Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Toxicol In Vitro ; 93: 105688, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37660999

RESUMO

Despite the fact that biotransformation in the liver plays an important role in the augmented toxicity and detoxification of chemicals, relatively little efforts have been made to incorporate biotransformation into in vitro neurotoxicity testing. Conventional in vitro systems for neurotoxicity tests lack the capability of investigating the qualitative and quantitative differences between parent chemicals and their metabolites in the human body. Therefore, there is a need for an in vitro toxicity screening system that can incorporate hepatic biotransformation of chemicals and predict the susceptibility of their metabolites to induce neurotoxicity. To address this need, we adopted 3D cultures of metabolically competent HepaRG cell line with ReNcell VM and established a high-throughput, metabolism-mediated neurotoxicity testing system. Briefly, spheroids of HepaRG cells were generated in an ultralow attachment (ULA) 384-well plate while 3D-cultured ReNcell VM was established on a 384-pillar plate with sidewalls and slits (384PillarPlate). Metabolically sensitive test compounds were added in the ULA 384-well plate with HepaRG spheroids and coupled with 3D-cultured ReNcell VM on the 384PillarPlate, which allowed us to generate metabolites in situ by HepaRG cells and test them against neural stem cells. We envision that this approach could be potentially adopted in pharmaceutical and chemical industries when high-throughput screening (HTS) is necessary to assess neurotoxicity of compounds and their metabolites.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Neurais , Humanos , Hepatócitos/metabolismo , Células Cultivadas , Fígado/metabolismo , Esferoides Celulares
2.
FEBS Open Bio ; 13(7): 1266-1277, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37317692

RESUMO

While useful for fundamental in vitro studies, monolayer cell cultures are not physiologically relevant. Spheroids, a complex three-dimensional (3D) structure, more closely resemble in vivo tumor growth. Spheroids allow the results obtained relating to proliferation, cell death, differentiation, metabolism, and various antitumor therapies to be more predictive of in vivo outcomes. In the protocol herein, a rapid and high-throughput method is discussed for the generation of single spheroids using various cancer cell lines, including brain cancer cells (U87 MG, SEBTA-027, SF188), prostate cancer cells (DU-145, TRAMP-C1), and breast cancer cells (BT-549, Py230) in 96-round bottom-well plates. The proposed method is associated with significantly low costs per plate without requiring refining or transferring. Homogeneous compact spheroid morphology was evidenced as early as 1 day after following this protocol. Proliferating cells were traced in the rim, while dead cells were found to be located inside the core region of the spheroid using confocal microscopy and the Incucyte® live imaging system. H&E staining of spheroid sections was utilized to investigate the tightness of the cell packaging. Through western blotting analyses, it was revealed that a stem cell-like phenotype was adopted by these spheroids. This method was also used to obtain the EC50 of the anticancer dipeptide carnosine on U87 MG 3D culture. This affordable, easy-to-follow five-step protocol allows for the robust generation of various uniform spheroids with 3D morphology characteristics.


Assuntos
Neoplasias Encefálicas , Esferoides Celulares , Humanos , Análise Custo-Benefício , Encéfalo , Morte Celular
3.
Methods Mol Biol ; 2679: 127-139, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37300612

RESUMO

Microfluidic technologies allow the generation of large datasets using smaller quantities of cells and reagents than with traditional well plate assays. Such miniaturized methods can also facilitate the generation of complex 3D preclinical models of solid tumors with controlled size and cell composition. This is particularly useful in the context of recreating the tumor microenvironment for preclinical screening of immunotherapies and combination therapies at a scale, to reduce the experimental costs during therapy development while using physiologically relevant 3D tumor models, and to assess the therapy's efficacy. Here, we describe the fabrication of microfluidic devices and the associated protocols to culture tumor-stromal spheroids for assessing the efficacy of anticancer immunotherapies as monotherapies and as part of combination therapy regimes.


Assuntos
Microfluídica , Esferoides Celulares , Técnicas de Cocultura , Microfluídica/métodos , Linhagem Celular Tumoral , Microambiente Tumoral
4.
Curr Protoc ; 2(10): e569, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36286844

RESUMO

When compared to two-dimensional (2D) cell cultures, 3D spheroids have been considered suitable in vitro models for drug discovery research and other studies of drug activity. Based on different 3D cell culture procedures, we describe procedures we have used to obtain 3D tumor spheroids by both the hanging-drop and ultra-low-attachment plate methods and to analyze the antiproliferative and antitumor efficacy of different chemotherapeutic agents, including a peptidomimetic. We have applied this method to breast and lung cancer cell lines such as BT-474, MCF-7, A549, and Calu-3. We also describe a proximity ligation assay of the cells from the spheroid model to detect protein-protein interactions of EGFR and HER2. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Growth of 3D spheroids using the hanging-drop method Basic Protocol 2: Growth of spheroids using ultra-low-attachment plates Support Protocol 1: Cell viability assay of tumor spheroids Support Protocol 2: Antiproliferative and antitumor study in 3D tumor spheroids Support Protocol 3: Proximity ligation assay on cells derived from 3D spheroids.


Assuntos
Neoplasias Pulmonares , Peptidomiméticos , Humanos , Esferoides Celulares , Técnicas de Cultura de Células/métodos , Neoplasias Pulmonares/tratamento farmacológico , Receptores ErbB
5.
Small ; 18(29): e2202112, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35754160

RESUMO

In biomaterials R&D, conventional monolayer cell culture on flat/planar material samples, such as films, is still commonly employed at early stages of the assessment of interactions of cells with candidate materials considered for a biomedical application. In this feasibility study, an approach for the assessment of 3D cell-material interactions through dispersed coaggregation of microparticles from biomaterials into tissue spheroids is presented. Biomaterial microparticles can be created comparatively quickly and easily, allow the miniaturization of the assessment platform, and enable an unhindered remodeling of the dynamic cell-biomaterial system at any time. The aggregation of the microsized biomaterials and the cells is supported by low-attachment round-bottom microwells from thin polymer films arranged in densely packed arrays. The study is conducted by the example of MG63 osteoblast-like and human mesenchymal stem/stromal cells, and a small library of model microbiomaterials related to bone repair and regeneration. For the proof of concept, example interactions including cell adhesion to the material, the hybrid spheroids' morphology, size, and shape, material-associated cell death, cell metabolic activity, cell proliferation, and (osteogenic) differentiation are investigated. The cells in the spheroids are shown to respond to differences in the microbiomaterials' properties, their amounts, and the duration of interaction with them.


Assuntos
Materiais Biocompatíveis , Células-Tronco Mesenquimais , Materiais Biocompatíveis/metabolismo , Técnicas de Cultura de Células/métodos , Humanos , Osteogênese/fisiologia , Esferoides Celulares , Engenharia Tecidual/métodos
6.
Drug Deliv Transl Res ; 12(9): 2157-2177, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35763196

RESUMO

Nanobiomaterials, or NBMs, have been used in medicine and bioimaging for decades, with wide-reaching applications ranging from their uses as carriers of genes and drugs, to acting as sensors and probes. When developing nanomedicine products, it is vitally important to evaluate their safety, ensuring that both biocompatibility and efficacy are achieved so their applications in these areas can be safe and effective. When discussing the safety of nanomedicine in general terms, it is foolish to make generalised statements due to the vast array of different manufactured nanomaterials, formulated from a multitude of different materials, in many shapes and sizes; therefore, NBM pre-clinical screening can be a significant challenge. Outside of their distribution in the various tissues, organs and cells in the body, a key area of interest is the impact of NBMs on the liver. A considerable issue for researchers today is accurately predicting human-specific liver toxicity prior to clinical trials, with hepatotoxicity not only the most cited reasons for withdrawal of approved drugs, but also a primary cause of attrition in pre-launched drug candidates. To date, no simple solution to adequately predict these adverse effects exists prior to entering human experimentation. The limitations of the current pre-clinical toolkit are believed to be one of the main reasons for this, with questions being raised on the relevance of animal models in pre-clinical assessment, and over the ability of conventional, simplified in vitro cell-based assays to adequately assess new drug candidates or NBMs. Common 2D cell cultures are unable to adequately represent the functions of 3D tissues and their complex cell-cell and cell-matrix interactions, as well as differences found in diffusion and transport conditions. Therefore, testing NBM toxicity in conventional 2D models may not be an accurate reflection of the actual toxicity these materials impart on the body. One such method of overcoming these issues is the use of 3D cultures, such as cell spheroids, to more accurately assess NBM-tissue interaction. In this study, we introduce a 3D hepatocellular carcinoma model cultured from HepG2 cells to assess both the cytotoxicity and viability observed following treatment with a variety of NBMs, namely a nanostructured lipid carrier (in the specific technical name = LipImage™ 815), a gold nanoparticle (AuNP) and a panel of polymeric (in the specific technical name = PACA) NBMs. This model is also in compliance with the 3Rs policy of reduction, refinement and replacement in animal experimentation [1], and meets the critical need for more advanced in vitro models for pre-clinical nanotoxicity assessment. Pipeline for the pre-clinical assessment of NBMs in liver spheroid model.


Assuntos
Ouro , Nanopartículas Metálicas , Animais , Técnicas de Cultura de Células/métodos , Ouro/farmacologia , Humanos , Fígado , Esferoides Celulares
7.
J Fluoresc ; 32(2): 521-531, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34989923

RESUMO

Tumor spheroid models have proven useful in the study of cancer cell responses to chemotherapeutic compounds by more closely mimicking the 3-dimensional nature of tumors in situ. Their advantages are often offset, however, by protocols that are long, complicated, and expensive. Efforts continue for the development of high-throughput assays that combine the advantages of 3D models with the convenience and simplicity of traditional 2D monolayer methods. Herein, we describe the development of a breast cancer spheroid image cytometry assay using T47D cells in Aggrewell™400 spheroid plates. Using the Celigo® automated imaging system, we developed a method to image and individually track thousands of spheroids within the Aggrewell™400 microwell plate over time. We demonstrate the use of calcein AM and propidium iodide staining to study the effects of known anti-cancer drugs Doxorubicin, Everolimus, Gemcitabine, Metformin, Paclitaxel and Tamoxifen. We use the image cytometry results to quantify the fluorescence of calcein AM and PI as well as spheroid size in a dose dependent manner for each of the drugs. We observe a dose-dependent reduction in spheroid size and find that it correlates well with the viability obtained from the CellTiter96® endpoint assay. The image cytometry method we demonstrate is a convenient and high-throughput drug-response assay for breast cancer spheroids under 400 µm in diameter, and may lay a foundation for investigating other three-dimensional spheroids, organoids, and tissue samples.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Ensaios de Triagem em Larga Escala/métodos , Citometria por Imagem/métodos , Esferoides Celulares/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Fluoresceínas , Corantes Fluorescentes , Humanos , Propídio
8.
Biotechnol Bioeng ; 118(12): 4687-4698, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34478150

RESUMO

Drug-induced liver injury (DILI) is a leading cause of therapy failure in the clinic and also contributes much to acute liver failure cases. Investigations of predictive sensitivity in animal models have limitations due to interspecies differences. Previously reported in vitro models of liver injury based on primary human hepatocytes (PHHs) cannot meet the requirements of high physiological fidelity, low cost, simple operation, and high throughput with improved sensitivity. Herein, we developed an integrated biomimetic array chip (iBAC) for establishing extracellular matrix (ECM)-based models. A collagen-based 3D PHH model was constructed on the iBAC as a case for the prediction of clinical DILI at throughput. The iBAC has a three-layer structure with a core component of 3D implanting holes. At an initial cell seeding numbers of 5000-10,000, the collagen-based 3D PHH model was optimized with improved and stabilized liver functionality, including cell viability, albumin, and urea production. Moreover, basal activities of most metabolic enzymes on the iBAC were maintained for at least 12 days. Next, a small-scale hepatotoxicity screening indicated that the 3D PHH model on the iBAC was more sensitive for predicting hepatotoxicity than the 2D PHH model on the plate. Finally, a large-scale screening of liver toxicity using 122 clinical drugs further demonstrated that the collagen-based 3D PHH model on the iBAC had superior predictive sensitivity compared to all previously reported in vitro models. These results indicated the importance of 3D collagen for liver physiological functionality and hepatotoxicity prediction. We anticipant it being a promising tool for risk assessment of drug-induced hepatotoxicity with a widespread acceptance in drug industry.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Hepatócitos , Dispositivos Lab-On-A-Chip , Modelos Biológicos , Biomimética , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/instrumentação , Avaliação Pré-Clínica de Medicamentos/métodos , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos
9.
Commun Biol ; 4(1): 1001, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34429505

RESUMO

Microphysiological in vitro systems are platforms for preclinical evaluation of drug effects and significant advances have been made in recent years. However, existing microfluidic devices are not yet able to deliver compounds to cell models in a way that reproduces the real physiological drug exposure. Here, we introduce a novel tumour-on-chip microfluidic system that mimics the pharmacokinetic profile of compounds on 3D tumour spheroids to evaluate their response to the treatments. We used this platform to test the response of SW620 colorectal cancer spheroids to irinotecan (SN38) alone and in combination with the ATM inhibitor AZD0156, using concentrations mimicking mouse plasma exposure profiles of both agents. We explored spheroid volume and viability as a measure of cancer cells response and changes in mechanistically relevant pharmacodynamic biomarkers (γH2AX, cleaved-caspase 3 and Ki67). We demonstrate here that our microfluidic tumour-on-chip platform can successfully predict the efficacy from in vivo studies and therefore represents an innovative tool to guide drug dose and schedules for optimal efficacy and pharmacodynamic assessment, while reducing the need for animal studies.


Assuntos
Antineoplásicos/farmacocinética , Irinotecano/farmacocinética , Piridinas/farmacocinética , Quinolinas/farmacocinética , Linhagem Celular Tumoral , Humanos , Técnicas Analíticas Microfluídicas , Esferoides Celulares
10.
Clin Transl Sci ; 14(5): 1659-1680, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33982436

RESUMO

Nonclinical testing has served as a foundation for evaluating potential risks and effectiveness of investigational new drugs in humans. However, the current two-dimensional (2D) in vitro cell culture systems cannot accurately depict and simulate the rich environment and complex processes observed in vivo, whereas animal studies present significant drawbacks with inherited species-specific differences and low throughput for increased demands. To improve the nonclinical prediction of drug safety and efficacy, researchers continue to develop novel models to evaluate and promote the use of improved cell- and organ-based assays for more accurate representation of human susceptibility to drug response. Among others, the three-dimensional (3D) cell culture models present physiologically relevant cellular microenvironment and offer great promise for assessing drug disposition and pharmacokinetics (PKs) that influence drug safety and efficacy from an early stage of drug development. Currently, there are numerous different types of 3D culture systems, from simple spheroids to more complicated organoids and organs-on-chips, and from single-cell type static 3D models to cell co-culture 3D models equipped with microfluidic flow control as well as hybrid 3D systems that combine 2D culture with biomedical microelectromechanical systems. This article reviews the current application and challenges of 3D culture systems in drug PKs, safety, and efficacy assessment, and provides a focused discussion and regulatory perspectives on the liver-, intestine-, kidney-, and neuron-based 3D cellular models.


Assuntos
Alternativas ao Uso de Animais/métodos , Técnicas de Cultura de Células em Três Dimensões , Avaliação Pré-Clínica de Medicamentos/métodos , Alternativas ao Uso de Animais/normas , Células Cultivadas , Técnicas de Cocultura , Avaliação Pré-Clínica de Medicamentos/normas , Humanos , Intestinos/citologia , Rim/citologia , Fígado/citologia , Neurônios , Esferoides Celulares , Testes de Toxicidade/métodos , Testes de Toxicidade/normas , Estados Unidos , United States Food and Drug Administration/normas
11.
Int J Radiat Biol ; 97(5): 695-703, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33617430

RESUMO

PURPOSE: The present study investigated the biological effects of spot scanning and passive scattering proton therapies at the distal end region of the spread-out Bragg peak (SOBP) using single cell and multicell spheroids. MATERIALS AND METHODS: The Geant4 Monte Carlo simulation was used to calculate linear energy transfer (LET) values in passive scattering and spot scanning beams. The biological doses of the two beam options at various points of the distal end region of SOBP were investigated using EMT6 single cells and 0.6-mm V79 spheroids irradiated with 6 and 15 Gy, respectively, by inserting the fractions surviving these doses onto dose-survival curves and reading the corresponding dose. RESULTS: LET values in the entrance region of SOBP were similar between the two beam options and increased at the distal end region of SOBP, where the LET value of spot scanning beams was higher than that of passive scattering beams. Increases in biological effects at the distal end region were similarly observed in single cells and spheroids; biological doses at 2-10 mm behind the distal end were 4.5-57% and 5.7-86% higher than physical doses in passive scattering and spot scanning beams, respectively, with the biological doses of spot scanning beams being higher than those of passive scattering beams (p < .05). CONCLUSIONS: In single cells and spheroids, the effects of proton irradiation were stronger than expected from measured physical doses at the distal end of SOBP and were correlated with LET increases.


Assuntos
Prótons , Espalhamento de Radiação , Esferoides Celulares/efeitos da radiação , Linhagem Celular , Transferência Linear de Energia , Método de Monte Carlo , Eficiência Biológica Relativa , Análise de Célula Única , Esferoides Celulares/citologia
12.
Phys Med ; 77: 194-203, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32882615

RESUMO

PURPOSE: To develop an on-lattice agent-based model describing the growth of multicellular tumor spheroids using simple Monte Carlo tools. METHODS: Cells are situated on the vertices of a cubic grid. Different cell states (proliferative, hypoxic or dead) and cell evolution rules, driven by 10 parameters, and the effects of the culture medium are included. About twenty spheroids of MCF-7 human breast cancer were cultivated and the experimental data were used for tuning the model parameters. RESULTS: Simulated spheroids showed adequate sizes of the necrotic nuclei and of the hypoxic and proliferative cell phases as a function of the growth time, mimicking the overall characteristics of the experimental spheroids. The relation between the radii of the necrotic nucleus and the whole spheroid obtained in the simulations was similar to the experimental one and the number of cells, as a function of the spheroid volume, was well reproduced. The statistical variability of the Monte Carlo model described the whole volume range observed for the experimental spheroids. Assuming that the model parameters vary within Gaussian distributions it was obtained a sample of spheroids that reproduced much better the experimental findings. CONCLUSIONS: The model developed allows describing the growth of in vitro multicellular spheroids and the experimental variability can be well reproduced. Its flexibility permits to vary both the agents involved and the rules that govern the spheroid growth. More general situations, such as, e. g., tumor vascularization, radiotherapy effects on solid tumors, or the validity of the tumor growth mathematical models can be studied.


Assuntos
Neoplasias da Mama , Esferoides Celulares , Feminino , Humanos , Cinética , Método de Monte Carlo , Necrose
13.
Eur J Cell Biol ; 99(5): 151095, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32646644

RESUMO

Differently of two-dimensional cell culture, three-dimensional (3D) multicellular spheroid model allows cells to establish cell-cell/cell-matrix interactions over the entire cell surface, more closely mimicking tumor microenvironments and cellular subpopulations with specific standards of morphology, differentiation and gene expression. Thenceforth several methodologies involving or the 3D cell aggregates generation or its histological processing and analysis have emerged, but in general they are laborious, expensive and complex to set up as a routine technique. Thus, we developed a complete methodology, detailing a simple, accessible and low-cost step by step, including 1) the 3D cell aggregate generation using hanging drop technique; 2) providing a simple way to assess morphological parameters of generated spheroids; followed by 3) a multiple and organized histological processing, keeping several individual spheroids inside an agarose apparatus, maintaining a known order and position of each ones, similar to tissue microarray principle; 4) until the last step, where it is allowed a simultaneous histological composition analysis of several spheroid slices, organized side by side, in a same block section, through conventional stainings or 5) immunostaining against different molecular markers. Therefore, the present methodology aims to popularize 3D cell culture, allowing to make this a regular technique in basic cell biology research, once all steps are performed without using onerous reagents, materials or equipment. In addition to bring the agarose apparatus as a simple low cost novelty, allowing high-throughput analysis of several spheroids simultaneously in an organized manner.


Assuntos
Técnicas de Cultura de Células/métodos , Neoplasias/patologia , Esferoides Celulares/citologia , Células A549 , Técnicas de Cultura de Células/economia , Linhagem Celular Tumoral , Humanos , Esferoides Celulares/metabolismo
14.
Toxicol In Vitro ; 67: 104921, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32599260

RESUMO

Silibinin (Sil) is used as hepatoprotective drug and is approved for therapeutic use in amanitin poisoning. In our study we compared Sil-bis-succinate (SilBS), a water-soluble drug approved for i.v.-administration, with Sil solved in ethanol (SilEtOH), which is normally used in research. We challenged monocultures or 3D-microtissues consisting of HepG2 cells or primary hepatocytes with α-amanitin and treated with SILBS, SILEtOH, penicillin and combinations thereof. Cell viability and the integrity of the microtissues was monitored. Finally, the expression of the transporters OATP1B1 and B3 was analyzed by qRT-PCR. We demonstrated that primary hepatocytes were more sensitive to α-amanitin compared to HepG2. Primary hepatocytes cultures were protected by SilBS and SilEtOH independent of penicillin from the cytotoxic effects of α-amanitin. Subsequent studies of the expression profile of the transporters OATP1B1/B3 revealed that primary hepatocytes do express both whereas in HepG2 cells they were hardly detectable. Our study showed that SilBS has significant advantage over SilEtOH with no additional benefit of penicillin. Moreover, HepG2 cells may not represent an appropriate model to investigate Amanita phalloides poisoning in vitro with focus on OATP transporters since these cells are lacking sensitivity towards α-amanitin probably due to missing cytotoxicity-associated transporters suggesting that primary hepatocytes should be preferred in this context.


Assuntos
Alfa-Amanitina/toxicidade , Penicilinas/farmacologia , Substâncias Protetoras/farmacologia , Silibina/farmacologia , Alternativas aos Testes com Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Modelos Biológicos , Substâncias Protetoras/química , Silibina/química , Esferoides Celulares
15.
Methods Mol Biol ; 2110: 83-97, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32002903

RESUMO

Endothelial cells (EC) play a crucial role in the pathophysiology of cardiovascular diseases, ischemia/reperfusion injury, and graft rejection in (xeno-)transplantation. In such nonphysiological conditions, EC are known to lose their quiescent phenotype and switch into an actively pro-inflammatory, procoagulant, and anti-fibrinolytic state. This case happens essentially because the endothelial glycocalyx-a layer of proteoglycans and glycoproteins covering the luminal surface of the endothelium-is shed. Heparan sulfate, one of the main components of the endothelial glycocalyx, contributes to its negative charge. In addition, many plasma proteins such as antithrombin III, superoxide dismutase, C1 inhibitor, and growth factors and cytokines bind to heparan sulfate and by this scenario contribute to the establishment of an anticoagulant and anti-inflammatory endothelial surface. Shedding of the glycocalyx results in a loss of plasma proteins from the endothelial surface, and this phenomenon causes the switch in phenotype. Particularly in xenotransplantation, both hyperacute and acute vascular rejection are characterized by coagulation dysregulation, a situation in which EC are the main players.Since many years, EC have been used in vitro in 2D flatbed cell culture models, with or without the application of shear stress. Such models have also been used to assess the effect of human transgenes on complement- and coagulation-mediated damage of porcine EC in the context of xenotransplantation. The methods described in this chapter include the analysis of endothelial cell-blood interactions without the necessity of using anticoagulants as the increased EC surface-to-volume ratio allows for natural anticoagulation of blood. Furthermore, this chapter contains the description of a novel microfluidic in vitro model carrying important features of small blood vessels, such as a 3D round-section geometry, shear stress, and pulsatile flow-all this in a closed circuit, recirculating system aiming at reproducing closely the in vivo situation in small vessels.


Assuntos
Anti-Inflamatórios/metabolismo , Anticoagulantes/metabolismo , Técnicas de Cultura de Células , Células Endoteliais/metabolismo , Animais , Bioensaio , Biomarcadores , Células Cultivadas , Imunofluorescência , Humanos , Microfluídica/métodos , Microesferas , Esferoides Celulares , Transplante Heterólogo
16.
Artigo em Inglês | MEDLINE | ID: mdl-31746269

RESUMO

Genotoxic compounds may be detoxified to non-genotoxic metabolites while many pro-carcinogens require metabolic activation to exert their genotoxicity in vivo. Standard genotoxicity assays were developed and utilized for risk assessment for over 40 years. Most of these assays are conducted in metabolically incompetent rodent or human cell lines. Deficient in normal metabolism and relying on exogenous metabolic activation systems, the current in vitro genotoxicity assays often have yielded high false positive rates, which trigger unnecessary and costly in vivo studies. Metabolically active cells such as hepatocytes have been recognized as a promising cell model in predicting genotoxicity of carcinogens in vivo. In recent years, significant advances in tissue culture and biological technologies provided new opportunities for using hepatocytes in genetic toxicology. This review encompasses published studies (both in vitro and in vivo) using hepatocytes for genotoxicity assessment. Findings from both standard and newly developed genotoxicity assays are summarized. Various liver cell models used for genotoxicity assessment are described, including the potential application of advanced liver cell models such as 3D spheroids, organoids, and engineered hepatocytes. An integrated strategy, that includes the use of human-based cells with enhanced biological relevance and throughput, and applying the quantitative analysis of data, may provide an approach for future genotoxicity risk assessment.


Assuntos
Carcinógenos/toxicidade , Fígado/citologia , Fígado/efeitos dos fármacos , Testes de Mutagenicidade/métodos , Mutagênicos/toxicidade , Animais , Células Cultivadas , Hepatócitos , Humanos , Organoides , Medição de Risco , Esferoides Celulares
17.
Biochem Biophys Res Commun ; 522(3): 736-742, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31787234

RESUMO

There is an increasing interest in studying the crosstalk between tumor-associated adipose tissue and tumor progression. In proximity to the primary site of kidney tumors, perinephric adipose tissue has direct contact with cancer cells when kidney cancer becomes invasive. To mimic the perinephric adipose tissue microenvironment, we applied the liquid overlay-based technique, which cost-effectively generated functional adipocyte spheroids using mesenchymal stem cells isolated from human perinephric adipose tissue. Thereafter, we co-cultured adipocyte spheroids with unpolarized macrophages and discovered an M2 phenotype skew in macrophages. Moreover, we discovered that, in the presence of adipocyte spheroids, M2 macrophages exhibited stronger invasive capacity than M1 macrophages. We further showed that the perinephric adipose tissue sampled from metastatic kidney cancer exhibited high expression of M2 macrophages. In conclusion, the liquid overlay-based technique can generate a novel three-dimensional platform enabling investigation of the interactions of adipocytes and other types of cells in a tumor microenvironment.


Assuntos
Adipócitos/citologia , Adipogenia , Tecido Adiposo/citologia , Técnicas de Cultura de Células/instrumentação , Células-Tronco Mesenquimais/citologia , Adipócitos/patologia , Tecido Adiposo/patologia , Técnicas de Cultura de Células/economia , Células Cultivadas , Microambiente Celular , Técnicas de Cocultura/economia , Técnicas de Cocultura/instrumentação , Humanos , Neoplasias Renais/patologia , Macrófagos/citologia , Macrófagos/patologia , Células-Tronco Mesenquimais/patologia , Esferoides Celulares/citologia , Esferoides Celulares/patologia , Células Tumorais Cultivadas
18.
Microsc Microanal ; 25(6): 1311-1322, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31571549

RESUMO

Spheroids-three-dimensional aggregates of cells grown from a cancer cell line-represent a model of living tissue for chemotherapy investigation. Distribution of chemotherapeutics in spheroid sections was determined using the matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI). Proliferating or apoptotic cells were immunohistochemically labeled and visualized by laser scanning confocal fluorescence microscopy (LSCM). Drug efficacy was evaluated by comparing coregistered MALDI MSI and LSCM data of drug-treated spheroids with LSCM only data of untreated control spheroids. We developed a fiducial-based workflow for coregistration of low-resolution MALDI MS with high-resolution LSCM images. To allow comparison of drug and cell distribution between the drug-treated and untreated spheroids of different shapes or diameters, we introduced a common diffusion-related coordinate, the distance from the spheroid boundary. In a procedure referred to as "peeling", we correlated average drug distribution at a certain distance with the average reduction in the affected cells between the untreated and the treated spheroids. This novel approach makes it possible to differentiate between peripheral cells that died due to therapy and the innermost cells which died naturally. Two novel algorithms-for MALDI MS image denoising and for weighting of MALDI MSI and LSCM data by the presence of cell nuclei-are also presented.


Assuntos
Antineoplásicos/farmacologia , Microscopia Confocal/métodos , Neoplasias/tratamento farmacológico , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Antineoplásicos/farmacocinética , Humanos , Modelos Teóricos , Esferoides Celulares/efeitos dos fármacos
19.
Eur Phys J E Soft Matter ; 42(8): 112, 2019 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-31456065

RESUMO

Computational models aiming at the spatio-temporal description of cancer evolution are a suitable framework for testing biological hypotheses from experimental data, and generating new ones. Building on our recent work (J. Theor. Biol. 389, 146 (2016)) we develop a 3D agent-based model, capable of tracking hundreds of thousands of interacting cells, over time scales ranging from seconds to years. Cell dynamics is driven by a Monte Carlo solver, incorporating partial differential equations to describe chemical pathways and the activation/repression of "genes", leading to the up- or down-regulation of specific cell markers. Each cell-agent of different kind (stem, cancer, stromal etc.) runs through its cycle, undergoes division, can exit to a dormant, senescent, necrotic state, or apoptosis, according to the inputs from its systemic network. The basic network at this stage describes glucose/oxygen/ATP cycling, and can be readily extended to cancer-cell specific markers. Eventual accumulation of chemical/radiation damage to each cell's DNA is described by a Markov chain of internal states, and by a damage-repair network, whose evolution is linked to the cell systemic network. Aimed at a direct comparison with experiments of tumorsphere growth from stem cells, the present model will allow to quantitatively study the role of transcription factors involved in the reprogramming and variable radio-resistance of simulated cancer-stem cells, evolving in a realistic computer simulation of a growing multicellular tumorsphere.


Assuntos
Carcinogênese/metabolismo , Evolução Clonal , Modelos Teóricos , Esferoides Celulares/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Dano ao DNA , Glucose/metabolismo , Humanos , Cadeias de Markov , Oxigênio/metabolismo , Esferoides Celulares/patologia , Células Tumorais Cultivadas
20.
Lab Chip ; 19(17): 2822-2833, 2019 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-31360969

RESUMO

The field of microfluidics-based three-dimensional (3D) cell culture system is rapidly progressing from academic proof-of-concept studies to valid solutions to real-world problems. Polydimethylsiloxane (PDMS)-based platform has been widely adopted as in vitro platforms for mimicking tumor microenvironment. However, PDMS has not been welcomed as a standardized commercial application for preclinical screening due to inherent material limitations that make it difficult to scale-up production. Here, we present an injection-molded plastic array 3D spheroid culture platform (Sphero-IMPACT). The platform is made of polystyrene (PS) in a standardized 96-well plate format with a user-friendly interface. This interface describes a simpler design that incorporates a tapered hole in the center of the rail to pattern a large spheroid with 3D extracellular matrix and various cell types. This hole is designed to accommodate standard pipette tip for automated system. The platform that mediate open microfluidics allows implement spontaneous fluid patterning with high repeatability from the end user. To demonstrate versatile use of the platform, we developed 3D perfusable blood vessel network and tumor spheroid assays. In addition, we established a tumor spheroid induced angiogenesis model that can be applicable for drug screening. Sphero-IMPACT has the potential to provide a robust and reproducible in vitro assay related to vascularized cancer research. This easy-to-use, ready-to-use platform can be translated into an enhanced preclinical model that faithfully reflects the complex tumor microenvironment.


Assuntos
Técnicas de Cultura de Células/normas , Glioblastoma/patologia , Técnicas Analíticas Microfluídicas/normas , Neovascularização Patológica/patologia , Esferoides Celulares/patologia , Técnicas de Cultura de Células/economia , Técnicas de Cultura de Células/instrumentação , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Técnicas Analíticas Microfluídicas/economia , Técnicas Analíticas Microfluídicas/instrumentação , Padrões de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA