Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 3: 63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26528478

RESUMEN

The interaction of hyaluronan (HA) with mesenchymal progenitor cells impacts trafficking and fate after tissue colonization during wound repair and these events contribute to diseases such as cancer. How this interaction occurs is poorly understood. Using 10T½ cells as a mesenchymal progenitor model and fluorescent (F-HA) or gold-labeled HA (G-HA) polymers, we studied the role of two HA receptors, RHAMM and CD44, in HA binding and uptake in non-adherent and adherent mesenchymal progenitor (10T½) cells to mimic aspects of cell trafficking and tissue colonization. We show that fluorescent labeled HA (F-HA) binding/uptake was high in non-adherent cells but dropped over time as cells became increasingly adherent. Non-adherent cells displayed both CD44 and RHAMM but only function-blocking anti-RHAMM and not anti-CD44 antibodies significantly reduced F-HA binding/uptake. Adherent cells, which also expressed CD44 and RHAMM, primarily utilized CD44 to bind to F-HA since anti-CD44 but not anti-RHAMM antibodies blocked F-HA uptake. RHAMM overexpression in adherent 10T½ cells led to increased F-HA uptake but this increased binding remained CD44 dependent. Further studies showed that RHAMM-transfection increased CD44 mRNA and protein expression while blocking RHAMM function reduced expression. Collectively, these results suggest that cellular microenvironments in which these receptors function as HA binding proteins differ significantly, and that RHAMM plays at least two roles in F-HA binding by acting as an HA receptor in non-attached cells and by regulating CD44 expression and display in attached cells. Our findings demonstrate adhesion-dependent mechanisms governing HA binding/ uptake that may impact development of new mesenchymal cell-based therapies.

2.
Hum Vaccin Immunother ; 11(8): 1936-44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26090875

RESUMEN

We report a biolistic technology platform for physical delivery of particle formulations of drugs or vaccines using parallel arrays of microchannels, which generate highly collimated jets of particles with high spatial resolution. Our approach allows for effective delivery of therapeutics sequentially or concurrently (in mixture) at a specified target location or treatment area. We show this new platform enables the delivery of a broad range of particles with various densities and sizes into both in vitro and ex vivo skin models. Penetration depths of ∼1 mm have been achieved following a single ejection of 200 µg high-density gold particles, as well as 13.6 µg low-density polystyrene-based particles into gelatin-based skin simulants at 70 psi inlet gas pressure. Ejection of multiple shots at one treatment site enabled deeper penetration of ∼3 mm in vitro, and delivery of a higher dose of 1 mg gold particles at similar inlet gas pressure. We demonstrate that particle penetration depths can be optimized in vitro by adjusting the inlet pressure of the carrier gas, and dosing is controlled by drug reservoirs that hold precise quantities of the payload, which can be ejected continuously or in pulses. Future investigations include comparison between continuous versus pulsatile payload deliveries. We have successfully delivered plasmid DNA (pDNA)-coated gold particles (1.15 µm diameter) into ex vivo murine and porcine skin at low inlet pressures of ∼30 psi. Integrity analysis of these pDNA-coated gold particles confirmed the preservation of full-length pDNA after each particle preparation and jetting procedures. This technology platform provides distinct capabilities to effectively deliver a broad range of particle formulations into skin with specially designed high-speed microarray ejector nozzles.


Asunto(s)
Biolística/métodos , Sistemas de Liberación de Medicamentos/métodos , Sistemas Microelectromecánicos/métodos , Preparaciones Farmacéuticas/administración & dosificación , Polvos/administración & dosificación , Vacunas de ADN/administración & dosificación , Animales , Humanos , Ratones Endogámicos BALB C , Porcinos
3.
Sci Transl Med ; 7(284): 284ra58, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25904742

RESUMEN

A fundamental problem in cancer drug development is that antitumor efficacy in preclinical cancer models does not translate faithfully to patient outcomes. Much of early cancer drug discovery is performed under in vitro conditions in cell-based models that poorly represent actual malignancies. To address this inconsistency, we have developed a technology platform called CIVO, which enables simultaneous assessment of up to eight drugs or drug combinations within a single solid tumor in vivo. The platform is currently designed for use in animal models of cancer and patients with superficial tumors but can be modified for investigation of deeper-seated malignancies. In xenograft lymphoma models, CIVO microinjection of well-characterized anticancer agents (vincristine, doxorubicin, mafosfamide, and prednisolone) induced spatially defined cellular changes around sites of drug exposure, specific to the known mechanisms of action of each drug. The observed localized responses predicted responses to systemically delivered drugs in animals. In pair-matched lymphoma models, CIVO correctly demonstrated tumor resistance to doxorubicin and vincristine and an unexpected enhanced sensitivity to mafosfamide in multidrug-resistant lymphomas compared with chemotherapy-naïve lymphomas. A CIVO-enabled in vivo screen of 97 approved oncology agents revealed a novel mTOR (mammalian target of rapamycin) pathway inhibitor that exhibits significantly increased tumor-killing activity in the drug-resistant setting compared with chemotherapy-naïve tumors. Finally, feasibility studies to assess the use of CIVO in human and canine patients demonstrated that microinjection of drugs is toxicity-sparing while inducing robust, easily tracked, drug-specific responses in autochthonous tumors, setting the stage for further application of this technology in clinical trials.


Asunto(s)
Antineoplásicos/química , Ensayos de Selección de Medicamentos Antitumorales/métodos , Linfoma/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Animales , Biomarcadores , Línea Celular Tumoral , Ciclofosfamida/análogos & derivados , Ciclofosfamida/química , Perros , Doxorrubicina/química , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Prednisolona/química , Serina-Treonina Quinasas TOR/metabolismo , Vincristina/química
4.
Proc Natl Acad Sci U S A ; 111(17): E1731-9, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24733940

RESUMEN

Tumor heterogeneity confounds cancer diagnosis and the outcome of therapy, necessitating analysis of tumor cell subsets within the tumor mass. Elevated expression of hyaluronan (HA) and HA receptors, receptor for HA-mediated motility (RHAMM)/HA-mediated motility receptor and cluster designation 44 (CD44), in breast tumors correlates with poor outcome. We hypothesized that a probe for detecting HA-HA receptor interactions may reveal breast cancer (BCa) cell heterogeneity relevant to tumor progression. A fluorescent HA (F-HA) probe containing a mixture of polymer sizes typical of tumor microenvironments (10-480 kDa), multiplexed profiling, and flow cytometry were used to monitor HA binding to BCa cell lines of different molecular subtypes. Formulae were developed to quantify binding heterogeneity and to measure invasion in vivo. Two subsets exhibiting differential binding (HA(-/low) vs. HA(high)) were isolated and characterized for morphology, growth, and invasion in culture and as xenografts in vivo. F-HA-binding amounts and degree of heterogeneity varied with BCa subtype, were highest in the malignant basal-like cell lines, and decreased upon reversion to a nonmalignant phenotype. Binding amounts correlated with CD44 and RHAMM displayed but binding heterogeneity appeared to arise from a differential ability of HA receptor-positive subpopulations to interact with F-HA. HA(high) subpopulations exhibited significantly higher local invasion and lung micrometastases but, unexpectedly, lower proliferation than either unsorted parental cells or the HA(-/low) subpopulation. Querying F-HA binding to aggressive tumor cells reveals a previously undetected form of heterogeneity that predicts invasive/metastatic behavior and that may aid both early identification of cancer patients susceptible to metastasis, and detection/therapy of invasive BCa subpopulations.


Asunto(s)
Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Ácido Hialurónico , Sondas Moleculares , Animales , Línea Celular Tumoral , Proliferación Celular , Pollos , Membrana Corioalantoides/metabolismo , Sistemas de Computación , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Fluorescencia , Humanos , Receptores de Hialuranos/metabolismo , Ratones , Ratones SCID , Invasividad Neoplásica , Fenotipo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Methods Mol Biol ; 916: 81-96, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22914934

RESUMEN

The unlimited differentiation and proliferation capacity of embryonic stem cells represents a great resource for regenerative medicine. Here, we describe a method for differentiating, isolating, and expanding endothelial cells (ECs) from mouse embryonic stem cells (mESCs). First, mESCs are expanded on a mouse embryonic fibroblast (mEF) feeder layer and partially differentiated into embryoid bodies (EBs) by growing the cells in an ultra-low attachment plate for up to 5 days. The EBs are then differentiated along the endothelial lineage using endothelial growth medium supplemented with 40 ng/mL vascular endothelial growth factor (VEGF). The differentiated endothelial population expresses both Fetal Liver Kinase 1 (Flk-1) and VE-Cadherin on the cell surface which can be further purified using a fluorescence-activated cell sorting (FACS) system and subsequently expanded on 0.1 % gelatin-coated plates. The differentiated cells can be analyzed by real-time PCR and flow cytometry to confirm enrichment of EC-specific genes and proteins.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Endoteliales/citología , Citometría de Flujo/métodos , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Cadherinas/metabolismo , Proliferación Celular , Cuerpos Embrioides/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Biomacromolecules ; 13(1): 12-22, 2012 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-22066590

RESUMEN

An increase in hyaluronan (HA) synthesis, cellular uptake, and metabolism occurs during the remodeling of tissue microenvironments following injury and during disease processes such as cancer. We hypothesized that multimodality HA-based probes selectively target and detectably accumulate at sites of high HA metabolism, thus providing a flexible imaging strategy for monitoring disease and repair processes. Kinetic analyses confirmed favorable available serum levels of the probe following intravenous (i.v.) or subcutaneous (s.c.) injection. Nuclear (technetium-HA, (99m)Tc-HA, and iodine-HA, (125)I-HA), optical (fluorescent Texas Red-HA, TR-HA), and magnetic resonance (gadolinium-HA, Gd-HA) probes imaged liver ((99m)Tc-HA), breast cancer cells/xenografts (TR-HA, Gd-HA), and vascular injury ((125)I-HA, TR-HA). Targeting of HA probes to these sites appeared to result from selective HA receptor-dependent localization. Our results suggest that HA-based probes, which do not require polysaccharide backbone modification to achieve favorable half-life and distribution, can detect elevated HA metabolism in homeostatic, injured, and diseased tissues.


Asunto(s)
Ácido Hialurónico/metabolismo , Neoplasias Hepáticas Experimentales , Imagen por Resonancia Magnética/métodos , Neoplasias Mamarias Experimentales , Sondas Moleculares , Tomografía Óptica/métodos , Enfermedades Vasculares , Animales , Línea Celular Tumoral , Femenino , Humanos , Ácido Hialurónico/química , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Sondas Moleculares/química , Sondas Moleculares/farmacología , Trasplante de Neoplasias , Ratas , Ratas Desnudas , Trasplante Heterólogo , Microambiente Tumoral , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
7.
Cell Adh Migr ; 5(2): 133-41, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21200152

RESUMEN

In pluripotent embryonic stem cells (ESCs), expression of the Hox master regulatory transcription factors that play essential roles in organogenesis, angiogenesis, and maintenance of differentiated tissues, is globally suppressed. We investigated whether differentiation of endothelial cells (ECs) from mouse ESCs was accompanied by activation of distinct Hox gene expression profiles. Differentiation was observed within 3 days, as indicated by the appearance of cells expressing specific endothelial marker genes (Flk-1+ /VE-Cadherin+ ). Expression of HoxA3 and HoxD3, which drive adult endothelial cell invasion and angiogenesis, peaked at day 3 and declined thereafter, whereas expression of HoxA5 and HoxD10, which maintain a mature quiescent EC phenotype, was low at day 3, but increased over time. The temporal and reciprocal changes in HoxD3 and HoxA5 expression were accompanied by corresponding changes in expression of established downstream target genes including integrin ß3 and Thrombospondin-2. Our results indicate that differentiation and maturation of ECs derived from cultured ESCs mimic changes in Hox gene expression that accompany maturation of immature angiogenic endothelium into differentiated quiescent endothelium in vivo.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/fisiología , Células Madre Pluripotentes/citología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/análisis , Cadherinas/genética , Cadherinas/metabolismo , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Endoteliales/citología , Silenciador del Gen , Integrina beta3/genética , Integrina beta3/metabolismo , Ratones , Células Madre Pluripotentes/metabolismo , ARN Interferente Pequeño/metabolismo , Trombospondinas/genética , Trombospondinas/metabolismo , Factores de Tiempo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
J Cell Biol ; 184(1): 57-66, 2009 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-19139262

RESUMEN

Epithelial cells, once dissociated and placed in two-dimensional (2D) cultures, rapidly lose tissue-specific functions. We showed previously that in addition to prolactin, signaling by laminin-111 was necessary to restore functional differentiation of mammary epithelia. Here, we elucidate two additional aspects of laminin-111 action. We show that in 2D cultures, the prolactin receptor is basolaterally localized and physically segregated from its apically placed ligand. Detachment of the cells exposes the receptor to ligation by prolactin leading to signal transducers and activators of transcription protein 5 (STAT5) activation, but only transiently and not sufficiently for induction of milk protein expression. We show that laminin-111 reorganizes mammary cells into polarized acini, allowing both the exposure of the prolactin receptor and sustained activation of STAT5. The use of constitutively active STAT5 constructs showed that the latter is necessary and sufficient for chromatin reorganization and beta-casein transcription. These results underscore the crucial role of continuous laminin signaling and polarized tissue architecture in maintenance of transcription factor activation, chromatin organization, and tissue-specific gene expression.


Asunto(s)
Ensamble y Desensamble de Cromatina , Glándulas Mamarias Animales/fisiología , Factor de Transcripción STAT5/fisiología , Acetilación , Animales , Caseínas/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Distroglicanos/metabolismo , Histonas/metabolismo , Janus Quinasa 2/metabolismo , Laminina/farmacología , Glándulas Mamarias Animales/citología , Ratones , Proteínas de la Leche/metabolismo , Fosforilación , Prolactina/farmacología , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Receptores de Prolactina/análisis , Receptores de Prolactina/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
9.
Nat Clin Pract Oncol ; 5(5): 280-90, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18349857

RESUMEN

Epithelial-mesenchymal transition (EMT) is a phenotypic conversion that facilitates organ morphogenesis and tissue remodeling in physiological processes, such as embryonic development and wound healing. A similar phenotypic conversion is also detected in fibrotic diseases and neoplasia, and is associated with disease progression. EMT in cancer epithelial cells often seems to be an incomplete and bidirectional process. In this Review, we discuss the phenomenon of EMT as it pertains to tumor development, focusing on exceptions to the commonly held rule that EMT promotes invasion and metastasis. We also highlight the role of RAS-controlled signaling mediators, ERK1, ERK2 and phosphatidylinositol 3-kinase, as microenvironmental responsive regulators of EMT.


Asunto(s)
Transformación Celular Neoplásica , Células Epiteliales/fisiología , Células Madre Mesenquimatosas/fisiología , Neoplasias/fisiopatología , Progresión de la Enfermedad , Humanos , Procesos Neoplásicos
10.
Biosens Bioelectron ; 23(8): 1307-13, 2008 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-18221863

RESUMEN

Impedance measurements of cell-based sensors are a primary characterization route for detection and analysis of cellular responses to chemical and biological agents in real time. The detection sensitivity and limitation depend on sensor impedance characteristics and thus on cell patterning techniques. This study introduces a cell patterning approach to bind cells on microarrays of gold electrodes and demonstrates that single-cell patterning can substantially improve impedance characteristics of cell-based sensors. Mouse fibroblast cells (NIH3T3) are immobilized on electrodes through a lysine-arginine-glycine-aspartic acid (KRGD) peptide-mediated natural cell adhesion process. Electrodes are made of three sizes and immobilized with either covalently bound or physically adsorbed KRGD (c-electrodes or p-electrodes). Cells attached to c-electrodes increase the measurable electrical signal strength by 48.4%, 24.2%, and 19.0% for three electrode sizes, respectively, as compared to cells attached to p-electrodes, demonstrating that both the electrode size and surface chemistry play a key role in cell adhesion and spreading and thus the impedance characteristics of cell-based sensors. Single cells patterned on c-electrodes with dimensions comparable to cell size exhibit well-spread cell morphology and substantially outperform cells patterned on electrodes of other configurations.


Asunto(s)
Bioensayo/instrumentación , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Electroquímica/instrumentación , Microelectrodos , Animales , Bioensayo/métodos , Impedancia Eléctrica , Electroquímica/métodos , Diseño de Equipo , Análisis de Falla de Equipo , Ratones , Células 3T3 NIH , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
11.
Cancer Res ; 67(14): 6882-8, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638899

RESUMEN

Toward the goal of developing an optical imaging contrast agent that will enable surgeons to intraoperatively distinguish cancer foci from adjacent normal tissue, we developed a chlorotoxin:Cy5.5 (CTX:Cy5.5) bioconjugate that emits near-IR fluorescent signal. The probe delineates malignant glioma, medulloblastoma, prostate cancer, intestinal cancer, and sarcoma from adjacent non-neoplastic tissue in mouse models. Metastatic cancer foci as small as a few hundred cells were detected in lymph channels. Specific binding to cancer cells is facilitated by matrix metalloproteinase-2 (MMP-2) as evidenced by reduction of CTX:Cy5.5 binding in vitro and in vivo by a pharmacologic blocker of MMP-2 and induction of CTX:Cy5.5 binding in MCF-7 cells following transfection with a plasmid encoding MMP-2. Mouse studies revealed that CTX:Cy5.5 has favorable biodistribution and toxicity profiles. These studies show that CTX:Cy5.5 has the potential to fundamentally improve intraoperative detection and resection of malignancies.


Asunto(s)
Carbocianinas/química , Neoplasias/metabolismo , Venenos de Escorpión/química , Animales , Neoplasias Encefálicas/metabolismo , Colorantes Fluorescentes/química , Glioma/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Microscopía Fluorescente/métodos , Neovascularización Patológica , Fotones , Ratas
12.
Biosens Bioelectron ; 23(2): 253-60, 2007 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-17560777

RESUMEN

Microarrays of single macrophage cell-based sensors were developed and demonstrated for potential real-time bacterium detection by synchrotron FTIR microscopy. The cells were patterned on gold electrodes of silicon oxide substrates by a surface engineering technique, in which the gold electrodes were immobilized with fibronectin to mediate cell adhesion and the silicon oxide background was passivated with polyethylene glycol (PEG) to resist protein adsorption and cell adhesion. Cell morphology and IR spectra of single, double, and triple cells on gold electrodes exposed to lipopolysaccharide (LPS) of different concentrations were compared to reveal the detection capability of this cell-based sensing platform. The single-cell-based system was found to generate the most significant and consistent IR spectrum shifts upon exposure to LPS, thus providing the highest detection sensitivity. Changes in cell morphology and IR shifts upon cell exposure to LPS were found to be dependent on the LPS concentration and exposure time, which established a method for the identification of LPS concentration and infected cell population. Possibility of using this single-cell system with conventional IR spectroscopy as well as its limitation was investigated by comparing IR spectra of single-cell arrays with gold electrode surface areas of 25, 100, and 400 microm2 using both synchrotron and conventional FTIR spectromicroscopes. This cell-based platform may potentially provide real-time, label-free, and rapid bacterial detection, and allow for high-throughput statistical analyses, and portability.


Asunto(s)
Bacterias/aislamiento & purificación , Bacterias/metabolismo , Técnicas Biosensibles/instrumentación , Recuento de Colonia Microbiana/instrumentación , Lipopolisacáridos/análisis , Macrófagos/microbiología , Espectroscopía Infrarroja por Transformada de Fourier/instrumentación , Animales , Línea Celular , Recuento de Colonia Microbiana/métodos , Diseño de Equipo , Análisis de Falla de Equipo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Sincrotrones/instrumentación , Integración de Sistemas
13.
J Biol Chem ; 282(22): 16667-80, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17392272

RESUMEN

CD44 is an integral hyaluronan receptor that can promote or inhibit motogenic signaling in tumor cells. Rhamm is a nonintegral cell surface hyaluronan receptor (CD168) and intracellular protein that promotes cell motility in culture. Here we describe an autocrine mechanism utilizing cell surface Rhamm-CD44 interactions to sustain rapid basal motility in invasive breast cancer cell lines that requires endogenous hyaluronan synthesis and the formation of Rhamm-CD44-ERK1,2 complexes. Motile/invasive MDA-MB-231 and Ras-MCF10A cells produce more endogenous hyaluronan, cell surface CD44 and Rhamm, an oncogenic Rhamm isoform, and exhibit more elevated basal activation of ERK1,2 than less invasive MCF7 and MCF10A breast cancer cells. Furthermore, CD44, Rhamm, and ERK1,2 uniquely co-immunoprecipitate and co-localize in MDA-MB-231 and Ras-MCF10A cells. Combinations of anti-CD44, anti-Rhamm antibodies, and a MEK1 inhibitor (PD098059) had less-than-additive blocking effects, suggesting the action of all three proteins on a common motogenic signaling pathway. Collectively, these results show that cell surface Rhamm and CD44 act together in a hyaluronan-dependent autocrine mechanism to coordinate sustained signaling through ERK1,2, leading to high basal motility of invasive breast cancer cells. Therefore, an effect of CD44 on tumor cell motility may depend in part on its ability to partner with additional proteins, such as cell surface Rhamm.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama/metabolismo , Movimiento Celular , Proteínas de la Matriz Extracelular/metabolismo , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/biosíntesis , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Anticuerpos Monoclonales/farmacología , Comunicación Autocrina/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Femenino , Flavonoides/farmacología , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , Mitosis/efectos de los fármacos , Invasividad Neoplásica , Isoformas de Proteínas/metabolismo
14.
Langmuir ; 23(8): 4472-9, 2007 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-17371055

RESUMEN

Single cell patterning holds important implications for biology, biochemistry, biotechnology, medicine, and bioinformatics. The challenge for single cell patterning is to produce small islands hosting only single cells and retaining their viability for a prolonged period of time. This study demonstrated a surface engineering approach that uses a covalently bound short peptide as a mediator to pattern cells with improved single cell adhesion and prolonged cellular viability on gold patterned SiO2 substrates. The underlying hypothesis is that cell adhesion is regulated by the type, availability, and stability of effective cell adhesion peptides, and thus covalently bound short peptides would promote cell spreading and, thus, single cell adhesion and viability. The effectiveness of this approach and the underlying mechanism for the increased probability of single cell adhesion and prolonged cell viability by short peptides were studied by comparing cellular behavior of human umbilical cord vein endothelial cells on three model surfaces whose gold electrodes were immobilized with fibronectin, physically adsorbed Arg-Glu-Asp-Val-Tyr, and covalently bound Lys-Arg-Glu-Asp-Val-Tyr, respectively. The surface chemistry and binding properties were characterized by reflectance Fourier transform infrared spectroscopy. Both short peptides were superior to fibronectin in producing adhesion of only single cells, whereas the covalently bound peptide also reduced apoptosis and necrosis of adhered cells. Controlling cell spreading by peptide binding domains to regulate apoptosis and viability represents a fundamental mechanism in cell-materials interaction and provides an effective strategy in engineering arrays of single cells.


Asunto(s)
Análisis por Micromatrices/instrumentación , Péptidos/química , Adhesión Celular , Supervivencia Celular , Electroquímica , Electrodos , Endotelio Vascular/citología , Fibronectinas/química , Humanos , Análisis por Micromatrices/métodos , Microscopía Confocal , Microscopía Fluorescente , Dióxido de Silicio/química , Especificidad por Sustrato , Propiedades de Superficie , Ingeniería de Tejidos
15.
J Am Chem Soc ; 128(4): 1197-203, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16433536

RESUMEN

Cellular patterning on silicon platforms is the basis for development of integrated cell-based biosensing devices, for which long-term cell selectivity and biostability remain a major challenge. We report the development of a silicon-based platform in a metal-insulator format capable of producing uniform and biostable cell patterns with long-term cell selectivity. Substrates patterned with arrays of gold electrodes were surface-engineered such that the electrodes were activated with fibronectin to mediate cell attachment and the silicon oxide background was passivated with PEG to resist protein adsorption and cell adhesion. Three types of oxide surfaces, i.e., native oxide, dry thermally grown oxide, and wet thermally grown oxide, were produced to illustrate the effect of oxide state of the surface on long-term cell selectivity. Results indicated that the cell selectivity over time differed dramatically among three patterned platforms and the best cell selectivity was found on the dry oxide surface for up to 10 days. Surface analysis results suggested that this enhancement in cell selectivity may be related to the presence of additional, more active oxide states on the dry oxide surface supporting the stability of PEG films and effectively suppressing the cell adhesion. This research offers a new strategy for development of stable and uniform cell-patterned surfaces, which is versatile for immobilization of silane-based chemicals for preparation of biostable interfaces.


Asunto(s)
Técnicas Citológicas/métodos , Oro/química , Dióxido de Silicio/química , Silicio/química , Animales , Técnicas Biosensibles/métodos , Carbocianinas/química , Línea Celular , Fibronectinas/química , Macrófagos/citología , Ratones , Microscopía Fluorescente , Oxidación-Reducción , Polietilenglicoles/química , Propiedades de Superficie
16.
Biosens Bioelectron ; 20(9): 1697-708, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15681184

RESUMEN

Clean silicon and gold-patterned silicon platforms were modified with methoxy-polyethylene glycol (M-PEG silane) via a self-assembly technique, which significantly improved their plasma protein resistance capability and cell patterning selectivity. Fibrinogen and IgG were used as model plasma proteins to study the efficacy of PEG layers in resisting protein adsorption. Selective cell patterning on the gold regions of a gold-patterned silicon substrate and tissue compatibility were studied with macrophage and fibroblast cells. The research also revealed how the presence of gold electrodes on a silicon substrate would influence the cell patterning selectivity. Our experimental results showed that the PEG-modified silicon surfaces had a high resistivity to protein and cell attachment and that the PEG-modified gold-patterned silicon surfaces nearly completely eliminated the protein adsorption and cell attachment on silicon. This study provides a new approach to developing biocompatible surfaces for silicon-based BioMEMS devices, particularly for biosensors where a metal-insulator format must be enforced.


Asunto(s)
Técnicas Biosensibles/métodos , Técnicas de Cultivo de Célula/métodos , Fibroblastos/fisiología , Oro/química , Macrófagos/fisiología , Silicio/química , Adsorción , Animales , Materiales Biocompatibles/análisis , Materiales Biocompatibles/química , Técnicas Biosensibles/instrumentación , Adhesión Celular/fisiología , Línea Celular , Supervivencia Celular , Fibrinógeno/química , Fibroblastos/citología , Inmunoglobulina G/química , Macrófagos/citología , Ensayo de Materiales , Ratones , Unión Proteica , Silanos/química , Propiedades de Superficie , Ingeniería de Tejidos/métodos
17.
Biomaterials ; 25(16): 3315-24, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-14980426

RESUMEN

We report an effective approach to patterning cells on gold-silicon dioxide substrates with high precision, selectivity, stability, and reproducibility. This technique is based on photolithography and surface molecular engineering and requires no cell positioning or delivery devices, thus significantly reducing the potential damage to cells. The cell patterning was achieved by activating the gold regions of the substrate with functionalized thiols that covalently bind proteins onto the gold regions to guide subsequent cell adhesion while passivating the silicon dioxide background with polyethylene glycol to resist cell adhesion. Fourier transform infrared reflectance spectroscopy verified the successful immobilization of proteins on gold surfaces. Protein patterns were visualized by tagging proteins with Rhodamine fluorescent probes. Time-of-flight secondary ion mass spectrometry was used to characterize the chemistry of both the cell-adhesive and cell-resistant regions of surfaces after each key chemical reaction occurring during the molecular surface engineering. The ability of the engineered surfaces to guide cell adhesion was illustrated by differential interference contrast (DIC) reflectance microscopy. The cell patterning technique introduced in this study is compatible with micro- and photo-electronics, and may have many medical, environmental, and defense applications.


Asunto(s)
Materiales Biocompatibles/química , Técnicas de Cultivo de Célula/métodos , Oro/química , Macrófagos/citología , Macrófagos/fisiología , Ingeniería de Tejidos/métodos , Animales , Adhesión Celular/fisiología , Técnicas de Cultivo de Célula/instrumentación , División Celular/fisiología , Línea Celular , Proteínas de la Matriz Extracelular/metabolismo , Ensayo de Materiales , Ratones , Dióxido de Silicio , Propiedades de Superficie , Ingeniería de Tejidos/instrumentación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA