Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioengineering (Basel) ; 11(4)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38671796

RESUMEN

Mesenchymal stromal cells (MSCs) have displayed potential in regenerating organ function due to their anti-fibrotic, anti-inflammatory, and regenerative properties. However, there is a need for delivery systems to enhance MSC retention while maintaining their anti-fibrotic characteristics. This study investigates the feasibility of using alginate hydrogel microstrands as a cell delivery vehicle to maintain MSC viability and phenotype. To accommodate cell implantation needs, we invented a Syringe-in-Syringe approach to reproducibly fabricate microstrands in small numbers with a diameter of around 200 µm and a porous structure, which would allow for transporting nutrients to cells by diffusion. Using murine NIH 3T3 fibroblasts and primary embryonic 16 (E16) salivary mesenchyme cells as primary stromal cell models, we assessed cell viability, growth, and expression of mesenchymal and fibrotic markers in microstrands. Cell viability remained higher than 90% for both cell types. To determine cell number within the microstrands prior to in vivo implantation, we have further optimized the alamarBlue assay to measure viable cell growth in microstrands. We have shown the effect of initial cell seeding density and culture period on cell viability and growth to accommodate future stromal cell delivery and implantation. Additionally, we confirmed homeostatic phenotype maintenance for E16 mesenchyme cells in microstrands.

2.
Exp Biol Med (Maywood) ; 247(23): 2103-2118, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36000165

RESUMEN

Alginate, a naturally occurring polysaccharide, has been widely used in cell encapsulation, 3D culture, cell therapy, tissue engineering, and regenerative medicine. Alginate's frequent use comes from its biocompatibility and ability to easily form hydrogel in a variety of forms (e.g. microcapsules, microfibers, and porous scaffolds), which can provide immunoprotection for cell therapy and mimic the extracellular matrix for tissue engineering. During the past 15 years, alginate hydrogel microfibers have attracted more and more attention due to its continuous thin tubular structures (diameter or shell thickness ⩽ 200 µm), high-density cell growth, high handleability and retrievability, and scalability. This review article provides a concise overview of alginate and its resultant hydrogel microfibers for the purpose of promoting multidisciplinary, collaborative, and convergent research in the field. It starts with a historical review of alginate as biomaterials and provides basics about alginate structure, properties, and mechanisms of hydrogel formation, followed by current challenges in effective cell delivery and functional tissue engineering. In particular, this work discusses how alginate microfiber technology could provide solutions to unmet needs with a focus on the current state of the art of alginate microfiber technology and its applications in 3D cell culture, cell delivery, and tissue engineering. At last, we discuss future directions in the perspective of alginate-based advanced technology development in biology and medicine.


Asunto(s)
Alginatos , Andamios del Tejido , Andamios del Tejido/química , Alginatos/química , Materiales Biocompatibles/química , Ingeniería de Tejidos , Hidrogeles/química
3.
Bioengineering (Basel) ; 9(1)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35049747

RESUMEN

Understanding the different regulatory functions of epithelial and mesenchymal cell types in salivary gland development and cellular organization is essential for proper organoid formation and salivary gland tissue regeneration. Here, we demonstrate a biocompatible platform using pre-formed alginate hydrogel microtubes to facilitate direct epithelial-mesenchymal cell interaction for 3D salivary gland cell organization, which allows for monitoring cellular organization while providing a protective barrier from cell-cluster loss during medium changes. Using mouse salivary gland ductal epithelial SIMS cells as the epithelial model cell type and NIH 3T3 fibroblasts or primary E16 salivary mesenchyme cells as the stromal model cell types, self-organization from epithelial-mesenchymal interaction was examined. We observed that epithelial and mesenchymal cells undergo aggregation on day 1, cavitation by day 4, and generation of an EpCAM-expressing epithelial cell layer as early as day 7 of the co-culture in hydrogel microtubes, demonstrating the utility of hydrogel microtubes to facilitate heterotypic cell-cell interactions to form cavitated organoids. Thus, pre-formed alginate microtubes are a promising co-culture method for further understanding epithelial and mesenchymal interaction during tissue morphogenesis and for future practical applications in regenerative medicine.

4.
Biofabrication ; 13(3)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33930885

RESUMEN

Alginate hydrogels in microtubular structures have great potential to advance three-dimensional (3D) culture, organoid formation, tissue engineering, and cell therapy. To address the need of fabricating consistent, stable hydrogel microtubes for efficient large organoid generation in a simple and quick manner, we have designed needle-in-needle devices to fabricate alginate hydrogel microtubes without any dead volume of the cell-alginate mixture and demonstrated the feasibility of injecting and culturing embryoid bodies in these pre-made hydrogel microtubes. We further used a reverse engineering approach to find out the optimal flow rates and alginate concentration for fabricating pre-made hydrogel microtubes with desired diameter using particular sets of needle-in-needle devices. We established the relationship of the alginate flow rate with diameter and wall thickness of the microtube using mathematic modeling. It offers a way to determine the flow rate for making microtubes with the desired dimension. Additionally, we evaluated the effect of CaCl2concentration on the diameter as well as stem cell viability. At last, we demonstrated the capacity of fabricating hydrogel microtubes of varying diameters using three sets of needle-in-needle devices and evaluated stem cell growth in these hydrogel microtubes. It provides a new avenue to accessible, repeatable, scalable, and easy to use pre-made 'off-the-shelf' hydrogel microtubes for 3D cell culture including, but not limiting to stem cells.


Asunto(s)
Hidrogeles , Alginatos , Técnicas de Cultivo de Célula , Supervivencia Celular , Cuerpos Embrioides , Células Madre , Ingeniería de Tejidos
5.
Adv Biosyst ; 4(9): e2000004, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32734694

RESUMEN

Age-related human trabecular meshwork (HTM) cell loss is suggested to affect its ability to regulate aqueous humor outflow in the eye. In addition, disease-related HTM cell loss is suggested to lead to elevated intraocular pressure in glaucoma. Induced pluripotent stem cell (iPSC)-derived trabecular meshwork (TM) cells are promising autologous cell sources that can be used to restore the declining TM cell population and function. Previously, an in vitro HTM model is bioengineered for understanding HTM cell biology and screening of pharmacological or biological agents that affect trabecular outflow facility. In this study, it is demonstrated that human iPSC-derived TM cells cultured on SU-8 scaffolds exhibit HTM-like cell morphology, extracellular matrix deposition, and drug responsiveness to dexamethasone treatment. These findings suggest that iPSC-derived TM cells behave like primary HTM cells and can thus serve as reproducible and scalable cell sources when using this in vitro system for glaucoma drug screening and further understanding of outflow pathway physiology, leading to personalized medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas , Modelos Biológicos , Malla Trabecular , Biomimética , Técnicas de Cultivo de Célula/métodos , Glaucoma/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Malla Trabecular/citología , Malla Trabecular/metabolismo
6.
Acta Biomater ; 105: 203-213, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31982588

RESUMEN

Human Schlemm's canal (HSC) cells are critical for understanding outflow physiology and glaucoma etiology. However, primary donor cells frequently used in research are difficult to isolate. HSC cells exhibit both vascular and lymphatic markers. Human adipose-derived stem cells (ADSCs) represent a potential source of HSC due to their capacity to differentiate into both vascular and lymphatic endothelial cells, via VEGF-A and VEGF-C. Shear stress plays a critical role in maintaining HSC integrity, function, and PROX1 expression. Additionally, the human trabecular meshwork (HTM) microenvironment could provide cues for HSC-like differentiation. We hypothesize that subjecting ADSCs to VEGF-A or VEGF-C, shear stress, and co-culture with HTM cells could provide biological, mechanical, and cellular cues necessary for HSC-like differentiation. To test this hypothesis, effects of VEGF-A, VEGF-C, and shear stress on ADSC differentiation were examined and compared to primary HSC cells in terms of cell morphology, and HSC marker expression using qPCR, immunoblotting, and immunocytochemistry analysis. Furthermore, the effect of co-culture with HTM cells on porous scaffolds on ADSC differentiation was studied. Treatment with VEGF-C under shear stress is effective in differentiating ADSCs into PROX1-expressing HSC-like cells. Co-culture with HTM cells on porous scaffolds leads to HTM/ADSC-derived HSC-like constructs that regulate through-flow and respond as expected to dexamethasone. STATEMENT OF SIGNIFICANCE: We successfully generated human Schlemm's canal (HSC) like cells from adipocyte-derived stem cells induced by biochemical and biomechanical cues as well as bioengineered human trabecular meshwork (HTM) on micropatterned, porous SU8 scaffolds. These stem cell-derived HSC-like cells co-cultured with HTM cells on SU8 scaffolds can regulate through-flow, and in particular, are responsive to steroid treatment as expected. These findings show that ADSC-derived HSC-like cells have the potential to recreate the ocular outflow pathway for in vitro glaucoma drug screening. To the best of our knowledge, it is the very first time to demonstrate derivation of Schlemm's canal-like cells from stem cells. It provides an important alternative source to primary Schlemm's canal cells that are very difficult to be isolated and cultured from human donors.


Asunto(s)
Bioingeniería , Diferenciación Celular , Evaluación Preclínica de Medicamentos , Glaucoma/tratamiento farmacológico , Células Madre/citología , Tejido Adiposo/citología , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , Dexametasona/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Glaucoma/patología , Humanos , Imagen Óptica , Perfusión , Células Madre/efectos de los fármacos , Malla Trabecular/citología
7.
Sci Rep ; 6: 31771, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27558173

RESUMEN

Glomerular hypertension is an important factor exacerbating glomerular diseases to end-stage renal diseases because, ultimately, it results in glomerular sclerosis (especially in hypertensive and diabetic nephropathy). The precise mechanism of glomerular sclerosis caused by glomerular hypertension is unclear, due partly to the absence of suitable in vitro or in vivo models capable of mimicking and regulating the complex mechanical forces and/or organ-level disease processes. We developed a "glomerulus-on-a-chip" (GC) microfluidic device. This device reconstitutes the glomerulus with organ-level glomerular functions to create a disease model-on-a chip that mimics hypertensive nephropathy in humans. It comprises two channels lined by closely opposed layers of glomerular endothelial cells and podocytes that experience fluid flow of physiological conditions to mimic the glomerular microenvironment in vivo. Our results revealed that glomerular mechanical forces have a crucial role in cellular cytoskeletal rearrangement as well as the damage to cells and their junctions that leads to increased glomerular leakage observed in hypertensive nephropathy. Results also showed that the GC could readily and flexibly meet the demands of a renal-disease model. The GC could provide drug screening and toxicology testing, and create potential new personalized and accurate therapeutic platforms for glomerular disease.


Asunto(s)
Hipertensión Renal/fisiopatología , Enfermedades Renales/fisiopatología , Glomérulos Renales/fisiopatología , Dispositivos Laboratorio en un Chip , Nefritis/fisiopatología , Actinas/metabolismo , Animales , Adhesión Celular , Técnicas de Cocultivo , Citoesqueleto/metabolismo , Humanos , Hipertensión/fisiopatología , Inmunoglobulina G/metabolismo , Técnicas In Vitro , Riñón/fisiopatología , Fallo Renal Crónico/fisiopatología , Podocitos/citología , Ratas , Ratas Endogámicas SHR , Microglobulina beta-2/metabolismo
8.
J Control Release ; 165(3): 226-33, 2013 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-23228849

RESUMEN

Herein we describe the development and implementation of a nanoporous cell-therapy device with controllable biodegradation. Dopamine-secreting PC12 cells were housed within newly formulated alginate-glutamine degradable polylysine (A-GD-PLL) microcapsules. The A-GD-PLL microcapsules provided a 3-D microenvironment for good spatial cell growth, viability and proliferation. The microcapsules were subsequently placed within a poly(ethylene glycol) (PEG)-coated poly(ε-caprolactone) (PCL) chamber covered with a PEG-grafted PCL nanoporous membrane formed by phase inversion. To enhance PC12 cell growth and to assist in controlled degradation of both the PC12 cells and the device construct, small PCL capsules containing neural growth factor (PCL-NGF) and a poly(lactic-co-glycolic acid) pellet containing glutamine (PLGA-GLN) were also placed within the PCL chamber. Release of NGF from the PCL-NGF capsules facilitated cell proliferation and viability, while the controlled release of GLN from the PLGA-GLN pellet resulted in A-GD-PLL microcapsule degradation and eventual PC12 cell death following a pre-specified period of time (4 weeks in this study). In vivo, our device was found to be well tolerated and we successfully demonstrated the controlled release of dopamine over a period of four weeks. This integrated biodegradable device holds great promise for the future treatment of a variety of diseases.


Asunto(s)
Trasplante de Células/instrumentación , Dopamina/administración & dosificación , Alginatos/química , Animales , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/química , Dopamina/química , Diseño de Equipo , Ácido Glucurónico/química , Glutamina/administración & dosificación , Glutamina/química , Ácidos Hexurónicos/química , Ácido Láctico/química , Ratones , Ratones Endogámicos BALB C , Nanoestructuras/administración & dosificación , Nanoestructuras/química , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/química , Células PC12 , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polilisina/química , Porosidad , Ratas
9.
Biomaterials ; 33(22): 5638-49, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22575837

RESUMEN

Although alginate-poly-L-lysine (AP(L)) encapsulation of cells producing bioactive peptides has been widely tested, it is unknown whether AP(L) supports lasting catabolic functions of encapsulated cells in adipose tissue, which are required for obesity reduction. We tested functions of AP(L)-encapsulated fibroblasts isolated from wild-type (WT) and aldehyde dehydrogenase 1a1 knockout mice (KO), which resist obesity on a high-fat (HF) diet, have a higher metabolic rate, and express increased levels of thermogenic uncoupling protein-1 (Ucp1) in their deleterious visceral fat depots compared to WT mice. To enable in vivo detection and quantification, fibroblasts were stably transfected with green-fluorescent protein. WT- or KO-containing microcapsules were injected into two visceral depots of WT mice fed an HF diet. Eighty days after transplantation, microcapsules were located in vivo using magnetic resonance imaging. KO microcapsules prevented weight gain in obese WT mice compared to a mock- and WT capsule-injected groups on an HF diet. The weight loss in KO-treated mice corresponded to lipid reduction and induction of thermogenesis in the injected visceral fat. The non-treated subcutaneous fat was not altered. Our data suggest that the AP(L) polymer supports long-term catabolic functions of genetically-modified fibroblasts, which can be potentially used for depot-specific obesity treatment.


Asunto(s)
Alginatos/química , Fibroblastos/citología , Fibroblastos/fisiología , Grasa Intraabdominal/citología , Grasa Intraabdominal/fisiología , Metabolismo de los Lípidos/fisiología , Polilisina/análogos & derivados , Termogénesis/fisiología , Animales , Técnicas de Cultivo de Célula/métodos , Supervivencia Celular , Femenino , Metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polilisina/química , Andamios del Tejido
10.
Nat Nanotechnol ; 6(11): 747-54, 2011 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22002097

RESUMEN

Many transfection techniques can deliver biomolecules into cells, but the dose cannot be controlled precisely. Delivering well-defined amounts of materials into cells is important for various biological studies and therapeutic applications. Here, we show that nanochannel electroporation can deliver precise amounts of a variety of transfection agents into living cells. The device consists of two microchannels connected by a nanochannel. The cell to be transfected is positioned in one microchannel using optical tweezers, and the transfection agent is located in the second microchannel. Delivering a voltage pulse between the microchannels produces an intense electric field over a very small area on the cell membrane, allowing a precise amount of transfection agent to be electrophoretically driven through the nanochannel, the cell membrane and into the cell cytoplasm, without affecting cell viability. Dose control is achieved by adjusting the duration and number of pulses. The nanochannel electroporation device is expected to have high-throughput delivery applications.


Asunto(s)
Electroporación/instrumentación , Electroporación/métodos , Modelos Biológicos , Nanotecnología/métodos , Transfección/métodos , Animales , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/fisiología , Supervivencia Celular/fisiología , Simulación por Computador , Diseño de Equipo , Humanos , Células Jurkat , Ratones , Microfluídica/métodos
11.
Biomicrofluidics ; 5(3): 32007-3200710, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22662030

RESUMEN

Cell-microscale pattern surface interactions are crucial to understand many fundamental biological questions and develop regenerative medicine and tissue engineering approaches. In this work, we demonstrated a simple method to pattern PDMS surface by sacrificing poly vinyl pyrrolidone (PVP) electrospinning nanofibers and investigated the growth profile of cells on the modified patterned surfaces using stroma cells. The stromal cells were observed to exhibit good viability on this modified surface and the patterned surface with alignment nanofibers could promote cell migration. Furthermore, the modified PDMS surface was integrated with microfluidic channels to create the microscale spatial factor and was used to explore the cell migration and orientation under this microsystem. Both spatial factor and patterned surfaces were found to contribute to the complex cell orientation under the combined dual effects. This established method is simple, fast, and easy for use, demonstrating the potential of this microsystem for applications in addressing biological questions in complex environment.

12.
Biosens Bioelectron ; 26(2): 778-83, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20630739

RESUMEN

Liposome nanoparticles (LNs) with a targeting ligand were used in a semi-continuous flow electroporation (SFE) device to enhance in vitro delivery of exogenous oligonucleotides (ODN). Nanoparticles comprising transferrin-targeted lipoplex encapsulating ODN G3139 were mixed with K562 cells (a chronic myeloid leukemia cell line) and incubated for half an hour to accomplish nanoparticle binding. The mixture was then flowed through a SFE channel where electric pulses were given. Better ODN delivery efficiency was achieved with an increase of ∼24% to the case in combination of non-targeted LNs and SFE, and ∼60% to the case using targeted LNs alone, respectively. The MTS assay results confirmed cell viability greater than 75%.


Asunto(s)
ADN sin Sentido/genética , ADN sin Sentido/farmacocinética , Electroporación/instrumentación , Análisis de Inyección de Flujo/instrumentación , Marcación de Gen/instrumentación , Transfección/instrumentación , ADN sin Sentido/administración & dosificación , Diseño de Equipo , Análisis de Falla de Equipo , Humanos , Células K562
13.
J Control Release ; 141(1): 62-9, 2010 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-19716852

RESUMEN

A multi-inlet microfluidic hydrodynamic focusing (MF) system to prepare lipopolyplex (LP) containing Bcl-2 antisense deoxyoligonucleotide (ODN) was developed and evaluated. The lipopolyplex nanoparticles consist of ODN:protamine:lipids (1:0.3:12.5wt/wt ratio) and the lipids included DC-Chol:egg PC:PEG-DSPE (40:58:2mol/mol%). Using K562 human erythroleukemia cells, which contain an abundance of Bcl-2 and overexpression of transferrin receptors (TfR), and G3139 (oblimerson sodium or Genasense(TM)) as a model cell line and drug, respectively, the Bcl-2 down-regulation at the mRNA and protein levels as well as cellular uptake and apoptosis was compared between the conventional bulk mixing (BM) method and the MF method. The lipopolyplex size and surface charge were characterized by dynamic light scattering (DLS) and zeta potential (zeta) measurement, respectively, while the ODN encapsulation efficiency was determined by gel electrophoresis. Cryogenic transmission electron microscopy (Cryo-TEM) was used to determine the morphology of LPs. Our results demonstrated that MF produced LP nanoparticles had similar structures but smaller size and size distribution compared to BM LP nanoparticles. MF LP nanoparticles had higher level of Bcl-2 antisense uptake and showed more efficient down-regulation of Bcl-2 protein level than BM LP nanoparticles.


Asunto(s)
Portadores de Fármacos/química , Microfluídica/métodos , Nanopartículas/química , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Apoptosis/efectos de los fármacos , Western Blotting , Microscopía por Crioelectrón , Regulación hacia Abajo , Fluoresceína-5-Isotiocianato , Humanos , Células K562 , Liposomas , Oligodesoxirribonucleótidos Antisentido/química , Oligodesoxirribonucleótidos Antisentido/genética , Tamaño de la Partícula , Receptores de Transferrina/biosíntesis , Propiedades de Superficie
14.
Mol Pharm ; 6(5): 1333-42, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19552481

RESUMEN

Polyethylenimine (PEI) and plasmid DNA (pDNA) complexes (PEI/pDNA) are nonviral vectors for gene delivery. The conventional method for producing these complexes involves bulk mixing (BM) of PEI and DNA followed by vortexing which at low N/P ratios results in large particle size distribution, low cytotoxicity, and poor gene transfection, while at high N/P ratios it results in small particle size and better gene transfection but high cytotoxicity. To improve size control, gene transfection efficiency, and cytotoxicity, in this study, we used a microfluidic hydrodynamic focusing (MF) device to prepare PEI/pDNA complexes at N/P = 3.3 and 6.7. We used bulk mixing as control, mouse NIH 3T3 fibroblast cells and mouse embryonic stem (mES) cells as model cell lines, plasmid encoding green fluorescent protein (pGFP) and secreted alkaline phosphatase (pSEAP) as the reporter gene, and commercially available Lipofectamine 2,000 as a positive control. The complexes were characterized by atomic force microscopy (AFM), dynamic light scattering (DLS), and zeta potential (zeta) measurement. Confocal laser scanning microscopy (CLSM) and fluorescent labeling techniques were used to visualize the complex size distribution, complexation uniformity, and cellular distribution. The results showed that MF produced complexes were smaller and more uniformly complexed and had higher cell viability and improved exogenous gene expression.


Asunto(s)
ADN Recombinante/administración & dosificación , Técnicas Analíticas Microfluídicas , Polietileneimina/química , Fosfatasa Alcalina/genética , Animales , Supervivencia Celular , Células Cultivadas , ADN Recombinante/genética , Sistemas de Liberación de Medicamentos , Colorantes Fluorescentes , Expresión Génica , Genes Reporteros , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Ratones , Microscopía de Fuerza Atómica , Microscopía Confocal , Células 3T3 NIH , Nanopartículas/química , Nanotecnología , Plásmidos/administración & dosificación , Plásmidos/genética , Dispersión de Radiación , Transfección
15.
Anal Chem ; 81(11): 4414-21, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19419195

RESUMEN

We have recently developed a semicontinuous flow electroporation (SFE) device for in vitro DNA delivery. Cells mixed with plasmid DNA continuously flowed through a serpentine channel, the side walls of which also serving as electrodes. With the use of pWizGFP plasmid and K562 cells as a model system, SFE showed better transgene expression (10-15%) compared to a commercial electroporation system. Quantitative results via MTS assay also revealed a 50% or higher cell viability. Similar observations were also found with pWizGFP transfection to mouse embryonic stem cells. Such improvements were attributed to less gas formation and Joule heating in SFE.


Asunto(s)
Electroporación/instrumentación , Técnicas de Transferencia de Gen/instrumentación , Técnicas Analíticas Microfluídicas/instrumentación , Animales , Línea Celular Tumoral , Supervivencia Celular , Electrodos , Embrión de Mamíferos/citología , Diseño de Equipo , Humanos , Ratones , Plásmidos/genética , Células Madre/citología , Temperatura , Transfección
16.
Biotechnol Prog ; 25(2): 535-42, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19334083

RESUMEN

Tissues are composed of multiple cell types in a well-organized three-dimensional (3D) microenvironment. To faithfully mimic the tissue in vivo, tissue-engineered constructs should have well-defined 3D chemical and spatial control over cell behavior to recapitulate developmental processes in tissue- and organ-specific differentiation and morphogenesis. It is a challenge to build a 3D complex from two-dimensional (2D) patterned structures with the presence of cells. In this study, embryonic stem (ES) cells grown on polymeric scaffolds with well-defined microstructure were constructed into a multilayer cell-scaffold complex using low pressure carbon dioxide (CO(2)) and nitrogen (N(2)). The mouse ES cells in the assembled constructs were viable, retained the ES cell-specific gene expression of Oct-4, and maintained the formation of embryoid bodies (EBs). In particular, cell viability was increased from 80% to 90% when CO(2) was replaced with N(2). The compressed gas-assisted bioassembly of stem cell-polymer constructs opens up a new avenue for tissue engineering and cell therapy.


Asunto(s)
Células Madre Embrionarias/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Supervivencia Celular , Gases , Ratones
17.
Biomed Microdevices ; 11(4): 795-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19288199

RESUMEN

A novel method of making microcapsules in a macrocapsule is demonstrated as a 3-D culture system in this article. Mouse embryonic stem (mES) cells as model cells were used in the 3-D culture space, and the cell viability and histological observation were conducted. Furthermore, Oct4 gene expression was evaluated for the undifferentiated status of mES cells in this 3-D model. The results showed that mES cells can grow in this 3-D model and retain their normal viability and morphology. This 3-D model allows mES cells to stay in the undifferentiated state better than 2-D culture systems. This work demonstrates a new 3-D tissue model which can provide an in vivo like microenvironment for non-differentiated mES cells with good immunoisolation. This approach may bridge the gap between traditional 2-D cell culture and animal models.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/citología , Modelos Biológicos , Ingeniería de Tejidos/métodos , Animales , Cápsulas , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Ratones
18.
Pharm Res ; 26(6): 1516-24, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19291371

RESUMEN

PURPOSE: Transferrin (Tf) conjugated lipopolyplexes (LPs) carrying G3139, an antisense oligonucleotide for Bcl-2, were synthesized and evaluated in Tf receptor positive K562 erythroleukemia cells and then in a murine K562 xenograft model. MATERIALS AND METHODS: Particle size and Zeta potentials of transferrin conjugated lipopolyplexs containing G3139 (Tf-LP-G3139) were measured by Dynamic Light Scattering and ZetaPALS. In vitro and in vivo sample's Bcl-2 downregulation was analyzed using Western blot and tumor tissue samples also exhibited by immunohistochemistry method. For athymic mice bearing with K562 xenograft tumors, tumor growth inhibition and survival rate were investigated. Nanoparticle distribution in 3-D cell cluster was observed by Laser scan confocal microscopy. IL-12 production in the plasma was measured by ELISA kit. RESULTS: In vitro, Tf-LP-G3139 was more effective in inducing down regulation of Bcl-2 in K562 cells than non-targeted LP-G3139, free G3139 and mismatched control ODN-G4126 in the same formulation. In vivo Tf-LP-G3139 was less effective than free G3139 in Bcl-2 down regulation. 3-D cell cluster model diffusion results indeed indicated limited penetration of the LPs into the cell cluster. Finally, the therapeutic efficacies of Tf-LP-G3139 and free G3139 were determined in the K562 xenograft model. Tf-LP-G3139 showed slower plasma clearance, higher AUC, and greater accumulation in the tumor compared to free G3139. In addition, Tf-LP-G3139 was found to be more effective in tumor growth inhibition and prolonging mouse survival than free G3139. This was associated with increased spleen weight and IL-12 production in the plasma. CONCLUSION: The role of the immune system in the therapeutic response obtained with the Tf-LPs is necessary and in vitro 3-D cell cluster model can be a potential tool to evaluate the nanoparticle distribution.


Asunto(s)
Lípidos , Neoplasias/terapia , Tionucleótidos/administración & dosificación , Tionucleótidos/uso terapéutico , Transferrina , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Portadores de Fármacos , Humanos , Ratones , Ratones Endogámicos ICR , Ratones Desnudos , Nanopartículas , Neoplasias/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Receptores de Transferrina/metabolismo , Tionucleótidos/inmunología , Tionucleótidos/farmacocinética , Transfección
19.
Biomaterials ; 29(31): 4253-9, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18694595

RESUMEN

To demonstrate the transplantation of drug-secreting cells with immunoprotection, a biodegradable delivery device combining two nanoporous capsules is developed using secretory alkaline phosphatase gene (SEAP) transfected mouse embryonic stem (mES) cells as a model system. The outer capsule is a poly (ethylene glycol) (PEG)-coated poly (epsilon-caprolactone) (PCL) chamber covered with a PEG grafted PCL nanoporous membrane made by phase inversion technique. SEAP gene transfected mES cells encapsulated in alginate-poly-L-lysine (AP) microcapsules are placed in the PCL capsule. Both nanoporous capsules showed good immunoprotection in the IgG solution. In microcapsules, mES cells could form a spheroid embryonic body (EB) and grow close to the microcapsule size. The secreted SEAP from encapsulated mES cells increased gradually to a maximum value before reaching a steady level, following the cell growth pattern in the microcapsule. Without microcapsules, mES cells only formed a monolayer in the large PCL capsule. The secreted SEAP release was very low. The integrated device showed a similar cell growth pattern to that in microcapsules alone, while the SEAP release rate could be regulated by the pore size of the large capsule. This integrated device can achieve multi-functionalities for cell-based therapy, i.e. a 3-D microenvironment provided by microcapsules for cell growth, superior immunoprotection and controllable release performance provided by the two nanoporous membranes, and good fibrosis prevention by PEG surface modification of the large capsule.


Asunto(s)
Materiales Biocompatibles/metabolismo , Trasplante de Células/métodos , Inmunoglobulina G/inmunología , Nanoestructuras , Fosfatasa Alcalina/metabolismo , Animales , Cápsulas , Forma de la Célula , Supervivencia Celular , Trasplante de Células/instrumentación , Difusión , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Membranas Artificiales , Ratones , Microscopía Electrónica de Rastreo , Poliésteres/metabolismo , Porosidad , Soluciones
20.
Hum Gene Ther ; 18(5): 474-81, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17518615

RESUMEN

Microencapsulation of recombinant cells is a novel alternative approach to tumor gene therapy. Therapeutic protein delivery can be sustained for systemic treatment of tumors because the recombinant cells are enclosed in microcapsules and the semipermeable membrane of the microcapsules protects the cells from host immune rejection and reduces the need for frequent injection. In this study, we describe a method to systemically inhibit tumor growth by in vivo culture of antiangiogenic endostatin-secreting Chinese hamster ovary (CHO) cells in microcapsules as small as 200 microm in diameter. Peritoneal administration of encapsulated endostatin-CHO cells inhibited melanoma growth to 66.4% and enhanced the survival of treated mice to 80% by 27 days posttreatment. Continuous systemic release of endostatin from microcapsules offers an effective therapeutic strategy to eradicate solid tumors.


Asunto(s)
Endostatinas/metabolismo , Melanoma Experimental/terapia , Animales , Células CHO , Bovinos , Proliferación Celular/efectos de los fármacos , Trasplante de Células , Embrión de Pollo , Cricetinae , Cricetulus , Composición de Medicamentos , Endostatinas/genética , Endostatinas/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Femenino , Terapia Genética/métodos , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...