Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
SLAS Discov ; 26(3): 364-372, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32914673

RESUMO

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been widely used for the assessment of drug proarrhythmic potential through multielectrode array (MEA). HiPSC-CM cultures beat spontaneously with a wide range of frequencies, however, which could affect drug-induced changes in repolarization. Pacing hiPSC-CMs at a physiological heart rate more closely resembles the state of in vivo ventricular myocytes and permits the standardization of test conditions to improve consistency. In this study, we systematically investigated the time window of stable ion currents in high-purity hiPSC-derived ventricular cardiomyocytes (hiPSC-vCMs) and confirmed that these cells could be used to correctly predict the proarrhythmic risk of Comprehensive In Vitro Proarrhythmia Assay (CiPA) reference compounds. To evaluate drug proarrhythmic potentials at a physiological beating rate, we used a MEA to electrically pace hiPSC-vCMs, and we recorded regular field potential waveforms in hiPSC-vCMs treated with DMSO and 10 CiPA reference drugs. Prolongation of field potential duration was detected in cells after exposure to high- and intermediate-risk drugs; in addition, drug-induced arrhythmia-like events were observed. The results of this study provide a simple and feasible method to investigate drug proarrhythmic potentials in hiPSC-CMs at a physiological beating rate.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Antiarrítmicos/farmacologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Fenetilaminas/efeitos adversos , Quinidina/efeitos adversos , Sulfonamidas/efeitos adversos , Potenciais de Ação/fisiologia , Arritmias Cardíacas/prevenção & controle , Cálcio/metabolismo , Cátions Bivalentes , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Transporte de Íons/efeitos dos fármacos , Microeletrodos , Modelos Biológicos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Nifedipino/farmacologia , Técnicas de Patch-Clamp , Cultura Primária de Células , Sotalol/efeitos adversos , Tetrodotoxina/antagonistas & inibidores , Tetrodotoxina/toxicidade , Verapamil/farmacologia
2.
Stem Cell Res ; 20: 21-29, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28249229

RESUMO

The combination of non-human primate animals and their induced pluripotent stem cell derived cardiomyocytes (iPSC-CMs) provides not only transplantation models for cell-based therapy of heart diseases, but also opportunities for heart-related drug research on both cellular and animal levels. However, the subtypes and electrophysiology properties of non-human primate iPSC-CMs hadn't been detailed characterized. In this study, we generated rhesus monkey induced pluripotent stem cells (riPSCs), and efficiently differentiated them into ventricular or atrial cardiomyocytes by modulating retinoic acid (RA) pathways. Our results revealed that the electrophysiological characteristics and response to canonical drugs of riPSC-CMs were similar with those of human pluripotent stem cell derived CMs. Therefore, rhesus monkeys and their iPSC-CMs provide a powerful and practicable system for heart related biomedical research.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Animais , Fator II de Transcrição COUP/genética , Fator II de Transcrição COUP/metabolismo , Sinalização do Cálcio/fisiologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Reprogramação Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Ventrículos do Coração/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Macaca mulatta , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia de Fluorescência , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/transplante , Técnicas de Patch-Clamp , Teratoma/metabolismo , Teratoma/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia
3.
Stem Cell Res ; 19: 94-103, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28110125

RESUMO

Most existing culture media for cardiac differentiation of human pluripotent stem cells (hPSCs) contain significant amounts of albumin. For clinical transplantation applications of hPSC-derived cardiomyocytes (hPSC-CMs), culturing cells in an albumin containing environment raises the concern of pathogen contamination and immunogenicity to the recipient patients. In addition, batch-to-batch variation of albumin may cause the inconsistent of hPSC cardiac differentiation. Here, we demonstrated that antioxidants l-ascorbic acid, trolox, N-acetyl-l-cysteine (NAC) and sodium pyruvate could functionally substitute albumin in the culture medium, and formulated an albumin-free, chemical-defined medium (S12 medium). We showed that S12 medium could support efficient hPSC cardiac differentiation with significantly improved reproducibility, and maintained long-term culture of hPSC-CMs. Furthermore, under chemical-defined and albumin-free conditions, human-induced pluripotent stem cells (hiPSCs) were established, and differentiated into highly homogenous atrial and ventricular myocytes in a scalable fashion with normal electrophysiological properties. Finally, we characterized the activity of three typical cardiac ion channels of those cells, and demonstrated that hPSC-derived ventricular cardiomyocytes (hPSC-vCMs) were suitable for drug cardiac safety evaluation. In summary, this simplified, chemical-defined and albumin-free culture medium supports efficient generation and maintaining of hPSC-CMs and facilitates both research and clinical applications of these cells.


Assuntos
Meios de Cultura/química , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Potenciais de Ação/efeitos dos fármacos , Antioxidantes/farmacologia , Fator II de Transcrição COUP/genética , Fator II de Transcrição COUP/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Microscopia de Fluorescência , Miócitos Cardíacos/metabolismo , Nifedipino/farmacologia , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Células-Tronco Pluripotentes/metabolismo , Piridinas/farmacologia , Tretinoína/farmacologia
4.
Stem Cells Dev ; 26(7): 528-540, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-27927069

RESUMO

The epicardium promotes neovascularization and cardiomyocyte regeneration by generating vascular smooth muscle cells (SMCs) and producing regenerative factors after adult heart infarction. It is therefore a potential cell resource for repair of the injured heart. However, the epicardium also participates in fibrosis and scarring of the injured heart, complicating its use in regenerative medicine. In this study, we report coexpression of TBX18 and WT1 in the majority of epicardial cells during mouse embryonic epicardial development. Furthermore, we describe a convenient chemically defined, immunogen-free, small molecule-based method for generating TBX18+/WT1+ epicardial-like cell populations with 80% homogeneity from human pluripotent stem cells by modulation of the WNT and retinoic acid signaling pathways. These epicardial-like cells exhibited characteristic epicardial cell morphology following passaging and differentiation into functional SMCs or cardiac fibroblast-like cells. Our findings add to existing understanding of human epicardial development and provide an efficient and stable method for generating both human epicardial-like cells and SMCs.


Assuntos
Diferenciação Celular/fisiologia , Miócitos Cardíacos/citologia , Miócitos de Músculo Liso/citologia , Pericárdio/citologia , Células-Tronco Pluripotentes/citologia , Animais , Fibroblastos/citologia , Humanos , Camundongos , Proteínas Repressoras/genética , Proteínas com Domínio T/genética , Proteínas WT1/genética
5.
Stem Cell Res Ther ; 3(2): 14, 2012 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-22512921

RESUMO

INTRODUCTION: Down syndrome (DS), a major cause of mental retardation, is caused by trisomy of some or all of human chromosome 21 and includes three basic karyotypes: trisomy 21, translocation, and mosaicism. The derivation of DS-specific induced pluripotent stem cells (iPSCs) provides us novel DS models that can be used to determine the DS mechanism and to devise therapeutic approaches for DS patients. METHODS: In the present study, fibroblasts from patients with DS of various karyotypes were reprogrammed into iPSCs via the overexpression of four factors: OCT4, SOX2, KLF4, and c-MYC, by using lentiviral vectors. The abilities of the iPSC-DS in the self-renewal and pluripotency in vitro and in vivo were then examined. RESULTS: The iPSC-DS showed characteristics similar to those of human embryonic stem cells, particularly the morphology, surface marker (SSEA4, TRA-1-60, and TRA-1-81) expression, pluripotent-specific transcription-factor expression levels, and methylation status of the OCT4 promoter. The pluripotency of iPSC-DS was also tested in vitro and in vivo. Embryoid bodies were formed and showed the expression of differentiated markers for three germ layers. Furthermore, iPSC-DS formed classic teratomas when injected into nonobese diabetic-severe combined immunodeficient (NOD-SCID) mice. CONCLUSIONS: iPSCs were generated from patients with DS. The iPSCs derived from different types of DS may be used in DS modeling, patient-care optimization, drug discovery, and eventually, autologous cell-replacement therapies.


