Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Microvasc Res ; 122: 60-70, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30472038

RESUMEN

Every organ demonstrates specific vascular characteristics and functions maintained by interactions of endothelial cells (ECs) and parenchymal cells. Particularly, brain ECs play a central role in the formation of a functional blood brain barrier (BBB). Organ-specific ECs have their own morphological features, and organ specificity must be considered when investigating interactions between ECs and other cell types constituting a target organ. Here we constructed angiogenesis-based microvascular networks with perivascular cells in a microfluidic device setting by coculturing ECs and mesenchymal stem cells (MSCs). Furthermore, we analyzed endothelial barrier functions as well as fundamental morphology, an essential step to build an in vitro BBB model. In particular, we used both brain microvascular ECs (BMECs) and human umbilical vein ECs (HUVECs) to test if organ specificity of ECs affects the formation processes and endothelial barrier functions of an engineered microvascular network. We found that microvascular formation processes differed by the source of ECs. HUVECs formed more extensive microvascular networks compared to BMECs while no differences were observed between BMECs and HUVECs in terms of both the microvascular diameter and the number of pericytes peripherally associated with the microvasculatures. To compare the endothelial barrier functions of each type of EC, we performed fluorescence dextran perfusion on constructed microvasculatures. The permeability coefficient of BMEC microvasculatures was significantly lower than that of HUVEC microvasculatures. In addition, there were significant differences in terms of tight junction protein expression. These results suggest that the organ source of ECs influences the properties of engineered microvasculature and thus is a factor to be considered in the design of organ-specific cell culture models.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Permeabilidad Capilar , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microvasos/metabolismo , Neovascularización Fisiológica , Pericitos/metabolismo , Barrera Hematoencefálica/citología , Comunicación Celular , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos , Dispositivos Laboratorio en un Chip , Microvasos/citología , Fenotipo , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
2.
Biol Pharm Bull ; 40(10): 1646-1653, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28966236

RESUMEN

The cross-linking of elastin by lysyl oxidase (LOX) family members is essential for the integrity and elasticity of elastic fibers, which play an important role in the characteristic resilience of various tissues. However, the temporal sequence of oxidation by LOX during elastic fiber formation is still incompletely understood. Here, we demonstrate that the cross-linking of tropoelastin molecules by LOX occurs concurrent with elastin deposition. Our data show that LOX deficiency or the inhibition of LOX enzyme activity leads to the loss of elastin deposition in skin fibroblast. Moreover, overexpression of LOX promotes the deposition and alignment of tropoelastin, whereas the addition of recombinant active-form of LOX in culture medium caused abnormal elastic fiber assembly. Immunoblotting and immunofluorescence show that LOX and tropoelastin are present together with fibronectin on the cell surface of preconfluent cultures. Further, fluorescence activated cell sorting (FACS) analysis for the localization of LOX on the cell surface reveals that the transfer of LOX to the extracellular space occurs in association with elastic fiber formation. In conclusion, our results support the view that LOX and tropoelastin are present on the cell surface and suggests the possibility that lysine oxidation by LOX precedes tropoelastin deposition onto microfibrils.


Asunto(s)
Proteína-Lisina 6-Oxidasa/metabolismo , Tropoelastina/metabolismo , Aminoácido Oxidorreductasas/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Células HEK293 , Humanos , Lisina/metabolismo , Oxidación-Reducción , Proteína-Lisina 6-Oxidasa/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tropoelastina/genética
3.
Genes Cells ; 20(12): 1077-87, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26610870

RESUMEN

Vascular calcification increases the risk of cardiovascular mortality. We previously reported that expression of elastin decreases with progression of inorganic phosphorus (Pi)-induced vascular smooth muscle cell (VSMC) calcification. However, the regulatory mechanisms of elastin mRNA expression during vascular calcification remain unclear. MicroRNA-29 family members (miR-29a, b and c) are reported to mediate elastin mRNA expression. Therefore, we aimed to determine the effect of miR-29 on elastin expression and Pi-induced vascular calcification. Calcification of human VSMCs was induced by Pi and evaluated measuring calcium deposition. Pi stimulation promoted Ca deposition and suppressed elastin expression in VSMCs. Knockdown of elastin expression by shRNA also promoted Pi-induced VSMC calcification. Elastin pre-mRNA measurements indicated that Pi stimulation suppressed elastin expression without changing transcriptional activity. Conversely, Pi stimulation increased miR-29a and miR-29b expression. Inhibition of miR-29 recovered elastin expression and suppressed calcification in Pi-treated VSMCs. Furthermore, over-expression of miR-29b promoted Pi-induced VSMC calcification. RT-qPCR analysis showed knockdown of elastin expression in VSMCs induced expression of osteoblast-related genes, similar to Pi stimulation, and recovery of elastin expression by miR-29 inhibition reduced their expression. Our study shows that miR-29-mediated suppression of elastin expression in VSMCs plays a pivotal role in osteoblastic differentiation leading to vascular calcification.


Asunto(s)
Elastina/genética , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Osteoblastos/efectos de los fármacos , Fósforo/farmacología , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Regulación hacia Abajo , Regulación de la Expresión Génica , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Osteoblastos/metabolismo , Calcificación Vascular/etiología
4.
Genes Cells ; 20(12): 982-91, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26419830

RESUMEN

Vascular calcification is known to reduce the elasticity of aorta. Several studies have suggested that autophagy-lysosomal pathway (ALP) in vascular smooth muscle cells (VSMCs) is associated with vascular calcification. A major component of oxidized low-density lipoproteins, 7-ketocholesterol (7-KC), has been reported to promote inorganic phosphorus (Pi)-induced vascular calcification and induce ALP. The aim of this study was to unravel the relationship between ALP and the progression of calcification by 7-KC. Calcification of human VSMCs was induced by Pi stimulation in the presence or absence of 7-KC. FACS analysis showed that 7-KC-induced apoptosis at a high concentration (30 µM), but not at a low concentration (15 µM). Interestingly, 7-KC promoted calcification in VSMCs regardless of apoptosis. Immunoblotting and immunostaining showed that 7-KC inhibits not only the fusion of autophagosomes and lysosomes but also causes a swell of lysosomes with the reduction of cathepsin B and D. Moreover, lysosomal protease inhibitors exacerbated the apoptosis-independent calcification by 7-KC although inhibition of autophagosome formation by Atg5 siRNA did not. Finally, the 7-KC-induced progression of calcification was alleviated by the treatment with antioxidant. Taken together, our data showed that 7-KC promotes VSMC calcification through lysosomal-dysfunction-dependent oxidative stress.


Asunto(s)
Cetocolesteroles/farmacología , Lisosomas/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Calcificación Vascular/inducido químicamente , Apoptosis/efectos de los fármacos , Catepsina B/metabolismo , Catepsina D/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lisosomas/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Fósforo/farmacología , Calcificación Vascular/metabolismo
5.
J Biol Chem ; 288(43): 30990-1001, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24036115

RESUMEN

Cell-matrix interactions are critical for tumor cell migration. Lutheran (Lu), also known as basal cell adhesion molecule (B-CAM), competes with integrins for binding to laminin α5, a subunit of LM-511, a major component of basement membranes. Here we show that the preferential binding of Lu/B-CAM to laminin α5 promotes tumor cell migration. The attachment of Lu/B-CAM transfectants to LM-511 was slightly weaker than that of control cells, and this was because Lu/B-CAM disturbed integrin binding to laminin α5. Lu/B-CAM induced a spindle cell shape with pseudopods and promoted cell migration on LM-511. In addition, blocking with an anti-Lu/B-CAM antibody led to a flat cell shape and inhibited migration on LM-511, similar to the effects of an activating integrin ß1 antibody. We conclude that tumor cell migration on LM-511 requires that Lu/B-CAM competitively modulates cell attachment through integrins. We suggest that this competitive interaction is involved in a balance between static and migratory cell behaviors.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Integrinas/metabolismo , Laminina/metabolismo , Sistema del Grupo Sanguíneo Lutheran/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Animales , Membrana Basal/metabolismo , Membrana Basal/patología , Adhesión Celular/genética , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Forma de la Célula , Células HEK293 , Humanos , Integrinas/genética , Laminina/genética , Sistema del Grupo Sanguíneo Lutheran/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología
6.
PLoS One ; 19(2): e0297285, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38359035

RESUMEN

Reconstruction of the biliary system is indispensable for the regeneration of transplantable liver grafts. Here, we report the establishment of the first continuous three-dimensional biliary system scaffold for bile acid excretion using a novel method. We confirmed the preservation of the liver-derived extracellular matrix distribution in the scaffold. In addition, hepatocyte progenitors decellularized via the bile duct by slow-speed perfusion differentiated into hepatocyte- and cholangiocyte-like cells, mimicking hepatic cords and bile ducts, respectively. Furthermore, qRT-PCR demonstrated increased ALB, BSEP, and AQP8 expression, revealing bile canaliculi- and bile duct-specific genetic patterns. Therefore, we concluded that locally preserved extracellular matrices in the scaffold stimulated hepatic progenitors and provided efficient differentiation, as well as regeneration of a three-dimensional continuous biliary system from hepatic cords through bile ducts. These findings suggest that organ-derived scaffolds can be utilized for the efficient reconstruction of functional biliary systems.


Asunto(s)
Sistema Biliar , Hígado , Hepatocitos , Conductos Biliares , Matriz Extracelular
7.
Adv Exp Med Biol ; 789: 419-425, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23852524

RESUMEN

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.


Asunto(s)
Arterias/anatomía & histología , Encéfalo/irrigación sanguínea , Circulación Cerebrovascular/fisiología , Animales , Arterias/fisiología , Endotelio Vascular/fisiología , Ratones , Estimulación Física/métodos , Corteza Somatosensorial/irrigación sanguínea , Vasodilatación/fisiología , Vibrisas/fisiología
8.
Acta Biomater ; 166: 301-316, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37164300

RESUMEN

Tissue engineers have utilised a variety of three-dimensional (3D) scaffolds for controlling multicellular dynamics and the resulting tissue microstructures. In particular, cutting-edge microfabrication technologies, such as 3D bioprinting, provide increasingly complex structures. However, unpredictable microtissue detachment from scaffolds, which ruins desired tissue structures, is becoming an evident problem. To overcome this issue, we elucidated the mechanism underlying collective cellular detachment by combining a new computational simulation method with quantitative tissue-culture experiments. We first quantified the stochastic processes of cellular detachment shown by vascular smooth muscle cells on model curved scaffolds and found that microtissue morphologies vary drastically depending on cell contractility, substrate curvature, and cell-substrate adhesion strength. To explore this mechanism, we developed a new particle-based model that explicitly describes stochastic processes of multicellular dynamics, such as adhesion, rupture, and large deformation of microtissues on structured surfaces. Computational simulations using the developed model successfully reproduced characteristic detachment processes observed in experiments. Crucially, simulations revealed that cellular contractility-induced stress is locally concentrated at the cell-substrate interface, subsequently inducing a catastrophic process of collective cellular detachment, which can be suppressed by modulating cell contractility, substrate curvature, and cell-substrate adhesion. These results show that the developed computational method is useful for predicting engineered tissue dynamics as a platform for prediction-guided scaffold design. STATEMENT OF SIGNIFICANCE: Microfabrication technologies aiming to control multicellular dynamics by engineering 3D scaffolds are attracting increasing attention for modelling in cell biology and regenerative medicine. However, obtaining microtissues with the desired 3D structures is made considerably more difficult by microtissue detachments from scaffolds. This study reveals a key mechanism behind this detachment by developing a novel computational method for simulating multicellular dynamics on designed scaffolds. This method enabled us to predict microtissue dynamics on structured surfaces, based on cell mechanics, substrate geometry, and cell-substrate interaction. This study provides a platform for the physics-based design of micro-engineered scaffolds and thus contributes to prediction-guided biomaterials design in the future.


Asunto(s)
Miocitos del Músculo Liso , Ingeniería de Tejidos , Ingeniería de Tejidos/métodos , Adhesión Celular , Microtecnología , Andamios del Tejido/química
9.
Tissue Eng Part A ; 28(13-14): 573-585, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34841881

RESUMEN

Glioblastoma (GBM) is the most common and lethal type of malignant primary brain tumor in adults. GBM displays heterogeneous tumor cell population comprising glioma-initiating cells (GICs) with stem cell-like characteristics and differentiated glioma cells. During GBM cell invasion into normal brain tissues, which is the hallmark characteristic of GBM, GICs at the invasion front retain stemness, while cells at the tumor core display cellular differentiation. However, the mechanism of cellular differentiation underlying the formation of spatial cellular heterogeneity in GBM remains unknown. In the present study, we first observed spatially heterogeneous GBM cell populations emerged from an isogenic clonal population of GICs during invasion into a 3D collagen hydrogel in a microfluidic device. Specifically, GICs at the invasion front maintained stemness, while trailing cells displayed astrocytic differentiation. The spatial cellular heterogeneity resulted from the difference in cell density between GICs at the invasion front and trailing cells. Trailing GICs at high cell density exhibited astrocytic differentiation through local accumulation of paracrine factors they secreted, while cells at the invasion front of low cell density retained stemness due to the lack of paracrine factors. In addition, we demonstrated that interstitial flow suppressed astrocytic differentiation of trailing GICs by the clearance of paracrine factors. Our findings suggest that intercellular crosstalk between tumor cells is an essential factor in developing the spatial cellular heterogeneity of GBM cells with various differentiation statuses. It also provides insights into the development of novel therapeutic strategies targeting GBM cells with stem cell characteristics at the invasion front. Impact Statement We elucidated the mechanism of cellular differentiation underlying the spatial cellular heterogeneity of glioblastoma composed of glioma-initiating cells (GICs) and differentiated glioma cells during invasion in a microfluidic device. Trailing cells at high cell density exhibited astrocytic differentiation through local accumulation of paracrine factors they produced, while cells at the invasion front of low cell density were shown to retain stemness due to the lack of paracrine factors. Our findings provide valuable knowledge for the development of effective therapeutic strategies targeting GICs at the invasion front.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Línea Celular Tumoral , Glioma/patología , Humanos , Células Madre Neoplásicas/patología , Comunicación Paracrina
10.
Methods Mol Biol ; 2206: 57-66, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32754811

RESUMEN

The construction of vascular networks is essential for developing functional organ/tissue constructs in terms of oxygen and nutrient supply. Although recent advances in microfluidic techniques have allowed for the construction of microvascular networks using microfluidic devices, their structures cannot be maintained for extended periods of time due to a lack of perivascular cells. To construct long-lasting microvascular networks, it is important that perivascular cells are present to provide structural support to vessels, because in vivo microvessels are covered by perivascular cells and stabilized. Here, we describe a microfluidic cell culture platform for the construction of microvascular networks with supportive perivascular cells. Our results showed that microvascular networks covered by pericyte-like perivascular cells formed in a microfluidic device and their structures were maintained for at least 3 weeks in vitro.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/citología , Células Madre Mesenquimatosas/citología , Microfluídica/métodos , Células Cultivadas , Técnicas de Cocultivo/métodos , Humanos , Microvasos/citología , Pericitos/citología
11.
Tissue Eng Part A ; 27(7-8): 467-478, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33403936

RESUMEN

Glioblastoma (GBM) is the most common and lethal type of malignant brain tumor. A deeper mechanistic understanding of the invasion of heterogeneous GBM cell populations is crucial to develop therapeutic strategies. A key regulator of GBM cell invasion is interstitial flow. However, the effect of an interstitial flow on the invasion of heterogeneous GBM cell populations composed of glioma initiating cells (GICs) and relatively differentiated progeny cells remains unclear. In the present study, we investigated how GICs invade three-dimensional (3D) hydrogels in response to an interstitial flow with respect to their differentiation status. Microfluidic culture systems were used to apply an interstitial flow to the cells migrating from the cell aggregates into the 3D hydrogel. Phase-contrast microscopy revealed that the invasion and protrusion formation of the GICs in differentiated cell conditions were significantly enhanced by a forward interstitial flow, whose direction was the same as that of the cell invasion, whereas those in stem cell conditions were not enhanced by the interstitial flow. The mechanism of flow-induced invasion was further investigated by focusing on differentiated cell conditions. Immunofluorescence images revealed that the expression of cell-extracellular matrix adhesion-associated molecules, such as integrin ß1, focal adhesion kinase, and phosphorylated Src, was upregulated in forward interstitial flow conditions. We then confirmed that cell invasion and protrusion formation were significantly inhibited by PP2, a Src inhibitor. Finally, we observed that the flow-induced cell invasion was preceded by nestin-positive immature GICs at the invasion front and followed by tubulin ß3-positive differentiated cells. Our findings provide insights into the development of novel therapeutic strategies to inhibit flow-induced glioma invasion. Impact statement A mechanistic understanding of heterogeneous glioblastoma cell invasion is crucial for developing therapeutic strategies. We observed that the invasion and protrusion formation of glioma initiating cells (GICs) were significantly enhanced by forward interstitial flow in differentiated cell conditions. The expression of integrin ß1, focal adhesion kinase, and phosphorylated Src was upregulated, and the flow-induced invasion was significantly inhibited by a Src inhibitor. The flow-induced heterogeneous cell invasion was preceded by nestin-positive GICs at the invasion front and followed by tubulin ß3-positive differentiated cells. Our findings provide insights into the development of novel therapeutic strategies to inhibit flow-induced glioma invasion.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Diferenciación Celular , Línea Celular Tumoral , Humanos , Células Madre Neoplásicas
12.
Nat Commun ; 12(1): 3390, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099675

RESUMEN

In the liver, the bile canaliculi of hepatocytes are connected to intrahepatic bile ducts lined with cholangiocytes, which remove cytotoxic bile from the liver tissue. Although liver organoids have been reported, it is not clear whether the functional connection between hepatocytes and cholangiocytes is recapitulated in those organoids. Here, we report the generation of a hepatobiliary tubular organoid (HBTO) using mouse hepatocyte progenitors and cholangiocytes. Hepatocytes form the bile canalicular network and secrete metabolites into the canaliculi, which are then transported into the biliary tubular structure. Hepatocytes in HBTO acquire and maintain metabolic functions including albumin secretion and cytochrome P450 activities, over the long term. In this study, we establish functional liver tissue incorporating a bile drainage system ex vivo. HBTO enable us to reproduce the transport of hepatocyte metabolites in liver tissue, and to investigate the way in which the two types of epithelial cells establish functional connections.


Asunto(s)
Conductos Biliares Intrahepáticos/citología , Comunicación Celular/fisiología , Hígado/citología , Organoides/fisiología , Cultivo Primario de Células/métodos , Animales , Conductos Biliares Intrahepáticos/fisiología , Diferenciación Celular , Células Cultivadas , Hepatocitos/fisiología , Hígado/fisiología , Ratones , Organoides/citología , Células Madre/fisiología
13.
FASEB J ; 23(7): 2155-64, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19246488

RESUMEN

Increasing interest has focused on capturing the complexity of tissues and organs in vitro as models of human pathophysiological processes. In particular, a need exists for a model that can investigate the interactions in three dimensions (3D) between epithelial tissues and a microvascular network since vascularization is vital for reconstructing functional tissues in vitro. Here, we implement a microfluidic platform to analyze angiogenesis in 3D cultures of rat primary hepatocytes and rat/human microvascular endothelial cells (rMVECs/hMVECs). Liver and vascular cells were cultured on each sidewall of a collagen gel scaffold between two microfluidic channels under static or flow conditions. Morphogenesis of 3D hepatocyte cultures was found to depend on diffusion and convection across the nascent tissue. Furthermore, rMVECs formed 3D capillary-like structures that extended across an intervening gel to the hepatocyte tissues in hepatocyte-rMVEC coculture while they formed 2D sheet-like structures in rMVEC monoculture. In addition, diffusion of fluorescent dextran across the gel scaffold was analyzed, demonstrating that secreted proteins from the hepatocytes and MVECs can be exchanged across the gel scaffold by diffusional transport. The experimental approach described here is useful more generally for investigating microvascular networks within 3D engineered tissues with multiple cell types in vitro.


Asunto(s)
Endotelio Vascular/citología , Células Epiteliales/citología , Hepatocitos/citología , Neovascularización Fisiológica , Comunicación Paracrina , Animales , Transporte Biológico , Técnicas de Cultivo de Célula , Técnicas de Cocultivo , Humanos , Proteínas/metabolismo , Ratas
14.
Anal Sci ; 36(8): 1015-1019, 2020 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-32201406

RESUMEN

We report on a novel microdevice to tune the curvature of a cell-adhering surface by controlling the air-pressure and micro-slit. Human aortic smooth muscle cells were cultured on demi-cylindrical concaves formed on a microdevice. Their shape-adapting behavior could be tracked when the groove direction was changed to the orthogonal direction. This microdevice demonstrated live observation of cells responding to dynamic changes of the anisotropic curvature of the adhering surface and could serve as a new platform to pursue mechanobiology on curved surfaces.


Asunto(s)
Presión del Aire , Técnicas de Cultivo de Célula/instrumentación , Dispositivos Laboratorio en un Chip , Anisotropía , Aorta , Calibración , Adhesión Celular , Diseño de Equipo , Humanos , Miocitos del Músculo Liso/citología
15.
Biofabrication ; 12(4): 045008, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32644945

RESUMEN

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.


Asunto(s)
Vasos Sanguíneos/anatomía & histología , Vasos Sanguíneos/fisiología , Hemorreología , Neovascularización Fisiológica , Remodelación Vascular , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/enzimología , Dextranos/química , Fluorescencia , Hemorreología/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Hidrodinámica , Metaloproteinasa 9 de la Matriz/metabolismo , Microesferas , NG-Nitroarginina Metil Éster/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/farmacología , Perfusión , Factores de Tiempo , Remodelación Vascular/efectos de los fármacos
16.
Lab Chip ; 9(2): 269-75, 2009 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19107284

RESUMEN

Capillary morphogenesis is a complex cellular process that occurs in response to external stimuli. A number of assays have been used to study critical regulators of the process, but those assays are typically limited by the inability to control biochemical gradients and to obtain images on the single cell level. We have recently developed a new microfluidic platform that has the capability to control the biochemical and biomechanical forces within a three dimensional scaffold coupled with accessible image acquisition. Here, the developed platform is used to evaluate and quantify capillary growth and endothelial cell migration from an intact cell monolayer. We also evaluate the endothelial cell response when placed in co-culture with physiologically relevant cell types, including cancer cells and smooth muscle cells. This resulted in the following observations: cancer cells can either attract (MTLn3 cancer cell line) endothelial cells and induce capillary formation or have minimal effect (U87MG cancer cell line) while smooth muscle cells (10T 1/2) suppress endothelial activity. Results presented demonstrate the capabilities of this platform to study cellular morphogenesis both qualitatively and quantitatively while having the advantage of enhanced imaging and internal biological controls. Finally, the platform has numerous applications in the study of angiogenesis, or migration of other cell types including tumor cells, into a three-dimensional scaffold or across an endothelial layer under precisely controlled conditions of mechanical, biochemical and co-culture environments.


Asunto(s)
Movimiento Celular , Técnicas Analíticas Microfluídicas/métodos , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Técnicas de Cocultivo , Células Endoteliales/fisiología , Humanos , Técnicas Analíticas Microfluídicas/instrumentación , Ratas
17.
Am J Pathol ; 173(2): 494-506, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18583317

RESUMEN

Formation of bile ducts in culture is important for reconstructing hepatic organoids with bile drainage systems. However, morphogenic factors of biliary epithelial cells (BECs) have been poorly understood because of the lack of experimental models. Here, we demonstrated that rat BECs formed bile ductular networks in dynamic culture, when culture conditions were sequentially controlled. BEC morphogenesis was achieved through two-dimensional culture on collagen gel, collagen gel sandwich configuration, and 1% dimethylsulfoxide stimulation. In this culture system, BECs developed into large bile duct structures (LBDs) that formed interconnected networks of continuous lumens. LBD luminal surfaces possessed well developed microvilli, consisted of 7 to 10 BECs, and their inner diameters measured 20 to 50 microm. Quantitative PCR analysis revealed that the cells in LBDs expressed apical and basal domain markers of BECs. Immunofluorescent staining identified apical domain markers such as Cl(-)/HCO(3)(-) anion exchanger 2 and cystic fibrosis transmembrane regulator on the luminal surface of LBDs, responding to secretin stimulation as well as laminin protein surrounding LBDs. Furthermore, the cells in LBDs transported metabolized fluorescein from the basal side to the luminal space, further demonstrating that the reconstructed LBDs were functionally and morphologically similar to the bile ducts in vivo. The culture model described here will be useful in reconstructing hepatic tissues as well as in understanding the mechanism of bile duct development and its disruption in disease.


Asunto(s)
Conductos Biliares/ultraestructura , Polaridad Celular/fisiología , Colágeno/fisiología , Dimetilsulfóxido/farmacología , Células Epiteliales/ultraestructura , Animales , Conductos Biliares/efectos de los fármacos , Conductos Biliares/fisiología , Técnicas de Cultivo de Célula , Células Cultivadas , Antiportadores de Cloruro-Bicarbonato/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Matriz Extracelular/fisiología , Geles , Ratas , Ratas Sprague-Dawley , Secretina/farmacología
18.
Exp Cell Res ; 314(14): 2579-90, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18635166

RESUMEN

Laminins are a diverse group of alpha/beta/gamma heterotrimers formed from five alpha, three beta and three gamma chains; they are major components of all basal laminae (BLs). One laminin chain that has garnered particular interest due to its widespread expression pattern and importance during development is laminin alpha 5. Little is known, however, about the expression and function of laminins containing the alpha 5 chain in human hepatocellular carcinoma (HCC). Here, using a specific antibody, we examined the expression of laminin alpha 5 in normal liver and in HCCs. In normal liver, although laminin alpha 5 was observed in hepatic BLs underlying blood vessels and bile ducts, it was absent from the parenchyma, which may be the origin of HCC. On the other hand, laminin alpha 5 deposition was observed throughout all HCCs tested, regardless of tumor grade. In well-differentiated HCCs, it localized along the trabecules of the tumor. In poorly-differentiated HCCs, it was present in surrounding tumor nodules. In HCC cell lines, laminin alpha 5 heterotrimerized with beta and gamma chains and was secreted into the culture media. To attempt to understand the function of laminins containing alpha 5, the expression of its receptors in HCCs was also determined. In this regard, alpha 3 beta 1/alpha 6 beta 1 integrins and Lutheran/basal cell adhesion molecule (Lu/B-CAM) were expressed in HCC cells. In vitro studies showed that HCC cells readily attached to laminin containing the alpha 5 chain, more so than did primary hepatocytes. In addition to alpha 3 beta 1/alpha 6 beta 1 integrins and Lu/B-CAM, laminin alpha 5 was recognized by integrin alpha 1 beta 1, which also was expressed in HCC cells. These results suggest that laminins containing alpha 5 serve as functional substrates regulating progression of HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Moléculas de Adhesión Celular/metabolismo , Integrinas/metabolismo , Laminina/metabolismo , Neoplasias Hepáticas/patología , Proteínas de Neoplasias/metabolismo , Adulto , Anciano , Animales , Carcinoma Hepatocelular/metabolismo , Adhesión Celular , Femenino , Humanos , Inmunohistoquímica , Integrina alfa6beta4/metabolismo , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Sistema del Grupo Sanguíneo Lutheran , Masculino , Persona de Mediana Edad , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley
19.
Methods Mol Biol ; 1905: 167-174, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30536099

RESUMEN

Construction of three-dimensional (3D) hepatic tissue structures is important for in vitro tissue engineering of the liver, because 3D culture of hepatocytes is critical for the maintenance of liver-specific functions. Although conventional 3D culture methods are useful for constructing 3D hepatic tissue structures, the precise control of culture microenvironments is required to construct more physiological tissues in vitro. Recent advances in microfluidics technologies have allowed us to utilize microfluidic devices for hepatic cell culture, which opened the door for creating more physiological 3D culture models of the liver. Here, we describe the method for the construction of hepatic tissue structures using a microfluidic device which has a 3D gel region with adjacent microchannels. Primary rat hepatocytes are seeded into a microchannel in a microfluidic device. The cells are then cultured in interstitial flow conditions, which leads to the construction of 3D tissue structures.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Hepatocitos/citología , Animales , Diferenciación Celular , Células Cultivadas , Diseño de Equipo , Dispositivos Laboratorio en un Chip , Modelos Biológicos , Ratas , Ingeniería de Tejidos
20.
APL Bioeng ; 3(3): 036102, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31431938

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA