Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Cell Dev Biol ; 26: 667-87, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20604707

RESUMO

The past few years have witnessed remarkable advances in stem cell biology and human genetics, and we have arrived at an era in which patient-specific cell and tissue models are now practical. The recent identification of cardiovascular progenitor cells, as well as the identification of genetic variants underlying congenital heart disorders and adult disease, opens the door to the development of human models of human cardiovascular disease. We review the current understanding of the contribution of progenitor cells to cardiogenesis and outline how pluripotent stem cells can be applied to the modeling of cardiovascular disorders of genetic origin. A key challenge will be to implement these models in an efficient manner to develop a molecular understanding of how genes lead to disease and to screen for genes and drugs that modify the disease process.


Assuntos
Cardiopatias/patologia , Coração/embriologia , Modelos Cardiovasculares , Miocárdio/citologia , Células-Tronco , Animais , Cardiopatias/genética , Proteínas de Homeodomínio/genética , Humanos , Proteínas com Homeodomínio LIM , Fatores de Transcrição
2.
Circ Res ; 123(9): 1066-1079, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30355156

RESUMO

RATIONALE: Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are a readily available, robustly reproducible, and physiologically appropriate human cell source for cardiac disease modeling, drug discovery, and toxicity screenings in vitro. However, unlike adult myocardial cells in vivo, hPSC-CMs cultured in vitro maintain an immature metabolic phenotype, where majority of ATP is produced through aerobic glycolysis instead of oxidative phosphorylation in the mitochondria. Little is known about the underlying signaling pathways controlling hPSC-CMs' metabolic and functional maturation. OBJECTIVE: To define the molecular pathways controlling cardiomyocytes' metabolic pathway selections and improve cardiomyocyte metabolic and functional maturation. METHODS AND RESULTS: We cultured hPSC-CMs in different media compositions including glucose-containing media, glucose-containing media supplemented with fatty acids, and glucose-free media with fatty acids as the primary carbon source. We found that cardiomyocytes cultured in the presence of glucose used primarily aerobic glycolysis and aberrantly upregulated HIF1α (hypoxia-inducible factor 1α) and its downstream target lactate dehydrogenase A. Conversely, glucose deprivation promoted oxidative phosphorylation and repressed HIF1α. Small molecule inhibition of HIF1α or lactate dehydrogenase A resulted in a switch from aerobic glycolysis to oxidative phosphorylation. Likewise, siRNA inhibition of HIF1α stimulated oxidative phosphorylation while inhibiting aerobic glycolysis. This metabolic shift was accompanied by an increase in mitochondrial content and cellular ATP levels. Furthermore, functional gene expressions, sarcomere length, and contractility were improved by HIF1α/lactate dehydrogenase A inhibition. CONCLUSIONS: We show that under standard culture conditions, the HIF1α-lactate dehydrogenase A axis is aberrantly upregulated in hPSC-CMs, preventing their metabolic maturation. Chemical or siRNA inhibition of this pathway results in an appropriate metabolic shift from aerobic glycolysis to oxidative phosphorylation. This in turn improves metabolic and functional maturation of hPSC-CMs. These findings provide key insight into molecular control of hPSC-CMs' metabolism and may be used to generate more physiologically mature cardiomyocytes for drug screening, disease modeling, and therapeutic purposes.


Assuntos
Aminoquinolinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Dissulfetos/farmacologia , Metabolismo Energético/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Alcaloides Indólicos/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , L-Lactato Desidrogenase/antagonistas & inibidores , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Sulfonamidas/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Glicólise/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Pluripotentes Induzidas/enzimologia , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Cardíacas/genética , Miócitos Cardíacos/enzimologia , Fosforilação Oxidativa/efeitos dos fármacos , Fenótipo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Biotechnol Bioeng ; 116(11): 3098-3111, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31317531

RESUMO

Here we propose a bio-MEMS device designed to evaluate contractile force and conduction velocity of cell sheets in response to mechanical and electrical stimulation of the cell source as it grows to form a cellular sheet. Moreover, the design allows for the incorporation of patient-specific data and cell sources. An optimized device would allow cell sheets to be cultured, characterized, and conditioned to be compatible with a specific patient's cardiac environment in vitro, before implantation. This design draws upon existing methods in the literature but makes an important advance by combining the mechanical and electrical stimulation into a single system for optimized cell sheet growth. The device has been designed to achieve cellular alignment, electrical stimulation, mechanical stimulation, conduction velocity readout, contraction force readout, and eventually cell sheet release. The platform is a set of comb electrical contacts consisting of three-dimensional walls made of polydimethylsiloxane and coated with electrically conductive metals on the tops of the walls. Not only do the walls serve as a method for stimulating cells that are attached to the top, but their geometry is tailored such that they are flexible enough to be bent by the cells and used to measure force. The platform can be stretched via a linear actuator setup, allowing for simultaneous electrical and mechanical stimulation that can be derived from patient-specific clinical data.


Assuntos
Sistemas Microeletromecânicos , Contração Miocárdica , Miocárdio/metabolismo , Engenharia Tecidual/instrumentação , Animais , Estimulação Elétrica , Humanos
4.
Biotechnol Bioeng ; 115(3): 630-644, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29178315

RESUMO

Three-dimensional (3D) cultures of human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) hold great promise for drug discovery, providing a better approximation to the in vivo physiology over standard two-dimensional (2D) monolayer cultures. However, the transition of CM differentiation protocols from 2D to 3D cultures is not straightforward. In this work, we relied on the aggregation of hPSC-derived cardiac progenitors and their culture under agitated conditions to generate highly pure cardiomyocyte aggregates. Whole-transcriptome analysis and 13 C-metabolic flux analysis allowed to demonstrate at both molecular and fluxome levels that such 3D culture environment enhances metabolic maturation of hiPSC-CMs. When compared to 2D, 3D cultures of hiPSC-CMs displayed down-regulation of genes involved in glycolysis and lipid biosynthesis and increased expression of genes involved in OXPHOS. Accordingly, 3D cultures of hiPSC-CMs had lower fluxes through glycolysis and fatty acid synthesis and increased TCA-cycle activity. Importantly, we demonstrated that the 3D culture environment reproducibly improved both CM purity and metabolic maturation across different hPSC lines, thereby providing a robust strategy to derive enriched hPSC-CMs with metabolic features closer to that of adult CMs.


Assuntos
Técnicas de Cultura de Células/métodos , Glicólise , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Metabolismo dos Lipídeos , Miócitos Cardíacos/metabolismo , Fosforilação Oxidativa , Linhagem Celular , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia
5.
Circulation ; 132(2): 109-21, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-25995316

RESUMO

BACKGROUND: Heart development is tightly regulated by signaling events acting on a defined number of progenitor and differentiated cardiac cells. Although loss of function of these signaling pathways leads to congenital malformation, the consequences of cardiac progenitor cell or embryonic cardiomyocyte loss are less clear. In this study, we tested the hypothesis that embryonic mouse hearts exhibit a robust mechanism for regeneration after extensive cell loss. METHODS AND RESULTS: By combining a conditional cell ablation approach with a novel blastocyst complementation strategy, we generated murine embryos that exhibit a full spectrum of cardiac progenitor cell or cardiomyocyte ablation. Remarkably, ablation of up to 60% of cardiac progenitor cells at embryonic day 7.5 was well tolerated and permitted embryo survival. Ablation of embryonic cardiomyocytes to a similar degree (50% to 60%) at embryonic day 9.0 could be fully rescued by residual myocytes with no obvious adult cardiac functional deficit. In both ablation models, an increase in cardiomyocyte proliferation rate was detected and accounted for at least some of the rapid recovery of myocardial cellularity and heart size. CONCLUSION: Our study defines the threshold for cell loss in the embryonic mammalian heart and reveals a robust cardiomyocyte compensatory response that sustains normal fetal development.


Assuntos
Proliferação de Células/fisiologia , Células-Tronco Embrionárias/fisiologia , Coração Fetal/citologia , Miócitos Cardíacos/fisiologia , Animais , Contagem de Células/métodos , Coração Fetal/crescimento & desenvolvimento , Técnicas de Introdução de Genes , Camundongos , Camundongos Transgênicos
6.
Development ; 140(20): 4165-76, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24026118

RESUMO

In mammals, cardiac development proceeds from the formation of the linear heart tube, through complex looping and septation, all the while increasing in mass to provide the oxygen delivery demands of embryonic growth. The developing heart must orchestrate regional differences in cardiomyocyte proliferation to control cardiac morphogenesis. During ventricular wall formation, the compact myocardium proliferates more vigorously than the trabecular myocardium, but the mechanisms controlling such regional differences among cardiomyocyte populations are not understood. Control of definitive cardiomyocyte proliferation is of great importance for application to regenerative cell-based therapies. We have used murine and human pluripotent stem cell systems to demonstrate that, during in vitro cellular differentiation, early ventricular cardiac myocytes display a robust proliferative response to ß-catenin-mediated signaling and conversely accelerate differentiation in response to inhibition of this pathway. Using gain- and loss-of-function murine genetic models, we show that ß-catenin controls ventricular myocyte proliferation during development and the perinatal period. We further demonstrate that the differential activation of the Wnt/ß-catenin signaling pathway accounts for the observed differences in the proliferation rates of the compact versus the trabecular myocardium during normal cardiac development. Collectively, these results provide a mechanistic explanation for the differences in localized proliferation rates of cardiac myocytes and point to a practical method for the generation of the large numbers of stem cell-derived cardiac myocytes necessary for clinical applications.


Assuntos
Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Ativação Enzimática , Regulação da Expressão Gênica no Desenvolvimento , Ventrículos do Coração/embriologia , Humanos , Camundongos , Morfogênese , Miócitos Cardíacos/metabolismo
7.
Circ Res ; 112(6): 900-10, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23307821

RESUMO

RATIONALE: Cardiogenesis is regulated by a complex interplay between transcription factors. However, little is known about how these interactions regulate the transition from mesodermal precursors to cardiac progenitor cells (CPCs). OBJECTIVE: To identify novel regulators of mesodermal cardiac lineage commitment. METHODS AND RESULTS: We performed a bioinformatic-based transcription factor binding site analysis on upstream promoter regions of genes that are enriched in embryonic stem cell-derived CPCs. From 32 candidate transcription factors screened, we found that Yin Yang 1 (YY1), a repressor of sarcomeric gene expression, is present in CPCs in vivo. Interestingly, we uncovered the ability of YY1 to transcriptionally activate Nkx2.5, a key marker of early cardiogenic commitment. YY1 regulates Nkx2.5 expression via a 2.1-kb cardiac-specific enhancer as demonstrated by in vitro luciferase-based assays, in vivo chromatin immunoprecipitation, and genome-wide sequencing analysis. Furthermore, the ability of YY1 to activate Nkx2.5 expression depends on its cooperative interaction with Gata4 at a nearby chromatin. Cardiac mesoderm-specific loss-of-function of YY1 resulted in early embryonic lethality. This was corroborated in vitro by embryonic stem cell-based assays in which we showed that the overexpression of YY1 enhanced the cardiogenic differentiation of embryonic stem cells into CPCs. CONCLUSIONS: These results demonstrate an essential and unexpected role for YY1 to promote cardiogenesis as a transcriptional activator of Nkx2.5 and other CPC-enriched genes.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Proteínas de Homeodomínio/metabolismo , Mioblastos Cardíacos/citologia , Fatores de Transcrição/metabolismo , Fator de Transcrição YY1/fisiologia , Animais , Diferenciação Celular/genética , Fator de Transcrição GATA4/metabolismo , Estudo de Associação Genômica Ampla/métodos , Proteína Homeobox Nkx-2.5 , Camundongos , Mioblastos Cardíacos/química , Ativação Transcricional/fisiologia , Fator de Transcrição YY1/análise , Fator de Transcrição YY1/genética
8.
Nature ; 460(7251): 113-7, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19571884

RESUMO

The generation and expansion of diverse cardiovascular cell lineages is a critical step during human cardiogenesis, with major implications for congenital heart disease. Unravelling the mechanisms for the diversification of human heart cell lineages has been hampered by the lack of genetic tools to purify early cardiac progenitors and define their developmental potential. Recent studies in the mouse embryo have identified a multipotent cardiac progenitor that contributes to all of the major cell types in the murine heart. In contrast to murine development, human cardiogenesis has a much longer onset of heart cell lineage diversification and expansion, suggesting divergent pathways. Here we identify a diverse set of human fetal ISL1(+) cardiovascular progenitors that give rise to the cardiomyocyte, smooth muscle and endothelial cell lineages. Using two independent transgenic and gene-targeting approaches in human embryonic stem cell lines, we show that purified ISL1(+) primordial progenitors are capable of self-renewal and expansion before differentiation into the three major cell types in the heart. These results lay the foundation for the generation of human model systems for cardiovascular disease and novel approaches for human regenerative cardiovascular medicine.


Assuntos
Linhagem da Célula , Proteínas de Homeodomínio/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Miocárdio/citologia , Diferenciação Celular , Divisão Celular , Linhagem Celular , Técnicas de Cocultura , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , Feto/citologia , Feto/embriologia , Coração/embriologia , Humanos , Proteínas com Homeodomínio LIM , Músculo Liso/citologia , Miócitos Cardíacos/citologia , Fatores de Transcrição , Proteínas Wnt/metabolismo , Proteína Wnt3
9.
Stem Cells ; 31(12): 2587-98, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23843322

RESUMO

Advanced heart failure represents a leading public health problem in the developed world. The clinical syndrome results from the loss of viable and/or fully functional myocardial tissue. Designing new approaches to augment the number of functioning human cardiac muscle cells in the failing heart serve as the foundation of modern regenerative cardiovascular medicine. A number of clinical trials have been performed in an attempt to increase the number of functional myocardial cells by the transplantation of a diverse group of stem or progenitor cells. Although there are some encouraging suggestions of a small early therapeutic benefit, to date, no evidence for robust cell or tissue engraftment has been shown, emphasizing the need for new approaches. Clinically meaningful cardiac regeneration requires the identification of the optimum cardiogenic cell types and their assembly into mature myocardial tissue that is functionally and electrically coupled to the native myocardium. We here review recent advances in stem cell biology and tissue engineering and describe how the convergence of these two fields may yield novel approaches for cardiac regeneration.


Assuntos
Células-Tronco Embrionárias/transplante , Miócitos Cardíacos/fisiologia , Miócitos Cardíacos/transplante , Engenharia Tecidual/métodos , Animais , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Humanos , Miócitos Cardíacos/citologia
10.
Nature ; 454(7200): 109-13, 2008 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-18568026

RESUMO

The heart is formed from cardiogenic progenitors expressing the transcription factors Nkx2-5 and Isl1 (refs 1 and 2). These multipotent progenitors give rise to cardiomyocyte, smooth muscle and endothelial cells, the major lineages of the mature heart. Here we identify a novel cardiogenic precursor marked by expression of the transcription factor Wt1 and located within the epicardium-an epithelial sheet overlying the heart. During normal murine heart development, a subset of these Wt1(+) precursors differentiated into fully functional cardiomyocytes. Wt1(+) proepicardial cells arose from progenitors that express Nkx2-5 and Isl1, suggesting that they share a developmental origin with multipotent Nkx2-5(+) and Isl1(+) progenitors. These results identify Wt1(+) epicardial cells as previously unrecognized cardiomyocyte progenitors, and lay the foundation for future efforts to harness the cardiogenic potential of these progenitors for cardiac regeneration and repair.


Assuntos
Linhagem da Célula , Coração/embriologia , Miócitos Cardíacos/citologia , Pericárdio/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Pericárdio/embriologia , Pericárdio/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo
11.
iScience ; 25(2): 103806, 2022 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-35198871

RESUMO

Dynamic profiling of changes in gene expression in response to stressors in specific microenvironments without requiring cellular destruction remains challenging. Current methodologies that seek to interrogate gene expression at a molecular level require sampling of cellular transcriptome and therefore lysis of the cell, preventing serial analysis of cellular transcriptome. To address this area of unmet need, we have recently developed a technology allowing transcriptomic analysis over time without cellular destruction. Our method, TRACE-seq (TRanscriptomic Analysis Captured in Extracellular vesicles using sequencing), is characterized by a cell-type specific transgene expression. It provides data on the transcriptome inside extracellular vesicles that provides an accurate representation of stress-responsive cellular transcriptomic changes. Thus, the transcriptome of cells expressing TRACE can be followed over time without destroying the source cell, which is a powerful tool for many fields of fundamental and translational biology research.

12.
iScience ; 23(3): 100959, 2020 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-32179481

RESUMO

Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfß/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV.

13.
Elife ; 82019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31591966

RESUMO

A fundamental goal in the biological sciences is to determine how individual cells with varied gene expression profiles and diverse functional characteristics contribute to development, physiology, and disease. Here, we report a novel strategy to assess gene expression and cell physiology in single living cells. Our approach utilizes fluorescently labeled mRNA-specific anti-sense RNA probes and dsRNA-binding protein to identify the expression of specific genes in real-time at single-cell resolution via FRET. We use this technology to identify distinct myocardial subpopulations expressing the structural proteins myosin heavy chain α and myosin light chain 2a in real-time during early differentiation of human pluripotent stem cells. We combine this live-cell gene expression analysis with detailed physiologic phenotyping to capture the functional evolution of these early myocardial subpopulations during lineage specification and diversification. This live-cell mRNA imaging approach will have wide ranging application wherever heterogeneity plays an important biological role.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Perfilação da Expressão Gênica/métodos , Microscopia Intravital/métodos , Análise de Célula Única/métodos , Diferenciação Celular , Humanos , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/fisiologia , Coloração e Rotulagem/métodos
14.
Acta Biomater ; 83: 211-220, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30352286

RESUMO

Post-cardiac surgical sternal and epicardial adhesions increase the risk and complexity of cardiac re-operative surgeries, which represent a significant challenge for patients with the congenital cardiac disease. Bioresorbable membranes can serve as barriers to prevent postoperative adhesions. Herein, we fabricated a bioresorbable gelatin/polycaprolactone (GT/PCL) composite membrane via electrospinning. The membrane was characterized in terms of morphology, mechanical properties, and biocompatibility. We then evaluated its efficacy as a physical barrier to prevent cardiac operative adhesions in a rabbit model. Our results showed that the membrane had a nanofibrous structure and was sturdy enough to be handled for the surgical procedures. In vitro studies with rabbit cardiac fibroblasts demonstrated that the membrane was biocompatible and inhibited cell infiltration. Further application of the membrane in a rabbit cardiac adhesion model revealed that the membrane was resorbed gradually and effectively resisted the sternal and epicardial adhesions. Interestingly, six months after the operation, the GT/PCL membrane was completely resorbed with simultaneous ingrowth of host cells to form a natural barrier. Collectively, these results indicated that the GT/PCL membrane might be a suitable barrier to prevent sternal and epicardial adhesions and might be utilized as a novel pericardial substitute for cardiac surgery. STATEMENT OF SIGNIFICANCE: Electrospinning is a versatile method to prepare nanofibrous membranes for tissue engineering and regenerative medicine applications. However, with the micro-/nano-scale structure and high porosity, the electrospun membrane might be an excellent candidate as a barrier to prevent postoperative adhesion. Here we prepared an electropun GT/PCL nanofibrous membrane and applied it as a barrier to prevent sternal and epicardial adhesions. Our results showed that the membrane had sufficient mechanical strength, good biocompatibility, and effectively resisted the sternal and epicardial adhesions. What's more, the membrane was bioresorbable and allowed simultaneous ingrowth of host cells to form a natural barrier. We believe that the current will inspire more research on nanomaterials to prevent postoperative adhesion applications.


Assuntos
Materiais Biocompatíveis , Gelatina , Membranas Artificiais , Nanofibras , Poliésteres , Aderências Teciduais/prevenção & controle , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Gelatina/química , Gelatina/farmacologia , Miocárdio/metabolismo , Miocárdio/patologia , Nanofibras/química , Nanofibras/uso terapêutico , Poliésteres/química , Poliésteres/farmacologia , Coelhos , Aderências Teciduais/metabolismo , Aderências Teciduais/patologia
15.
PLoS One ; 13(10): e0205108, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30281646

RESUMO

Understanding the molecular pathways regulating cardiogenesis is crucial for the early diagnosis of heart diseases and improvement of cardiovascular disease. During normal mammalian cardiac development, collagen and calcium-binding EGF domain-1 (Ccbe1) is expressed in the first and second heart field progenitors as well as in the proepicardium, but its role in early cardiac commitment remains unknown. Here we demonstrate that during mouse embryonic stem cell (ESC) differentiation Ccbe1 is upregulated upon emergence of Isl1- and Nkx2.5- positive cardiac progenitors. Ccbe1 is markedly enriched in Isl1-positive cardiac progenitors isolated from ESCs differentiating in vitro or embryonic hearts developing in vivo. Disruption of Ccbe1 activity by shRNA knockdown or blockade with a neutralizing antibody results in impaired differentiation of embryonic stem cells along the cardiac mesoderm lineage resulting in a decreased expression of mature cardiomyocyte markers. In addition, knockdown of Ccbe1 leads to smaller embryoid bodies. Collectively, our results show that CCBE1 is essential for the commitment of cardiac mesoderm and consequently, for the formation of cardiac myocytes in differentiating mouse ESCs.


Assuntos
Proteínas de Ligação ao Cálcio/deficiência , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias Murinas/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Supressoras de Tumor/deficiência , Animais , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Coração/embriologia , Proteína Homeobox Nkx-2.5/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , RNA Interferente Pequeno , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética
16.
PLoS One ; 13(4): e0196697, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29698509

RESUMO

AIMS: Atrial natriuretic peptide (ANP), secreted primarily by atrial cardiomyocytes, decreases blood pressure by raising cyclic 3',5'-guanosine monophosphate (cGMP) levels and inducing vasorelaxation, natriuresis, and diuresis. Raising the level of ANP has been shown to be an effective treatment for hypertension. To advance the future development of an anti-microRNA (miR) approach to increasing expression of ANP, we investigated the regulation of NPPA expression by two miRs: miR-425 and miR-155. We examined whether miR-425 and miR-155 have an additive effect on the expression and function of ANP. METHODS AND RESULTS: Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were transfected with miR-425, miR-155, or a combination of the two miRs. Two days later, NPPA expression was measured using real time qPCR. Each of the miRs decreased NPPA expression over a wide range of concentrations, with a significant reduction at concentrations as low as 1 nM. The combination of miR-425 and miR-155 reduced NPPA expression to a greater extent than either miR-425 or miR-155 alone. An in vitro assay was developed to study the potential biological significance of the miR-induced decrease in NPPA expression. The cooperative effect of miR-425 and miR-155 on NPPA expression was associated with a significant decrease in cGMP levels. CONCLUSIONS: These data demonstrate that miR-425 and miR-155 regulate NPPA expression in a cooperative manner. Targeting both miRNAs with anti-miRs (possibly at submaximal concentrations) might prove to be a more effective strategy to modulate ANP levels, and thus blood pressure, than targeting either miRNA alone.


Assuntos
Fator Natriurético Atrial/metabolismo , GMP Cíclico/metabolismo , MicroRNAs/metabolismo , Animais , Fator Natriurético Atrial/genética , Células COS , Linhagem Celular , Chlorocebus aethiops , Células-Tronco Embrionárias Humanas/citologia , Humanos , MicroRNAs/genética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Transfecção
17.
Trends Cell Biol ; 27(5): 352-364, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28007424

RESUMO

The advent of human pluripotent stem cell (hPSC) biology has opened unprecedented opportunities for the use of tissue engineering to generate human cardiac tissue for in vitro study. Engineering cardiac constructs that recapitulate human development and disease requires faithful recreation of the cardiac niche in vitro. Here we discuss recent progress in translating the in vivo cardiac microenvironment into PSC models of the human heart. We review three key physiologic features required to recreate the cardiac niche and facilitate normal cardiac differentiation and maturation: the biochemical, biophysical, and bioelectrical signaling cues. Finally, we discuss key barriers that must be overcome to fulfill the promise of stem cell biology in preclinical applications and ultimately in clinical practice.


Assuntos
Microambiente Celular , Doença , Modelos Biológicos , Fenômenos Fisiológicos , Células-Tronco Pluripotentes/citologia , Humanos
18.
Adv Healthc Mater ; 6(2)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27774763

RESUMO

In this essay the authors argue that chamber pressure dominates the biomechanics of the contraction cycle of the heart, while tissue stiffness dominates the relaxation cycle. This appears to be an under-recognized challenge in cardiac tissue engineering. Optimal approaches will involve constructing chambers or modulating the stiffness of the scaffold/substrate in synchrony with the beating cycle.


Assuntos
Miocárdio , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Humanos
19.
Curr Protoc Stem Cell Biol ; 43: 1F.20.1-1F.20.9, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29140569

RESUMO

Human pluripotent stem-cell derived cardiomyocytes (hPSC-CMs) hold great promise for applications in human disease modeling, drug discovery, cardiotoxicity screening, and, ultimately, regenerative medicine. The ability to study multiple parameters of hPSC-CM function, such as contractile and electrical activity, calcium cycling, and force generation, is therefore of paramount importance. hPSC-CMs cultured on stiff substrates like glass or polystyrene do not have the ability to shorten during contraction, making them less suitable for the study of hPSC-CM contractile function. Other approaches require highly specialized hardware and are difficult to reproduce. Here we describe a protocol for the preparation of hPSC-CMs on soft substrates that enable shortening, and subsequently the simultaneous quantitative analysis of their contractile and electrical activity, calcium cycling, and force generation at single-cell resolution. This protocol requires only affordable and readily available materials and works with standard imaging hardware. © 2017 by John Wiley & Sons, Inc.


Assuntos
Técnicas de Cultura de Células/métodos , Miócitos Cardíacos , Células-Tronco Pluripotentes , Vidro/química , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Poliestirenos/química
20.
Sci Rep ; 7(1): 8590, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28819274

RESUMO

The immature phenotype of human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) constrains their potential in cell therapy and drug testing. In this study, we report that shifting hPSC-CMs from glucose-containing to galactose- and fatty acid-containing medium promotes their fast maturation into adult-like CMs with higher oxidative metabolism, transcriptional signatures closer to those of adult ventricular tissue, higher myofibril density and alignment, improved calcium handling, enhanced contractility, and more physiological action potential kinetics. Integrated "-Omics" analyses showed that addition of galactose to culture medium improves total oxidative capacity of the cells and ameliorates fatty acid oxidation avoiding the lipotoxicity that results from cell exposure to high fatty acid levels. This study provides an important link between substrate utilization and functional maturation of hPSC-CMs facilitating the application of this promising cell type in clinical and preclinical applications.


Assuntos
Carbono/farmacologia , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/metabolismo , Biomarcadores/metabolismo , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Ácidos Graxos/toxicidade , Galactose/farmacologia , Glucose/deficiência , Glicólise/efeitos dos fármacos , Ventrículos do Coração/citologia , Humanos , Cinética , Lactose/farmacologia , Modelos Biológicos , Miócitos Cardíacos/efeitos dos fármacos , Oxirredução , Fosforilação Oxidativa/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/ultraestrutura , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA