Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Small ; 17(15): e2004889, 2021 04.
Article in English | MEDLINE | ID: mdl-33150735

ABSTRACT

Hemostasis is an innate protective mechanism that plays a central role in maintaining the homeostasis of the vascular system during vascular injury. Studying this essential physiological process is often challenged by the difficulty of modeling and probing the complex dynamics of hemostatic responses in the native context of human blood vessels. To address this major challenge, this paper describes a microengineering approach for in vitro modeling of hemostasis. This microphysiological model replicates the living endothelium, multilayered microarchitecture, and procoagulant activity of human blood vessels, and is also equipped with a microneedle that is actuated with spatial precision to simulate penetrating vascular injuries. The system recapitulates key features of the hemostatic response to acute vascular injury as observed in vivo, including i) thrombin-driven accumulation of platelets and fibrin, ii) formation of a platelet- and fibrin-rich hemostatic plug that halts blood loss, and iii) matrix deformation driven by platelet contraction for wound closure. Moreover, the potential use of this model for drug testing applications is demonstrated by evaluating the effects of anticoagulants and antiplatelet agents that are in current clinical use. The vascular injury-on-a-chip may serve as an enabling platform for preclinical investigation of hematological disorders and emerging therapeutic approaches against them.


Subject(s)
Thrombosis , Vascular System Injuries , Fibrin , Hemostasis , Humans , Lab-On-A-Chip Devices
2.
J Immunol ; 189(5): 2266-73, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844118

ABSTRACT

T cells navigate a wide variety of tissues and organs for immune surveillance and effector functions. Although nanoscale topographical structures of extracellular matrices and stromal/endothelial cell surfaces in local tissues may guide the migration of T cells, there has been little opportunity to study how nanoscale topographical features affect T cell migration. In this study, we systematically investigated mechanisms of nanotopography-guided migration of T cells using nanoscale ridge/groove surfaces. The velocity and directionality of T cells on these nanostructured surfaces were quantitatively assessed with and without confinement, which is a key property of three-dimensional interstitial tissue spaces for leukocyte motility. Depending on the confinement, T cells exhibited different mechanisms for nanotopography-guided migration. Without confinement, actin polymerization-driven leading edge protrusion was guided toward the direction of nanogrooves via integrin-mediated adhesion. In contrast, T cells under confinement appeared to migrate along the direction of nanogrooves purely by mechanical effects, and integrin-mediated adhesion was dispensable. Therefore, surface nanotopography may play a prominent role in generating migratory patterns for T cells. Because the majority of cells in periphery migrate along the topography of extracellular matrices with much lower motility than T cells, nanotopography-guided migration of T cells would be an important strategy to efficiently perform cell-mediated immune responses by increasing chances of encountering other cells within a given amount of time.


Subject(s)
Cell Movement/immunology , Nanotechnology/instrumentation , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Amino Acid Sequence , Animals , Cell Communication/immunology , Cell Membrane/immunology , Cell Membrane/ultrastructure , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microscopy, Interference , Molecular Sequence Data , Nanotechnology/methods , Surface Properties , T-Lymphocyte Subsets/ultrastructure
3.
Langmuir ; 27(7): 3238-43, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21348500

ABSTRACT

We present new methods that enable the fabrication of multiscale, multicomponent protein-patterned surfaces and multiscale topologically structured surfaces by exploiting the merits of two well-established techniques: capillary force lithography (CFL) and microscope projection photolithography (MPP) based on a protein-friendly photoresist. We further demonstrate that, when hierarchically organized micro- and nanostructures were used as a cell culture platform, human colon cancer cells (cell line SW480) preferentially adhere and migrate onto the area with nanoscale topography over the one with microscale topography. These methods will provide many exciting opportunities for the study of cellular responses to multiscale physicochemical cues.


Subject(s)
Nanostructures/chemistry , Nanotechnology/methods , Proteins/chemistry , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Humans , Microscopy, Electron, Scanning , Nanostructures/ultrastructure
4.
Anal Chem ; 82(7): 3016-22, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20218573

ABSTRACT

We present a simple analytical method to measure adhesion of human umbilical vein endothelial cells (HUVECs) and calf pulmonary artery endothelial cells (CPAEs) using nanopatterned, biodegradable poly(lactic-co-glycolic acid) (PLGA) surfaces for potential applications to artificial tissue-engineered blood vessel. Various nanostructured PLGA surfaces (350 nm wide ridges/350 nm grooves, 350 nm ridges/700 nm grooves, 350 nm ridges/1750 nm grooves, 700 nm ridges/350 nm grooves, 1050 nm ridges/350 nm grooves, 1750 nm ridges/350 nm grooves) and flat (unpatterned) surfaces were fabricated on the bottom of polydimethylsiloxane (PDMS) microfluidic channel of 2 mm width and 60 microm height by using thermal imprinting and irreversible channel bonding. To measure adhesion strength of HUVECs and CPAEs, the cells were exposed to a range of shear stress (12, 40, and 80 dyn/cm(2)) within the channels for 20 min after a preculture for 3 days and the remaining cells were counted under each condition. The highest adhesion strength was found on the surface of 700 nm wide ridges, 350 nm wide grooves for both cell types. The enhanced adhesion on nanopatterned surfaces can be attributed to two aspects: (i) contact guidance along the line direction and (ii) clustered focal adhesions. In particular, the contact guidance induced cell alignment along the line directions, which in turn lowers wall shear stress applied to the cell surface, as supported by a simple hydrodynamic model based on cell morphology.


Subject(s)
Cell Adhesion , Endothelial Cells/cytology , Microfluidic Analytical Techniques/methods , Nanostructures/chemistry , Cells, Cultured , Dimethylpolysiloxanes/chemistry , Fluorescent Dyes/chemistry , Humans , Lactic Acid/chemistry , Microscopy, Electron, Scanning , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Surface Properties
5.
Adv Funct Mater ; 19(10): 1579-1586, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-20046799

ABSTRACT

This work reports the design of and experimentation with a topographically patterned cell culture substrate of variable local density and anisotropy as a facile and efficient platform to guide the organization and migration of cells in spatially desirable patterns. Using UV-assisted capillary force lithography, an optically transparent microstructured layer of a UV curable poly(urethane acrylate) resin is fabricated and employed as a cell-culture substrate after coating with fibronectin. With variable local pattern density and anisotropy present in a single cell-culture substrate, the differential polarization of cell morphology and movement in a single experiment is quantitatively characterized. It is found that cell shape and velocity are exquisitely sensitive to variation in the local anisotropy of the two-dimensional rectangular lattice arrays, with cell elongation and speed decreasing on symmetric lattice patterns. It is also found that cells could integrate orthogonal spatial cues when determining the direction of cell orientation and movement. Furthermore, cells preferentially migrate toward the topographically denser areas from sparser ones. Consistent with these results, it is demonstrated that systematic variation of local densities of rectangular lattice arrays enable a planar assembly of cells into a specified location. It is envisioned that lithographically defined substrates of variable local density and anisotropy not only provide a new route to tailoring the cell-material interface but could serve as a template for advanced tissue engineering.

6.
ACS Appl Mater Interfaces ; 11(27): 23919-23925, 2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31199616

ABSTRACT

Diverse biological processes in the body rely on the ability of cells to exert contractile forces on their extracellular matrix (ECM). In three-dimensional (3D) cell culture, however, this intrinsic cellular property can cause unregulated contraction of ECM hydrogel scaffolds, leading to a loss of surface anchorage and the resultant structural failure of in vitro tissue constructs. Despite advances in the 3D culture technology, this issue remains a significant challenge in the development and long-term maintenance of physiological 3D in vitro models. Here, we present a simple yet highly effective and accessible solution to this problem. We leveraged a single-step surface functionalization technique based on polydopamine to drastically increase the strength of adhesion between hydrogel scaffolds and cell culture substrates. Our method is compatible with different types of ECM and polymeric surfaces and also permits prolonged shelf storage of functionalized culture substrates. The proof-of-principle of this technique was demonstrated by the stable long-term (1 month) 3D culture of human lung fibroblasts. Furthermore, we showed the robustness and advanced application of the method by constructing a dynamic cell stretching system and performing over 100 000 cycles of mechanical loading on 3D multicellular constructs for visualization and quantitative analysis of stretch-induced tissue alignment. Finally, we demonstrated the potential of our technique for the development of microphysiological in vitro models by establishing microfluidic 3D co-culture of vascular endothelial cells and fibroblasts to engineer self-assembled, perfusable 3D microvascular beds.


Subject(s)
Cell Culture Techniques , Endothelial Cells/metabolism , Extracellular Matrix/chemistry , Fibroblasts/metabolism , Hydrogels/chemistry , Indoles/chemistry , Polymers/chemistry , Tissue Scaffolds/chemistry , Endothelial Cells/cytology , Fibroblasts/cytology , Humans , Printing, Three-Dimensional , Time Factors
7.
ACS Nano ; 13(7): 7627-7643, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31194909

ABSTRACT

The vasculature is an essential component of the circulatory system that plays a vital role in the development, homeostasis, and disease of various organs in the human body. The ability to emulate the architecture and transport function of blood vessels in the integrated context of their associated organs represents an important requirement for studying a wide range of physiological processes. Traditional in vitro models of the vasculature, however, largely fail to offer such capabilities. Here we combine microfluidic three-dimensional (3D) cell culture with the principle of vasculogenic self-assembly to engineer perfusable 3D microvascular beds in vitro. Our system is created in a micropatterned hydrogel construct housed in an elastomeric microdevice that enables coculture of primary human vascular endothelial cells and fibroblasts to achieve de novo formation, anastomosis, and controlled perfusion of 3D vascular networks. An open-top chamber design adopted in this hybrid platform also makes it possible to integrate the microengineered 3D vasculature with other cell types to recapitulate organ-specific cellular heterogeneity and structural organization of vascularized human tissues. Using these capabilities, we developed stem cell-derived microphysiological models of vascularized human adipose tissue and the blood-retinal barrier. Our approach was also leveraged to construct a 3D organotypic model of vascularized human lung adenocarcinoma as a high-content drug screening platform to simulate intravascular delivery, tumor-killing effects, and vascular toxicity of a clinical chemotherapeutic agent. Furthermore, we demonstrated the potential of our platform for applications in nanomedicine by creating microengineered models of vascular inflammation to evaluate a nanoengineered drug delivery system based on active targeting liposomal nanocarriers. These results represent a significant improvement in our ability to model the complexity of native human tissues and may provide a basis for developing predictive preclinical models for biopharmaceutical applications.


Subject(s)
Adenocarcinoma of Lung/pathology , Cell Culture Techniques , Cell Engineering , Endothelial Cells/cytology , Fibroblasts/cytology , Microfluidic Analytical Techniques , Adenocarcinoma of Lung/blood supply , Humans , Hydrogels/chemistry , Microcirculation
8.
Lab Chip ; 7(11): 1461-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17960272

ABSTRACT

A label-free microfluidic method for separation and enrichment of human breast cancer cells is presented using cell adhesion as a physical marker. To maximize the adhesion difference between normal epithelial and cancer cells, flat or nanostructured polymer surfaces (400 nm pillars, 400 nm perpendicular, or 400 nm parallel lines) were constructed on the bottom of polydimethylsiloxane (PDMS) microfluidic channels in a parallel fashion using a UV-assisted capillary moulding technique. The adhesion of human breast epithelial cells (MCF10A) and cancer cells (MCF7) on each channel was independently measured based on detachment assays where the adherent cells were counted with increasing flow rate after a pre-culture for a period of time (e.g., one, two, and four hours). It was found that MCF10A cells showed higher adhesion than MCF7 cells regardless of culture time and surface nanotopography at all flow rates, resulting in label-free separation and enrichment of cancer cells. For the cell types used in our study, an optimum separation was found for 2 hours pre-culture on the 400 nm perpendicular line pattern followed by flow-induced detachment at a flow rate of 200 microl min(-1). The fraction of MCF7 cells was increased from 0.36 +/- 0.04 to 0.83 +/- 0.04 under these optimized conditions.


