Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2081: 191-201, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31721126

RESUMO

In vivo small animal bioluminescent imaging has become an indispensable technique for interrogating the localization, health, and functionality of implanted cells within the complex environment of a living organism. However, this task can be daunting for even the most experienced researchers because it requires multiple animal handling steps and produces differential output signal characteristics in response to a number of experimental design variables. The recent emergence of autobioluminescent cells, which autonomously and continuously produce bioluminescent output signals without external stimulation, has the potential to simplify this process, reduce variability by removing human-induced error, and improve animal welfare by reducing the number of required needlesticks per procedure. This protocol details the implantation and imaging of autobioluminescent cells within a mouse model to demonstrate how cells implanted from a single injection can be imaged repeatedly across any post-implantation timescale without the need for further human-animal interaction or signal activation steps. This approach provides a facile means to continuously monitor implanted cellular output signals in real-time for extended time periods.


Assuntos
Medições Luminescentes/métodos , Imagem Molecular , Imagem com Lapso de Tempo , Animais , Linhagem Celular , Análise de Dados , Descoberta de Drogas , Feminino , Expressão Gênica , Genes Reporter , Humanos , Camundongos , Imagem Molecular/métodos , Software
2.
J Biomed Mater Res A ; 105(8): 2119-2128, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28371246

RESUMO

In this study, a new 3D liver model was developed using biomimetic nanofiber scaffolds and co-culture system consisting of hepatocytes and fibroblasts for the maintenance of long-term liver functions. The chitosan nanofiber scaffolds were fabricated by the electrospinning technique. To enhance cellular adhesion and spreading, the surfaces of the chitosan scaffolds were coated with fibronectin (FN) by adsorption and evaluated for various cell types. Cellular phenotype, protein expression, and liver-specific functions were extensively characterized by immunofluorescent and histochemical stainings, albumin enzyme-linked immunosorbent assay and Cytochrome p450 detoxification assays, and scanning electron microscopy. The electrospun chitosan scaffolds exhibited a highly porous and randomly oriented nanofibrous structure. The FN coating on the surface of the chitosan nanofibers significantly enhanced cell attachment and spreading, as expected, as surface modification with this cell adhesion molecule on the chitosan surface is important for focal adhesion formation and integrin binding. Comparison of hepatocyte mono-cultures and co-cultures in 3D culture systems indicated that the hepatocytes in co-cultures formed colonies and maintained their morphologies and functions for prolonged periods of time. The 3D liver tissue model developed in this study will provide useful tools toward the development of engineered liver tissues for drug screening and tissue engineering applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2119-2128, 2017.


Assuntos
Quitosana/química , Materiais Revestidos Biocompatíveis/química , Fibronectinas/química , Hepatócitos/citologia , Nanofibras/química , Alicerces Teciduais/química , Adesão Celular , Técnicas de Cocultura , Sistema Enzimático do Citocromo P-450/metabolismo , Células Hep G2 , Hepatócitos/metabolismo , Humanos
3.
J Vis Exp ; (120)2017 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-28287599

RESUMO

Substrate and cell patterning techniques are widely used in cell biology to study cell-to-cell and cell-to-substrate interactions. Conventional patterning techniques work well only with simple shapes, small areas and selected bio-materials. This article describes a method to distribute cell suspensions as well as substrate solutions into complex, long, closed (dead-end) polydimethylsiloxane (PDMS) microchannels using negative pressure. This method enables researchers to pattern multiple substrates including fibronectin, collagen, antibodies (Sal-1), poly-D-lysine (PDL), and laminin. Patterning of substrates allows one to indirectly pattern a variety of cells. We have tested C2C12 myoblasts, the PC12 neuronal cell line, embryonic rat cortical neurons, and amphibian retinal neurons. In addition, we demonstrate that this technique can directly pattern fibroblasts in microfluidic channels via brief application of a low vacuum on cell suspensions. The low vacuum does not significantly decrease cell viability as shown by cell viability assays. Modifications are discussed for application of the method to different cell and substrate types. This technique allows researchers to pattern cells and proteins in specific patterns without the need for exotic materials or equipment and can be done in any laboratory with a vacuum.


Assuntos
Células PC12/citologia , Células PC12/metabolismo , Proteínas/metabolismo , Animais , Adesão Celular , Contagem de Células , Células Cultivadas , Fibroblastos/citologia , Modelos Animais , Ratos
4.
Biofabrication ; 6(3): 035016, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24989641

RESUMO

Substrate and cell patterning are widely used techniques in cell biology to study cell-to-cell and cell-substrate interactions. Conventional patterning techniques work well only with simple shapes, small areas and selected bio-materials. This paper describes a method to distribute cell suspensions as well as substrate solutions into complex, long, closed (dead-end) polydimethylsiloxane (PDMS) microchannels using negative pressure. Our method builds upon a previous vacuum-assisted method used for micromolding (Jeon et al 1999 Adv. Mater 11 946) and successfully patterned collagen-I, fibronectin and Sal-1 substrates on glass and polystyrene surfaces, filling microchannels with lengths up to 120 mm and covering areas up to 13 × 10 mm(2). Vacuum-patterned substrates were subsequently used to culture mammalian PC12 and fibroblast cells and amphibian neurons. Cells were also patterned directly by injecting cell suspensions into microchannels using vacuum. Fibroblast and neuronal cells patterned using vacuum showed normal growth and minimal cell death indicating no adverse effects of vacuum on cells. Our method fills reversibly sealed PDMS microchannels. This enables the user to remove the PDMS microchannel cast and access the patterned biomaterial or cells for further experimental purposes. Overall, this is a straightforward technique that has broad applicability for cell biology.


Assuntos
Fibroblastos/citologia , Neurônios/citologia , Engenharia Tecidual/métodos , Animais , Adesão Celular , Humanos , Células PC12 , Ratos , Engenharia Tecidual/instrumentação
5.
Biochem Biophys Res Commun ; 433(3): 327-32, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23501105

RESUMO

Two-dimensional (2D) monolayer cultures are the standard in vitro model for cancer research. However, they fail to recapitulate the three-dimensional (3D) environment and quickly lose their function. In this study, we developed a new 3D multicellular heterospheroid tumor model in a collagen hydrogel culture system that more closely mimics the in vivo tumor microenvironment for anti-cancer drug testing. Three aspects of cancer were chosen to be modeled based on their ability to resist anti-cancer drugs: 3D, multicellularity, and extracellular matrix (ECM) barrier. The hanging drop method and co-culture of liver carcinoma with stromal fibroblasts were used to form controlled and uniform heterospheroids. These heterospheroids were then encapsulated in collagen gel in order to create a 3D model of liver cancer that would act more similarly to in vivo ECM conditions. The 3D heterospheroid tumor model was tested with an anti-cancer drug to determine how each of the above aspects affects drug resistance. The results demonstrate that the 3D heterospheroid model is more resistant to drug over 2D monolayer and homospheroid cultures, indicating stromal fibroblasts and collagen hydrogel culture system provides more resistance to anti-cancer drug. This study will provide useful information toward the development of improved biomimetic tumor models in vitro for cancer research in pre-clinical drug development.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Colágeno/química , Doxorrubicina/farmacologia , Fibroblastos/efeitos dos fármacos , Células Hep G2/efeitos dos fármacos , Hidrogéis/química , Esferoides Celulares/efeitos dos fármacos , Animais , Carcinoma/tratamento farmacológico , Carcinoma/patologia , Linhagem Celular , Técnicas de Cocultura , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/ultraestrutura , Fibroblastos/citologia , Células Hep G2/patologia , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Camundongos , Microscopia de Fluorescência , Mitomicina/farmacologia , Conformação Molecular , Esferoides Celulares/ultraestrutura , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Alicerces Teciduais
6.
Biotechnol Bioeng ; 110(2): 637-47, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22991229

RESUMO

The in vitro generation of a three-dimensional (3-D) myocardial tissue-like construct employing cells, biomaterials, and biomolecules is a promising strategy in cardiac tissue regeneration, drug testing, and tissue engineering applications. Despite significant progress in this field, current cardiac tissue models are not yet able to stably maintain functional characteristics of cardiomyocytes for long-term culture and therapeutic purposes. The objective of this study was to fabricate bioactive 3-D chitosan nanofiber scaffolds using an electrospinning technique and exploring its potential for long-term cardiac function in the 3-D co-culture model. Chitosan is a natural polysaccharide biomaterial that is biocompatible, biodegradable, non-toxic, and cost effective. Electrospun chitosan was utilized to provide structural scaffolding characterized by scale and architectural resemblance to the extracellular matrix (ECM) in vivo. The chitosan fibers were coated with fibronectin via adsorption in order to enhance cellular adhesion to the fibers and migration into the interfibrous milieu. Ventricular cardiomyocytes were harvested from neonatal rats and studied in various culture conditions (i.e., mono- and co-cultures) for their viability and function. Cellular morphology and functionality were examined using immunofluorescent staining for alpha-sarcomeric actin (SM-actin) and gap junction protein, Connexin-43 (Cx43). Scanning electron microscopy (SEM) and light microscopy were used to investigate cellular morphology, spatial organization, and contractions. Calcium indicator was used to monitor calcium ion flux of beating cardiomyocytes. The results demonstrate that the chitosan nanofibers retained their cylindrical morphology in long-term cell cultures and exhibited good cellular attachment and spreading in the presence of adhesion molecule, fibronectin. Cardiomyocyte mono-cultures resulted in loss of cardiomyocyte polarity and islands of non-coherent contractions. However, the cardiomyocyte-fibroblast co-cultures resulted in polarized cardiomyocyte morphology and retained their morphology and function for long-term culture. The Cx43 expression in the fibroblast co-culture was higher than the cardiomyocytes mono-culture and endothelial cells co-culture. In addition, fibroblast co-cultures demonstrated synchronized contractions involving large tissue-like cellular networks. To our knowledge, this is the first attempt to test chitosan nanofiber scaffolds as a 3-D cardiac co-culture model. Our results demonstrate that chitosan nanofibers can serve as a potential scaffold that can retain cardiac structure and function. These studies will provide useful information to develop a strategy that allows us to generate engineered 3-D cardiac tissue constructs using biocompatible and biodegradable chitosan nanofiber scaffolds for many tissue engineering applications.


Assuntos
Quitosana/química , Técnicas de Cocultura/métodos , Miócitos Cardíacos/citologia , Nanofibras/química , Alicerces Teciduais/química , Actinas/metabolismo , Adsorção , Animais , Cálcio/metabolismo , Células Cultivadas , Conexina 43/metabolismo , Citoesqueleto/ultraestrutura , Técnicas Eletroquímicas , Células Endoteliais/citologia , Fibroblastos/citologia , Fibronectinas/metabolismo , Adesões Focais/metabolismo , Proteínas Imobilizadas/metabolismo , Miócitos Cardíacos/metabolismo , Ratos , Ratos Wistar , Engenharia Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...