Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Healthc Eng ; 2017: 5740975, 2017.
Article in English | MEDLINE | ID: mdl-29317995

ABSTRACT

Disruption of the blood-brain barrier (BBB) is the hallmark of many neurovascular disorders, making it a critically important focus for therapeutic options. However, testing the effects of either drugs or pathological agents is difficult due to the potentially damaging consequences of altering the normal brain microenvironment. Recently, in vitro coculture tissue models have been developed as an alternative to animal testing. Despite low cost, these platforms use synthetic scaffolds which prevent normal barrier architecture, cellular crosstalk, and tissue remodeling. We created a biodegradable electrospun gelatin mat "biopaper" (BP) as a scaffold material for an endothelial/astrocyte coculture model allowing cell-cell contact and crosstalk. To compare the BP and traditional models, we investigated the expression of 27 genes involved in BBB permeability, cellular function, and endothelial junctions at different time points. Gene expression levels demonstrated higher expression of transcripts involved in endothelial junction formation, including TJP2 and CDH5, in the BP model. The traditional model had higher expression of genes associated with extracellular matrix-associated proteins, including SPARC and COL4A1. Overall, the results demonstrate that the BP coculture model is more representative of a healthy BBB state, though both models have advantages that may be useful in disease modeling.


Subject(s)
Biocompatible Materials , Blood-Brain Barrier , Gene Expression , Models, Biological , Biological Transport , Coculture Techniques , Humans , Permeability
2.
Acta Biomater ; 35: 32-41, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26945632

ABSTRACT

Here, we describe an in vitro strategy to model vascular morphogenesis where human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) are encapsulated in peptide-functionalized poly(ethylene glycol) (PEG) hydrogels, either on standard well plates or within a passive pumping polydimethylsiloxane (PDMS) tri-channel microfluidic device. PEG hydrogels permissive towards cellular remodeling were fabricated using thiol-ene photopolymerization to incorporate matrix metalloproteinase (MMP)-degradable crosslinks and CRGDS cell adhesion peptide. Time lapse microscopy, immunofluorescence imaging, and RNA sequencing (RNA-Seq) demonstrated that iPSC-ECs formed vascular networks through mechanisms that were consistent with in vivo vasculogenesis and angiogenesis when cultured in PEG hydrogels. Migrating iPSC-ECs condensed into clusters, elongated into tubules, and formed polygonal networks through sprouting. Genes upregulated for iPSC-ECs cultured in PEG hydrogels relative to control cells on tissue culture polystyrene (TCP) surfaces included adhesion, matrix remodeling, and Notch signaling pathway genes relevant to in vivo vascular development. Vascular networks with lumens were stable for at least 14days when iPSC-ECs were encapsulated in PEG hydrogels that were polymerized within the central channel of the microfluidic device. Therefore, iPSC-ECs cultured in peptide-functionalized PEG hydrogels offer a defined platform for investigating vascular morphogenesis in vitro using both standard and microfluidic formats. STATEMENT OF SIGNIFICANCE: Human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) cultured in synthetic hydrogels self-assemble into capillary networks through mechanisms consistent with in vivo vascular morphogenesis.


Subject(s)
Blood Vessels/physiology , Endothelial Cells/cytology , Hydrogels/pharmacology , Induced Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Blood Vessels/drug effects , Capillaries/drug effects , Capillaries/physiology , Cell Adhesion/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism
3.
J Biomed Mater Res A ; 104(4): 901-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26650896

ABSTRACT

Gaining a greater understanding of the blood-brain barrier (BBB) is critical for improvement in drug delivery, understanding pathologies that compromise the BBB, and developing therapies to protect the BBB. In vitro human tissue models are valuable tools for studying these issues. The standard in vitro BBB models use commercially available cell culture inserts to generate bilayer co-cultures of astrocytes and endothelial cells (EC). Electrospinning can be used to produce customized cell culture substrates with optimized material composition and mechanical properties with advantages over off-the-shelf materials. Electrospun gelatin is an ideal cell culture substrate because it is a natural polymer that can aid cell attachment and be modified and degraded by cells. Here, we have developed a method to produce cell culture inserts with electrospun gelatin "biopaper" membranes. The electrospun fiber diameter and cross-linking method were optimized for the growth of primary human endothelial cell and primary human astrocyte bilayer co-cultures to model human BBB tissue. BBB co-cultures on biopaper were characterized via cell morphology, trans-endothelial electrical resistance (TEER), and permeability to FITC-labeled dextran and compared to BBB co-cultures on standard cell culture inserts. Over longer culture periods (up to 21 days), cultures on the optimized electrospun gelatin biopapers were found to have improved TEER, decreased permeability, and permitted a smaller separation between co-cultured cells when compared to standard PET inserts.


Subject(s)
Astrocytes/cytology , Biocompatible Materials/chemistry , Blood-Brain Barrier/cytology , Endothelial Cells/cytology , Gelatin/chemistry , Brain/cytology , Cell Line , Coculture Techniques/methods , Cross-Linking Reagents/chemistry , Electricity , Humans , Membranes, Artificial , Paper
4.
BMC Cancer ; 15: 12, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25605670

ABSTRACT

BACKGROUND: Ductal carcinoma in situ (DCIS) is a non-invasive form of breast cancer that is thought to be a precursor to most invasive and metastatic breast cancers. Understanding the mechanisms regulating the invasive transition of DCIS is critical in order to better understand how some types of DCIS become invasive. While significant insights have been gained using traditional in vivo and in vitro models, existing models do not adequately recapitulate key structure and functions of human DCIS well. In addition, existing models are time-consuming and costly, limiting their use in routine screens. Here, we present a microscale DCIS model that recapitulates key structures and functions of human DCIS, while enhancing the throughput capability of the system to simultaneously screen numerous molecules and drugs. METHODS: Our microscale DCIS model is prepared in two steps. First, viscous finger patterning is used to generate mammary epithelial cell-lined lumens through extracellular matrix hydrogels. Next, DCIS cells are added to fill the mammary ducts to create a DCIS-like structure. For coculture experiments, human mammary fibroblasts (HMF) are added to the two side channels connected to the center channel containing DCIS. To validate the invasive transition of the DCIS model, the invasion of cancer cells and the loss of cell-cell junctions are then examined. A student t-test is conducted for statistical analysis. RESULTS: We demonstrate that our DCIS model faithfully recapitulates key structures and functions of human mammary DCIS and can be employed to study the mechanisms involved in the invasive progression of DCIS. First, the formation of cell-cell junctions and cell polarity in the normal mammary duct, and the structure of the DCIS model are characterized. Second, coculture with HMF is shown to induce the invasion of DCIS. Third, multiple endpoint analyses are demonstrated to validate the invasion. CONCLUSIONS: We have developed and characterized a novel in vitro model of normal and DCIS-inflicted mammary ducts with 3D lumen structures. These models will enable researchers to investigate the role of microenvironmental factors on the invasion of DCIS in more in vivo-like conditions.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , In Vitro Techniques/methods , Models, Biological , Cell Line, Tumor , Coculture Techniques , Female , Humans , In Vitro Techniques/instrumentation , Microfluidics , Neoplasm Invasiveness
5.
FASEB J ; 28(11): 4583-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25077562

ABSTRACT

Advances in tissue engineering and microtechnology have enabled researchers to more easily generate in vitro tissue models that mimic the tissue geometry and spatial organization found in vivo (e.g., vessel or mammary duct models with tubular structures). However, the widespread adoption of these models for biological studies has been slow, in part due to the lack of direct comparisons between existing 2-dimensional and 3-dimensional cell culture models and new organotypic models that better replicate tissue structure. Using previously developed vessel and mammary duct models with 3-dimensional lumen structures, we have begun to explore this question. In a direct comparison between these next generation organotypic models and more traditional methods, we observed differences in the levels of several secreted growth factors and cytokines. In addition, endothelial vessel geometry profoundly affects the phenotypic behavior of carcinoma cells, suggesting that more traditional in vitro assays may not capture in vivo events. Here, we seek to review and add to the increasing evidence supporting the hypothesis that using cell culture models with more relevant tissue structure influences cell fate and behavior, potentially increasing the relevance of biological findings.


Subject(s)
Blood Vessels/cytology , Cell Differentiation/physiology , Cell Culture Techniques/methods , Cytokines/biosynthesis , Endothelial Cells/cytology , Humans , Models, Biological , Tissue Engineering/methods
6.
Integr Biol (Camb) ; 6(6): 627-635, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24791272

ABSTRACT

In prostate cancer, bone is a frequent site of metastasis; however, the molecular mechanisms of this tumor tropism remain unclear. Here, we integrate a microfluidic coculture platform with multi-photon imaging based techniques to assess both phenotypic cell behavior and FAD fluorescence intensity and fluorescence lifetime in the same cell. This platform combines two independent assays normally performed with two different cell populations into a single device, allowing us to simultaneously assess both phenotypic cell behavior and enzyme activity. We observed that the osteotropic prostate cancer cell line (C4-2B), when in a coculture with bone marrow stromal cells (MC3T3-E1), has increased protrusive phenotype and increased total and protein-bound FAD compared to its parent cell line (LNCaP). We hypothesized that an increase in ROS-generating APAO activity may be responsible for these effects, and found that the effects were decreased in the presence of the antioxidant N-Acetyl Cysteine (NAC). This suggests that an ROS-related signaling mechanism at the bone metastatic site may be correlated with and play a role in increased invasion of metastasizing prostate cancer cells. The studies performed using this combined platform will lead to new insights into the mechanisms that drive prostate cancer metastasis.


Subject(s)
Flavin-Adenine Dinucleotide/metabolism , Mesenchymal Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Cell Line, Tumor , Coculture Techniques , Humans , Male , Mesenchymal Stem Cells/cytology , Microfluidics , Microscopy, Fluorescence, Multiphoton , Reactive Oxygen Species/antagonists & inhibitors
7.
Proc Natl Acad Sci U S A ; 110(25): 10111-6, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23729815

ABSTRACT

Although the field of microfluidics has made significant progress in bringing new tools to address biological questions, the accessibility and adoption of microfluidics within the life sciences are still limited. Open microfluidic systems have the potential to lower the barriers to adoption, but the absence of robust design rules has hindered their use. Here, we present an open microfluidic platform, suspended microfluidics, that uses surface tension to fill and maintain a fluid in microscale structures devoid of a ceiling and floor. We developed a simple and ubiquitous model predicting fluid flow in suspended microfluidic systems and show that it encompasses many known capillary phenomena. Suspended microfluidics was used to create arrays of collagen membranes, mico Dots (µDots), in a horizontal plane separating two fluidic chambers, demonstrating a transwell platform able to discern collective or individual cellular invasion. Further, we demonstrated that µDots can also be used as a simple multiplexed 3D cellular growth platform. Using the µDot array, we probed the combined effects of soluble factors and matrix components, finding that laminin mitigates the growth suppression properties of the matrix metalloproteinase inhibitor GM6001. Based on the same fluidic principles, we created a suspended microfluidic metabolite extraction platform using a multilayer biphasic system that leverages the accessibility of open microchannels to retrieve steroids and other metabolites readily from cell culture. Suspended microfluidics brings the high degree of fluidic control and unique functionality of closed microfluidics into the highly accessible and robust platform of open microfluidics.


Subject(s)
Microfluidic Analytical Techniques , Microfluidics/instrumentation , Microfluidics/methods , Models, Biological , Adrenal Cortex/cytology , Breast Neoplasms/pathology , Capillaries/metabolism , Cell Biology/instrumentation , Cell Line, Tumor , Cell Membrane/physiology , Cell Movement , Collagen Type I/metabolism , Computer Simulation , Female , Humans , Hydrocortisone/analysis , Hydrocortisone/metabolism , Male , Matrix Metalloproteinase Inhibitors/pharmacology , Metabolomics/instrumentation , Metabolomics/methods , Prostatic Neoplasms/pathology , Steroids/analysis , Steroids/metabolism , Toxicology/instrumentation , Toxicology/methods
8.
Lab Chip ; 13(9): 1732-6, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23503983

ABSTRACT

The zebrafish has emerged as a useful model system for a variety of studies, including the investigation of inflammation and immunity. However, current zebrafish imaging techniques, such as agraose mounting, can be time-consuming and detrimental for long-term imaging. Alternatively, automated sorting and imaging systems can be costly and/or complicated to assemble. Here we describe the Zebrafish Entrapment by Restriction Array (ZEBRA) device, a microfluidic device that can be used to quickly and repeatably position zebrafish embryos in a predictable array using only a pipette. This technique is well suited for use with automated microscope stages leading to decreased imaging time and increased throughput compared to traditional methods. The addition of access ports above the embryo can be used to administer treatments, and potentially wounding or injections. We demonstrate the effectiveness of this device for a neutrophil migration screening application using larvae 3 days post fertilization (dpf) Tg(mpx:dendra2). Larvae were loaded into ZEBRA devices and treated with a neutrophil attractant (LTB4) or LTB4 with and without a PI3K inhibitor, LY294002. Treatment with LY294002 impaired neutrophil motility into the fin induced by LTB4 treatment. The findings report the development of ZEBRA a device that can be used to screen for small molecules that affect leukocyte motility and inflammation using live zebrafish.


Subject(s)
Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Zebrafish/embryology , Animals , Cell Movement/drug effects , Cell Movement/physiology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Leukotriene B4/pharmacology , Morpholines/pharmacology , Neutrophils/cytology , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
9.
Biomaterials ; 34(5): 1471-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23191982

ABSTRACT

The study of angiogenesis is important to understanding a variety of human pathologies including cancer, cardiovascular and inflammatory diseases. In vivo angiogenesis assays can be costly and time-consuming, limiting their application in high-throughput studies. While traditional in vitro assays may overcome these limitations, they lack the ability to accurately recapitulate the main elements of the tissue microenvironment found in vivo, thereby limiting our ability to draw physiologically relevant biological conclusions. To bridge the gap between in vivo and in vitro angiogenesis assays, several microfluidic methods have been developed to generate in vitro assays that incorporate blood vessel models with physiologically relevant three-dimensional (3D) lumen structures. However, these models have not seen widespread adoption, which can be partially attributed to the difficulty in fabricating these structures. Here, we present a simple, accessible method that takes advantage of basic fluidic principles to create 3D lumens with circular cross-sectional geometries through ECM hydrogels that are lined with endothelial monolayers to mimic the structure of blood vessels in vitro. This technique can be used to pattern endothelial cell-lined lumens in different microchannel geometries, enabling increased flexibility for a variety of studies. We demonstrate the implementation and application of this technique to the study of angiogenesis in a physiologically relevant in vitro setting.


Subject(s)
Biological Assay/instrumentation , Biomimetic Materials/chemical synthesis , Endothelial Cells/physiology , Microfluidics/instrumentation , Microvessels/physiology , Neovascularization, Physiologic/physiology , Batch Cell Culture Techniques/instrumentation , Cells, Cultured , Endothelial Cells/cytology , Equipment Design , Equipment Failure Analysis , Humans , Materials Testing , Microvessels/cytology , Tissue Engineering/instrumentation
10.
J Lab Autom ; 17(2): 96-103, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22357560

ABSTRACT

Extracellular matrix (ECM) hydrogels with patterned lumens have been used as a framework to generate more physiologically relevant models of tissues, such as vessels and mammary ducts, for biological investigations. However, these models have not found widespread use in research labs or in high-throughput screening applications in large part because the basic methods for generating the lumen structures are generally cumbersome and slow. Here we present viscous finger patterning, a technique to generate lumens through ECM hydrogels in microchannels that can be accomplished using manual or automated pipetting. Passive pumping is used to flow culture media through an unpolymerized hydrogel, creating a lumen through the hydrogel that is subsequently polymerized. Viscous finger patterning takes advantage of viscous fingering, the fluid dynamics phenomenon where a less viscous fluid will flow through and displace a more viscous fluid. We have characterized the technique and used it to create a variety of channel geometries and ECM hydrogel compositions, as well as for the generation of lumens surrounded by multiple hydrogel layers. Because viscous finger patterning can be performed with automated liquid handling systems, high-throughput generation of ECM hydrogels with patterned lumen is enabled. The ability to rapidly and cost-effectively create large numbers of lumens in natural polymers overcomes a critical barrier to the use of more physiologically relevant tissue models in a variety of biological studies and drug screening applications.


Subject(s)
Extracellular Matrix , Hydrogels , Tissue Scaffolds , Animals , Culture Media/chemistry , Humans , Organ Culture Techniques/methods
SELECTION OF CITATIONS
SEARCH DETAIL