Assuntos
Diferenciação Celular , Síndrome de Down/genética , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas , Cariótipo Anormal , Animais , Diferenciação Celular/genética , Células Cultivadas , Pré-Escolar , Síndrome de Down/terapia , Fibroblastos/metabolismo , Expressão Gênica , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Teratoma
6.
Protein Cell ; 3(1): 51-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22259120

RESUMO

While human induced pluripotent stem cells (hiPSCs) have promising applications in regenerative medicine, most of the hiPSC lines available today are not suitable for clinical applications due to contamination with nonhuman materials, such as sialic acid, and potential pathogens from animal-product-containing cell culture systems. Although several xeno-free cell culture systems have been established recently, their use of human fibroblasts as feeders reduces the clinical potential of hiPSCs due to batch-to-batch variation in the feeders and time-consuming preparation processes. In this study, we have developed a xeno-free and feeder-cell-free human embryonic stem cell (hESC)/hiPSC culture system using human plasma and human placenta extracts. The system maintains the self-renewing capacity and pluripotency of hESCs for more than 40 passages. Human iPSCs were also derived from human dermal fibroblasts using this culture system by overexpressing three transcription factors-Oct4, Sox2 and Nanog. The culture system developed here is inexpensive and suitable for large scale production.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura , Células-Tronco Pluripotentes/citologia , Diferenciação Celular , Reprogramação Celular , Proteínas da Matriz Extracelular/isolamento & purificação , Feminino , Fibroblastos/citologia , Humanos , Lentivirus/genética , Placenta/química , Células-Tronco Pluripotentes/metabolismo , Gravidez , Cloreto de Sódio/química , Fatores de Transcrição/genética
7.
Cell Res ; 21(4): 579-87, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21102549

RESUMO

Although myocyte cell transplantation studies have suggested a promising therapeutic potential for myocardial infarction, a major obstacle to the development of clinical therapies for myocardial repair is the difficulties associated with obtaining relatively homogeneous ventricular myocytes for transplantation. Human embryonic stem cells (hESCs) are a promising source of cardiomyocytes. Here we report that retinoid signaling regulates the fate specification of atrial versus ventricular myocytes during cardiac differentiation of hESCs. We found that both Noggin and the pan-retinoic acid receptor antagonist BMS-189453 (RAi) significantly increased the cardiac differentiation efficiency of hESCs. To investigate retinoid functions, we compared Noggin+RAi-treated cultures with Noggin+RA-treated cultures. Our results showed that the expression levels of the ventricular-specific gene IRX-4 were radically elevated in Noggin+RAi-treated cultures. MLC-2V, another ventricular-specific marker, was expressed in the majority of the cardiomyocytes in Noggin+RAi-treated cultures, but not in the cardiomyocytes of Noggin+RA-treated cultures. Flow cytometry analysis and electrophysiological studies indicated that with 64.7 ± 0.88% (mean ±s.e.m) cardiac differentiation efficiency, 83% of the cardiomyocytes in Noggin+RAi-treated cultures had embryonic ventricular-like action potentials (APs). With 50.7 ± 1.76% cardiac differentiation efficiency, 94% of the cardiomyocytes in Noggin+RA-treated cultures had embryonic atrial-like APs. These results were further confirmed by imaging studies that assessed the patterns and properties of the Ca(2+) sparks of the cardiomyocytes from the two cultures. These findings demonstrate that retinoid signaling specifies the atrial versus ventricular differentiation of hESCs. This study also shows that relatively homogeneous embryonic atrial- and ventricular-like myocyte populations can be efficiently derived from hESCs by specifically regulating Noggin and retinoid signals.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Retinoides/metabolismo , Transdução de Sinais , Potenciais de Ação/efeitos dos fármacos , Sequência de Bases , Western Blotting , Miosinas Cardíacas/biossíntese , Miosinas Cardíacas/genética , Proteínas de Transporte/farmacologia , Linhagem Celular , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Imunofluorescência , Átrios do Coração/embriologia , Ventrículos do Coração/embriologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Reação em Cadeia da Polimerase , Retinoides/farmacologia
8.
Mol Reprod Dev ; 74(1): 28-34, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16941671

RESUMO

Our and other previous studies have shown that telophase enucleation is an efficient method for preparing recipient cytoplasts in nuclear transfer. Conventional methods of somatic cell nuclear transfer either by electro-fusion or direct nucleus injection have very low efficiency in animal somatic cell cloning. To simplify the manipulation procedure and increase the efficiency of somatic cell nuclear transfer, this study was designed to study in vitro and in vivo development of Asian yellow goat cloned embryos reconstructed by direct whole cell intracytoplasmic injection (WCICI) into in vitro matured oocytes enucleated at telophase II stage. Our results demonstrated that the rates of cleavage and blastocyst development of embryos reconstructed by WCICI were slightly higher than in conventional subzonal injection (SUZI) group, but no statistic difference (P > 0.05) existed between these two methods. However, the percentage of successful embryonic reconstruction in WCICI group was significantly higher than that in SUZI group (P < 0.05). After embryo transfer at 4-cell stage, the foster in both groups gave birth to offspring. Therefore, the present study suggests that the telophase ooplasm could properly reprogram the genome of somatic cells, produce Asian yellow goat cloned embryos and viable kids, and whole cell intracytoplasmic injection is an efficient protocol for goat somatic cell nuclear transfer.


Assuntos
Clonagem de Organismos/métodos , Cabras/genética , Técnicas de Transferência Nuclear , Animais , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Microinjeções , Oócitos/metabolismo , Telófase
9.
Mol Reprod Dev ; 74(4): 412-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17034044

RESUMO

Interspecies nuclear transfer is an invalulable tool for studying nucleus-cytoplasm interactions; and at the same time, it provides a possible alternative to clone endangered animals whose oocytes are difficult to obtain. In the present study, we investigated the possibility of cloning Tibetan antelope embryos using abattoir-derived caprine oocytes as recipients. Effects of culture conditions, enucleation timing, and donor cell passages on the in vitro development of Tibetan antelope-goat cloned embryos were studied. Maternal to zygotic transition timing of interspecies Tibetan antelope embryos was also investigated using two types of cloned embryos, Tibetan antelope-rabbit and Tibetan antelope-goat embryos. Our results indicate that: (1) goat oocyte is able to reprogram somatic cells of different genus and supports development to blastocyst in vitro. (2) Coculture system supported the development of Tibetan antelope-goat embryos to blastocyst rate stage (4.0%), while CR1aa alone did not. (3) When MII phase enucleated caprine cytoplast and TII phase enucleated caprine cytoplast were used as recipients, the fusion rate and blastocyst rate of hybrid embryos were not statistically different (73.9% vs. 67.4%; 4.0% vs. 1.1%). (4) When donor cells at 3-8 passages were used, 2.9% hybrid embryos developed to blastocysts, while none developed to blastocysts when cells at 10-17 passages were used. (5) There may be a morula-to-blastocyst block for Tibetan antelope-goat, while there may be an 8- to 16-cell block for Tibetan antelope-rabbit embryos.


Assuntos
Antílopes , Clonagem de Organismos/métodos , Cabras , Técnicas de Transferência Nuclear , Animais , Células Cultivadas , Cromossomos de Mamíferos/química , Técnicas de Cultura Embrionária , Transferência Embrionária , Feminino , Oócitos/efeitos dos fármacos , Oócitos/crescimento & desenvolvimento , Gravidez , Prenhez , Coelhos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA