Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 189: 52-65, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38346641

RESUMO

Adipocytes normally accumulate in the epicardial and pericardial layers around the human heart, but their infiltration into the myocardium can be proarrhythmic. METHODS AND RESULTS: Human adipose derived stem/stromal cells and human induced pluripotent stem cells (hiPSC) were differentiated, respectively into predominantly white fat-like adipocytes (hAdip) and ventricular cardiomyocytes (CMs). Adipocytes cultured in CM maintenance medium (CM medium) maintained their morphology, continued to express adipogenic markers, and retained clusters of intracellular lipid droplets. In contrast, hiPSC-CMs cultivated in adipogenic growth medium displayed abnormal cell morphologies and more clustering across the monolayer. Pre-plated hiPSC-CMs co-cultured in direct contact with hAdips in CM medium displayed prolonged action potential durations, increased triangulation, slowed conduction velocity, increased conduction velocity heterogeneity, and prolonged calcium transients. When hAdip-conditioned medium was added to monolayer cultures of hiPSC-CMs, results similar to those recorded with direct co-cultures were observed. Both co-culture and conditioned medium experiments resulted in increases in transcript abundance of SCN10A, CACNA1C, SLC8A1, and RYR2, with a decrease in KCNJ2. Human adipokine immunoblots revealed the presence of cytokines that were elevated in adipocyte-conditioned medium, including MCP-1, IL-6, IL-8 and CFD that could induce electrophysiological changes in cultured hiPSC-CMs. CONCLUSIONS: Co-culture of hiPSC-CMs with hAdips reveals a potentially pathogenic role of infiltrating human adipocytes on myocardial tissue. In the absence of structural changes, hAdip paracrine release alone is sufficient to cause CM electrophysiological dysfunction mirroring the co-culture conditions. These effects, mediated largely by paracrine mechanisms, could promote arrhythmias in the heart.


Assuntos
Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Humanos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Diferenciação Celular/fisiologia , Adipócitos , Potenciais de Ação
2.
Genes (Basel) ; 14(10)2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37895213

RESUMO

Cardiomyopathies (CMPs) represent a significant healthcare burden and are a major cause of heart failure leading to premature death. Several CMPs are now recognized to have a strong genetic basis, including arrhythmogenic cardiomyopathy (ACM), which predisposes patients to arrhythmic episodes. Variants in one of the five genes (PKP2, JUP, DSC2, DSG2, and DSP) encoding proteins of the desmosome are known to cause a subset of ACM, which we classify as desmosome-related ACM (dACM). Phenotypically, this disease may lead to sudden cardiac death in young athletes and, during late stages, is often accompanied by myocardial fibrofatty infiltrates. While the pathogenicity of the desmosome genes has been well established through animal studies and limited supplies of primary human cells, these systems have drawbacks that limit their utility and relevance to understanding human disease. Human induced pluripotent stem cells (hiPSCs) have emerged as a powerful tool for modeling ACM in vitro that can overcome these challenges, as they represent a reproducible and scalable source of cardiomyocytes (CMs) that recapitulate patient phenotypes. In this review, we provide an overview of dACM, summarize findings in other model systems linking desmosome proteins with this disease, and provide an up-to-date summary of the work that has been conducted in hiPSC-cardiomyocyte (hiPSC-CM) models of dACM. In the context of the hiPSC-CM model system, we highlight novel findings that have contributed to our understanding of disease and enumerate the limitations, prospects, and directions for research to consider towards future progress.


Assuntos
Cardiomiopatias , Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Cardiomiopatias/metabolismo , Fenótipo
3.
Integr Biol (Camb) ; 13(10): 246-257, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34875067

RESUMO

The actomyosin cytoskeleton enables cells to resist deformation, crawl, change their shape and sense their surroundings. Despite decades of study, how its molecular constituents can assemble together to form a network with the observed mechanics of cells remains poorly understood. Recently, it has been shown that the actomyosin cortex of quiescent cells can undergo frequent, abrupt reconfigurations and displacements, called cytoquakes. Notably, such fluctuations are not predicted by current physical models of actomyosin networks, and their prevalence across cell types and mechanical environments has not previously been studied. Using micropost array detectors, we have performed high-resolution measurements of the dynamic mechanical fluctuations of cells' actomyosin cortex and stress fiber networks. This reveals cortical dynamics dominated by cytoquakes-intermittent events with a fat-tailed distribution of displacements, sometimes spanning microposts separated by 4 µm, in all cell types studied. These included 3T3 fibroblasts, where cytoquakes persisted over substrate stiffnesses spanning the tissue-relevant range of 4.3 kPa-17 kPa, and primary neonatal rat cardiac fibroblasts and myofibroblasts, human embryonic kidney cells and human bone osteosarcoma epithelial (U2OS) cells, where cytoquakes were observed on substrates in the same stiffness range. Overall, these findings suggest that the cortex self-organizes into a marginally stable mechanical state whose physics may contribute to cell mechanical properties, active behavior and mechanosensing.


Assuntos
Citoesqueleto de Actina , Actomiosina , Animais , Citoesqueleto , Microtúbulos , Ratos , Fibras de Estresse
4.
Dis Model Mech ; 14(8)2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34378773

RESUMO

Clinically pertinent electrocardiogram (ECG) data from model systems, such as zebrafish, are crucial for illuminating factors contributing to human cardiac electrophysiological abnormalities and disease. Current zebrafish ECG collection strategies have not adequately addressed the consistent acquisition of high-quality traces or sources of phenotypic variation that could obscure data interpretation. Thus, we developed a novel platform to ensure high-quality recording of in vivo subdermal adult zebrafish ECGs and zebrafish ECG reading GUI (zERG), a program to acquire measurements from traces that commercial software cannot examine owing to erroneous peak calling. We evaluate normal ECG trait variation, revealing highly reproducible intervals and wave amplitude variation largely driven by recording artifacts, and identify sex and body size as potential confounders to PR, QRS and QT intervals. With this framework, we characterize the effect of the class I anti-arrhythmic drug flecainide acetate on adults, provide support for the impact of a Long QT syndrome model, and establish power calculations for this and other studies. These results highlight our pipeline as a robust approach to evaluate zebrafish models of human cardiac electrophysiological phenotypes.


Assuntos
Síndrome do QT Longo , Peixe-Zebra , Animais , Eletrocardiografia/métodos , Peixe-Zebra/genética
5.
J Clin Med ; 10(14)2021 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-34300226

RESUMO

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a progressive heart condition which causes fibro-fatty myocardial scarring, ventricular arrhythmias, and sudden cardiac death. Most cases of ARVC can be linked to pathogenic mutations in the cardiac desmosome, but the pathophysiology is not well understood, particularly in early phases when arrhythmias can develop prior to structural changes. Here, we created a novel human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) model of ARVC from a patient with a c.2358delA variant in desmoglein-2 (DSG2). These DSG2-mutant (DSG2Mut) hiPSC-CMs were compared against two wildtype hiPSC-CM lines via immunostaining, RT-qPCR, Western blot, RNA-Seq, cytokine expression and optical mapping. Mutant cells expressed reduced DSG2 mRNA and had altered localization of desmoglein-2 protein alongside thinner, more disorganized myofibrils. No major changes in other desmosomal proteins were noted. There was increased pro-inflammatory cytokine expression that may be linked to canonical and non-canonical NFκB signaling. Action potentials in DSG2Mut CMs were shorter with increased upstroke heterogeneity, while time-to-peak calcium and calcium decay rate were reduced. These were accompanied by changes in ion channel and calcium handling gene expression. Lastly, suppressing DSG2 in control lines via siRNA allowed partial recapitulation of electrical anomalies noted in DSG2Mut cells. In conclusion, the aberrant cytoskeletal organization, cytokine expression, and electrophysiology found DSG2Mut hiPSC-CMs could underlie early mechanisms of disease manifestation in ARVC patients.

6.
Semin Cell Dev Biol ; 119: 49-60, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33952430

RESUMO

Cardiomyocyte (CM) maturation is the transformation of differentiated fetal CMs into adult CMs that involves changes in morphology, cell function and metabolism, and the transcriptome. This process is, however, incomplete and ultimately arrested in pluripotent stem cell-derived CMs (PSC-CMs) in culture, which hinders their broad biomedical application. For this reason, enormous efforts are currently being made with the goal of generating mature PSC-CMs. In this review, we summarize key aspects of maturation observed in native CMs and discuss recent findings on the factors and mechanisms that regulate the process. Particular emphasis is put on transcriptional regulation and single-cell RNA-sequencing analysis that has emerged as a key tool to study time-series gene regulation and to determine the maturation state. We then discuss different biomimetic strategies to enhance PSC-CM maturation and discuss their effects at the single cell transcriptomic and functional levels.


Assuntos
Miócitos Cardíacos/fisiologia , Engenharia Tecidual/métodos , Transcriptoma/fisiologia , Diferenciação Celular , Humanos
7.
Sci Rep ; 11(1): 4430, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33627695

RESUMO

Interactions between cardiac myofibroblasts and myocytes may slow conduction and generate spontaneous beating in fibrosis, increasing the chance of life-threatening arrhythmia. While co-culture studies have shown that myofibroblasts can affect cardiomyocyte electrophysiology in vitro, the extent of myofibroblast-myocyte electrical conductance in a syncytium is unknown. In this neonatal rat study, cardiac myofibroblasts were transduced with Channelrhodopsin-2, which allowed acute and selective increase of myofibroblast current, and plated on top of cardiomyocytes. Optical mapping revealed significantly decreased conduction velocity (- 27 ± 6%, p < 10-3), upstroke rate (- 13 ± 4%, p = 0.002), and action potential duration (- 14 ± 7%, p = 0.004) in co-cultures when 0.017 mW/mm2 light was applied, as well as focal spontaneous beating in 6/7 samples and a decreased cycle length (- 36 ± 18%, p = 0.002) at 0.057 mW/mm2 light. In silico modeling of the experiments reproduced the experimental findings and suggested the light levels used in experiments produced excess current similar in magnitude to endogenous myofibroblast current. Fitting the model to experimental data predicted a tissue-level electrical conductance across the 3-D interface between myofibroblasts and cardiomyocytes of ~ 5 nS/cardiomyocyte, and showed how increased myofibroblast-myocyte conductance, increased myofibroblast/myocyte capacitance ratio, and increased myofibroblast current, which occur in fibrosis, can work in tandem to produce pro-arrhythmic increases in conduction and spontaneous beating.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Miócitos Cardíacos/patologia , Miofibroblastos/patologia , Potenciais de Ação/fisiologia , Animais , Arritmias Cardíacas/fisiopatologia , Eletrofisiologia Cardíaca/métodos , Células Cultivadas , Técnicas de Cocultura/métodos , Fibrose/fisiopatologia , Frequência Cardíaca/fisiologia , Optogenética/métodos , Ratos
8.
Sci Rep ; 10(1): 8387, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32433563

RESUMO

Cardiac tissue engineering strategies have the potential to regenerate functional myocardium following myocardial infarction. In this study, we utilized novel electrospun fibrin microfiber sheets of different stiffnesses (50.0 ± 11.2 kPa and 90.0 ± 16.4 kPa) to engineer biomimetic models of vascularized cardiac tissues. We characterized tissue assembly, electrophysiology, and contractility of neonatal rat ventricular cardiomyocytes (NRVCMs) cultured on these sheets. NRVCMs cultured on the softer substrates displayed higher conduction velocities (CVs) and improved electrophysiological properties. Human umbilical vein endothelial cells (HUVECs) formed dense networks on the sheets when co-cultured with human adipose-derived stem/stromal cells (hASCs). To achieve vascularized cardiac tissues, we tested various tri-culture protocols of NRVCM:hASC:HUVEC and found that a ratio of 1,500,000:37,500:150,000 cells/cm2 enabled the formation of robust endothelial networks while retaining statistically identical electrophysiological characteristics to NRVCM-only cultures. Tri-cultures at this ratio on 90 kPa substrates exhibited average CVs of 14 ± 0.6 cm/s, Action Potential Duration (APD)80 and APD30 of 152 ± 11 ms and 71 ± 6 ms, respectively, and maximum capture rate (MCR) of 3.9 ± 0.7 Hz. These data indicate the significant potential of generating densely packed endothelial networks together with electrically integrated cardiac cells in vitro as a physiologic 3D cardiac model.


Assuntos
Eletrofisiologia Cardíaca/métodos , Adipócitos/citologia , Animais , Biomimética/métodos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Ratos , Células-Tronco/citologia , Células-Tronco/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo
9.
Nano Lett ; 20(3): 1561-1570, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-31845810

RESUMO

Matrix nanotopographical cues are known to regulate the structure and function of somatic cells derived from human pluripotent stem cell (hPSC) sources. High-throughput electrophysiological analysis of excitable cells derived from hPSCs is possible via multielectrode arrays (MEAs) but conventional MEA platforms use flat substrates and do not reproduce physiologically relevant tissue-specific architecture. To address this issue, we developed a high-throughput nanotopographically patterned multielectrode array (nanoMEA) by integrating conductive, ion-permeable, nanotopographic patterns with 48-well MEA plates, and investigated the effect of substrate-mediated cytoskeletal organization on hPSC-derived cardiomyocyte and neuronal function at scale. Using our nanoMEA platform, we found patterned hPSC-derived cardiac monolayers exhibit both enhanced structural organization and greater sensitivity to treatment with calcium blocking or conduction inhibiting compounds when subjected to high-throughput dose-response studies. Similarly, hPSC-derived neurons grown on nanoMEA substrates exhibit faster migration and neurite outgrowth speeds, greater colocalization of pre- and postsynaptic markers, and enhanced cell-cell communication only revealed through examination of data sets derived from multiple technical replicates. The presented data highlight the nanoMEA as a new tool to facilitate high-throughput, electrophysiological analysis of ordered cardiac and neuronal monolayers, which can have important implications for preclinical analysis of excitable cell function.


Assuntos
Diferenciação Celular , Fenômenos Eletrofisiológicos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Eletrodos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Neurônios/citologia
10.
Circulation ; 140(18): 1491-1505, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31533459

RESUMO

BACKGROUND: Inflammation is a prominent feature of arrhythmogenic cardiomyopathy (ACM), but whether it contributes to the disease phenotype is not known. METHODS: To define the role of inflammation in the pathogenesis of ACM, we characterized nuclear factor-κB signaling in ACM models in vitro and in vivo and in cardiac myocytes from patient induced pluripotent stem cells. RESULTS: Activation of nuclear factor-κB signaling, indicated by increased expression and nuclear accumulation of phospho-RelA/p65, occurred in both an in vitro model of ACM (expression of JUP2157del2 in neonatal rat ventricular myocytes) and a robust murine model of ACM (homozygous knock-in of mutant desmoglein-2 [Dsg2mut/mut]) that recapitulates the cardiac manifestations seen in patients with ACM. Bay 11-7082, a small-molecule inhibitor of nuclear factor-κB signaling, prevented the development of ACM disease features in vitro (abnormal redistribution of intercalated disk proteins, myocyte apoptosis, release of inflammatory cytokines) and in vivo (myocardial necrosis and fibrosis, left ventricular contractile dysfunction, electrocardiographic abnormalities). Hearts of Dsg2mut/mut mice expressed markedly increased levels of inflammatory cytokines and chemotactic molecules that were attenuated by Bay 11-7082. Salutary effects of Bay 11-7082 correlated with the extent to which production of selected cytokines had been blocked. Nuclear factor-κB signaling was also activated in cardiac myocytes derived from a patient with ACM. These cells produced and secreted abundant inflammatory cytokines under basal conditions, and this was also greatly reduced by Bay 11-7082. CONCLUSIONS: Inflammatory signaling is activated in ACM and drives key features of the disease. Targeting inflammatory pathways may be an effective new mechanism-based therapy for ACM.


Assuntos
Arritmias Cardíacas/metabolismo , Cardiomiopatias/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Animais , Arritmias Cardíacas/patologia , Cardiomiopatias/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Ratos Transgênicos , Ratos Wistar , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia
11.
J Pharmacol Toxicol Methods ; 99: 106612, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31319140

RESUMO

INTRODUCTION: Voltage-sensitive optical (VSO) sensors offer a minimally invasive method to study the time course of repolarization of the cardiac action potential (AP). This Comprehensive in vitro Proarrhythmia Assay (CiPA) cross-platform study investigates protocol design and measurement variability of VSO sensors for preclinical cardiac electrophysiology assays. METHODS: Three commercial and one academic laboratory completed a limited study of the effects of 8 blinded compounds on the electrophysiology of 2 commercial lines of human induced pluripotent stem-cell derived cardiomyocytes (hSC-CMs). Acquisition technologies included CMOS camera and photometry; fluorescent voltage sensors included di-4-ANEPPS, FluoVolt and genetically encoded QuasAr2. The experimental protocol was standardized with respect to cell lines, plating and maintenance media, blinded compounds, and action potential parameters measured. Serum-free media was used to study the action of drugs, but the exact composition and the protocols for cell preparation and drug additions varied among sites. RESULTS: Baseline AP waveforms differed across platforms and between cell types. Despite these differences, the relative responses to four selective ion channel blockers (E-4031, nifedipine, mexiletine, and JNJ 303 blocking IKr, ICaL, INa, and IKs, respectively) were similar across all platforms and cell lines although the absolute changes differed. Similarly, four mixed ion channel blockers (flecainide, moxifloxacin, quinidine, and ranolazine) had comparable effects in all platforms. Differences in repolarisation time course and response to drugs could be attributed to cell type and experimental method differences such as composition of the assay media, stimulated versus spontaneous activity, and single versus cumulative compound addition. DISCUSSION: In conclusion, VSOs represent a powerful and appropriate method to assess the electrophysiological effects of drugs on iPSC-CMs for the evaluation of proarrhythmic risk. Protocol considerations and recommendations are provided toward standardizing conditions to reduce variability of baseline AP waveform characteristics and drug responses.

12.
Br J Pharmacol ; 176(15): 2780-2794, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31074016

RESUMO

BACKGROUND AND PURPOSE: The electrophysiological properties of human pluripotent stem cell-derived cardiomyocytes (CMs) have not yet been characterized in a syncytial context. This study systematically characterized the contributions of different repolarizing potassium currents in human embryonic stem cell-derived CMs (hESC-CMs) during long-term culture as cell monolayers. EXPERIMENTAL APPROACH: The H9 hESC line was differentiated to CMs and plated to form confluent cell monolayers. Optical mapping was used to record the action potentials (APs) and conduction velocity (CV) during electrophysiological and pharmacological experiments. RT-PCR and Western blot were used to detect the presence and expression levels of ion channel subunits. KEY RESULTS: Long-term culture of hESC-CMs led to shortened AP duration (APD), faster repolarization rate, and increased CV. Selective block of IKr , IKs , IK1 , and IKur significantly affected AP repolarization and APD in a concentration- and culture time-dependent manner. Baseline variations in APD led to either positive or negative APD dependence of drug response. Chromanol 293B produced greater relative AP prolongation in mid- and late-stage cultures, while DPO-1 had more effect in early-stage cultures. CV in cell monolayers in early- and late-stage cultures was most susceptible to slowing by E-4031 and BaCl2 respectively. CONCLUSIONS AND IMPLICATIONS: IKr , IKs , IK1 , and IKur all play an essential role in the regulation of APD and CV in hESC-CMs. During time in culture, increased expression of IKr and IK1 helps to accelerate repolarization, shorten APD, and increase CV. We identified a new pro-arrhythmic parameter, positive APD dependence of ion channel block, which can increase APD and repolarization gradients.


Assuntos
Potenciais de Ação/fisiologia , Miócitos Cardíacos/fisiologia , Canais de Potássio/fisiologia , Linhagem Celular , Células-Tronco Embrionárias Humanas/citologia , Humanos
13.
Stem Cell Reports ; 12(5): 982-995, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31056480

RESUMO

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) hold great promise for cardiac studies, but their structural and functional immaturity precludes their use as faithful models of adult myocardium. Here we describe engineered heart slices (EHS), preparations of decellularized porcine myocardium repopulated with hiPSC-CMs that exhibit structural and functional improvements over standard culture. EHS exhibited multicellular, aligned bundles of elongated CMs with organized sarcomeres, positive inotropic responses to isoproterenol, anisotropic conduction of action potentials, and electrophysiological functionality for more than 200 days. We developed a new drug assay, GRIDS, that serves as a "fingerprint" of cardiac drug sensitivity for a range of pacing rates and drug concentrations. GRIDS maps characterized differences in drug sensitivity between EHS and monolayers more clearly than changes in action potential durations or conduction velocities. EHS represent a tissue-like model for long-term culture, structural, and functional improvement, and higher fidelity drug response of hiPSC-CMs.


Assuntos
Coração/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Cardiotônicos/farmacologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células Cultivadas , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Isoproterenol/farmacologia , Suínos
14.
Tissue Eng Part C Methods ; 25(4): 243-252, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30913987

RESUMO

Ischemic cardiomyopathy poses a significant public health burden due to the irreversible loss of functional cardiac tissue. Alternative treatment strategies include creation of three-dimensional (3D) cardiac tissues to both replace and augment injured native tissue. In this study, we utilize a net mold-based method to create a biomaterial-free 3D cardiac tissue and compare it to current methods using biomaterials. Cardiomyocytes, fibroblasts, and endothelial cells were combined using a hanging drop method to create spheroids. For the net mold patch method, spheroids were seeded into a net mold-based system to create biomaterial-free 3D cardiac patches. For the gel patch, spheroids were embedded in a collagen gel. Immunohistochemistry revealed increased alignment, vascularization, collagen I expression, cell viability, and higher density of cells in the net mold patch compared with the gel patch. Furthermore, in vivo testing in a left anterior descending artery ligation rat model found increased ejection fraction and smaller scar area following implantation of the net mold patch. We present a novel and simple reproducible method to create biomaterial-free 3D net mold patches that may potentially improve the treatment of heart failure in the future.


Assuntos
Materiais Biocompatíveis/farmacologia , Coração/fisiologia , Engenharia Tecidual/métodos , Animais , Artérias/cirurgia , Linhagem Celular , Tamanho Celular , Sobrevivência Celular/efeitos dos fármacos , Colágeno/farmacologia , Eletrocardiografia , Exossomos/metabolismo , Feminino , Coração/diagnóstico por imagem , Coração/efeitos dos fármacos , Humanos , Ligadura , Ratos , Ratos Endogâmicos Lew , Ratos Nus , Esferoides Celulares/citologia
15.
Tissue Eng Part A ; 25(9-10): 725-735, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30520705

RESUMO

IMPACT STATEMENT: Genetic heart diseases such as arrhythmogenic cardiomyopathy (AC), a common genetic cause of sudden cardiac death, can be modeled using patient-specific induced pluripotent stem cell-derived cardiac myocytes (CMs). However, it is important to culture these cells in a multicellular syncytium with exposure to surrounding matrix cues to create more accurate and robust models of the disease due to the importance of cell-cell and cell-matrix interactions. The engineered heart slice, constructed by seeding CMs on intact decellularized matrix slices, allows molecular and functional studies on an aligned multilayered syncytium of CMs. This study reveals the potential for an improved disease-in-a-dish model of AC.


Assuntos
Arritmias Cardíacas , Cardiomiopatias , Células-Tronco Pluripotentes Induzidas , Modelos Cardiovasculares , Mutação , Miocárdio , Placofilinas , Engenharia Tecidual , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Placofilinas/genética , Placofilinas/metabolismo , Suínos
16.
J Tissue Eng Regen Med ; 12(2): e962-e972, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28103423

RESUMO

Cardiac tissue engineering approaches have the potential to regenerate functional myocardium with intrinsic vascular networks. This study compared the relative effects of human adipose-derived stem/stromal cells (hASCs) and human dermal fibroblasts (hDFs) in cocultures with neonatal rat ventricular cardiomyocytes (NRVCMs) and human umbilical vein endothelial cells (HUVECs). At the same ratios of NRVCM:hASC and NRVCM:hDF, the hASC cocultures displayed shorter action potentials and maintained capture at faster pacing rates. Similarly, in coculture with HUVECs, hASC:HUVEC exhibited superior ability to support vascular capillary network formation relative to hDF:HUVEC. Based on these studies, a range of suitable cell ratios were determined to develop a triculture system. Six seeding ratios of NRVCM:hASC:HUVEC were tested and it was found that a ratio of 500:50:25 cells (i.e. 250,000:25,000:12,500 cells/cm2 ) resulted in the formation of robust vascular networks while retaining action potential durations and propagation similar to pure NRVCM cultures. Tricultures in this ratio exhibited an average conduction velocity of 20 ± 2 cm/s, action potential durations at 80% repolarization (APD80 ) and APD30 of 122 ± 5 ms and 59 ± 4 ms, respectively, and maximum capture rate of 7.4 ± 0.6 Hz. The NRVCM control groups had APD80 and APD30 of 120 ± 9 ms and 51 ± 5 ms, with a maximum capture rate of 7.3 ± 0.2 Hz. In summary, the combination of hASCs in the appropriate ratios with NRVCMs and HUVECs can facilitate the formation of densely vascularized cardiac tissues that appear not to impact the electrophysiological function of cardiomyocytes negatively. Copyright © 2017 John Wiley & Sons, Ltd.


Assuntos
Prótese Vascular , Coração/fisiologia , Células-Tronco Mesenquimais/citologia , Regeneração/fisiologia , Engenharia Tecidual/métodos , Animais , Fenômenos Eletrofisiológicos , Feminino , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Miócitos Cardíacos/citologia , Neovascularização Fisiológica , Ratos Sprague-Dawley
17.
Sci Rep ; 7(1): 4566, 2017 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-28676704

RESUMO

We have developed a novel method to deliver stem cells using 3D bioprinted cardiac patches, free of biomaterials. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), fibroblasts (FB) and endothelial cells (EC) were aggregated to create mixed cell spheroids. Cardiac patches were created from spheroids (CM:FB:EC = 70:15:15, 70:0:30, 45:40:15) using a 3D bioprinter. Cardiac patches were analyzed with light and video microscopy, immunohistochemistry, immunofluorescence, cell viability assays and optical electrical mapping. Cardiac tissue patches of all cell ratios beat spontaneously after 3D bioprinting. Patches exhibited ventricular-like action potential waveforms and uniform electrical conduction throughout the patch. Conduction velocities were higher and action potential durations were significantly longer in patches containing a lower percentage of FBs. Immunohistochemistry revealed staining for CM, FB and EC markers, with rudimentary CD31+ blood vessel formation. Immunofluorescence revealed the presence of Cx43, the main cardiac gap junction protein, localized to cell-cell borders. In vivo implantation suggests vascularization of 3D bioprinted cardiac patches with engraftment into native rat myocardium. This constitutes a significant step towards a new generation of stem cell-based treatment for heart failure.


Assuntos
Materiais Biocompatíveis , Bioimpressão , Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais , Animais , Materiais Biocompatíveis/química , Diferenciação Celular , Células Cultivadas , Fenômenos Eletrofisiológicos , Células Endoteliais , Fibroblastos/citologia , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Ratos , Esferoides Celulares , Transplante de Tecidos
18.
J Vis Exp ; (125)2017 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-28715377

RESUMO

This protocol describes 3D bioprinting of cardiac tissue without the use of biomaterials, using only cells. Cardiomyocytes, endothelial cells and fibroblasts are first isolated, counted and mixed at desired cell ratios. They are co-cultured in individual wells in ultra-low attachment 96-well plates. Within 3 days, beating spheroids form. These spheroids are then picked up by a nozzle using vacuum suction and assembled on a needle array using a 3D bioprinter. The spheroids are then allowed to fuse on the needle array. Three days after 3D bioprinting, the spheroids are removed as an intact patch, which is already spontaneously beating. 3D bioprinted cardiac patches exhibit mechanical integration of component spheroids and are highly promising in cardiac tissue regeneration and as 3D models of heart disease.


Assuntos
Bioimpressão/métodos , Miócitos Cardíacos/citologia , Esferoides Celulares/citologia , Engenharia Tecidual/métodos , Humanos , Miócitos Cardíacos/metabolismo , Esferoides Celulares/metabolismo
19.
Cell Stem Cell ; 19(1): 95-106, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27320040

RESUMO

Neurons derived from human pluripotent stem cells (hPSCs) are powerful tools for studying human neural development and diseases. Robust functional coupling of hPSC-derived neurons with target tissues in vitro is essential for modeling intercellular physiology in a dish and to further translational studies, but it has proven difficult to achieve. Here, we derive sympathetic neurons from hPSCs and show that they can form physical and functional connections with cardiac muscle cells. Using multiple hPSC reporter lines, we recapitulated human autonomic neuron development in vitro and successfully isolated PHOX2B::eGFP+ neurons that exhibit sympathetic marker expression and electrophysiological properties and norepinephrine secretion. Upon pharmacologic and optogenetic manipulation, PHOX2B::eGFP+ neurons controlled beating rates of cardiomyocytes, and the physical interactions between these cells increased neuronal maturation. This study provides a foundation for human sympathetic neuron specification and for hPSC-based neuronal control of organs in a dish.


Assuntos
Diferenciação Celular , Miócitos Cardíacos/citologia , Neurônios/citologia , Células-Tronco Pluripotentes/citologia , Sistema Nervoso Simpático/citologia , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Ventrículos do Coração/citologia , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Optogenética , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/genética
20.
Integr Biol (Camb) ; 8(2): 230-242, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26822672

RESUMO

Patterns of cellular organization in diverse tissues frequently display a complex geometry and topology tightly related to the tissue function. Progressive disorganization of tissue morphology can lead to pathologic remodeling, necessitating the development of experimental and theoretical methods of analysis of the tolerance of normal tissue function to structural alterations. A systematic way to investigate the relationship of diverse cell organization to tissue function is to engineer two-dimensional cell monolayers replicating key aspects of the in vivo tissue architecture. However, it is still not clear how this can be accomplished on a tissue level scale in a parameterized fashion, allowing for a mathematically precise definition of the model tissue organization and properties down to a cellular scale with a parameter dependent gradual change in model tissue organization. Here, we describe and use a method of designing precisely parameterized, geometrically complex patterns that are then used to control cell alignment and communication of model tissues. We demonstrate direct application of this method to guiding the growth of cardiac cell cultures and developing mathematical models of cell function that correspond to the underlying experimental patterns. Several anisotropic patterned cultures spanning a broad range of multicellular organization, mimicking the cardiac tissue organization of different regions of the heart, were found to be similar to each other and to isotropic cell monolayers in terms of local cell-cell interactions, reflected in similar confluency, morphology and connexin-43 expression. However, in agreement with the model predictions, different anisotropic patterns of cell organization, paralleling in vivo alterations of cardiac tissue morphology, resulted in variable and novel functional responses with important implications for the initiation and maintenance of cardiac arrhythmias. We conclude that variations of tissue geometry and topology can dramatically affect cardiac tissue function even if the constituent cells are themselves similar, and that the proposed method can provide a general strategy to experimentally and computationally investigate when such variation can lead to impaired tissue function.


Assuntos
Arritmias Cardíacas/metabolismo , Coração/fisiologia , Miócitos Cardíacos/citologia , Algoritmos , Animais , Anisotropia , Comunicação Celular , Células Cultivadas , Simulação por Computador , Conexina 43/metabolismo , Fibronectinas/química , Modelos Teóricos , Miocárdio/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...