to study the disease biology and treatment of human cardiomyopathies. Here, we review the major strategies and workflow of using the patient-specific iPSCs derived cardiomyocytes to model inherited human cardiomyopathies. The creation of patient-specific iPSC lines in patients with hypertrophic cardiomyopathy and dilated cardiomyopathy have been reported and served as important human models of inherited diseases to improve our understanding of the disease mechanisms and enable drug screening.


Assuntos
Cardiomiopatias/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Biológicos , Cardiomiopatias/fisiopatologia , Diferenciação Celular , Reprogramação Celular , Humanos , Contração Miocárdica
19.
J Cardiovasc Transl Res ; 6(6): 989-99, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24081385

RESUMO

While human embryonic stem cells (hESCs) can differentiate into functional cardiomyocytes, their immature phenotypes limit their therapeutic application for myocardial regeneration. We sought to determine whether electrical stimulation could enhance the differentiation and maturation of hESC-derived cardiomyocytes. Cardiac differentiation was induced in a HES3 hESC line via embryoid bodies formation treated with a p38 MAP kinase inhibitor. Detailed molecular and functional analysis were performed in those hESC-derived cardiomyocytes cultured for 4 days in the absence or presence of electrical field stimulation (6.6 V/cm, 1 Hz, and 2 ms pulses) using an eight-channel C-Pace stimulator (Ion-Optics Co., MA). Upon electrical stimulation, quantitative polymerase chain reaction demonstrated significant upregulation of cardiac-specific gene expression including HCN1, MLC2V, SCN5A, SERCA, Kv4.3, and GATA4; immunostaining and flow cytometry analysis revealed cellular elongation and an increased proportion of troponin-T positive cells (6.3 ± 1.2% vs. 15.8 ± 2.1%; n = 3, P < 0.01). Electrophysiological studies showed an increase in the proportion of ventricular-like hESC-derived cardiomyocytes (48 vs. 29%, P < 0.05) with lengthening of their action potential duration at 90% repolarization (387.7 ± 35.35; n = 11 vs. 291.8 ± 20.82; n = 10, P < 0.05) and 50% repolarization (313.9 ± 27.94; n = 11 vs. 234.0 ± 16.10; n = 10, P < 0.05) after electrical stimulation. Nonetheless, the membrane diastolic potentials and action potential upstrokes of different hESC-derived cardiomyocyte phenotypes, and the overall beating rate remained unchanged (all P > 0.05). Fluorescence confocal imaging revealed that electrical stimulation significantly increased both spontaneous and caffeine-induced calcium flux in the hESC-derived cardiomyocytes (approximately 1.6-fold for both cases; P < 0.01). In conclusion, electrical field stimulation increased the expression of cardiac-specific genes and the yield of differentiation, promoted ventricular-like phenotypes, and improved the calcium handling of hESC-derived cardiomyocytes.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Miócitos Cardíacos/fisiologia , Sinalização do Cálcio , Linhagem Celular , Linhagem da Célula , Estimulação Elétrica , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/enzimologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Potenciais da Membrana , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Hum Mol Genet ; 22(7): 1395-403, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23300193

RESUMO

In this paper, we report a novel heterozygous mutation of A285V codon conversion on exon 4 of the desmin (DES), using whole exome sequencing (WES) in an isolated proband with documented dilated cardiomyopathy (DCM). This mutation is predicted to cause three-dimensional structure changes of DES. Immunohistological and electron microscopy studies demonstrated diffuse abnormal DES aggregations in DCM-induced-pluripotent stem cell (iPSC)-derived cardiomyocytes, and control-iPSC-derived cardiomyocytes transduced with A285V-DES. DCM-iPSC-derived cardiomyocytes also exhibited functional abnormalities in vitro. This is the first demonstration that patient-specific iPSC-derived cardiomyocytes can be used to provide histological and functional confirmation of a suspected genetic basis for DCM identified by WES.


Assuntos
Cardiomiopatia Dilatada/genética , Desmina/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/metabolismo , Adulto , Sequência de Aminoácidos , Sequência de Bases , Cardiomiopatia Dilatada/diagnóstico por imagem , Cardiomiopatia Dilatada/fisiopatologia , Diferenciação Celular , Desmina/química , Desmina/metabolismo , Exoma , Éxons , Células HEK293 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Análise de Sequência de DNA , Volume Sistólico/genética , Ultrassonografia , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...