Subject(s)
Breast Neoplasms/pathology , Cell Adhesion , Microfluidics/methods , Cell Line, Tumor , Humans , Nanostructures
9.
Lab Chip ; 6(8): 988-94, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16874367

ABSTRACT

We present a simple cell docking method induced by receding meniscus to capture non-adherent yeast cells onto microwells inside a microfluidic channel. Microwells were fabricated either by capillary moulding of UV curable polyurethane acrylate (PUA) onto glass substrate or direct replica moulding of poly(dimethyl siloxane) (PDMS). A cell suspension of the budding yeast, Saccharomyces cerevisiae, was introduced into the microfluidic channel by surface tension driven capillary flow and a receding meniscus was subsequently generated by evaporation. As the meniscus progressed, one to multiple yeast cells were spontaneously captured onto microwells by lateral capillary force created at the bottom of the meniscus. Using this cell-based platform, we observed the response of yeast cells upon stimulation by a mating pheromone (alpha-factor) by monitoring the expression of green fluorescent protein (GFP) with time. It was observed that alpha-factor triggered the expression of GFP at 60 min after stimulation and the fluorescence intensity was sustained for an additional 60 min without changes.


Subject(s)
Microfluidic Analytical Techniques , Microfluidics , Saccharomyces cerevisiae/cytology , Cells, Immobilized , Dimethylpolysiloxanes , Gene Expression Regulation, Fungal/drug effects , Green Fluorescent Proteins/biosynthesis , Mating Factor , Peptides/chemistry , Peptides/pharmacology , Saccharomyces cerevisiae/metabolism , Time Factors
10.
Sci Rep ; 6: 23412, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26996137

ABSTRACT

Stiff nuclei in cell-dense microenvironments may serve as distinct biomechanical cues for cell migration, but such a possibility has not been tested experimentally. As a first step addressing this question, we altered nuclear stiffness of endothelial cells (ECs) by reducing the expression of A-type lamins using siRNA, and investigated the migration of T cells on and under EC layers. While most T cells crawling on control EC layers avoided crossing over EC nuclei, a significantly higher fraction of T cells on EC layers with reduced expression of A-type lamins crossed over EC nuclei. This result suggests that stiff EC nuclei underlying T cells may serve as "duro-repulsive" cues to direct T cell migration toward less stiff EC cytoplasm. During subendothelial migration under EC layers with reduced expression of A-type lamins, T cells made prolonged contact and substantially deformed EC nuclei, resulting in reduced speed and directional persistence. This result suggests that EC nuclear stiffness promotes fast and directionally persistent subendothelial migration of T cells by allowing minimum interaction between T cells and EC nuclei.


Subject(s)
Cell Movement , Endothelial Cells/physiology , Lamin Type A/physiology , T-Lymphocytes/physiology , Animals , Cell Adhesion , Cell Nucleus/metabolism , Cells, Cultured , Endothelial Cells/ultrastructure , Lamin Type A/metabolism , Membrane Glycoproteins , Mice , T-Lymphocytes/cytology
11.
JCI Insight ; 1(19): e90064, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27882353

ABSTRACT

Clinical monitoring of adoptive T cell transfer (ACT) utilizes serial blood analyses to discern T cell activity. While useful, these data are 1-dimensional and lack spatiotemporal information related to treatment efficacy or toxicity. We utilized a human genetic reporter, somatostatin receptor 2 (SSTR2), and PET, to quantitatively and longitudinally visualize whole-body T cell distribution and antitumor dynamics using a clinically approved radiotracer. Initial evaluations determined that SSTR2-expressing T cells were detectable at low densities with high sensitivity and specificity. SSTR2-based PET was applied to ACT of chimeric antigen receptor (CAR) T cells targeting intercellular adhesion molecule-1, which is overexpressed in anaplastic thyroid tumors. Timely CAR T cell infusions resulted in survival of tumor-bearing mice, while later infusions led to uniform death. Real-time PET imaging revealed biphasic T cell expansion and contraction at tumor sites among survivors, with peak tumor burden preceding peak T cell burden by several days. In contrast, nonsurvivors displayed unrelenting increases in tumor and T cell burden, indicating that tumor growth was outpacing T cell killing. Thus, longitudinal PET imaging of SSTR2-positive ACT dynamics enables prognostic, spatiotemporal monitoring with unprecedented clarity and detail to facilitate comprehensive therapy evaluation with potential for clinical translation.


Subject(s)
Immunotherapy, Adoptive , Neoplasms, Experimental/therapy , Positron-Emission Tomography , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , Animals , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic , Genes, Reporter , Humans , Jurkat Cells , Mice , Transduction, Genetic , Xenograft Model Antitumor Assays
12.
PLoS One ; 10(4): e0124440, 2015.
Article in English | MEDLINE | ID: mdl-25901755

ABSTRACT

The ability to use a systemically injected agent to image tumor is influenced by tumor characteristics such as permeability and vascularity, and the size, shape, and affinity of the imaging agent. In this study, six different imaging biomolecules, with or without specificity to tumor, were examined for tumor uptake and internalization at the whole body, ex-vivo tissue, and cellular levels: antibodies, antibody fragments (Fab), serum albumin, and streptavidin. The time of peak tumor uptake was dependent solely on the size of molecules, suggesting that molecular size is the major factor that influences tumor uptake by its effect on systemic clearance and diffusion into tumor. Affinity to tumor antigen failed to augment tumor uptake of Fab above non-specific accumulation, which suggests that Fab fragments of typical monoclonal antibodies may fall below an affinity threshold for use as molecular imaging agents. Despite abundant localization into the tumor, albumin and streptavidin were not found on cell surface or inside cells. By comparing biomolecules differing in size and affinity, our study highlights that while pharmacokinetics are a dominant factor in tumor uptake for biomolecules, affinity to tumor antigen is required for tumor binding and internalization.


Subject(s)
Endocytosis , Neoplasms/metabolism , Proteins/metabolism , Animals , Diagnostic Imaging , HEK293 Cells , HeLa Cells , Humans , Immunoglobulin G/blood , Intercellular Adhesion Molecule-1/metabolism , Mice, SCID , Models, Biological , Molecular Weight , Protein Binding , Proteins/pharmacokinetics , Tissue Distribution , Whole Body Imaging , Xenograft Model Antitumor Assays
13.
Integr Biol (Camb) ; 6(4): 450-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24599186

ABSTRACT

Intraluminal crawling is considered to be important for extravasation of leukocytes in blood vessels, but biochemical/biophysical cues guiding the crawling of leukocytes have not been clearly understood. Here we provide evidence that T cells sense the topography of luminal surfaces and the nuclei of endothelial cells (ECs) using lamellipodia and filopodia, respectively, to optimize path finding during intraluminal crawling. Well-aligned EC layers or replicas of EC layers, which exhibit topography similar to that of EC layers, were fabricated, and flow was applied either parallel or perpendicular to the orientation of EC alignment. T cells crawled along the valleys of the topographical landscapes of the EC layers, while avoiding nuclei of ECs regardless of flow direction. Pharmacological inhibitor treatments revealed that sensing of topography and nuclei of EC layers was mediated by lamellipodia and filopodia, respectively. Lamellipodia or filopodia-inhibited T cells crawled significantly longer distances for extravasation than did normal T cells, indicating that sensing biophysical cues are critical for optimizing routes for extravasation.


Subject(s)
Adherens Junctions/physiology , Cell Movement/physiology , Endothelial Cells/physiology , Pseudopodia/physiology , T-Lymphocytes/physiology , Adherens Junctions/ultrastructure , Animals , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/physiology , Endothelial Cells/ultrastructure , Indoles/pharmacology , Mice , Mice, Transgenic , Microscopy, Electron, Scanning , Microscopy, Phase-Contrast , Pseudopodia/ultrastructure , Statistics, Nonparametric , T-Lymphocytes/ultrastructure , Thiophenes/pharmacology
14.
PLoS One ; 8(9): e73960, 2013.
Article in English | MEDLINE | ID: mdl-24069255

ABSTRACT

T cells navigate complex microenvironments to initiate and modulate antigen-specific immune responses. While recent intravital microscopy study revealed that migration of T cells were guided by various tissue microstructures containing unique nanoscale topographical structures, the effects of complex nanotopographical structures on the migration of T cells have not been systematically studied. In this study, we fabricated surfaces containing nanoscale zigzag structures with various side lengths and turning angles using UV-assisted capillary force lithography and motility of T cells on zigzag patterned surfaces was studied. Motility of T cells was mostly affected by the turning angle, not by the side length, of the zigzag structures. In particular, motility behaviors of T cells near interfaces formed by turning points of zigzag patterns were significantly affected by turning angles. For obtuse turning angles, most of the T cells smoothly crossed the interfaces, but as the turning angle decreased, a substantial fraction of the T cells migrated along the interfaces. When the formation of lamellipodia, thin sheet-like structures typically generated at the leading edges of migrating cells by actin polymerization-driven membrane protrusion, was inhibited by an Arp2/3 inhibitor CK-636, a substantial fraction of T cells on those surfaces containing zigzag patterns with an acute turning angle were trapped at the interfaces formed by the turning points of the zigzag patterns. This result suggests that thin, wide lamellipodia at the leading edges of T cells play critical roles in motility of T cells in complex topographical microenvironments.


Subject(s)
Cell Movement/physiology , T-Lymphocytes/physiology , Actin-Related Protein 2-3 Complex/pharmacology , Cell Movement/drug effects , Cells, Cultured , Humans , Pseudopodia/physiology , Surface Properties , T-Lymphocytes/cytology , T-Lymphocytes/ultrastructure
15.
Biomaterials ; 33(7): 2007-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22189145

ABSTRACT

In this work, well-aligned endothelial cell (EC) layers were prepared by culturing ECs on surfaces containing nanoscale ridges/grooves fabricated by UV-assisted capillary force lithography. Then, the dynamics of T cells on well-aligned ECs were compared with that on randomly oriented ECs cultured on flat surfaces. With this experimental setting, we demonstrated for the first time that EC alignment is important for the regulation of transendothelial migration (TEM) of T cells, a critical step for leukocyte infiltration; T cells preferentially underwent TEM at the junctions surrounded by more than three ECs only if ECs surrounding those junctions were poorly aligned. As a result, TEM of T cells occurred more quickly and frequently on randomly oriented ECs cultured on flat surfaces than on well-aligned ECs cultured on nanostructured surfaces. This result will suggest a new strategy for the design of synthetic small diameter vascular grafts and extend our current knowledge of leukocyte dynamics on an inflamed endothelium.


Subject(s)
Cell Culture Techniques , Endothelial Cells/metabolism , Nanostructures , T-Lymphocytes/metabolism , Animals , Blood Vessel Prosthesis , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Inflammation/pathology , Leukocytes/cytology , Leukocytes/metabolism , Mice , Surface Properties , T-Lymphocytes/cytology
16.
Biomaterials ; 31(15): 4360-6, 2010 May.
Article in English | MEDLINE | ID: mdl-20202681

ABSTRACT

Human embryonic stem cells (hESCs) are pluripotent cells that have the potential to be used for tissue engineering and regenerative medicine. Repairing nerve injury by differentiating hESCs into a neuronal lineage is one important application of hESCs. Biochemical and biological agents are widely used to induce hESC differentiation. However, it would be better if we could induce differentiation of hESCs without such agents because these factors are expensive and it is difficult to control the optimal concentrations for efficient differentiation with reduced side effects. Moreover, the mechanism of differentiation induced by these factors is still not fully understood. In this study, we present evidence that nanoscale ridge/groove pattern arrays alone can effectively and rapidly induce the differentiation of hESCs into a neuronal lineage without the use any differentiation-inducing agents. Using UV-assisted capillary force lithography, we constructed nanoscale ridge/groove pattern arrays with a dimension and alignment that were finely controlled over a large area. Human embryonic stem cells seeded onto the 350-nm ridge/groove pattern arrays differentiated into neuronal lineage after five days, in the absence differentiation-inducing agents. This nanoscale technique could be used for a new neuronal differentiation protocol of hESCs and may also be useful for nanostructured scaffolding for nerve injury repair.


Subject(s)
Cell Culture Techniques , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Neurons/physiology , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biomarkers/metabolism , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/cytology , Humans , Materials Testing , Neurons/cytology , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL