Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Biofabrication ; 12(4): 045008, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32644945

ABSTRACT

Vascular networks consist of hierarchical structures of various diameters and are necessary for efficient blood distribution. Recent advances in vascular tissue engineering and bioprinting have allowed us to construct large vessels, such as arteries, small vessels, such as capillaries and microvessels, and intermediate-scale vessels, such as arterioles, individually. However, little is known about the control of vessel diameters between small vessels and intermediate-scale vessels. Here, we focus on vascular remodeling, which creates lasting structural changes in the vessel wall in response to hemodynamic stimuli, to regulate vessel diameters in vitro. The purpose of this study is to control the vessel diameter at an intermediate scale by inducing outward remodeling of microvessels in vitro. Human umbilical vein endothelial cells and mesenchymal stem cells were cocultured in a microfluidic device to construct microvessels, which were then perfused with a culture medium to induce outward vascular remodeling. We successfully constructed vessels with diameters of 40-150 µm in perfusion culture, whereas vessels with diameters of <20 µm were maintained in static culture. We also revealed that the in vitro vascular remodeling was mediated by NO pathways and MMP-9. These findings provide insight into the regulation of diameters of tissue-engineered blood vessels. This is an important step toward the construction of hierarchical vascular networks within biofabricated three-dimensional systems.


Subject(s)
Blood Vessels/anatomy & histology , Blood Vessels/physiology , Hemorheology , Neovascularization, Physiologic , Vascular Remodeling , Blood Vessels/drug effects , Blood Vessels/enzymology , Dextrans/chemistry , Fluorescence , Hemorheology/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Hydrodynamics , Matrix Metalloproteinase 9/metabolism , Microspheres , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide/pharmacology , Perfusion , Time Factors , Vascular Remodeling/drug effects
2.
APL Bioeng ; 3(3): 036102, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31431938

ABSTRACT

Hemodynamic and biochemical factors play important roles in critical steps of angiogenesis. In particular, interstitial flow has attracted attention as an important hemodynamic factor controlling the angiogenic process. Here, we applied a wide range of interstitial flow magnitudes to an in vitro three-dimensional (3D) angiogenesis model in a microfluidic device. This study aimed to investigate the effect of interstitial flow magnitude in combination with the vascular endothelial growth factor (VEGF) concentration on 3D microvascular network formation. Human umbilical vein endothelial cells (HUVECs) were cultured in a series of interstitial flow generated by 2, 8, and 25 mmH2O. Our findings indicated that interstitial flow significantly enhanced vascular sprout formation, network extension, and the development of branching networks in a magnitude-dependent manner. Furthermore, we demonstrated that the proangiogenic effect of interstitial flow application could not be substituted by the increased VEGF concentration. In addition, we found that HUVECs near vascular sprouts significantly elongated in >8 mmH2O conditions, while activation of Src was detected even in 2 mmH2O conditions. Our results suggest that the balance between the interstitial flow magnitude and the VEGF concentration plays an important role in the regulation of 3D microvascular network formation in vitro.

3.
Biofabrication ; 11(3): 035007, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31025629

ABSTRACT

Recent advances in microfabrication technologies have enabled us to construct collagen gel microbeads, which can be cultured with hepatocytes. However, little is known about the hepatocyte-collagen gel microbead interactions. Here, we aimed to clarify the effects of the balance between cell-cell and cell-collagen gel microbead interactions on hepatocyte morphogenesis and functions. The magnitude of cell-microbead interactions was controlled by changing the size of the microbeads, which were smaller than, comparable to, and larger than hepatocytes. These small, medium, and large microbeads were cultured separately with primary hepatocytes. Phase-contrast and time-lapse imaging revealed that the medium microbeads significantly induced the construction of 3D structures composed of the microbeads and hepatocytes in a self-organizing manner, whereas hepatocytes formed 2D monolayers with the small or large microbeads. These results suggest that only the medium microbeads induced the 3D tissue formation of hepatocytes. Furthermore, liver-specific functions, such as albumin secretion and ammonia clearance, were significantly upregulated in the 3D structures. These findings are critical to understand how to control the construction of 3D hepatocyte tissues with hydrogel microbeads in the context of biofabrication.


Subject(s)
Collagen/pharmacology , Hepatocytes/cytology , Microspheres , Morphogenesis , Animals , Cattle , Cells, Cultured , Hepatocytes/drug effects , Liver/drug effects , Liver/physiology , Male , Morphogenesis/drug effects , Rats, Sprague-Dawley , Swine , Tissue Engineering
4.
Tissue Eng Part A ; 25(5-6): 499-510, 2019 03.
Article in English | MEDLINE | ID: mdl-30234439

ABSTRACT

IMPACT STATEMENT: Construction of capillary networks is a fundamental challenge for the development of three-dimensional (3D) tissue engineering. However, it is not well understood how to construct stable capillary networks that maintain a luminal size similar to that of capillary structures in vivo (i.e., <10 µm diameter). In this study, we demonstrated the construction of stable capillary networks covered by pericyte-like perivascular cells using an in vitro 3D angiogenesis model by optimizing interactions between endothelial cells and perivascular cells. Our 3D angiogenesis model can be combined with 3D culture of epithelial cells in the context of vascularization of 3D tissue-engineered constructs.


Subject(s)
Capillaries/cytology , Pericytes/cytology , Tissue Engineering/methods , Basement Membrane/metabolism , Cell Proliferation , Human Umbilical Vein Endothelial Cells/cytology , Humans , Imaging, Three-Dimensional , Mesenchymal Stem Cells/cytology , Microfluidics , Neovascularization, Physiologic
5.
Acta Biomater ; 95: 307-318, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30593886

ABSTRACT

There is a great deal of demand for the construction of transplantable liver grafts. Over the last decade, decellularization techniques have been developed to construct whole liver tissue grafts as potential biomaterials. However, the lack of intact vascular networks, especially sinusoids, in recellularized liver scaffolds leads to hemorrhage and thrombosis after transplantation, which is a major obstacle to the development of transplantable liver grafts. In the present study, we hypothesized that both mechanical (e.g., fluid shear stress) and chemical factors (e.g., fibronectin coating) can enhance the formation of hierarchical vascular networks including sinusoid-scale microvessels. We demonstrated that perfusion culture promoted formation of sinusoid-scale microvessels in recellularized liver scaffolds, which was not observed in static culture. In particular, perfusion culture at 4.7 ml/min promoted the formation of sinusoid-scale microvessels compared to perfusion culture at 2.4 and 9.4 ml/min. In addition, well-aligned endothelium was observed in perfusion culture, suggesting that endothelial cells sensed the flow-induced shear stress. Moreover, fibronectin coating of decellularized liver scaffolds enhanced the formation of sinusoid-scale microvessels in perfusion culture at 4.7 ml/min. This study represents a critical step in the development of functional recellularized liver scaffolds, which can be used not only for transplantation but also for drug screening and disease-modeling studies. STATEMENT OF SIGNIFICANCE: Decellularized liver scaffolds are promising biomaterials that allow production of large-scale tissue-engineered liver grafts. However, it is difficult to maintain recellularized liver grafts after transplantation due to hemorrhage and thrombosis. To overcome this obstacle, construction of an intact vascular network including sinusoid-scale microvessels is essential. In the present study, we succeeded in constructing sinusoid-scale microvessels in decellularized liver scaffolds via a combination of perfusion culture and surface coating. We further confirmed that endothelial cells in decellularized liver scaffolds responded to flow-derived mechanical stress by aligning actin filaments. Our strategy to construct sinusoid-scale microvessels is critical for the development of intact vascular networks, and addresses the limitations of recellularized liver scaffolds after transplantation.


Subject(s)
Liver/cytology , Microvessels/cytology , Perfusion , Animals , Fibronectins/pharmacology , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley , Stress, Mechanical , Tissue Scaffolds/chemistry
6.
J Atheroscler Thromb ; 22(10): 1091-9, 2015.
Article in English | MEDLINE | ID: mdl-25972025

ABSTRACT

AIM: von Willebrand factor (VWF) plays an important role in the regulation of hemostasis and thrombosis formation, particularly under a high shear rate. However, the adhesive force due to the molecular interaction between VWF and glycoprotein Ibα (GPIbα) has not been fully explored. Thus, we employed atomic force microscopy to directly measure the adhesive force between VWF and GPIbα. METHODS: We measured the adhesive force between VWF and GPIbα at the molecular level using an atomic force microscope (AFM). An AFM cantilever was coated with recombinant N-terminus VWF binding site of GPIbα, whereas a cover glass was coated with native VWF. RESULTS: The adhesive force at the molecular level was measured using an AFM. In the presence of 1 µg/mL VWF, the adhesion force was nearly 200 pN. As per the Gaussian fit analysis, the adhesive force of a single bond could have been 54 or 107 pN. CONCLUSION: Our consideration with the Gaussian fit analysis proposed that the adhesive force of a single bond could be 54 pN, which is very close to that obtained by optical tweezers (50 pN).


Subject(s)
Blood Platelets/metabolism , Hemostasis/physiology , Microscopy, Atomic Force/methods , Platelet Glycoprotein GPIb-IX Complex/chemistry , Thrombosis/blood , von Willebrand Factor/chemistry , Blood Platelets/ultrastructure , Humans , Molecular Structure , Platelet Glycoprotein GPIb-IX Complex/ultrastructure , Thrombosis/diagnosis , von Willebrand Factor/ultrastructure
7.
J Tissue Eng Regen Med ; 9(3): 247-56, 2015 Mar.
Article in English | MEDLINE | ID: mdl-23086892

ABSTRACT

In liver sinusoids, hepatic stellate cells (HSCs) locate the outer surface of microvessels to form a functional unit with endothelia and hepatocytes. To reconstruct functional liver tissue in vitro, formation of the HSC-incorporated sinusoidal structure is essential. We previously demonstrated capillary formation of endothelial cells (ECs) in tri-culture, where a polyethylene terephthalate (PET) microporous membrane was intercalated between the ECs and hepatic organoids composed of small hepatocytes (SHs), i.e. hepatic progenitor cells, and HSCs. However, the high thickness and low porosity of the membranes limited heterotypic cell-cell interactions, which are essential to form HSC-EC hybrid structures. Here, we focused on the effective use of the thin and highly porous poly( d, l-lactide-co-glycolide) (PLGA) microporous membranes in SH-HSC-EC tri-culture to reconstruct the HSC-incorporated liver capillary structures in vitro. First, the formation of EC capillary-like structures was induced on Matrigel-coated PLGA microporous membranes. Next, the membranes were stacked on hepatic organoids composed of small SHs and HSCs. When the pore size and porosity of the membranes were optimized, HSCs selectively migrated to the EC capillary-like structures. This process was mediated in part by platelet-derived growth factor (PDGF) signalling. In addition, the HSCs were located along the outer surface of the EC capillary-like structures with their long cytoplasmic processes. In the HSC-incorporated capillary tissues, SHs acquired high levels of differentiated functions, compared to those without ECs. This model will provide a basis for the construction of functional, thick, vascularized liver tissues in vitro.


Subject(s)
Coated Materials, Biocompatible/chemistry , Endothelial Cells/metabolism , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Lactic Acid/chemistry , Membranes, Artificial , Polyglycolic Acid/chemistry , Animals , Cells, Cultured , Coculture Techniques/methods , Endothelial Cells/cytology , Hepatic Stellate Cells/cytology , Hepatocytes/cytology , Male , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity , Rats , Rats, Sprague-Dawley
8.
J Cereb Blood Flow Metab ; 34(11): 1761-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25074744

ABSTRACT

The pial and penetrating arteries have a crucial role in regulating cerebral blood flow (CBF) to meet neural demand in the cortex. Here, we examined the longitudinal effects of chronic hypoxia on the arterial diameter responses to single whisker stimulation in the awake mouse cortex, where activity-induced responses of CBF were gradually attenuated. The vasodilation responses to whisker stimulation under prehypoxia normal conditions were 8.1% and 12% relative to their baselines in the pial arteries and penetrating arterioles, respectively. After 3 weeks of hypoxia, however, these responses were significantly reduced to 5.5% and 4.1%, respectively. The CBF response, measured using laser-Doppler flowmetry (LDF), induced by the same whisker stimulation was also attenuated (14% to 2.6%). A close linear correlation was found for the responses between the penetrating arteriolar diameter and LDF, and their temporal dynamics. After 3 weeks of chronic hypoxia, the initiation of vasodilation in the penetrating arterioles was significantly extended, but the pial artery responses remained unchanged. These results show that vasodilation of the penetrating arterioles followed the pial artery responses, which are not explainable in terms of proximal integration signaling. The findings therefore indicate an additional mechanism for triggering pial artery dilation in the neurovascular coupling.


Subject(s)
Cerebral Arteries/physiology , Cerebrovascular Circulation/physiology , Somatosensory Cortex/blood supply , Synaptic Transmission/physiology , Vasodilation/physiology , Wakefulness/physiology , Animals , Arterioles/physiology , Blood Flow Velocity/physiology , Mice , Mice, Transgenic , Neurons/metabolism , Somatosensory Cortex/physiology
9.
Neurol Res ; 36(8): 752-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24620967

ABSTRACT

OBJECTIVES: Biomechanical stress distribution correlates with the biological responses after stenting. Computational analyses have contributed to the optimization of stent geometry. In particular, structural analysis based on pre-operative angiography can be used to predict the stent-artery interaction before endovascular treatments. However, the simulated results need to be validated. In this report, we compared the simulated arterial structure with post-operative images after an intracranial Wingspan stent. METHODS: A Wingspan stent was deployed at a slightly curved ascending pharyngeal artery (APA) in the swine. Using a finite element method (FEM), the configuration after stenting was simulated and quantitatively compared with post-procedural 3D angiography. RESULTS: The finite element analysis demonstrated arterial straightening after stenting. The simulated images were similar to the experimental results with respect to the curvature index of the center line and the cross-sectional areas. CONCLUSION: We assessed the simulated structural deformation after Wingspan stenting, by comparison with experimental results.


Subject(s)
Computer Simulation , Imaging, Three-Dimensional , Stents , Stress, Mechanical , Alloys , Angiography , Animals , Biomechanical Phenomena , Carotid Artery, External/diagnostic imaging , Carotid Artery, External/surgery , Finite Element Analysis , Swine
10.
PLoS One ; 8(12): e82085, 2013.
Article in English | MEDLINE | ID: mdl-24312630

ABSTRACT

Endothelial progenitor cell (EPC) transplantation induces the formation of new blood-vessel networks to supply nutrients and oxygen, and is feasible for the treatment of ischemia and cardiovascular diseases. However, the role of EPCs as a source of proangiogenic cytokines and consequent generators of an extracellular growth factor microenvironment in three-dimensional (3D) microvessel formation is not fully understood. We focused on the contribution of EPCs as a source of proangiogenic cytokines on 3D microvessel formation using an in vitro 3D network model. To create a 3D network model, EPCs isolated from rat bone marrow were sandwiched with double layers of collagen gel. Endothelial cells (ECs) were then cultured on top of the upper collagen gel layer. Quantitative analyses of EC network formation revealed that the length, number, and depth of the EC networks were significantly enhanced in a 3D model with ECs and EPCs compared to an EC monoculture. In addition, conditioned medium (CM) from the 3D model with ECs and EPCs promoted network formation compared to CM from an EC monoculture. We also confirmed that EPCs secreted vascular endothelial growth factor (VEGF). However, networks cultured with the CM were shallow and did not penetrate the collagen gel in great depth. Therefore, we conclude that EPCs contribute to 3D network formation at least through indirect incorporation by generating a local VEGF gradient. These results suggest that the location of EPCs is important for controlling directional 3D network formation in the field of tissue engineering.


Subject(s)
Endothelial Cells/cytology , Stem Cells/cytology , Stem Cells/metabolism , Tissue Engineering/methods , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone Marrow Cells/cytology , Cattle , Culture Media, Conditioned/pharmacology , Fibroblast Growth Factor 2/pharmacology , Microvessels/cytology , Microvessels/physiology , Neovascularization, Physiologic/drug effects , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects
11.
Adv Exp Med Biol ; 789: 419-425, 2013.
Article in English | MEDLINE | ID: mdl-23852524

ABSTRACT

The present study reports a semiautomatic image analysis method for measuring the spatiotemporal dynamics of the vessel dilation that was fluorescently imaged with either confocal or two-photon microscope. With this method, arterial dilation induced by whisker stimulation was compared between cortical surface and parenchymal tissue in the vibrissae area of somatosensory cortex in awake Tie2-GFP mice in which the vascular endothelium had genetically expressed green fluorescent protein. We observed that a mean arterial diameter during a pre-stimulus baseline state was 39 ± 7, 19 ± 1, 16 ± 4, 17 ± 4, and 14 ± 3 µm at depths of 0, 100, 200, 300, and 400 µm, respectively. The stimulation-evoked dilation induced by mechanical whisker deflection (10 Hz for 5 s) was 3.4 ± 0.8, 1.8 ± 0.8, 1.8 ± 0.9, 1.6 ± 0.9, and 1.5 ± 0.6 µm at each depth, respectively. Consequently, no significant differences were observed for the vessel dilation rate between the cortical surface and parenchymal arteries: 8.8 %, 9.9 %, 10.9 %, 9.2 %, and 10.3 % relative to their baseline diameters, respectively. These preliminary results demonstrate that the present method is useful to further investigate the quantitative relationships between the spatiotemporally varying arterial tone and the associated blood flow changes in the parenchymal microcirculation to reveal the regulatory mechanism of the cerebral blood flow.


Subject(s)
Arteries/anatomy & histology , Brain/blood supply , Cerebrovascular Circulation/physiology , Animals , Arteries/physiology , Endothelium, Vascular/physiology , Mice , Physical Stimulation/methods , Somatosensory Cortex/blood supply , Vasodilation/physiology , Vibrissae/physiology
12.
J Cereb Blood Flow Metab ; 33(9): 1440-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23756692

ABSTRACT

To clarify mechanisms through which activation of the nucleus basalis of Meynert (NBM) increases cerebral cortical blood flow, we examined whether cortical parenchymal arteries dilate during NBM stimulation in anesthetized mice. We used two-photon microscopy to measure the diameter of single penetrating arteries at different depths (~800 µm, layers I to V) of the frontal cortex, and examined changes in the diameter during focal electrical stimulation of the NBM (0.5 ms at 30 to 50 µA and 50 Hz) and hypercapnia (3% CO2 inhalation). Stimulation of the NBM caused diameter of penetrating arteries to increase by 9% to 13% of the prestimulus diameter throughout the different layers of the cortex, except at the cortical surface and upper part of layer V, where the diameter of penetrating arteries increased only slightly during NBM stimulation. Hypercapnia caused obvious dilation of the penetrating arteries in all cortical layers, including the surface arteries. The diameters began to increase within 1 second after the onset of NBM stimulation in the upper cortical layers, and later in lower layers. Our results indicate that activation of the NBM dilates cortical penetrating arteries in a layer-specific manner in magnitude and latency, presumably related to the density of cholinergic nerve terminals from the NBM.


Subject(s)
Basal Nucleus of Meynert , Cerebral Arteries , Cerebrovascular Circulation/physiology , Vasodilation/physiology , Animals , Basal Nucleus of Meynert/anatomy & histology , Basal Nucleus of Meynert/blood supply , Cerebral Arteries/anatomy & histology , Cerebral Arteries/physiology , Male , Mice
13.
J Neurointerv Surg ; 5 Suppl 3: iii38-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23048176

ABSTRACT

OBJECT: Little is known about how much protection a flow diversion stent provides to a non-thrombosed aneurysm without the adjunctive use of coils. METHODS: A three-dimensional anatomically realistic computation aneurysm model was created from the digital subtraction angiogram of a large internal carotid artery-ophthalmic artery aneurysm which could have been treated with either a neck bridging stent or a flow diversion stent. Three-dimensional computational models of the Neuroform EZ neck bridging stent and Pipeline embolization device were created based on measurements with a stereo-microscope. Each stent was placed in the computational aneurysm model and intra-aneurysmal flow structures were compared before and after placement of the stents. Computational fluid dynamics were performed by numerically solving the continuity and Navier-Stokes momentum equations for a steady blood flow based on the finite volume method. Blood was assumed as an incompressible Newtonian fluid. Vessel walls were assumed to be rigid, and no-slip boundary conditions were applied at the lumens. To estimate the change in the intra-aneurysmal pressures we assumed that, at the inlets, the intra-arterial pressure at peak systole was 120 mm Hg both before and after stent placement RESULTS: Without any stent, the blood flow entered into the aneurysm dome from the mid to proximal neck area and ascended along the distal wall of the aneurysm. The flow then changed its direction anteriorly and moved along the proximal wall of the aneurysm dome. In addition to the primary intra-aneurysmal circulation pattern, a counterclockwise vortex was observed in the aneurysm dome. The placement of a Neuroform EZ stent induced a mean reduction in flow velocity of 14% and a small change in the overall intra-aneurysmal flow pattern. The placement of a Pipeline device induced a mean reduction in flow velocity of 74% and a significant change in flow pattern. Despite the flow velocity changes, Neuroform EZ and Pipeline devices induced reductions in intra-aneurysmal pressure of only 4 mm Hg and 8 mm Hg, respectively. CONCLUSIONS: The flow diversion effects of both stents were limited to flow velocity reduction. In a non-thrombosed aneurysm or an aneurysm with delayed thrombosis, the intra-aneurysmal pressure remains essentially unchanged regardless of the level of the intra-aneurysmal flow velocity reduction induced by the stents.


Subject(s)
Cerebrovascular Circulation/physiology , Intracranial Aneurysm/physiopathology , Intracranial Aneurysm/therapy , Stents , Angiography, Digital Subtraction , Blood Flow Velocity/physiology , Blood Pressure/physiology , Carotid Artery, Internal/pathology , Carotid Artery, Internal/surgery , Computer Simulation , Equipment Design , Humans , Software
14.
Int J Numer Method Biomed Eng ; 28(2): 214-28, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22548127

ABSTRACT

Hemodynamics is thought to be a fundamental factor in the formation, progression, and rupture of cerebral aneurysms. Understanding these mechanisms is important to improve their rupture risk assessment and treatment. In this study, we analyze the blood flow field in a growing cerebral aneurysm using experimental particle image velocimetry (PIV) and computational fluid dynamics (CFD) techniques. Patient-specific models were constructed from longitudinal 3D computed tomography angiography images acquired at 1-y intervals. Physical silicone models were constructed from the computed tomography angiography images using rapid prototyping techniques, and pulsatile flow fields were measured with PIV. Corresponding CFD models were created and run under matching flow conditions. Both flow fields were aligned, interpolated, and compared qualitatively by inspection and quantitatively by defining similarity measures between the PIV and CFD vector fields. Results showed that both flow fields were in good agreement. Specifically, both techniques provided consistent representations of the main intra-aneurysmal flow structures and their change during the geometric evolution of the aneurysm. Despite differences observed mainly in the near wall region, and the inherent limitations of each technique, the information derived is consistent and can be used to study the role of hemodynamics in the natural history of intracranial aneurysms.


Subject(s)
Computer Simulation , Hemodynamics/physiology , Intracranial Aneurysm/physiopathology , Models, Cardiovascular , Hemorheology , Humans , Male , Middle Aged
15.
Biomaterials ; 33(9): 2693-700, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22236830

ABSTRACT

There is great demand for constructing well-organized three-dimensional (3D) tissues in vitro. Here, we developed a 3D stacked culture method using biodegradable poly(d,l-lactide-co- glycolide) (PLGA) membranes with defined topography. Pore size and porosity of the membranes can be controlled by changing the moisture content during fabrication. The optimized membrane served as a scaffold to manipulate small hepatocyte (SH) layers when they were stacked, while it degraded after stacking, resulting in the reorganization of the cells into a 3D stacked structure. Immunofluorescent staining for domain markers of cell polarity and electron microscopy confirmed that the cells in the 3D stacked structures recovered polarity. Furthermore, the cells exhibited improved liver-specific function as compared with cells in a monolayer. This 3D stacked culture may enable reconstruction of multilayered hepatic tissues with highly differentiated functions in vitro.


Subject(s)
Biocompatible Materials/pharmacology , Hepatocytes/cytology , Lactic Acid/pharmacology , Membranes, Artificial , Polyglycolic Acid/pharmacology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Biodegradation, Environmental/drug effects , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Shape/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Liver/cytology , Microscopy, Electron, Scanning , Organ Specificity/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
16.
Tissue Eng Part A ; 18(9-10): 1045-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22220631

ABSTRACT

Vascularization of engineered tissues in vitro remains a major challenge in liver tissue engineering. Liver microvessels, termed liver sinusoids, have highly specialized structures, and recapturing these sinusoidal structures is essential for reconstruction of functional liver tissue in vitro. Liver sinusoids are composed of hepatocytes, hepatic stellate cells (HSCs), and endothelial cells (ECs). Direct HSC-EC contacts are increasingly recognized for their roles in EC capillary morphogenesis. However, the hypothetical role of HSC-EC contacts in morphogenesis remains unclear in hepatocyte-HSC-EC triculture. In the present study, we first determined the effects of direct HSC-EC contacts on EC capillary morphogenesis using a hepatocyte-HSC-EC triculture model where HSC behavior was spatially controlled to achieve HSC-mediated proximal layers of hepatocytes and ECs. EC capillary morphogenesis was induced by overlaying Matrigel on an EC layer. Direct HSC-EC contacts inhibited EC capillary morphogenesis, suggesting that the HSC-EC contacts may be an important factor in capillary formation. We next tested the hypothesis that, in addition to spatial control, temporal control of HSC behavior is also important in achieving capillary morphogenesis in the triculture. ECs responded to the induction of capillary morphogenesis before the formation of direct HSC-EC contacts, while the ECs remained to form monolayers when capillary morphogenesis was induced after the HSC-EC contacts were established. When capillary morphogenesis was successfully achieved in the triculture, HSCs tended to preferably localize near the preformed capillary-like structures, resulting in the reconstruction of liver sinusoidal structures. In these structures, hepatocyte maturation was induced. Our findings indicate that control, both spatial and temporal, of HSC behavior is a key engineering strategy for the vascularization of engineered liver tissue in vitro.


Subject(s)
Endothelial Cells/cytology , Hepatic Stellate Cells/cytology , Liver/cytology , Tissue Engineering/methods , Animals , Cell Communication/physiology , Cells, Cultured , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley
17.
Biomatter ; 2(4): 290-5, 2012.
Article in English | MEDLINE | ID: mdl-23507893

ABSTRACT

To meet the increasing demand for liver tissue engineering, various three-dimensional (3D) liver cell culture techniques have been developed. Nevertheless, conventional liver cell culture techniques involving the suspending cells in extracellular matrix (ECM) components and the seeding of cells into 3D biodegradable scaffolds have an intrinsic shortcoming, low cell-scaffold ratios. We have developed a microporous membrane-based liver cell culture technique. Cell behaviors and tissue organization can be controlled by membrane geometry, and cell-dense thick tissues can be reconstructed by layering cells cultured on biodegradable microporous membranes. Applications extend from liver parenchymal cell monoculture to multi-cell type cultures for the reconstruction of 3D functional liver tissue. This review focuses on the expanding role for microporous membranes in liver tissue engineering, primarily from our research.


Subject(s)
Liver, Artificial , Liver/pathology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Bile Ducts/pathology , Biocompatible Materials/chemistry , Biodegradation, Environmental , Cell Culture Techniques , Extracellular Matrix/metabolism , Hepatocytes/cytology , Humans , Imaging, Three-Dimensional , Liver Diseases/metabolism , Liver Failure/therapy , Porosity , Regeneration
18.
Tissue Eng Part A ; 17(3-4): 361-70, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20799907

ABSTRACT

Hepatic stellate cells (HSCs) form a functional unit with endothelia and hepatocytes in the liver to play a pivotal role in heterotypic cellular communication. To investigate this role of HSCs, it is of great benefit to establish a triculture model that forms the functional unit from proximal layers of hepatocytes, HSCs, and endothelial cells (ECs). Here, we established a three-dimensional triculture model, using a microporous membrane to create the functional unit. HSC behavior was controlled by the membrane pore size, which was critical for achieving proximal cell layers. With a specific pore size, the HSCs intercalated between layers of hepatocytes and ECs, due to the limitation on HSC behavior. When only cytoplasmic processes of quiescent HSCs were adjacent to ECs, while the HSC bodies remained on the side of the hepatocytes, the ECs changed morphologically and were capable of long-term survival. We confirmed that HSCs mediated the communication between hepatocytes and ECs in terms of EC morphogenesis. This triculture model allows us to investigate the roles of HSCs as both facilitators and integrators of cell-cell communication between hepatocytes and ECs, and is useful for investigating heterotypic cellular communication in vitro.


Subject(s)
Cell Communication/physiology , Cell Culture Techniques/methods , Coculture Techniques/methods , Endothelial Cells/cytology , Hepatic Stellate Cells/cytology , Hepatocytes/cytology , Membranes, Artificial , Animals , Cells, Cultured , Endothelial Cells/physiology , Hepatic Stellate Cells/physiology , Hepatocytes/physiology , Male , Porosity , Rats , Rats, Sprague-Dawley
19.
Med Eng Phys ; 32(9): 1015-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20675176

ABSTRACT

Endovascular treatment of cerebral aneurysms using stents has advanced markedly in recent years. Mechanically, a cerebrovascular stent must be very flexible longitudinally and have low radial stiffness. However, no study has examined the stress distribution and deformation of cerebrovascular stents using the finite element method (FEM) and experiments. Stents can have open- and closed-cell structures, and open-cell stents are used clinically in the cerebrovasculature because of their high flexibility. However, the open-cell structure confers a risk of in-stent stenosis due to protrusion of stent struts into the normal parent artery. Therefore, a flexible stent with a closed-cell structure is required. To design a clinically useful, highly flexible, closed-cell stent, one must examine the mechanical properties of the closed-cell structure. In this study, we investigated the relationship between mesh patterns and the mechanical properties of closed-cell stents. Several mesh patterns were designed and their characteristics were studied using numerical simulation. The results showed that the bending stiffness of a closed-cell stent depends on the geometric configuration of the stent cell. It decreases when the stent cell is stretched in the circumferential direction. Mechanical flexibility equal to an open-cell structure was obtained in a closed-cell structure by varying the geometric configuration of the stent cell.


Subject(s)
Intracranial Aneurysm/surgery , Mechanical Phenomena , Prosthesis Design/methods , Stents , Compressive Strength , Finite Element Analysis , Materials Testing , Models, Molecular , Molecular Conformation , Reproducibility of Results
20.
J Biomech Eng ; 131(7): 074002, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19640134

ABSTRACT

Oxygen is essential to maintaining normal brain function. A large body of evidence suggests that the partial pressure of oxygen (pO(2)) in brain tissue is physiologically maintained within a narrow range in accordance with region-specific brain activity. Since the transportation of oxygen in the brain tissue is mainly driven by a diffusion process caused by a concentration gradient of oxygen from blood to cells, the spatial organization of the vascular system, in which the oxygen content is higher than in tissue, is a key factor for maintaining effective transportation. In addition, a local mechanism that controls energy demand and blood flow supply plays a critical role in moment-to-moment adjustment of tissue pO(2) in response to dynamically varying brain activity. In this review, we discuss the spatiotemporal structures of brain tissue oxygen transport in relation to local brain activity based on recent reports of tissue pO(2) measurements with polarographic oxygen microsensors in combination with simultaneous recordings of neural activity and local cerebral blood flow in anesthetized animal models. Although a physiological mechanism of oxygen level sensing and control of oxygen transport remains largely unknown, theoretical models of oxygen transport are a powerful tool for better understanding the short-term and long-term effects of local changes in oxygen demand and supply. Finally, emerging new techniques for three-dimensional imaging of the spatiotemporal dynamics of pO(2) map may enable us to provide a whole picture of how the physiological system controls the balance between demand and supply of oxygen during both normal and pathological brain activity.


Subject(s)
Brain Chemistry , Brain/metabolism , Models, Neurological , Oxygen Consumption/physiology , Oxygen/chemistry , Oxygen/metabolism , Animals , Biological Transport, Active/physiology , Diffusion , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...