Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Sci Rep ; 10(1): 6918, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332853

RESUMO

Three-dimensional (3D) in vitro models of human skeletal muscle mimic aspects of native tissue structure and function, thereby providing a promising system for disease modeling, drug discovery or pre-clinical validation, and toxicity testing. Widespread adoption of this research approach is hindered by the lack of easy-to-use platforms that are simple to fabricate and that yield arrays of human skeletal muscle micro-tissues (hMMTs) in culture with reproducible physiological responses that can be assayed non-invasively. Here, we describe a design and methods to generate a reusable mold to fabricate a 96-well platform, referred to as MyoTACTIC, that enables bulk production of 3D hMMTs. All 96-wells and all well features are cast in a single step from the reusable mold. Non-invasive calcium transient and contractile force measurements are performed on hMMTs directly in MyoTACTIC, and unbiased force analysis occurs by a custom automated algorithm, allowing for longitudinal studies of function. Characterizations of MyoTACTIC and resulting hMMTs confirms the capability of the device to support formation of hMMTs that recapitulate biological responses. We show that hMMT contractile force mirrors expected responses to compounds shown by others to decrease (dexamethasone, cerivastatin) or increase (IGF-1) skeletal muscle strength. Since MyoTACTIC supports hMMT long-term culture, we evaluated direct influences of pancreatic cancer chemotherapeutics agents on contraction competent human skeletal muscle myotubes. A single application of a clinically relevant dose of Irinotecan decreased hMMT contractile force generation, while clear effects on myotube atrophy were observed histologically only at a higher dose. This suggests an off-target effect that may contribute to cancer associated muscle wasting, and highlights the value of the MyoTACTIC platform to non-invasively predict modulators of human skeletal muscle function.


Assuntos
Músculo Esquelético/fisiologia , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Antineoplásicos/farmacologia , Fenômenos Biomecânicos , Cálcio/metabolismo , Humanos
2.
Sci Rep ; 10(1): 6919, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332814

RESUMO

To accelerate the cardiac drug discovery pipeline, we set out to develop a platform that would be capable of quantifying tissue-level functions such as contractile force and be amenable to standard multiwell-plate manipulations. We report a 96-well-based array of 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues - termed Cardiac MicroRings (CaMiRi) - in custom 3D-print-molded multiwell plates capable of contractile force measurement. Within each well, two elastomeric microcantilevers are situated above a circumferential ramp. The wells are seeded with cell-laden collagen, which, in response to the gradual slope of the circumferential ramp, self-organizes around tip-gated microcantilevers to form contracting CaMiRi. The contractile force exerted by the CaMiRi is measured and calculated using the deflection of the cantilevers. Platform responses were robust and comparable across wells, and we used it to determine an optimal tissue formulation. We validated the contractile force response of CaMiRi using selected cardiotropic compounds with known effects. Additionally, we developed automated protocols for CaMiRi seeding, image acquisition, and analysis to enable the measurement of contractile force with increased throughput. The unique tissue fabrication properties of the platform, and the consequent effects on tissue function, were demonstrated upon adding hPSC-derived epicardial cells to the system. This platform represents an open-source contractile force screening system useful for drug screening and tissue engineering applications.


Assuntos
Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Animais , Automação , Cardiotônicos/farmacologia , Células Cultivadas , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Camundongos , Contração Miocárdica/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Impressão Tridimensional
3.
Can J Cardiol ; 30(11): 1307-22, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25442432

RESUMO

In recent years, the development of 3-dimensional engineered heart tissue (EHT) has made large strides forward because of advances in stem cell biology, materials science, prevascularization strategies, and nanotechnology. As a result, the role of tissue engineering in cardiac regenerative medicine has become multifaceted as new applications become feasible. Cardiac tissue engineering has long been established to have the potential to partially or fully restore cardiac function after cardiac injury. However, EHTs may also serve as surrogate human cardiac tissue for drug-related toxicity screening. Cardiotoxicity remains a major cause of drug withdrawal in the pharmaceutical industry. Unsafe drugs reach the market because preclinical evaluation is insufficient to weed out cardiotoxic drugs in all their forms. Bioengineering methods could provide functional and mature human myocardial tissues, ie, physiologically relevant platforms, for screening the cardiotoxic effects of pharmaceutical agents and facilitate the discovery of new therapeutic agents. Finally, advances in induced pluripotent stem cells have made patient-specific EHTs possible, which opens up the possibility of personalized medicine. Herein, we give an overview of the present state of the art in cardiac tissue engineering, the challenges to the field, and future perspectives.


Assuntos
Materiais Biocompatíveis , Cardiopatias/terapia , Coração/fisiologia , Medicina Regenerativa/métodos , Engenharia Tecidual , Animais , Humanos
4.
Lab Chip ; 14(5): 869-82, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24352498

RESUMO

Tissue engineering enables the generation of three-dimensional (3D) functional cardiac tissue for pre-clinical testing in vitro, which is critical for new drug development. However, current tissue engineering methods poorly recapitulate the architecture of oriented cardiac bundles with supporting capillaries. In this study, we designed a microfabricated bioreactor to generate 3D micro-tissues, termed biowires, using both primary neonatal rat cardiomyocytes and human embryonic stem cell (hESC) derived cardiomyocytes. Perfusable cardiac biowires were generated with polytetrafluoroethylene (PTFE) tubing template, and were integrated with electrical field stimulation using carbon rod electrodes. To demonstrate the feasibility of this platform for pharmaceutical testing, nitric oxide (NO) was released from perfused sodium nitroprusside (SNP) solution and diffused through the tubing. The NO treatment slowed down the spontaneous beating of cardiac biowires based on hESC derived cardiomyocytes and degraded the myofibrillar cytoskeleton of the cardiomyocytes within the biowires. The biowires were also integrated with electrical stimulation using carbon rod electrodes to further improve phenotype of cardiomyocytes, as indicated by organized contractile apparatus, higher Young's modulus, and improved electrical properties. This microfabricated platform provides a unique opportunity to assess pharmacological effects on cardiac tissue in vitro by perfusion in a cardiac bundle model, which could provide improved physiological relevance.


Assuntos
Materiais Biocompatíveis/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Engenharia Tecidual , Animais , Materiais Biocompatíveis/química , Reatores Biológicos , Células Cultivadas , Módulo de Elasticidade , Estimulação Elétrica , Eletrodos , Células-Tronco Embrionárias/citologia , Géis/química , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Perfusão , Politetrafluoretileno/química , Ratos
5.
Proc Natl Acad Sci U S A ; 110(49): E4698-707, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24255110

RESUMO

Access to robust and information-rich human cardiac tissue models would accelerate drug-based strategies for treating heart disease. Despite significant effort, the generation of high-fidelity adult-like human cardiac tissue analogs remains challenging. We used computational modeling of tissue contraction and assembly mechanics in conjunction with microfabricated constraints to guide the design of aligned and functional 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues that we term cardiac microwires (CMWs). Miniaturization of the platform circumvented the need for tissue vascularization and enabled higher-throughput image-based analysis of CMW drug responsiveness. CMW tissue properties could be tuned using electromechanical stimuli and cell composition. Specifically, controlling self-assembly of 3D tissues in aligned collagen, and pacing with point stimulation electrodes, were found to promote cardiac maturation-associated gene expression and in vivo-like electrical signal propagation. Furthermore, screening a range of hPSC-derived cardiac cell ratios identified that 75% NKX2 Homeobox 5 (NKX2-5)+ cardiomyocytes and 25% Cluster of Differentiation 90 OR (CD90)+ nonmyocytes optimized tissue remodeling dynamics and yielded enhanced structural and functional properties. Finally, we demonstrate the utility of the optimized platform in a tachycardic model of arrhythmogenesis, an aspect of cardiac electrophysiology not previously recapitulated in 3D in vitro hPSC-derived cardiac microtissue models. The design criteria identified with our CMW platform should accelerate the development of predictive in vitro assays of human heart tissue function.


Assuntos
Microambiente Celular/fisiologia , Miocárdio/citologia , Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Fenômenos Biomecânicos , Estimulação Elétrica , Análise de Elementos Finitos , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Humanos , Antígenos Thy-1/metabolismo , Fatores de Transcrição/metabolismo
6.
Nat Methods ; 10(8): 781-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23793239

RESUMO

Directed differentiation protocols enable derivation of cardiomyocytes from human pluripotent stem cells (hPSCs) and permit engineering of human myocardium in vitro. However, hPSC-derived cardiomyocytes are reflective of very early human development, limiting their utility in the generation of in vitro models of mature myocardium. Here we describe a platform that combines three-dimensional cell cultivation with electrical stimulation to mature hPSC-derived cardiac tissues. We used quantitative structural, molecular and electrophysiological analyses to explain the responses of immature human myocardium to electrical stimulation and pacing. We demonstrated that the engineered platform allows for the generation of three-dimensional, aligned cardiac tissues (biowires) with frequent striations. Biowires submitted to electrical stimulation had markedly increased myofibril ultrastructural organization, elevated conduction velocity and improved both electrophysiological and Ca(2+) handling properties compared to nonstimulated controls. These changes were in agreement with cardiomyocyte maturation and were dependent on the stimulation rate.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Diferenciação Celular/fisiologia , Estimulação Elétrica , Fenômenos Eletrofisiológicos , Humanos , Microscopia Eletrônica de Transmissão , Miocárdio/ultraestrutura
7.
Stem Cell Res Ther ; 4(1): 14, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23425700

RESUMO

Tissue engineering has developed many paradigms and techniques on how to best integrate cells and extracellular matrix to create in vitro structures that replicate native tissue. The strategy best suited for building these constructs depends mainly on the target cells, tissues, and organ of interest, and how readily their respective niches can be recapitulated in vitro with available technologies. In this review we examine engineered heart tissue and two techniques that can be used to induce tissue morphogenesis in artificial niches in vitro: engineered surface topology and electrical control of the system. For both the differentiation of stem cells into heart cells and further assembly of these cells into engineered tissues, these two techniques are effective in inducing in vivo like structure and function. Biophysical modulation through the control of topography and manipulation of the electrical microenvironment has been shown to have effects on cell growth and differentiation, expression of mature cardiac-related proteins and genes, cell alignment via cytoskeletal organization, and electrical and contractile properties. Lastly, we discuss the evolution and potential of these techniques, and bridges to regenerative therapies.


Assuntos
Diferenciação Celular/fisiologia , Coração/fisiologia , Engenharia Tecidual/métodos , Animais , Humanos , Miocárdio/metabolismo , Células-Tronco/fisiologia
8.
Tissue Eng Part A ; 18(9-10): 910-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22092279

RESUMO

Engineered myocardial tissues can be used to elucidate fundamental features of myocardial biology, develop organotypic in vitro model systems, and as engineered tissue constructs for replacing damaged heart tissue in vivo. However, a key limitation is an inability to test the wide range of parameters (cell source, mechanical, soluble and electrical stimuli) that might impact the engineered tissue in a high-throughput manner and in an environment that mimics native heart tissue. Here we used microelectromechanical systems technology to generate arrays of cardiac microtissues (CMTs) embedded within three-dimensional micropatterned matrices. Microcantilevers simultaneously constrain CMT contraction and report forces generated by the CMTs in real time. We demonstrate the ability to routinely produce ~200 CMTs per million cardiac cells (<1 neonatal rat heart) whose spontaneous contraction frequency, duration, and forces can be tracked. Independently varying the mechanical stiffness of the cantilevers and collagen matrix revealed that both the dynamic force of cardiac contraction as well as the basal static tension within the CMT increased with boundary or matrix rigidity. Cell alignment is, however, reduced within a stiff collagen matrix; therefore, despite producing higher force, CMTs constructed from higher density collagen have a lower cross-sectional stress than those constructed from lower density collagen. We also study the effect of electrical stimulation on cell alignment and force generation within CMTs and we show that the combination of electrical stimulation and auxotonic load strongly improves both the structure and the function of the CMTs. Finally, we demonstrate the suitability of our technique for high-throughput monitoring of drug-induced changes in spontaneous frequency or contractility in CMTs as well as high-speed imaging of calcium dynamics using fluorescent dyes. Together, these results highlight the potential for this approach to quantitatively demonstrate the impact of physical parameters on the maturation, structure, and function of cardiac tissue and open the possibility to use high-throughput, low volume screening for studies on engineered myocardium.


Assuntos
Coração/fisiologia , Microtecnologia/métodos , Miocárdio/citologia , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Animais , Fenômenos Biomecânicos , Células Cultivadas , Imunofluorescência , Ratos , Ratos Sprague-Dawley
9.
Nanotechnology ; 22(49): 494003, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22101261

RESUMO

While in nature the formation of complex tissues is gradually shaped by the long journey of development, in tissue engineering constructing complex tissues relies heavily on our ability to directly manipulate and control the micro-cellular environment in vitro. Not surprisingly, advancements in both microfabrication and nanofabrication have powered the field of tissue engineering in many aspects. Focusing on cardiac tissue engineering, this paper highlights the applications of fabrication techniques in various aspects of tissue engineering research: (1) cell responses to micro- and nanopatterned topographical cues, (2) cell responses to patterned biochemical cues, (3) controlled 3D scaffolds, (4) patterned tissue vascularization and (5) electromechanical regulation of tissue assembly and function.


Assuntos
Microtecnologia/métodos , Miocárdio/citologia , Nanotecnologia/métodos , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Animais , Humanos , Técnicas Analíticas Microfluídicas/métodos , Alicerces Teciduais/química
10.
Tissue Eng Part A ; 17(15-16): 1901-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21417693

RESUMO

Although it has been observed that aggregate size affects cardiac development, an incomplete understanding of the cellular mechanisms underlying human pluripotent stem cell-derived cardiomyogenesis has limited the development of robust defined-condition cardiac cell generation protocols. Our objective was thus to elucidate cellular and molecular mechanisms underlying the endogenous control of human embryonic stem cell (hESC) cardiac tissue development, and to test the hypothesis that hESC aggregate size influences extraembryonic endoderm (ExE) commitment and cardiac inductive properties. hESC aggregates were generated with 100, 1000, or 4000 cells per aggregate using microwells. The frequency of endoderm marker (FoxA2 and GATA6)-expressing cells decreased with increasing aggregate size during early differentiation. Cardiogenesis was maximized in aggregates initiated from 1000 cells, with frequencies of 0.49±0.06 cells exhibiting a cardiac progenitor phenotype (KDR(low)/C-KIT(neg)) on day 5 and 0.24±0.06 expressing cardiac Troponin T on day 16. A direct relationship between ExE and cardiac differentiation efficiency was established by forming aggregates with varying ratios of SOX7 (a transcription factor required for ExE development) overexpressing or knockdown hESCs to unmanipulated hESCs. We demonstrate, in a defined, serum-free cardiac induction system, that robust and efficient cardiac differentiation is a function of endogenous ExE cell concentration, a parameter that can be directly modulated by controlling hESC aggregate size.


Assuntos
Forma Celular , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Agregação Celular , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Tamanho Celular , Corpos Embrioides/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Humanos , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/metabolismo
11.
Biotechnol Bioeng ; 108(3): 704-19, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21246514

RESUMO

Embryonic stem cell (ESC) derivatives are a promising cell source for cardiac cell therapy. Mechanistic studies upon cell injection in conventional animal models are limited by inefficient delivery and poor cell survival. As an alternative, we have used an engineered heart tissue (EHT) based on neonatal rat cardiomyocytes (CMs) cultivated with electrical field stimulation as an in vitro model to study cell injection. We injected (0.001, 0.01, and 0.1 million) and tracked (by qPCR and histology) undifferentiated yellow-fluorescent protein transgenic mouse ESCs and Flk1 + /PDGFRα+ cardiac progenitor (CPs) cells, to investigate the effect of the cardiac environment on cell differentiation, as well as to test whether our in vitro model system could recapitulate the formation of teratoma-like structures commonly observed upon in vivo ESC injection. By 8 days post-injection, ESCs were spatially segregated from the cardiac cell population; however, ESC injection increased survival of CMs. The presence of ESCs blocked electrical conduction through the tissue, resulting in a 46% increase in the excitation threshold. Expression of mouse cardiac troponin I, was markedly increased in CP injected constructs compared to ESC injected constructs at all time points and cell doses tested. As early as 2 weeks, epithelial and ganglion-like structures were observed in ESC injected constructs. By 4 weeks of ESC injection, teratoma-like structures containing neural, epithelial, and connective tissue were observed in the constructs. Non-cardiac structures were observed in the CP injected constructs only after extended culture (4 weeks) and only at high cell doses, suggesting that these cells require further enrichment or differentiation prior to transplantation. Our data indicate that the cardiac environment of host tissue and electrical field stimulation did not preferentially guide the differentiation of ESCs towards the cardiac lineage. In the same environment, injection of CP resulted in a more robust cardiac differentiation than injection of ESC. Our data demonstrate that the model-system developed herein can be used to study the functional effects of candidate stem cells on the host myocardium, as well as to measure the residual activity of undifferentiated cells present in the mixture.


Assuntos
Células-Tronco Embrionárias/fisiologia , Miocárdio/citologia , Miócitos Cardíacos/fisiologia , Engenharia Tecidual/métodos , Animais , Camundongos , Camundongos Transgênicos , Ratos
12.
Biotechnol Bioeng ; 102(2): 493-507, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18767184

RESUMO

The ability to generate human pluripotent stem cell-derived cell types at sufficiently high numbers and in a reproducible manner is fundamental for clinical and biopharmaceutical applications. Current experimental methods for the differentiation of pluripotent cells such as human embryonic stem cells (hESC) rely on the generation of heterogeneous aggregates of cells, also called "embryoid bodies" (EBs), in small scale static culture. These protocols are typically (1) not scalable, (2) result in a wide range of EB sizes and (3) expose cells to fluctuations in physicochemical parameters. With the goal of establishing a robust bioprocess we first screened different scalable suspension systems for their ability to support the growth and differentiation of hESCs. Next homogeneity of initial cell aggregates was improved by employing a micro-printing strategy to generate large numbers of size-specified hESC aggregates. Finally, these technologies were integrated into a fully controlled bioreactor system and the impact of oxygen concentration was investigated. Our results demonstrate the beneficial effects of stirred bioreactor culture, aggregate size-control and hypoxia (4% oxygen tension) on both cell growth and cell differentiation towards cardiomyocytes. QRT-PCR data for markers such as Brachyury, LIM domain homeobox gene Isl-1, Troponin T and Myosin Light Chain 2v, as well as immunohistochemistry and functional analysis by response to chronotropic agents, documented the impact of these parameters on cardiac differentiation. This study provides an important foundation towards the robust generation of clinically relevant numbers of hESC derived cells.


Assuntos
Reatores Biológicos , Células-Tronco Embrionárias/fisiologia , Mesoderma/citologia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/fisiologia , Engenharia Tecidual/métodos , Anaerobiose , Técnicas de Cultura de Células/métodos , Diferenciação Celular/genética , Tamanho Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Humanos , Oxigênio/farmacologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA