Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biotechnol Bioeng ; 117(3): 763-775, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31736056

RESUMO

Oxygen is vital to the function of all tissues including the liver and lack of oxygen, that is, hypoxia can result in both acute and chronic injuries to the liver in vivo and ex vivo. Furthermore, a permanent oxygen gradient is naturally present along the liver sinusoid, which plays a role in the metabolic zonation and the pathophysiology of liver diseases. Accordingly, here, we introduce an in vitro microfluidic platform capable of actively creating a series of oxygen concentrations on a single continuous microtissue, ranging from normoxia to severe hypoxia. This range approximately captures both the physiologically relevant oxygen gradient generated from the portal vein to the central vein in the liver, and the severe hypoxia occurring in ischemia and liver diseases. Primary rat hepatocytes cultured in this microfluidic platform were exposed to an oxygen gradient of 0.3-6.9%. The establishment of an ascending hypoxia gradient in hepatocytes was confirmed in response to the decreasing oxygen supply. The hepatocyte viability in this platform decreased to approximately 80% along the hypoxia gradient. Simultaneously, a progressive increase in accumulation of reactive oxygen species and expression of hypoxia-inducible factor 1α was observed with increasing hypoxia. These results demonstrate the induction of distinct metabolic and genetic responses in hepatocytes upon exposure to an oxygen (/hypoxia) gradient. This progressive hypoxia-on-a-chip platform can be used to study the role of oxygen and hypoxia-associated molecules in modeling healthy and injured liver tissues. Its use can be further expanded to the study of other hypoxic tissues such as tumors as well as the investigation of drug toxicity and efficacy under oxygen-limited conditions.


Assuntos
Hepatócitos/metabolismo , Hipóxia/metabolismo , Dispositivos Lab-On-A-Chip , Hepatopatias/metabolismo , Oxigênio/metabolismo , Animais , Células Cultivadas , Fígado/citologia , Fígado/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
2.
Biotechnol Bioeng ; 112(12): 2571-82, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25994312

RESUMO

We describe the generation of microfluidic platforms for the co-culture of primary hepatocytes and endothelial cells; these platforms mimic the architecture of a liver sinusoid. This paper describes a progressional study of creating such a liver sinusoid on a chip system. Primary rat hepatocytes (PRHs) were co-cultured with primary or established endothelial cells in layers in single and dual microchannel configurations with or without continuous perfusion. Cell viability and maintenance of hepatocyte functions were monitored and compared for diverse experimental conditions. When primary rat hepatocytes were co-cultured with immortalized bovine aortic endothelial cells (BAECs) in a dual microchannel with continuous perfusion, hepatocytes maintained their normal morphology and continued to produce urea for at least 30 days. In order to demonstrate the utility of our microfluidic liver sinusoid platform, we also performed an analysis of viral replication for the hepatotropic hepatitis B virus (HBV). HBV replication, as measured by the presence of cell-secreted HBV DNA, was successfully detected. We believe that our liver model closely mimics the in vivo liver sinusoid and supports long-term primary liver cell culture. This liver model could be extended to diverse liver biology studies and liver-related disease research such as drug induced liver toxicology, cancer research, and analysis of pathological effects and replication strategies of various hepatotropic infectious agents. .


Assuntos
Células Endoteliais/fisiologia , Hepatócitos/fisiologia , Fígado Artificial , Microfluídica/métodos , Animais , Bovinos , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , DNA Viral/análise , Vírus da Hepatite B/fisiologia , Ratos , Fatores de Tempo , Ureia/metabolismo , Replicação Viral
3.
Sci Rep ; 9(1): 7377, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31089162

RESUMO

Poly(dimethylsiloxane) (PDMS) is likely the most popular material for microfluidic devices in lab-on-a-chip and other biomedical applications. However, the hydrophobicity of PDMS leads to non-specific adsorption of proteins and other molecules such as therapeutic drugs, limiting its broader use. Here, we introduce a simple method for preparing PDMS materials to improve hydrophilicity and decrease non-specific protein adsorption while retaining cellular biocompatibility, transparency, and good mechanical properties without the need for any post-cure surface treatment. This approach utilizes smart copolymers comprised of poly(ethylene glycol) (PEG) and PDMS segments (PDMS-PEG) that, when blended with PDMS during device manufacture, spontaneously segregate to surfaces in contact with aqueous solutions and reduce the hydrophobicity without any added manufacturing steps. PDMS-PEG-modified PDMS samples showed contact angles as low as 23.6° ± 1° and retained this hydrophilicity for at least twenty months. Their improved wettability was confirmed using capillary flow experiments. Modified devices exhibited considerably reduced non-specific adsorption of albumin, lysozyme, and immunoglobulin G. The modified PDMS was biocompatible, displaying no adverse effects when used in a simple liver-on-a-chip model using primary rat hepatocytes. This PDMS modification method can be further applied in analytical separations, biosensing, cell studies, and drug-related studies.


Assuntos
Dimetilpolisiloxanos/química , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/instrumentação , Polímeros Responsivos a Estímulos/química , Animais , Células Cultivadas , Hepatócitos , Interações Hidrofóbicas e Hidrofílicas , Teste de Materiais , Polietilenoglicóis/química , Cultura Primária de Células , Ratos , Propriedades de Superfície
4.
Lab Chip ; 19(18): 3022-3031, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31465069

RESUMO

Non-alcoholic fatty liver disease (NAFLD) and its progressive form non-alcoholic steatohepatitis (NASH) affect 25% of the world population. NAFLD is predicted to soon become the main cause of liver morbidity and transplantation. The disease is characterized by a progressive increase of lipid accumulation in hepatocytes, which eventually induce fibrosis and inflammation, and can ultimately cause cirrhosis and hepatic carcinoma. Here, we created a patterned model of NAFLD on a chip using free fatty acid gradients to recapitulate a spectrum of disease conditions in a single continuous liver tissue. We established the NAFLD progression via quantification of intracellular lipid accumulation and transcriptional levels of fatty acid transporters and NAFLD pathogenesis markers. We then used this platform to create oxygen driven steatosis zonation mimicking the sinusoidal lipid distribution on a single continuous tissue and showed that this fat zonation disappears under progressed steatosis, in agreement with in vivo observations and recent computational studies. While we focus on free fatty acids and oxygen as the drivers of NAFLD, the microfluidic platform here is extensible to simultaneous use of other drivers.


Assuntos
Técnicas Analíticas Microfluídicas , Modelos Biológicos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Oxigênio/metabolismo , Animais , Células Cultivadas , Progressão da Doença , Feminino , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Ratos , Ratos Endogâmicos Lew
5.
Sci Rep ; 8(1): 8951, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29895900

RESUMO

An important number of healthy and diseased tissues shows spatial variations in their metabolic capacities across the tissue. The liver is a prime example of such heterogeneity where the gradual changes in various metabolic activities across the liver sinusoid is termed as "zonation" of the liver. Here, we introduce the Metabolic Patterning on a Chip (MPOC) platform capable of dynamically creating metabolic patterns across the length of a microchamber of liver tissue via actively enforced gradients of various metabolic modulators such as hormones and inducers. Using this platform, we were able to create continuous liver tissues of both rat and human origin with gradually changing metabolic activities. The gradients we have created in nitrogen, carbohydrate and xenobiotic metabolisms recapitulated an in vivo like zonation and zonal toxic response. Beyond its application in recapitulation of liver zonation in vitro as we demonstrate here, the MPOC platform can be used and expanded for a variety of purposes including better understanding of heterogeneity in many different tissues during developmental and adult stages.


Assuntos
Metabolismo dos Carboidratos , Hepatócitos/metabolismo , Fígado/metabolismo , Xenobióticos/metabolismo , Animais , Feminino , Hepatócitos/citologia , Humanos , Fígado/citologia , Cultura Primária de Células , Ratos , Ratos Endogâmicos Lew
6.
Biofabrication ; 5(4): 045008, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24280542

RESUMO

This paper presents a novel liver model that mimics the liver sinusoid where most liver activities occur. A key aspect of our current liver model is a layered co-culture of primary rat hepatocytes (PRHs) and primary rat liver sinusoidal endothelial cells (LSECs) or bovine aortic endothelial cells (BAECs) on a transwell membrane. When a layered co-culture was attempted with a thin Matrigel layer placed between hepatocytes and endothelial cells to mimic the space of Disse, the cells did not form completely separated monolayers. However, when hepatocytes and endothelial cells were cultured on the opposite sides of a transwell membrane, PRHs co-cultured with LSECs or BAECs maintained their viability and normal morphology for 39 and 57 days, respectively. We assessed the presence of hepatocyte-specific differentiation markers to verify that PRHs remained differentiated in the long-term co-culture and analyzed hepatocyte function by monitoring urea synthesis. We also noted that the expression of cytochrome P-450 remained similar in the co-cultured system from day 1 to day 48. Thus, our novel liver model system demonstrated that primary hepatocytes can be cultured for extended times and retain their hepatocyte-specific functions when layered with endothelial cells.


Assuntos
Técnicas de Cocultura/instrumentação , Células Endoteliais/citologia , Hepatócitos/citologia , Modelos Biológicos , Engenharia Tecidual/instrumentação , Animais , Bovinos , Diferenciação Celular , Sobrevivência Celular , Técnicas de Cocultura/métodos , Citocromo P-450 CYP2E1/metabolismo , Fígado/citologia , Fígado/metabolismo , Membranas Artificiais , Ratos , Ratos Sprague-Dawley , Engenharia Tecidual/métodos
7.
Nanoscale Res Lett ; 8(1): 487, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24252646

RESUMO

While flow-induced voltage over a graphene layer has been reported, its origin remains unclear. In our previous study, we suggested different mechanisms for different experimental configurations: phonon dragging effect for the parallel alignment and an enhanced out-of-plane phonon mode for the perpendicular alignment (Appl. Phys. Lett. 102:063116, 2011). In order to further examine the origin of flow-induced voltage, we introduced a transverse flow component by integrating staggered herringbone grooves in the microchannel. We found that the flow-induced voltage decreased significantly in the presence of herringbone grooves in both parallel and perpendicular alignments. These results support our previous interpretation.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa