Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Opt Lett ; 46(9): 2168-2171, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33929445

RESUMO

Fluorescence lifetime imaging microscopy (FLIM) is a powerful technique, capable of label-free assessment of the metabolic state and function within single cells. The FLIM measurements of autofluorescence were recently shown to be sensitive to the functional state and subtype of T cells. Therefore, autofluorescence FLIM could improve cell manufacturing technologies for adoptive immunotherapy, which currently require a time-intensive process of cell labeling with fluorescent antibodies. However, current autofluorescence FLIM implementations are typically too slow, bulky, and prohibitively expensive for use in cell manufacturing pipelines. Here we report a single photon-excited confocal whole-cell autofluorescence system that uses fast field-programmable gate array-based time tagging electronics to achieve time-correlated single photon counting (TCSPC) of single-cell autofluorescence. The system includes simultaneous near-infrared bright-field imaging and is sensitive to variations in the fluorescence decay profile of the metabolic coenzyme NAD(P)H in human T cells due to the activation state. The classification of activated and quiescent T cells achieved high accuracy and precision (area under the receiver operating characteristic curve, AUC = 0.92). The lower-cost, higher acquisition speed, and resistance to pile-up effects at high photon flux compared to traditional multiphoton-excited FLIM and TCSPC implementations with similar SNR make this system attractive for integration into flow cytometry, sorting, and quality control in cell manufacturing.


Assuntos
Microscopia de Fluorescência por Excitação Multifotônica , Linfócitos T/citologia , Humanos
2.
Methods ; 101: 93-102, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26518252

RESUMO

The blood-brain barrier (BBB) is a critical component of the central nervous system (CNS) that regulates the flux of material between the blood and the brain. Because of its barrier properties, the BBB creates a bottleneck to CNS drug delivery. Human in vitro BBB models offer a potential tool to screen pharmaceutical libraries for CNS penetration as well as for BBB modulators in development and disease, yet primary and immortalized models respectively lack scalability and robust phenotypes. Recently, in vitro BBB models derived from human pluripotent stem cells (hPSCs) have helped overcome these challenges by providing a scalable and renewable source of human brain microvascular endothelial cells (BMECs). We have demonstrated that hPSC-derived BMECs exhibit robust structural and functional characteristics reminiscent of the in vivo BBB. Here, we provide a detailed description of the methods required to differentiate and functionally characterize hPSC-derived BMECs to facilitate their widespread use in downstream applications.


Assuntos
Diferenciação Celular , Células Endoteliais/fisiologia , Células-Tronco Pluripotentes/fisiologia , Barreira Hematoencefálica/citologia , Encéfalo/irrigação sanguínea , Técnicas de Cultura de Células , Linhagem Celular , Humanos , Microvasos/citologia
3.
Mol Pharm ; 13(9): 3341-9, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27421304

RESUMO

Cancer-targeting alkylphosphocholine (APC) analogues are being clinically developed for diagnostic imaging, intraoperative visualization, and therapeutic applications. These APC analogues derived from chemically synthesized phospholipid ethers were identified and optimized for cancer-targeting specificity using extensive structure-activity studies. While they strongly label human brain cancers associated with disrupted blood-brain barriers (BBB), APC permeability across intact BBB remains unknown. Three of our APC analogues, CLR1404 (PET radiotracer), CLR1501 (green fluorescence), and CLR1502 (near-infrared fluorescence), were tested for permeability across a BBB model composed of human induced pluripotent stem cell-derived brain microvascular endothelial cells (iPSC-derived BMECs). This in vitro BBB system has reproducibly consistent high barrier integrity marked by high transendothelial electrical resistance (TEER > 1500 Ω-cm(2)) and functional expression of drug efflux transporters. The radioiodinated and fluorescent APC analogues demonstrated fairly low permeability across the iPSC-BMEC (35 ± 5.7 (CLR1404), 54 ± 3.2 (CLR1501), and 26 ± 4.9 (CLR1502) × 10(-5) cm/min) compared with BBB-impermeable sucrose (13 ± 2.5) and BBB-permeable diazepam (170 ± 29). Only the fluorescent APC analogues (CLR1501, CLR1502) underwent BCRP and MRP polarized drug efflux transport in the brain-to-blood direction of the BBB model, and this efflux can be specifically blocked with pharmacological inhibition. None of the tested APC analogues appeared to undergo substantial P-gp transport. Limited permeability of the APC analogues across an intact BBB into normal brain likely contributes to the high tumor to background ratios observed in initial human trials. Moreover, addition of fluorescent moieties to APCs resulted in greater BMEC efflux via MRP and BCRP, and may affect fluorescence-guided applications. Overall, the characterization of APC analogue permeability across human BBB is significant for advancing future brain tumor-targeted applications of these agents.


Assuntos
Barreira Hematoencefálica/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Fosforilcolina/análogos & derivados , Antineoplásicos/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/citologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-37885458

RESUMO

Human stem cells provide emerging methods for drug screening, disease modeling, and personalized patient therapies. To meet this growing demand for scale-up, stem cell manufacturing methods must be streamlined with continuous monitoring technologies and automated feedback to optimize growth conditions for high production and consistency. Label-free optical imaging and sensing, including multiphoton microscopy, Raman spectroscopy, and low-cost methods such as phase and transmitted light microscopy, can provide rapid, repeatable, and non-invasive monitoring of stem cells throughout cell differentiation and maturation. Machine learning algorithms trained on label-free optical imaging and sensing features could identify viable cells and predict optimal manufacturing conditions. These techniques have the potential to streamline stem cell manufacturing and accelerate their use in regenerative medicine.

5.
Nat Commun ; 12(1): 4580, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321477

RESUMO

Human pluripotent stem cell (hPSC)-derived cardiomyocytes provide a promising regenerative cell therapy for cardiovascular patients and an important model system to accelerate drug discovery. However, cost-effective and time-efficient platforms must be developed to evaluate the quality of hPSC-derived cardiomyocytes during biomanufacturing. Here, we develop a non-invasive label-free live cell imaging platform to predict the efficiency of hPSC differentiation into cardiomyocytes. Autofluorescence imaging of metabolic co-enzymes is performed under varying differentiation conditions (cell density, concentration of Wnt signaling activator) across five hPSC lines. Live cell autofluorescence imaging and multivariate classification models provide high accuracy to separate low (< 50%) and high (≥ 50%) differentiation efficiency groups (quantified by cTnT expression on day 12) within 1 day after initiating differentiation (area under the receiver operating characteristic curve, 0.91). This non-invasive and label-free method could be used to avoid batch-to-batch and line-to-line variability in cell manufacturing from hPSCs.


Assuntos
Diferenciação Celular , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Técnicas de Cultura de Células , Linhagem Celular , Hepatócitos , Humanos , Controle de Qualidade , Via de Sinalização Wnt
6.
Lab Chip ; 20(20): 3744-3756, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33048070

RESUMO

Endothelial cells (EC) in vivo are continuously exposed to a mechanical microenvironment from blood flow, and fluidic shear stress plays an important role in EC behavior. New approaches to generate physiologically and pathologically relevant pulsatile flows are needed to understand EC behavior under different shear stress regimes. Here, we demonstrate an adaptable pump (Adapt-Pump) platform for generating pulsatile flows from human pluripotent stem cell-derived cardiac spheroids (CS) via quantitative imaging-based signal transduction. Pulsatile flows generated from the Adapt-Pump system can recapitulate unique CS contraction characteristics, accurately model responses to clinically relevant drugs, and simulate CS contraction changes in response to fluidic mechanical stimulation. We discovered that ECs differentiated under a long QT syndrome derived pathological pulsatile flow exhibit abnormal EC monolayer organization. This Adapt-Pump platform provides a powerful tool for modeling the cardiovascular system and improving our understanding of EC behavior under different mechanical microenvironments.


Assuntos
Células Endoteliais , Miócitos Cardíacos , Humanos , Fluxo Pulsátil , Transdução de Sinais , Células-Tronco , Estresse Mecânico
7.
Neuron ; 108(5): 937-952.e7, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-32979312

RESUMO

The blood vessels in the central nervous system (CNS) have a series of unique properties, termed the blood-brain barrier (BBB), which stringently regulate the entry of molecules into the brain, thus maintaining proper brain homeostasis. We sought to understand whether neuronal activity could regulate BBB properties. Using both chemogenetics and a volitional behavior paradigm, we identified a core set of brain endothelial genes whose expression is regulated by neuronal activity. In particular, neuronal activity regulates BBB efflux transporter expression and function, which is critical for excluding many small lipophilic molecules from the brain parenchyma. Furthermore, we found that neuronal activity regulates the expression of circadian clock genes within brain endothelial cells, which in turn mediate the activity-dependent control of BBB efflux transport. These results have important clinical implications for CNS drug delivery and clearance of CNS waste products, including Aß, and for understanding how neuronal activity can modulate diurnal processes.


Assuntos
Barreira Hematoencefálica/fisiologia , Relógios Circadianos/genética , Ritmo Circadiano/genética , Células Endoteliais/fisiologia , Neurônios/fisiologia , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Barreira Hematoencefálica/efeitos dos fármacos , Relógios Circadianos/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Drogas Desenhadas/administração & dosagem , Células Endoteliais/efeitos dos fármacos , Feminino , Homeostase/efeitos dos fármacos , Homeostase/genética , Locomoção/efeitos dos fármacos , Locomoção/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos
8.
Sci Rep ; 9(1): 2765, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808965

RESUMO

A major cause of chronic kidney disease (CKD) is glomerular disease, which can be attributed to a spectrum of podocyte disorders. Podocytes are non-proliferative, terminally differentiated cells. Thus, the limited supply of primary podocytes impedes CKD research. Differentiation of human pluripotent stem cells (hPSCs) into podocytes has the potential to produce podocytes for disease modeling, drug screening, and cell therapies. In the podocyte differentiation process described here, hPSCs are first induced to primitive streak-like cells by activating canonical Wnt signaling. Next, these cells progress to mesoderm precursors, proliferative nephron progenitors, and eventually become mature podocytes by culturing in a serum-free medium. Podocytes generated via this protocol adopt podocyte morphology, express canonical podocyte markers, and exhibit podocyte phenotypes, including albumin uptake and TGF-ß1 triggered cell death. This study provides a simple, defined strategy to generate podocytes for in vitro modeling of podocyte development and disease or for cell therapies.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Podócitos/citologia , Células Cultivadas , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Podócitos/metabolismo , Linha Primitiva/citologia , Linha Primitiva/metabolismo , Proteínas Wnt/metabolismo
9.
Nat Protoc ; 12(9): 1890-1900, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28817124

RESUMO

Here, we describe how to efficiently direct human pluripotent stem cells (hPSCs) differentiation into self-renewing epicardial cells in a completely defined, xeno-free system by temporal modulation of regulators of canonical Wnt signaling. Appropriate differentiation-stage-specific application of Gsk3 inhibitor, Wnt inhibitor, and Gsk3 inhibitor (GiWiGi) is sufficient to produce cells expressing epicardial markers and exhibiting epicardial phenotypes with a high yield and purity from multiple hPSC lines in 16 d. Characterization of differentiated cells is performed via flow cytometry and immunostaining to assess quantitative expression and localization of epicardial cell-specific proteins. In vitro differentiation into fibroblasts and smooth muscle cells (SMCs) is also described. In addition, culture in the presence of transforming growth factor (TGF)-ß inhibitors allows long-term expansion of hPSC-derived epicardial cells (for at least 25 population doublings). Functional human epicardial cells differentiated via this protocol may constitute a potential cell source for heart disease modeling, drug screening, and cell-based therapeutic applications.


Assuntos
Diferenciação Celular/fisiologia , Engenharia Celular/métodos , Pericárdio/citologia , Células-Tronco Pluripotentes/citologia , Técnicas de Cultura de Células , Humanos , Via de Sinalização Wnt/fisiologia
10.
Bioeng Transl Med ; 2(2): 191-201, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-29170757

RESUMO

During heart development, epicardial progenitors contribute various cardiac lineages including smooth muscle cells, cardiac fibroblasts, and endothelial cells. However, their specific contribution to the human endothelium has not yet been resolved, at least in part due to the inability to expand and maintain human primary or pluripotent stem cell (hPSC)-derived epicardial cells. Here we first generated CDH5-2A-eGFP knock-in hPSC lines and differentiated them into self-renewing WT1+ epicardial cells, which gave rise to endothelial cells upon VEGF treatment in vitro. In addition, we found that the percentage of endothelial cells correlated with WT1 expression in a WT1-2A-eGFP reporter line. The resulting endothelial cells displayed many endocardium-like endothelial cell properties, including high expression levels of endocardial-specific markers, nutrient transporters and well-organized tight junctions. These findings suggest that human epicardial progenitors may have the capacity to form endocardial endothelium during development and have implications for heart regeneration and cardiac tissue engineering.

11.
Sci Adv ; 3(11): e1701679, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29134197

RESUMO

The blood-brain barrier (BBB) is composed of specialized endothelial cells that are critical to neurological health. A key tool for understanding human BBB development and its role in neurological disease is a reliable and scalable source of functional brain microvascular endothelial cells (BMECs). Human pluripotent stem cells (hPSCs) can theoretically generate unlimited quantities of any cell lineage in vitro, including BMECs, for disease modeling, drug screening, and cell-based therapies. We demonstrate a facile, chemically defined method to differentiate hPSCs to BMECs in a developmentally relevant progression via small-molecule activation of key signaling pathways. hPSCs are first induced to mesoderm commitment by activating canonical Wnt signaling. Next, these mesoderm precursors progress to endothelial progenitors, and treatment with retinoic acid leads to acquisition of BBB-specific markers and phenotypes. hPSC-derived BMECs generated via this protocol exhibit endothelial properties, including tube formation and low-density lipoprotein uptake, as well as efflux transporter activities characteristic of BMECs. Notably, these cells exhibit high transendothelial electrical resistance above 3000 ohm·cm2. These hPSC-derived BMECs serve as a robust human in vitro BBB model that can be used to study brain disease and inform therapeutic development.


Assuntos
Barreira Hematoencefálica/metabolismo , Diferenciação Celular , Linhagem da Célula , Claudina-5/metabolismo , Técnicas de Cocultura , Impedância Elétrica , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Fenótipo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma
12.
Cell Stem Cell ; 20(6): 831-843.e5, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28526555

RESUMO

Inactivating mutations in the thyroid hormone (TH) transporter Monocarboxylate transporter 8 (MCT8) cause severe psychomotor retardation in children. Animal models do not reflect the biology of the human disease. Using patient-specific induced pluripotent stem cells (iPSCs), we generated MCT8-deficient neural cells that showed normal TH-dependent neuronal properties and maturation. However, the blood-brain barrier (BBB) controls TH entry into the brain, and reduced TH availability to neural cells could instead underlie the diseased phenotype. To test potential BBB involvement, we generated an iPSC-based BBB model of MCT8 deficiency, and we found that MCT8 was necessary for polarized influx of the active form of TH across the BBB. We also found that a candidate drug did not appreciably cross the mutant BBB. Our results therefore clarify the underlying physiological basis of this disorder, and they suggest that circumventing the diseased BBB to deliver active TH to the brain could be a viable therapeutic strategy.


Assuntos
Barreira Hematoencefálica/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Transportadores de Ácidos Monocarboxílicos/deficiência , Neurônios/metabolismo , Transtornos Psicomotores/metabolismo , Barreira Hematoencefálica/patologia , Linhagem Celular , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Neurônios/patologia , Transtornos Psicomotores/genética , Transtornos Psicomotores/patologia , Simportadores
13.
Artigo em Inglês | MEDLINE | ID: mdl-28462012

RESUMO

The epicardium contributes both multi-lineage descendants and paracrine factors to the heart during cardiogenesis and cardiac repair, underscoring its potential for cardiac regenerative medicine. Yet little is known about the cellular and molecular mechanisms that regulate human epicardial development and regeneration. Here, we show that the temporal modulation of canonical Wnt signaling is sufficient for epicardial induction from 6 different human pluripotent stem cell (hPSC) lines, including a WT1-2A-eGFP knock-in reporter line, under chemically-defined, xeno-free conditions. We also show that treatment with transforming growth factor beta (TGF-ß)-signalling inhibitors permitted long-term expansion of the hPSC-derived epicardial cells, resulting in a more than 25 population doublings of WT1+ cells in homogenous monolayers. The hPSC-derived epicardial cells were similar to primary epicardial cells both in vitro and in vivo, as determined by morphological and functional assays, including RNA-seq. Our findings have implications for the understanding of self-renewal mechanisms of the epicardium and for epicardial regeneration using cellular or small-molecule therapies.

14.
Curr Opin Genet Dev ; 34: 54-60, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26313850

RESUMO

Microfluidic devices employ submillimeter length scale control of flow to achieve high-resolution spatial and temporal control over the microenvironment, providing powerful tools to elucidate mechanisms of human pluripotent stem cell (hPSC) regulation and to elicit desired hPSC fates. In addition, microfluidics allow control of paracrine and juxtracrine signaling, thereby enabling fabrication of microphysiological systems comprised of multiple cell types organized into organs-on-a-chip. Microfluidic cell culture systems can also be integrated with actuators and sensors, permitting construction of high-density arrays of cell-based biosensors for screening applications. This review describes recent advances in using microfluidics to understand mechanisms by which the microenvironment regulates hPSC fates and applications of microfluidics to realize the potential of hPSCs for in vitro modeling and screening applications.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/genética , Microfluídica/métodos , Células-Tronco Pluripotentes/citologia , Microambiente Celular/genética , Humanos , Comunicação Parácrina/genética
15.
Sci Rep ; 5: 12963, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26261043

RESUMO

Migration of endothelial cells is essential for wound healing and angiogenesis. Src kinase activity plays important roles at the protrusions of migrating endothelial cells. However, the spatiotemporal coordination between Src kinase activity and the protrusion of cell edge remains unclear. Therefore, we investigate these coordinated molecular events at the initiation of cell migration, by integrating microfabrication, fluorescence resonance energy transfer (FRET)-based biosensors, and automated computational image analysis. We demonstrate that the physical release of restrictive micropattern triggered a significant decrease of Src activity at the protrusive edge of endothelial cells. Computational cross-correlation analysis reveals that the decrease of Src activity occurred earlier in time, and was well-coordinated with the protrusion of cell edge in polarized cells, but not in non-polarized cells. These results suggest that the spatiotemporal control of Src kinase activity is well-coordinated with cell polarization and protrusion in endothelial cells upon the release of physical constraint, as that experienced by endothelial cells sprouting from stiff tumor micro-environment during angiogenesis. Therefore, our integrative approach enabled the discovery of a new model where Src is de-activated in coordination with membrane protrusion, providing important insights into the regulation of endothelial migration and angiogenesis.


Assuntos
Microambiente Celular/genética , Células Endoteliais/citologia , Cicatrização/genética , Quinases da Família src/metabolismo , Movimento Celular/genética , Polaridade Celular/genética , Extensões da Superfície Celular/genética , Células Endoteliais/metabolismo , Transferência Ressonante de Energia de Fluorescência , Humanos , Neovascularização Fisiológica/genética , Quinases da Família src/genética
16.
Stem Cell Res ; 15(1): 122-129, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26042795

RESUMO

Human pluripotent stem cell (hPSC)-derived endothelial cells and their progenitors are important for vascular research and therapeutic revascularization. Here, we report a completely defined endothelial progenitor differentiation platform that uses a minimalistic medium consisting of Dulbecco's modified eagle medium and ascorbic acid, lacking of albumin and growth factors. Following hPSC treatment with a GSK-3ß inhibitor and culture in this medium, this protocol generates more than 30% multipotent CD34+ CD31+ endothelial progenitors that can be purified to >95% CD34+ cells via magnetic activated cell sorting (MACS). These CD34+ progenitors are capable of differentiating into endothelial cells in serum-free inductive media. These hPSC-derived endothelial cells express key endothelial markers including CD31, VE-cadherin, and von Willebrand factor (vWF), exhibit endothelial-specific phenotypes and functions including tube formation and acetylated low-density lipoprotein (Ac-LDL) uptake. This fully defined platform should facilitate production of proliferative, xeno-free endothelial progenitor cells for both research and clinical applications.


Assuntos
Albuminas/química , Diferenciação Celular , Células Progenitoras Endoteliais/citologia , Células-Tronco Pluripotentes/citologia , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Meios de Cultura/farmacologia , Células Progenitoras Endoteliais/efeitos dos fármacos , Humanos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos
17.
Integr Biol (Camb) ; 5(2): 431-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23250282

RESUMO

The microenvironment has been shown to regulate cellular functions including cell growth, differentiation, proliferation, migration, cancer development and metastasis. However, the underlying molecular mechanism remains largely unclear. We have integrated micro-pattern technology and molecular biosensors based on fluorescence resonance energy transfer (FRET) to visualize calcium responses in cells constrained to grow on a micro-patterned surface. Upon ATP stimulation, human umbilical vein endothelial cells (HUVECs) cultured on different surface micro-patterns had a shorter decay time and a reduced peak of a transient intracellular calcium rise compared to control cells without constraints. The decay time is regulated by the plasma membrane and the membrane calcium channels, while the peak by endoplasmic reticulum (ER) calcium release. Further results revealed that voltage operated channels (VOCs), coupling the plasma membrane and ER, can affect both the decay time and the peak of calcium response. The inhibition of VOCs can eliminate the effect of different micro-patterns on calcium signals. When two connected HUVECs were constrained to grow on a micro-pattern, drastically distinct calcium responses upon ATP stimulation can be observed, in contrast to the similar responses of two connected cells cultured without patterns. Interestingly, the inhibition of VOCs also blocked this difference of calcium responses between two connected cells on micro-patterns. These results indicate that a micro-patterned surface can have a profound effect on the calcium responses of HUVECs under ATP stimulation, largely mediated by VOCs. Therefore, our results shed new light on the molecular mechanism by which HUVECs perceive the microenvironment and regulate intracellular calcium signals.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Microambiente Celular/fisiologia , Células Endoteliais/fisiologia , Transferência Ressonante de Energia de Fluorescência/métodos , Microscopia de Fluorescência/métodos , Imagem Molecular/métodos , Células Cultivadas , Humanos
18.
Med Biol Eng Comput ; 48(10): 1023-32, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20490938

RESUMO

Micro/nano-fabrication techniques, such as soft lithography and electrospinning, have been well-developed and widely applied in many research fields in the past decade. Due to the low costs and simple procedures, these techniques have become important and popular for biological studies. In this review, we focus on the studies integrating micro/nano-fabrication work to elucidate the molecular mechanism of signaling transduction in cell biology. We first describe different micro/nano-fabrication technologies, including techniques generating three-dimensional scaffolds for tissue engineering. We then introduce the application of these technologies in manipulating the physical or chemical micro/nano-environment to regulate the cellular behavior and response, such as cell life and death, differentiation, proliferation, and cell migration. Recent advancement in integrating the micro/nano-technologies and live cell imaging are also discussed. Finally, potential schemes in cell biology involving micro/nano-fabrication technologies are proposed to provide perspectives on the future research activities.


Assuntos
Microtecnologia/métodos , Engenharia Tecidual/métodos , Fenômenos Fisiológicos Celulares/fisiologia , Humanos , Nanotecnologia/métodos , Alicerces Teciduais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA