Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Proc Natl Acad Sci U S A ; 113(12): 3215-20, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26951667

ABSTRACT

Understanding the forces controlling vascular network properties and morphology can enhance in vitro tissue vascularization and graft integration prospects. This work assessed the effect of uniaxial cell-induced and externally applied tensile forces on the morphology of vascular networks formed within fibroblast and endothelial cell-embedded 3D polymeric constructs. Force intensity correlated with network quality, as verified by inhibition of force and of angiogenesis-related regulators. Tensile forces during vessel formation resulted in parallel vessel orientation under static stretching and diagonal orientation under cyclic stretching, supported by angiogenic factors secreted in response to each stretch protocol. Implantation of scaffolds bearing network orientations matching those of host abdominal muscle tissue improved graft integration and the mechanical properties of the implantation site, a critical factor in repair of defects in this area. This study demonstrates the regulatory role of forces in angiogenesis and their capacities in vessel structure manipulation, which can be exploited to improve scaffolds for tissue repair.


Subject(s)
Blood Vessels/physiology , Morphogenesis , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Physiologic , Tensile Strength , Tissue Scaffolds
2.
Anal Chem ; 90(12): 7480-7488, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29772178

ABSTRACT

A multitude of cell screening assays for diagnostic and research applications rely on quantitative measurements of a sample in the presence of different reagent concentrations. Standard methods rely on microtiter plates of varying well density, which provide simple and standardized sample addressability. However, testing hundreds of chemical dilutions requires complex automation, and typical well volumes of microtiter plates are incompatible with the analysis of a small number of cells. Here, we present a microfluidic device for creating a high-resolution chemical gradient spanning 200 nanoliter wells. Using air-based shearing, we show that the individual wells can be compartmentalized without altering the concentration gradient, resulting in a large set of isolated nanoliter cell culture wells. We provide an analytical and numerical model for predicting the concentration within each culture chamber and validate it against experimental results. We apply our system for the investigation of yeast cell metabolic gene regulation in the presence of different ratios of galactose/glucose concentrations and successfully resolve the nutrient threshold at which the cells activate the galactose pathway.


Subject(s)
Cell Culture Techniques , Galactose/chemistry , Glucose/chemistry , Microfluidic Analytical Techniques , Nanotechnology , Cell Culture Techniques/instrumentation , Galactose/metabolism , Glucose/metabolism , Microfluidic Analytical Techniques/instrumentation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
4.
Biotechnol Bioeng ; 105(3): 645-54, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19787638

ABSTRACT

Novel tissue-culture bioreactors employ flow-induced shear stress as a means of mechanical stimulation of cells. We developed a computational fluid dynamics model of the complex three-dimensional (3D) microstructure of a porous scaffold incubated in a direct perfusion bioreactor. Our model was designed to predict high shear-stress values within the physiological range of those naturally sensed by vascular cells (1-10 dyne/cm(2)), and will thereby provide suitable conditions for vascular tissue-engineering experiments. The model also accounts for cellular growth, which was designed as an added cell layer grown on all scaffold walls. Five model variants were designed, with geometric differences corresponding to cell-layer thicknesses of 0, 50, 75, 100, and 125 microm. Four inlet velocities (0.5, 1, 1.5, and 2 cm/s) were applied to each model. Wall shear-stress distribution and overall pressure drop calculations were then used to characterize the relation between flow rate, shear stress, cell-layer thickness, and pressure drop. The simulations showed that cellular growth within 3D scaffolds exposes cells to elevated shear stress, with considerably increasing average values in correlation to cell growth and inflow velocity. Our results provide in-depth analysis of the microdynamic environment of cells cultured within 3D environments, and thus provide advanced control over tissue development in vitro.


Subject(s)
Blood Vessels/growth & development , Stress, Mechanical , Tissue Engineering/methods , Computer Simulation
5.
Commun Biol ; 2: 158, 2019.
Article in English | MEDLINE | ID: mdl-31069267

ABSTRACT

The key to understanding, harnessing, and manipulating natural biological processes for the benefit of tissue engineering lies in providing a controllable dynamic environment for tissue development in vitro while being able to track cell activity in real time. This work presents a multi-channel bioreactor specifically designed to enable on-line imaging of fluorescently labeled cells embedded in replicated 3D engineered constructs subjected to different flow conditions. The images are acquired in 3D using a standard upright confocal microscope and further analyzed and quantified by computer vision. The platform is used to characterize and quantify the pace and directionality of angiogenic processes induced by flow. The presented apparatus bears considerable potential to advance scientific research, from basic research pursuing the effect of flow versus static conditions on 3D scaffolds and cell types, to clinically oriented modeling in drug screening and cytotoxicity assays.


Subject(s)
Bioreactors , Cell Culture Techniques/methods , Endothelial Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds , Cell Culture Techniques/instrumentation , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/methods , Microscopy, Confocal , Neovascularization, Physiologic , Perfusion , Rheology , Tissue Engineering/instrumentation , Vascular Endothelial Growth Factor A/pharmacology
6.
ACS Biomater Sci Eng ; 4(4): 1265-1271, 2018 Apr 09.
Article in English | MEDLINE | ID: mdl-33418657

ABSTRACT

Engineered three-dimensional (3D) constructs have received much attention as in vitro tools for the study of cell-cell and cell-matrix interactions, and have been explored for potential use as experimental models or therapeutic human tissue substitutes. Yet, due to diffusion limitations, the lack of stable and perfusable blood vessel networks jeopardizes cell viability once the tissue dimensions extend beyond several hundred microns. Direct perfusion of 3D scaffold cultures has been shown to enhance oxygen and nutrient availability. Additionally, flow-induced shear stress at physiologically relevant levels, positively impacted endothelial cell migration and alignment in various two-dimensional (2D) culture models and promoted angiogenic sprouting in microfluidic systems. However, little is known about the effect of flow on vascularization in implantable 3D engineered tissue models. The present study investigated the effect of direct flow-induced shear stress on vascularization in implantable 3D tissue. The differential effect of various levels of shear stress, applied while maintaining constant culture conditions, on vascular parameters was measured. Samples grown under direct flow conditions showed significant increases (>100%) in vessel network morphogenesis parameters and increases in vessel and extracellular matrix (ECM) protein depth distribution, as compared to those grown under static conditions. Enhanced vascular network morphogenesis parameters and higher colocalization of alpha-smooth muscle actin (α-SMA) with endothelial vessel networks characterized the specific contribution of direct flow to vessel network complexity and maturation. These observations suggest that flow conditions promote 3D neovascularization and may be advantageous in attempts to create large-volume, clinically relevant tissue substitutes.

7.
Sci Rep ; 5: 17840, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26648270

ABSTRACT

Implantable 3D engineered vascular tissue constructs can be formed by co-culturing endothelial and fibroblast cells on macroporous scaffolds. Here we show that these constructs can be used for studying the dynamics of neovascular formation in-vitro by a combination of live confocal imaging and an array of image processing and analysis tools, revealing multiple distinct stages of morphogenesis. We show that this process involves both vasculogenic and angiogenic elements, including an initial endothelial multicellular cluster formation followed by rapid extensive sprouting, ultimately resulting in a stable interconnected endothelial network morphology. This vascular morphogenesis is time-correlated with the deposition and formation of an extensive extra-cellular matrix environment. We further show that endothelial network junctions are formed by two separate morphogenic mechanisms of anastomosis and cluster thinning.


Subject(s)
Neovascularization, Physiologic , Tissue Engineering , Tissue Scaffolds , Cell Culture Techniques , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Humans , Tissue Engineering/methods , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Biomaterials ; 32(31): 7856-69, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21816465

ABSTRACT

Sufficient vascularization in engineered tissues can be achieved through coordinated application of improved biomaterial systems with proper cell types. In this study, we employed 3D fibrin gels alone or in combination with the synthetic poly(l-lactic acid) (PLLA)/polylactic-glycolic acid (PLGA) sponges to support in-vitro construct vascularization and to enhance neovascularization upon implantation. Two multicellular assays were embedded in these constructs: (a) co-culture of endothelial (EC) and fibroblast cells, and (b) a tri-culture combination of ECs, fibroblasts and tissue specific skeletal myoblast cells. In-vitro vessel network formation was examined under advanced confocal microscopy in various time points from cell seeding. Vessel network maturity levels and morphology were found to be highly regulated by fibrinogen concentrations in-vitro. Combination of PLLA/PLGA sponges with fibrin matrices provided added mechanical strength and featured highly mature vessels-like networks. Implantation studies revealed that the implanted ECs developed into 3D interconnected vessel-like networks in-vivo. The PLLA/PLGA scaffold proved to be a key stimulator of neovascularization and perfusion of implanted grafts. Our findings demonstrate that complex biomaterial platform involving fibrin and PLLA/PLGA synthetic scaffold provide a way to enhancing vascularization in-vitro and in-vivo.


Subject(s)
Blood Vessel Prosthesis , Cells/cytology , Fibrin/pharmacology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Differentiation/drug effects , Cells/drug effects , Cells/metabolism , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mice , Microscopy, Confocal , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Neovascularization, Physiologic , Perfusion , Thrombin/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL