Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurodev Disord ; 16(1): 27, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783199

RESUMEN

BACKGROUND: Tuberous sclerosis complex (TSC) is a multi-system genetic disease that causes benign tumors in the brain and other vital organs. The most debilitating symptoms result from involvement of the central nervous system and lead to a multitude of severe symptoms including seizures, intellectual disability, autism, and behavioral problems. TSC is caused by heterozygous mutations of either the TSC1 or TSC2 gene and dysregulation of mTOR kinase with its multifaceted downstream signaling alterations is central to disease pathogenesis. Although the neurological sequelae of the disease are well established, little is known about how these mutations might affect cellular components and the function of the blood-brain barrier (BBB). METHODS: We generated TSC disease-specific cell models of the BBB by leveraging human induced pluripotent stem cell and microfluidic cell culture technologies. RESULTS: Using microphysiological systems, we demonstrate that a BBB generated from TSC2 heterozygous mutant cells shows increased permeability. This can be rescued by wild type astrocytes or by treatment with rapamycin, an mTOR kinase inhibitor. CONCLUSION: Our results demonstrate the utility of microphysiological systems to study human neurological disorders and advance our knowledge of cell lineages contributing to TSC pathogenesis and informs future therapeutics.


Asunto(s)
Barrera Hematoencefálica , Células Madre Pluripotentes Inducidas , Proteína 2 del Complejo de la Esclerosis Tuberosa , Esclerosis Tuberosa , Esclerosis Tuberosa/fisiopatología , Esclerosis Tuberosa/genética , Humanos , Barrera Hematoencefálica/fisiopatología , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Sirolimus/farmacología , Astrocitos/metabolismo
2.
Lab Chip ; 24(6): 1794-1807, 2024 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-38362777

RESUMEN

Human microphysiological systems, such as organs on chips, are an emerging technology for modeling human physiology in a preclinical setting to understand the mechanism of action of drugs, to evaluate the efficacy of treatment options for human disease and impairment, and to assess drug toxicity. By using human cells co-cultured in three-dimensional constructs, organ chips can provide greater fidelity to the human cellular condition than their two-dimensional predecessors. However, with the rise of SARS-CoV-2 and the global COVID-19 pandemic, it became clear that many microphysiological systems were not compatible with or optimized for studies of infectious disease and operation in a Biosafety Level 3 (BSL-3) environment. Given that one of the early sites of SARS-CoV-2 infection is the airway, we created a human airway organ chip that could operate in a BSL-3 space with high throughput and minimal manipulation, while retaining the necessary physical and physiological components to recapitulate tissue response to infectious agents and the immune response to infection.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Carga Viral , Pandemias , Inmunohistoquímica , Citocinas , Dispositivos Laboratorio en un Chip
3.
bioRxiv ; 2023 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-38168450

RESUMEN

Tuberous sclerosis complex (TSC) is a multi-system genetic disease that causes benign tumors in the brain and other vital organs. The most debilitating symptoms result from involvement of the central nervous system and lead to a multitude of severe symptoms including seizures, intellectual disability, autism, and behavioral problems. TSC is caused by heterozygous mutations of either the TSC1 or TSC2 gene. Dysregulation of mTOR kinase with its multifaceted downstream signaling alterations is central to disease pathogenesis. Although the neurological sequelae of the disease are well established, little is known about how these mutations might affect cellular components and the function of the blood-brain barrier (BBB). We generated disease-specific cell models of the BBB by leveraging human induced pluripotent stem cell and microfluidic cell culture technologies. Using these microphysiological systems, we demonstrate that the BBB generated from TSC2 heterozygous mutant cells shows increased permeability which can be rescued by wild type astrocytes and with treatment with rapamycin, an mTOR kinase inhibitor. Our results further demonstrate the utility of microphysiological systems to study human neurological disorders and advance our knowledge of the cell lineages contributing to TSC pathogenesis.

4.
Fluids Barriers CNS ; 17(1): 38, 2020 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493346

RESUMEN

BACKGROUND: The United States faces a national crisis involving opioid medications, where currently more than 130 people die every day. To combat this epidemic, a better understanding is needed of how opioids penetrate into the central nervous system (CNS) to facilitate pain relief and, potentially, result in addiction and/or misuse. Animal models, however, are a poor predictor of blood-brain barrier (BBB) transport and CNS drug penetration in humans, and many traditional 2D cell culture models of the BBB and neurovascular unit have inadequate barrier function and weak or inappropriate efflux transporter expression. Here, we sought to better understand opioid transport mechanisms using a simplified microfluidic neurovascular unit (NVU) model consisting of human brain microvascular endothelial cells (BMECs) co-cultured with astrocytes. METHODS: Human primary and induced pluripotent stem cell (iPSC)-derived BMECs were incorporated into a microfluidic NVU model with several technical improvements over our previous design. Passive barrier function was assessed by permeability of fluorescent dextrans with varying sizes, and P-glycoprotein function was assessed by rhodamine permeability in the presence or absence of inhibitors; quantification was performed with a fluorescent plate reader. Loperamide, morphine, and oxycodone permeability was assessed in the presence or absence of P-glycoprotein inhibitors and cortisol; quantification was performed with mass spectrometry. RESULTS: We first report technical and methodological optimizations to our previously described microfluidic model using primary human BMECs, which results in accelerated barrier formation, decreased variability, and reduced passive permeability relative to Transwell models. We then demonstrate proper transport and efflux of loperamide, morphine, and oxycodone in the microfluidic NVU containing BMECs derived from human iPSCs. We further demonstrate that cortisol can alter permeability of loperamide and morphine in a divergent manner. CONCLUSIONS: We reveal a novel role for the stress hormone cortisol in modulating the transport of opioids across the BBB, which could contribute to their abuse or overdose. Our updated BBB model represents a powerful tool available to researchers, clinicians, and drug manufacturers for understanding the mechanisms by which opioids access the CNS.


Asunto(s)
Analgésicos Opioides/farmacocinética , Astrocitos/fisiología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Células Endoteliales/fisiología , Hidrocortisona/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Modelos Neurológicos , Astrocitos/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Microvasos/citología
5.
Stem Cell Reports ; 12(3): 474-487, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30773484

RESUMEN

There is a profound need for functional, biomimetic in vitro tissue constructs of the human blood-brain barrier and neurovascular unit (NVU) to model diseases and identify therapeutic interventions. Here, we show that induced pluripotent stem cell (iPSC)-derived human brain microvascular endothelial cells (BMECs) exhibit robust barrier functionality when cultured in 3D channels within gelatin hydrogels. We determined that BMECs cultured in 3D under perfusion conditions were 10-100 times less permeable to sodium fluorescein, 3 kDa dextran, and albumin relative to human umbilical vein endothelial cell and human dermal microvascular endothelial cell controls, and the BMECs maintained barrier function for up to 21 days. Analysis of cell-cell junctions revealed expression patterns supporting barrier formation. Finally, efflux transporter activity was maintained over 3 weeks of perfused culture. Taken together, this work lays the foundation for development of a representative 3D in vitro model of the human NVU constructed from iPSCs.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Endotelio/efectos de los fármacos , Hidrogeles/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Albúminas/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Dextranos/metabolismo , Células Endoteliales/metabolismo , Endotelio/metabolismo , Fluoresceína/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Microvasos/efectos de los fármacos , Microvasos/metabolismo
6.
Int J Mol Sci ; 19(9)2018 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158434

RESUMEN

Endothelial dysfunction is a known consequence of bone morphogenetic protein type II receptor (BMPR2) mutations seen in pulmonary arterial hypertension (PAH). However, standard 2D cell culture models fail to mimic the mechanical environment seen in the pulmonary vasculature. Hydrogels have emerged as promising platforms for 3D disease modeling due to their tunable physical and biochemical properties. In order to recreate the mechanical stimuli seen in the pulmonary vasculature, we have created a novel 3D hydrogel-based pulmonary vasculature model ("artificial arteriole") that reproduces the pulsatile flow rates and pressures seen in the human lung. Using this platform, we studied both Bmpr2R899X and WT endothelial cells to better understand how the addition of oscillatory flow and physiological pressure influenced gene expression, cell morphology, and cell permeability. The addition of oscillatory flow and pressure resulted in several gene expression changes in both WT and Bmpr2R899X cells. However, for many pathways with relevance to PAH etiology, Bmpr2R899X cells responded differently when compared to the WT cells. Bmpr2R899X cells were also found not to elongate in the direction of flow, and instead remained stagnant in morphology despite mechanical stimuli. The increased permeability of the Bmpr2R899X layer was successfully reproduced in our artificial arteriole, with the addition of flow and pressure not leading to significant changes in permeability. Our artificial arteriole is the first to model many mechanical properties seen in the lung. Its tunability enables several new opportunities to study the endothelium in pulmonary vascular disease with increased control over environmental parameters.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Células Endoteliales/fisiología , Hipertensión Pulmonar/fisiopatología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Línea Celular , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Hipertensión Pulmonar/metabolismo , Ratones , Análisis de Secuencia de ARN
7.
SLAS Technol ; 23(6): 592-598, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29787331

RESUMEN

The fabrication of engineered vascularized tissues and organs requiring sustained, controlled perfusion has been facilitated by the development of several pump systems. Currently, researchers in the field of tissue engineering require the use of pump systems that are in general large, expensive, and generically designed. Overall, these pumps often fail to meet the unique demands of perfusing clinically useful tissue constructs. Here, we describe a pumping platform that overcomes these limitations and enables scalable perfusion of large, three-dimensional hydrogels. We demonstrate the ability to perfuse multiple separate channels inside hydrogel slabs using a preprogrammed schedule that dictates pumping speed and time. The use of this pump system to perfuse channels in large-scale engineered tissue scaffolds sustained cell viability over several weeks.


Asunto(s)
Hidrogeles , Perfusión/métodos , Técnicas de Cultivo de Tejidos/métodos , Ingeniería de Tejidos/métodos , Costos y Análisis de Costo , Perfusión/economía , Perfusión/instrumentación , Técnicas de Cultivo de Tejidos/economía , Técnicas de Cultivo de Tejidos/instrumentación , Ingeniería de Tejidos/economía , Ingeniería de Tejidos/instrumentación
8.
ACS Omega ; 2(7): 3583-3594, 2017 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-28782050

RESUMEN

In this study, we demonstrate the theranostic capability of actively targeted, site-specific multibranched gold nanoantennas (MGNs) in triple-negative breast cancer (TNBC) cells in vitro. By utilizing multiplexed surface-enhanced Raman scattering (SERS) imaging, enabled by the narrow peak widths of Raman signatures, we simultaneously targeted immune checkpoint receptor programmed death ligand 1 (PDL1) and the epidermal growth factor receptor (EGFR) overexpressed in TNBC cells. A 1:1 mixture of MGNs functionalized with anti-PDL1 antibodies and Raman tag 5,5-dithio-bis-(2-nitrobenzoic acid) (DTNB) and MGNs functionalized with anti-EGFR antibodies and Raman tag para-mercaptobenzoic acid (pMBA) were incubated with the cells. SERS imaging revealed a cellular traffic map of MGN localization by surface binding and receptor-mediated endocytosis, enabling targeted diagnosis of both biomarkers. Furthermore, cells incubated with anti-EGFR-pMBA-MGNs and illuminated with an 808 nm laser for 15 min at 4.7 W/cm2 exhibited photothermal cell death only within the laser spot (indicated by live/dead cell fluorescence assay). Therefore, this study not only provides an optical imaging platform that can track immunomarkers with spatiotemporal control but also demonstrates an externally controlled light-triggered therapeutic approach enabling receptor-specific treatment with biocompatible theranostic nanoprobes.

9.
Artículo en Inglés | MEDLINE | ID: mdl-29441348

RESUMEN

The neurovascular unit (NVU) is composed of neurons, astrocytes, pericytes, and endothelial cells that form the blood-brain barrier (BBB). The NVU regulates material exchange between the bloodstream and the brain parenchyma, and its dysfunction is a primary or secondary cause of many cerebrovascular and neurodegenerative disorders. As such, there are substantial research thrusts in academia and industry toward building NVU models that mimic endogenous organization and function, which could be used to better understand disease mechanisms and assess drug efficacy. Human pluripotent stem cells, which can self-renew indefinitely and differentiate to almost any cell type in the body, are attractive for these models because they can provide a limitless source of individual cells from the NVU. In addition, human-induced pluripotent stem cells (iPSCs) offer the opportunity to build NVU models with an explicit genetic background and in the context of disease susceptibility. Herein, we review how iPSCs are being used to model neurovascular and neurodegenerative diseases, with particular focus on contributions of the BBB, and discuss existing technologies and emerging opportunities to merge these iPSC progenies with biomaterials platforms to create complex NVU systems that recreate the in vivo microenvironment.

10.
ACS Omega ; 1(2): 234-243, 2016 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-27656689

RESUMEN

In this work, we demonstrate controlled drug delivery from low-temperature-sensitive liposomes (LTSLs) mediated by photothermal heating from multibranched gold nanoantennas (MGNs) in triple-negative breast cancer (TNBC) cells in vitro. The unique geometry of MGNs enables the generation of mild hyperthermia (∼42 °C) by converting near-infrared light to heat and effectively delivering doxorubicin (DOX) from the LTSLs in breast cancer cells. We confirmed the cellular uptake of MGNs by using both fluorescence confocal Z-stack imaging and transmission electron microscopy (TEM) imaging. We performed a cellular viability assay and live/dead cell fluorescence imaging of the combined therapeutic effects of MGNs with DOX-loaded LTSLs (DOX-LTSLs) and compared them with free DOX and DOX-loaded non-temperature-sensitive liposomes (DOX-NTSLs). Imaging of fluorescent live/dead cell indicators and MTT assay outcomes both demonstrated significant decreases in cellular viability when cells were treated with the combination therapy. Because of the high phase-transition temperature of NTSLs, no drug delivery was observed from the DOX-NTSLs. Notably, even at a low DOX concentration of 0.5 µg/mL, the combination treatment resulted in a higher (33%) cell death relative to free DOX (17% cell death). The results of our work demonstrate that the synergistic therapeutic effect of photothermal hyperthermia of MGNs with drug delivery from the LTSLs can successfully eradicate aggressive breast cancer cells with higher efficacy than free DOX by providing a controlled light-activated approach and minimizing off-target toxicity.

11.
Adv Healthc Mater ; 5(7): 781-5, 2016 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-26844941

RESUMEN

A 3D microvascularized gelatin hydrogel is produced using thermoresponsive sacrificial poly(N-isopropylacrylamide) microfibers. The capillary-like microvascular network allows constant perfusion of media throughout the thick hydrogel, and significantly improves the viability of human neonatal dermal fibroblasts encapsulated within the gel at a high density.


Asunto(s)
Gelatina/farmacología , Hidrogeles/farmacología , Microvasos/efectos de los fármacos , Temperatura , Resinas Acrílicas/farmacología , Dermis/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Imagenología Tridimensional , Recién Nacido , Microscopía Confocal , Perfusión
12.
Biomicrofluidics ; 9(3): 036501, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26045731

RESUMEN

Biomimetic scaffolds approaching physiological scale, whose size and large cellular load far exceed the limits of diffusion, require incorporation of a fluidic means to achieve adequate nutrient/metabolite exchange. This need has driven the extension of microfluidic technologies into the area of biomaterials. While construction of perfusable scaffolds is essentially a problem of microfluidic device fabrication, functional implementation of free-standing, thick-tissue constructs depends upon successful integration of external pumping mechanisms through optimized connective assemblies. However, a critical analysis to identify optimal materials/assembly components for hydrogel substrates has received little focus to date. This investigation addresses this issue directly by evaluating the efficacy of a range of adhesive and mechanical fluidic connection methods to gelatin hydrogel constructs based upon both mechanical property analysis and cell compatibility. Results identify a novel bioadhesive, comprised of two enzymatically modified gelatin compounds, for connecting tubing to hydrogel constructs that is both structurally robust and non-cytotoxic. Furthermore, outcomes from this study provide clear evidence that fluidic interconnect success varies with substrate composition (specifically hydrogel versus polydimethylsiloxane), highlighting not only the importance of selecting the appropriately tailored components for fluidic hydrogel systems but also that of encouraging ongoing, targeted exploration of this issue. The optimization of such interconnect systems will ultimately promote exciting scientific and therapeutic developments provided by microfluidic, cell-laden scaffolds.

13.
Integr Biol (Camb) ; 4(4): 368-73, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22344285

RESUMEN

The oncogenic fusion protein BCR-ABL is produced by chronic myeloid leukemia (CML) cells and functions as an abnormal, constitutively active tyrosine kinase that interferes with normal migratory and apoptotic behaviour of cells. Small molecule tyrosine kinase inhibitors (TKIs), such as dasatinib, eliminate CML progenitor cells, but fail to target the stem cell fraction resulting in persistent disease. In order to achieve a cure for CML in the majority of patients, we need an improved understanding of intracellular signalling dynamics, including the shuttling of BCR-ABL between cytosolic and nuclear compartments. In the past, the instability of BCR-ABL in assays using conventional immunohistochemical techniques has made this difficult and has not allowed for reliable analysis at the single cell level. Here we show how the utilization of rapid on-chip cell fixation within a microfluidic platform provides a means to immunofluorescently analyze the spatiotemporal localization of both BCR-ABL and c-ABL, as well as the linked apoptosis mediator, BCL-XL, in arrays of single CD34+ CML stem/progenitor cells, without cell loss. We demonstrate this proceeds up to 4 times faster than benchtop methods. Our results indicate that whilst both BCR-ABL and c-ABL shuttle from the cytoplasm to the nucleus following dasatinib treatment, the temporal dynamics are not synchronized. The microfluidic platform has the potential to provide insights into the intracellular signalling events in single cells. The ability to examine signalling events and assess BCR-ABL expression/activity in isolated cells in "real-time" may help elucidate the characteristics of rare CML stem cell events, which lead to the resistance of CML stem cells to TKIs.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Técnicas Analíticas Microfluídicas/métodos , Células Madre Neoplásicas/metabolismo , Transporte de Proteínas/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Dasatinib , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Cinética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Técnicas Analíticas Microfluídicas/instrumentación , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/farmacología , Tiazoles/farmacología , Proteína bcl-X/metabolismo
14.
Biomicrofluidics ; 5(2): 24106, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21673844

RESUMEN

We discuss the ability to perform fluorescent immunocytochemistry, following cell fixation, using a microfluidic array of primary, nonadherent, single CD34+ stem cells. The technique requires small cell samples and proceeds with no cell loss, making it well-suited to monitoring these rare patient-derived cells. The chip allows us to correlate live cell dynamics across arrays of individual cells with post-translational modifications of intracellular proteins, following their exposure to drug treatments. Results also show that due to the microfluidic environment, the time scale of cell fixation was significantly reduced compared to conventional methods, leading to greater confidence in the status of the protein modifications studied.

15.
Methods Mol Biol ; 731: 285-91, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21516415

RESUMEN

Validation of new therapeutic targets calls for the advance in innovative assays that probe both spatial and temporal relationships in signaling networks. Cell death assays have already found a widespread use in pharmacological profiling of anticancer drugs. Such assays are, however, predominantly restricted to end point DEAD/LIVE parameter that provides only a snapshot of inherently stochastic process such as tumor cell death. Development of new methods that can offer kinetic real-time analysis would be highly advantageous for the pharmacological screening and predictive toxicology. In the present work we outline innovative protocols for the real-time analysis of tumor cell death, based on propidium iodide (PI) and SYTOX Green probes. These can be readily adapted to both flow cytometry and time-lapse fluorescence imaging. Considering vast time savings and kinetic data acquisition such assays have the potential to be applied in a number of areas including accelerated anticancer drug discovery and high-throughput screening routines.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales/instrumentación , Citometría de Flujo , Humanos , Imagen Molecular , Factores de Tiempo
16.
Chem Commun (Camb) ; 46(42): 7921-3, 2010 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-20859575

RESUMEN

We report for the first time the time-resolved mapping of intracellular nanoparticle labels from within living cells retained in a microstructured trap using Raman spectroscopy. The methods employed here also demonstrate the ability to rapidly discriminate between cell populations containing different SERS labels.


Asunto(s)
Microfluídica/instrumentación , Nanopartículas , Animales , Células CHO , Cricetinae , Cricetulus , Espectrometría Raman
17.
Exp Biol Med (Maywood) ; 235(6): 777-83, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20511682

RESUMEN

Microfluidic devices are well-suited for the study of metabolism and paracrine and autocrine signaling because they allow steady or intermittent perfusion of biological cells at cell densities that approach those in living tissue. They also enable the study of small populations of rare cells. However, it can be difficult to introduce the cells into a microfluidic device to achieve and control such densities without damaging or clumping the cells. We describe simple procedures that address the problem of efficient introduction of cells and cell culture media into microfluidic devices using small bore polyetheretherketone (PEEK) tubing and Hamilton gastight syringes. Suspension or adherent cells grown in cell culture flasks are centrifuged and extracted directly from the centrifuge pellet into the end of the PEEK tubing by aspiration. The tube end is then coupled to prepunched channels in the polydimethylsiloxane microfluidic device by friction fitting. Controlled depression of the syringe plunger expels the cells into the microfluidic device only seconds following aspiration. The gastight syringes and PEEK tubing with PEEK fittings provide a non-compliant source of pressure and suction with a rapid response time that is well suited for short-term intramicrofluidic cellular studies. The benefits of this method are its simplicity, modest expense, the short preparation time required for loading appropriate numbers of cells and the applicability of the technique to small quantities of rare or expensive cells. This should in turn lead to new applications of microfluidic devices to biology and medicine.


Asunto(s)
Microfluídica , Técnicas de Cultivo de Célula/métodos , Línea Celular , Células Cultivadas , Centrifugación
18.
Anal Chem ; 81(23): 9828-33, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19902928

RESUMEN

Lab-on-a-chip technologies have the potential to deliver significant technological advances in modern biomedicine, through the ability to provide appropriate low-cost microenvironments for screening cells. However, to date, few studies have investigated the suitability of poly(dimethylsiloxane) (PDMS) for live cell culture. Here, we describe an inexpensive method for production of reusable, optical-grade PDMS microculture chips which provide a static and self-contained microwell system analogous to conventional polystyrene multiwell plates. We use these structures to probe the effects of PDMS upon live cell culture bioassays, using time-lapse fluorescence imaging to explore the toxicity of the substrate. We use three model systems to explore the efficacy of the microstructured devices: (i) live cell culture, (ii) adenoviral gene delivery to mammalian cells, and (iii) gravity enforced formation of multicellular tumor spheroids (MCTS). Results show that PDMS is nontoxic to cells, as their viability and growth characteristic in PDMS-based platforms is comparable to that of their polystyrene counterparts.


Asunto(s)
Bioensayo/instrumentación , Bioensayo/métodos , Técnicas de Cultivo de Célula/métodos , Dimetilpolisiloxanos , Dispositivos Laboratorio en un Chip , Procedimientos Analíticos en Microchip/métodos , Adenoviridae/genética , Animales , Técnicas de Cultivo de Célula/economía , Técnicas de Cultivo de Célula/instrumentación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dimetilpolisiloxanos/química , Dimetilpolisiloxanos/farmacología , Genes Virales , Vidrio/química , Humanos , Espacio Intracelular/metabolismo , Neoplasias/patología , Esferoides Celulares/metabolismo , Factores de Tiempo
19.
Lab Chip ; 9(18): 2659-64, 2009 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-19704981

RESUMEN

Stem cells hold great promise as a means of treating otherwise incurable, degenerative diseases due to their ability both to self-renew and differentiate. However, stem cell damage can also play a role in the disease with the formation of solid tumors and leukaemias such as chronic myeloid leukaemia (CML), a hematopoietic stem cell (HSC) disorder. Despite recent medical advances, CML remains incurable by drug therapy. Understanding the mechanisms which govern chemoresistance of individual stem cell leukaemias may therefore require analysis at the single cell level. This task is not trivial using current technologies given that isolating HSCs is difficult, expensive, and inefficient due to low cell yield from patients. In addition, hematopoietic cells are largely non-adherent and thus difficult to study over time using conventional cell culture techniques. Hence, there is a need for new microfluidic platforms that allow the functional interrogation of hundreds of non-adherent single cells in parallel. We demonstrate the ability to perform assays, normally performed on the macroscopic scale, within the microfluidic platform using minimal reagents and low numbers of primary cells. We investigated normal and CML stem cell responses to the tyrosine kinase inhibitor, dasatinib, a drug approved for the treatment of CML. Dynamic, on-chip three-color cell viability assays revealed that differences in the responses of normal and CML stem/progenitor cells to dasatinib were observed even in the early phases of exposure, during which time normal cells exhibit a significantly elevated cell death rate, as compared to both controls and CML cells. Further studies show that dasatinib does, however, markedly reduce CML stem/progenitor cell migration in situ.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Microfluídica/métodos , Anexina A5/metabolismo , Apoptosis/fisiología , División Celular/fisiología , Supervivencia Celular , Células Cultivadas , Dasatinib , Humanos , Cinética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Microscopía Fluorescente , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Transducción de Señal/fisiología , Tiazoles/farmacología , Tiazoles/uso terapéutico
20.
Anal Chem ; 81(16): 6952-9, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19572560

RESUMEN

Cell cytotoxicity tests are among the most common bioassays using flow cytometry and fluorescence imaging analysis. The permeability of plasma membranes to charged fluorescent probes serves, in these assays, as a marker distinguishing live from dead cells. Since it is generally assumed that probes, such as propidium iodide (PI) or 7-amino-actinomycin D (7-AAD), are themselves cytotoxic, they are currently generally used only as the end-point markers of assays for live versus dead cells. In the current study, we provide novel insights into potential applications of these classical plasma membrane integrity markers in the dynamic tracking of drug-induced cytotoxicity. We show that treatment of a number of different human tumor cell lines in cultures for up to 72 h with the PI, 7-AAD, SYTOX Green (SY-G), SYTOX Red (SY-R), TO-PRO, and YO-PRO had no effect on cell viability assessed by the integrity of plasma membrane, cell cycle progression, and rate of proliferation. We subsequently explore the potential of dynamic labeling with these markers in real-time analysis, by comparing results from both conventional cytometry and microfluidic chips. Considering the simplicity of the staining protocols and their low cost combined with the potential for real-time data collection, we show how that real-time fluorescent imaging and Lab-on-a-Chip platforms have the potential to be used for automated drug screening routines.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Línea Celular Tumoral , Proliferación Celular , Dactinomicina/análogos & derivados , Dactinomicina/química , Citometría de Flujo , Humanos , Microfluídica , Propidio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA