Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmaceuticals (Basel) ; 17(4)2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38675489

RESUMO

No standardized in vitro cell culture models for glioblastoma (GBM) have yet been established, excluding the traditional two-dimensional culture. GBM tumorspheres (TSs) have been highlighted as a good model platform for testing drug effects and characterizing specific features of GBM, but a detailed evaluation of their suitability and comparative performance is lacking. Here, we isolated GBM TSs and extracellular matrices (ECM) from tissues obtained from newly diagnosed IDH1 wild-type GBM patients and cultured GBM TSs on five different culture platforms: (1) ordinary TS culture liquid media (LM), (2) collagen-based three-dimensional (3D) matrix, (3) patient typical ECM-based 3D matrix, (4) patient tumor ECM-based 3D matrix, and (5) mouse brain. For evaluation, we obtained transcriptome data from all cultured GBM TSs using microarrays. The LM platform exhibited the most similar transcriptional program to paired tissues based on GBM genes, stemness- and invasiveness-related genes, transcription factor activity, and canonical signaling pathways. GBM TSs can be cultured via an easy-to-handle and cost- and time-efficient LM platform while preserving the transcriptional program of the originating tissues without supplementing the ECM or embedding it into the mouse brain. In addition to applications in basic cancer research, GBM TSs cultured in LM may also serve as patient avatars in drug screening and pre-clinical evaluation of targeted therapy and as standardized and clinically relevant models for precision medicine.

2.
ACS Biomater Sci Eng ; 6(2): 813-821, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33464850

RESUMO

Human iPSC-derived mesenchymal stem cells (iMSCs) are an alternative to primary mesenchymal stem cells (MSCs), which have been a limited supply, and have attracted a great deal of interest as a promising cell source in cell-based therapy. However, despite their enormous therapeutic potential, it has been difficult to translate this potential into clinical applications due to the short viability duration of transplanted iMSCs. Therefore, to maximize the therapeutic effects of iMSCs, it is extremely important to extend their retention rate during and even after the transplantation. In this study, we developed a new extracellular matrix (ECM)-coating method involving the mild reduction of the cell surface. The reduction of disulfide bonds around the cell membrane enhanced the coating efficiency without a decrease in the viability and differentiation potential of iMSCs. We then induced ECM-coated single iMSCs to form three-dimensional spheroids via self-assembly of the aggregates within a physically confined microenvironment. The spheroids exhibited longer maintenance of the survival rate. Nanometric ECM coating of the cell membrane is a new approach as a key for resolving the conventional challenges of cell-based therapy.


Assuntos
Matriz Extracelular , Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Diferenciação Celular , Humanos , Fosfinas
3.
Biomater Res ; 22: 32, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30323947

RESUMO

BACKGROUND: Human mesenchymal stem cells (hMSCs) are, due to their pluripotency, useful sources of cells for stem cell therapy and tissue regeneration. The phenotypes of hMSCs are strongly influenced by their microenvironment, in particular the extracellular matrix (ECM), the composition and structure of which are important in regulating stem cell fate. In reciprocal manner, the properties of ECM are remodeled by the hMSCs, but the mechanism involved in ECM remodeling by hMSCs under topographical stimulus is unclear. In this study, we therefore examined the effect of nanotopography on the expression of ECM proteins by hMSCs by analyzing the quantity and structure of the ECM on a nanogrooved surface. METHODS: To develop the nanoengineered, hMSC-derived ECM, we fabricated the nanogrooves on a coverglass using a UV-curable polyurethane acrylate (PUA). Then, hMSCs were cultivated on the nanogrooves, and the cells at the full confluency were decellularized. To analyze the effect of nanotopography on the hMSCs, the hMSCs were re-seeded on the nanoengineered, hMSC-derived ECM. RESULTS: hMSCs cultured within the nano-engineered hMSC-derived ECM sheet showed a different pattern of expression of ECM proteins from those cultured on ECM-free, nanogrooved surface. Moreover, hMSCs on the nano-engineered ECM sheet had a shorter vinculin length and were less well-aligned than those on the other surface. In addition, the expression pattern of ECM-related genes by hMSCs on the nanoengineered ECM sheet was altered. Interestingly, the expression of genes for osteogenesis-related ECM proteins was downregulated, while that of genes for chondrogenesis-related ECM proteins was upregulated, on the nanoengineered ECM sheet. CONCLUSIONS: The nanoengineered ECM influenced the phenotypic features of hMSCs, and that hMSCs can remodel their ECM microenvironment in the presence of a nanostructured ECM to guide differentiation into a specific lineage.

4.
Biomater Sci ; 6(10): 2704-2713, 2018 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-30151505

RESUMO

During gastric cancer (GC) progression, increased extracellular matrix (ECM) deposition, notably collagen type I, correlates with an overall increase in expression of the mesenchymal phenotype. In GC tissue, the intestinal epithelium exhibits impaired cell-cell adhesion and enhanced cell-ECM adhesion. The alteration of intercellular integrity is one of tumorigenesis feature including tumor invasion and metastasis. Using a density-varying ECM, we studied the effect of ECM density on both intercellular- and ECM-interactions according to alterations of ECM-mediated signaling. A dense collagen matrix increases integrin-mediated cell-ECM interactions with phosphorylated FAK and ERK signaling in human gastric adenocarcinoma cells (AGS, MKN74), which regulates GC proliferation and the chemotherapeutic response. In addition, GC cells exhibited a disrupted membranous E-cadherin/ß-catenin complex and, remarkably, showed cytoplasmic or nucleic localization of ß-catenin in response to collagen density. Furthermore, we found that membranous E-cadherin/ß-catenin complex could be recovered by inhibiting the phosphorylation of FAK, which in turn influences the chemotherapeutic effect. These results provide insight into how matrix density differentially regulates cancer cell phenotype and may have significant implications for the design of biomaterials with appropriate physical properties for in vitro tumor models.


Assuntos
Caderinas/metabolismo , Matriz Extracelular/metabolismo , Neoplasias Gástricas/metabolismo , beta Catenina/metabolismo , Antígenos CD , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/metabolismo , Humanos , Cadeias beta de Integrinas/metabolismo , Microambiente Tumoral
5.
Sci Rep ; 8(1): 4608, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29545552

RESUMO

Glioblastoma multiforme (GBM) is the most common brain tumor with very aggressive and infiltrative. Extracellular matrix (ECM) plays pivotal roles in the infiltrative characteristics of GBM. To understand the invasive characteristic of GBM, it is necessary to study cell-ECM interaction in the physiologically relevant biomimetic model that recapitulates the GBM-specific ECM microenvironment. Here, we propose biomimetic GBM-specific ECM microenvironment for studying mode and dynamics of glioblastoma cell invasion. Using tissue decellularization process, we constructed a patient tissue-derived ECM (pdECM)-based three-dimensional in vitro model. In our model, GBM cells exhibited heterogeneous morphology and altered the invasion routes in a microenvironment-adaptive manner. We further elucidate the effects of inhibition of ECM remodeling-related enzymatic activity (Matrix metalloproteinase (MMP) 2/9, hyaluronan synthase (HAS)) on GBM cell invasion. Interestingly, after blocking both enzyme activity, GBM cells underwent morphological transition and switch the invasion mode. Such adaptability could render cell invasion resistant to anti-cancer target therapy. There results provide insight of how organ-specific matrix differentially regulates cancer cell phenotype, and have significant implications for the design of matrix with appropriate physiologically relevant properties for in vitro tumor model.


Assuntos
Neoplasias Encefálicas/patologia , Movimento Celular , Matriz Extracelular/patologia , Glioblastoma/patologia , Microambiente Tumoral , Neoplasias Encefálicas/metabolismo , Técnicas de Cultura de Células , Proliferação de Células , Matriz Extracelular/metabolismo , Glioblastoma/metabolismo , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Células Tumorais Cultivadas
6.
Sci Rep ; 7: 41541, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128310

RESUMO

Gastric cancer (GC) is a common aggressive malignant tumor with high incidence and mortality worldwide. GC is classified into intestinal and diffuse types according to the histo-morphological features. Because of distinctly different clinico-pathological features, new cancer therapy strategies and in vitro preclinical models for the two pathological variants of GC is necessary. Since extracellular matrix (ECM) influence the biological behavior of tumor cells, we hypothesized that GC might be more similarly modeled in 3D with matrix rather than in 2D. Herein, we developed a microfluidic-based a three-dimensional (3D) in vitro gastric cancer model, with subsequent drug resistance assay. AGS (intestinal type) and Hs746T (diffuse type) gastric cancer cell lines were encapsulated in collagen beads with high cellular viability. AGS exhibited an aggregation pattern with expansive growth, whereas Hs746T showed single-cell-level infiltration. Importantly, in microtumor models, epithelial-mesenchymal transition (EMT) and metastatic genes were upregulated, whereas E-cadherin was downregulated. Expression of ß-catenin was decreased in drug-resistant cells, and chemosensitivity toward the anticancer drug (5-FU) was observed in microtumors. These results suggest that in vitro microtumor models may represent a biologically relevant platform for studying gastric cancer cell biology and tumorigenesis, and for accelerating the development of novel therapeutic targets.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Matriz Extracelular/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Matriz Extracelular/genética , Imunofluorescência , Humanos , Técnicas Analíticas Microfluídicas , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , beta Catenina/genética , beta Catenina/metabolismo
7.
ACS Nano ; 11(1): 730-741, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28051852

RESUMO

We present a method to induce cell directional behavior using slanted nanocilia arrays. NIH-3T3 fibroblasts demonstrated bidirectional polarization in a rectangular arrangement on vertical nanocilia arrays and exhibited a transition from a bidirectional to a unidirectional polarization pattern when the angle of the nanocilia was decreased from 90° to 30°. The slanted nanocilia guided and facilitated spreading by allowing the cells to contact the sidewalls of the nanocilia, and the directional migration of the cells opposed the direction of the slant due to the anisotropic bending stiffness of the slanted nanocilia. Although the cells recognized the underlying anisotropic geometry when the nanocilia were coated with fibronectin, collagen type I, and Matrigel, the cells lost their directionality when the nanocilia were coated with poly-d-lysine and poly-l-lysine. Furthermore, although the cells recognized geometrical anisotropy on fibronectin coatings, pharmacological perturbation of PI3K-Rac signaling hindered the directional elongation of the cells on both the slanted and vertical nanocilia. Furthermore, myosin light chain II was required for the cells to obtain polarized morphologies. These results indicated that the slanted nanocilia array provided anisotropic contact guidance cues to the interacting cells. The polarization of cells was controlled through two steps: the recognition of underlying geometrical anisotropy and the subsequent directional spreading according to the guidance cues.

8.
ACS Biomater Sci Eng ; 3(12): 3546-3552, 2017 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-33445389

RESUMO

The extracellular matrix (ECM) provides physical and chemical support to the surrounding cells. During cell growth, ECM secretion and network formation influence cell morphology, cell adhesion, cell-to-cell interactions, and cell migration. Microfluidics-based cell culture systems are limited by the integration of structural ECM into the device. We report the development of a cell-derived ECM-incorporated microfluidic device that can provide structural characteristics and biochemical components of cell-derived ECM. Using an on-chip decellularization process, we constructed an ECM sheet, secreted and deposited from monolayer-cultured mouse embryonic fibroblasts (NIH/3T3), inside the microfluidic device. ECM components (including collagens, fibronectin, laminin, and elastin) and mesh-type fibronectin fibrous architecture were maintained on the surface of the porous membrane of the microfluidic device after decellularization. To verify the usability of the fibroblast-derived ECM sheet integrated microfluidic device in a cell culture platform, we tested the recellularization of human umbilical vein endothelial cells (HUVEC) and analyzed HUVEC-ECM and HUVEC-HUVEC interactions. On the ECM sheet, HUVECs exhibited morphologies and focal adhesion features that were markedly different from those of other groups. We then explored the effect of the ECM sheet on HUVEC mechanosensitivity. An increase in fluid shear stresses led to focal adhesion and the polymerization and reorganization of HUVEC adherens junctions, similar to natural junctional development, whereas the control group exhibited stimuli-insensitive behaviors. We conclude that the decellularized ECM sheet-incorporated microfluidic device provides an in vivo-like physical and biochemical ECM microenvironment for microfluidics-based cell culture.

9.
Neuro Oncol ; 19(2): 197-207, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27571886

RESUMO

Background: Deprivation of tumor bioenergetics by inhibition of multiple energy pathways has been suggested as an effective therapeutic approach for various human tumors. However, this idea has not been evaluated in glioblastoma (GBM). We hypothesized that dual inhibition of glycolysis and oxidative phosphorylation could effectively suppress GBM tumorspheres (TS). Methods: Effects of 2-deoxyglucose (2DG) and metformin, alone and in combination, on GBM-TS were evaluated. Viability, cellular energy metabolism status, stemness, invasive properties, and GBM-TS transcriptomes were examined. In vivo efficacy was tested in a mouse orthotopic xenograft model. Results: GBM-TS viability was decreased by the combination of 2DG and metformin. ATP assay and PET showed that cellular energy metabolism was also decreased by this combination. Sphere formation, expression of stemness-related proteins, and invasive capacity of GBM-TS were also significantly suppressed by combined treatment with 2DG and metformin. A transcriptome analysis showed that the expression levels of stemness- and epithelial mesenchymal transition-related genes were also significantly downregulated by combination of 2DG and metformin. Combination treatment also prolonged survival of tumor-bearing mice and decreased invasiveness of GBM-TS. Conclusion: The combination of 2DG and metformin effectively decreased the stemness and invasive properties of GBM-TS and showed a potential survival benefit in a mouse orthotopic xenograft model. Our findings suggest that targeting TS-forming cells by this dual inhibition of cellular bioenergetics warrants expedited clinical evaluation for the treatment of GBM.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Desoxiglucose/farmacologia , Glioblastoma/tratamento farmacológico , Metformina/farmacologia , Animais , Antimetabólitos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Metabolismo Energético/efeitos dos fármacos , Glioblastoma/metabolismo , Glioblastoma/patologia , Glicólise/efeitos dos fármacos , Humanos , Hipoglicemiantes/farmacologia , Camundongos , Camundongos Nus , Fosforilação Oxidativa/efeitos dos fármacos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncotarget ; 7(40): 65643-65659, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27582539

RESUMO

Studies have investigated biguanide-derived agents for the treatment of cancers and have reported their effects against tumorspheres (TSs). The purpose of this study was determining the effects of HL156A, a newly designed biguanide with improved pharmacokinetics, on glioblastoma TSs (GMB TSs) and assess the feasibility of this drug as a new line of therapy against glioblastoma, alone or combined with a conventional therapeutic agent, temozolomide(TMZ). The effects of HL156A, alone and combined with TMZ, on the stemness and invasive properties of GBM TSs and survival of orthotopic xenograft animals were assessed. HL156A, combined with TMZ, inhibited the stemness of GBM TSs, proven by neurosphere formation assay and marker expression. Three-dimensional collagen matrix invasion assays provided evidence that combined treatment inhibited invasive properties, compared with control and TMZ-alone treatment groups. TMZ alone and combined treatment repressed the expression of epithelial-mesenchymal transition-related genes. A gene ontology comparison of TMZ and combination-treatment groups revealed altered expression of genes encoding proteins involved in cellular adhesion and migration. Combined treatment with HL156A and TMZ showed survival benefits in an orthotopic xenograft mouse model. The inhibitory effect of combination treatment on the stemness and invasive properties of GBM TSs suggest the potential usage of this regimen as a novel strategy for the treatment of GBM.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/patologia , Dacarbazina/análogos & derivados , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/patologia , Guanidinas/farmacologia , Células-Tronco Neoplásicas/patologia , Pirrolidinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Dacarbazina/farmacologia , Quimioterapia Combinada , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Invasividade Neoplásica , Células-Tronco Neoplásicas/efeitos dos fármacos , Temozolomida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Adv Healthc Mater ; 4(3): 405-11, 2015 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25230171

RESUMO

Understanding the effects of topographic characteristics on tumor cell migration is important for the development of new anti-migratory therapies. However, simplified in vitro culture systems often lead to inaccurate results regarding the efficacy of drugs. Histopathologically, glioblastoma multiform (GBM) cells migrate along the orientation of thin, elongated anatomical structures, such as white-matter tracts. Here, a tapered microtract array platform which mimics the anatomical features of brain tissue is introduced. This platform enables optimization of design for platform fabrication depending on topographic effects. By monitoring the migration of GBM cells on a simple tapered microtract, a saltatory migration resembling the migratory phenotype of human GBM cells in vivo is observed. The platform effectively induces the native characteristics and behavior of cells by topographic cues, allowing to observe the critical point for crawling to saltatory transition. Furthermore, this platform can be applied to efficiently screen anti-cancer drug by inhibiting associated signaling pathways on GBM cells. In conclusion, the microtract array platform reported here may provide a better understanding of the effects of topographic characteristics on cell migration, and may also be useful to determine the efficacy of antimigratory drugs for glioblastoma cells with cellular and molecular research and high-throughput screening.


Assuntos
Neoplasias Encefálicas/patologia , Ensaios de Migração Celular/métodos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Glioblastoma/patologia , Ensaios de Triagem em Larga Escala/métodos , Androstadienos/farmacologia , Compostos de Anilina/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Butadienos/farmacologia , Linhagem Celular , Ensaios de Migração Celular/instrumentação , Movimento Celular , Ensaios de Seleção de Medicamentos Antitumorais/instrumentação , Glioblastoma/tratamento farmacológico , Humanos , Nitrilas/farmacologia , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...