Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
FASEB J ; 36(1): e22067, 2022 01.
Article in English | MEDLINE | ID: mdl-34914140

ABSTRACT

The objective of the current study was to examine the drug-induced effects of the EP2 agonist, omidenapag (OMD), on human corneal stroma, two- and three-dimensional (2D and 3D) cultures of human corneal stroma fibroblasts (HCSFs). The drug-induced effects on 2D monolayers and 3D spheroids were characterized by examining the ultrastructures by scanning electron microscope (SEM), transendothelial electrical resistance (TEER) measurements, and fluorescein isothiocyanate (FITC)-dextran permeability. The physical properties of 3D spheroids with respect to size and stiffness were also examined. In addition, the gene expressions of extracellular matrix (ECM) molecules, including collagen (COL) 1, 4, and 6, and fibronectin (FN), a tissue inhibitor of metalloproteinase (TIMP) 1-4, matrix metalloproteinase (MMP) 2, 9, and 14, aquaporin1 (AQP1), and several endoplasmic reticulum (ER) stress-related factors were evaluated. In the 2D HCSFs, OMD induced (1) a significant increase in ECM deposits, as evidenced by SEM, the mRNA expression of COL4 and FN, and (2) a decrease in TEER values and a concentration-dependent increase in FITC-dextran permeability. In the case of 3D spheroids, OMD had no effect on size but a substantial increase in stiffness was observed. Furthermore, such OMD-induced effects on stiffness were dramatically modulated by the osmotic pressure of the system. In contrast to the above 2D cultures, among the ECM molecules and the modulators of 3D spheroids, namely, TIMPS and MMPs, the down-regulation of COL1, TIMP1 and 2 and the up-regulation of MMP9 were observed. Interestingly, such diversity in terms of OMD-induced gene expressions between 2D and 3D cultures was also recognized in AQP1 (2D; no significant change, 3D; significant up-regulation) and ER stress-related genes. The findings presented herein suggest that the EP2 agonist, OMD, alters the physical stiffness of 3D spheroids obtained from human corneal stroma fibroblasts and this alteration is dependent on the osmotic pressures. 2D and 3D cell cultures may be useful for evaluating the drug induced effects of OMD toward human corneal stroma.


Subject(s)
Cornea/metabolism , Fibroblasts/metabolism , Osmotic Pressure/drug effects , Receptors, Prostaglandin E, EP2 Subtype , Spheroids, Cellular/metabolism , Cornea/ultrastructure , Endoplasmic Reticulum Stress , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Eye Proteins/metabolism , Female , Fibroblasts/ultrastructure , Humans , Male , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Spheroids, Cellular/ultrastructure
2.
Pancreas ; 50(7): 982-989, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34629448

ABSTRACT

OBJECTIVES: To facilitate exploring a link between pancreatic ductal adenocarcinoma (PDAC) and diabetes mellitus, we constructed a novel 3-dimensional (3D) in vitro coculturing system for studying interactions between PDAC and islet cells. METHODS: Adopting a 3D rotary cell culture system, we have cocultured several PDAC cell lines and MIN6 islet ß cells. The cellular morphology and viability of both cell types were investigated by time-lapse imaging, confocal and scanning electron microscopy, and immunohistochemistry. RESULTS: The developed coculture method enabled the formation of 3D PDAC and ß-cell spheroids (pseudo islets). We showed that surface morphology and growth of cultured cells mimicked their in vivo appearance. In addition, the coculture demonstrated the affinity of the PDAC cells to grow around and invade the pseudo islets. CONCLUSIONS: Using rotary cell culture system, we have established a simple in vitro 3D pancreatic model. It is a flexible culture system that can easily be expanded with the addition of various stromal/neural components to further mimic in vivo conditions, thus enabling holistic investigation of the endocrine and exocrine pancreas.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Communication , Cell Culture Techniques, Three Dimensional/methods , Coculture Techniques/methods , Insulin-Secreting Cells/metabolism , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Survival , Humans , Immunohistochemistry , Insulin-Secreting Cells/pathology , Mice , Microscopy, Confocal , Microscopy, Electron, Scanning , Pancreatic Neoplasms/pathology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Spheroids, Cellular/ultrastructure , Time-Lapse Imaging/methods
3.
Int J Mol Sci ; 22(13)2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34202056

ABSTRACT

Mesenchymal stem cells (MSC) are known for their vascular regeneration capacity by neoangiogenesis. Even though, several delivery approaches exist, particularly in the case of intravascular delivery, only limited number of cells reach the targeted tissue and are not able to remain on site. Applicated cells exhibit poor survival accompanied with a loss of functionality. Moreover, cell application techniques lead to cell death and impede the overall MSC function and survival. 3D cell spheroids mimic the physiological microenvironment, thus, overcoming these limitations. Therefore, in this study we aimed to evaluate and assess the feasibility of 3D MSCs spheroids for endovascular application, for treatment of ischemic peripheral vascular pathologies. Multicellular 3D MSC spheroids were generated at different cell seeding densities, labelled with ultra-small particles of iron oxide (USPIO) and investigated in vitro in terms of morphology, size distribution, mechanical stability as well as ex vivo with magnetic resonance imaging (MRI) to assess their trackability and distribution. Generated 3D spheroids were stable, viable, maintained stem cell phenotype and were easily trackable and visualized via MRI. MSC 3D spheroids are suitable candidates for endovascular delivery approaches in the context of ischemic peripheral vascular pathologies.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Spheroids, Cellular , Animals , Cell Culture Techniques , Cell Differentiation , Humans , Ischemia/diagnosis , Ischemia/etiology , Ischemia/metabolism , Ischemia/therapy , Magnetic Resonance Imaging , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/etiology , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/therapy , Spheroids, Cellular/cytology , Spheroids, Cellular/ultrastructure , Staining and Labeling
4.
BMC Mol Cell Biol ; 22(1): 37, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34225662

ABSTRACT

BACKGROUND: Organ culture models have been used over the past few decades to study development and disease. The in vitro three-dimensional (3D) culture system of organoids is well known, however, these 3D systems are both costly and difficult to culture and maintain. As such, less expensive, faster and less complex methods to maintain 3D cell culture models would complement the use of organoids. Chick embryos have been used as a model to study human biology for centuries, with many fundamental discoveries as a result. These include cell type induction, cell competence, plasticity and contact inhibition, which indicates the relevance of using chick embryos when studying developmental biology and disease mechanisms. RESULTS: Here, we present an updated protocol that enables time efficient, cost effective and long-term expansion of fetal organ spheroids (FOSs) from chick embryos. Utilizing this protocol, we generated FOSs in an anchorage-independent growth pattern from seven different organs, including brain, lung, heart, liver, stomach, intestine and epidermis. These three-dimensional (3D) structures recapitulate many cellular and structural aspects of their in vivo counterpart organs and serve as a useful developmental model. In addition, we show a functional application of FOSs to analyze cell-cell interaction and cell invasion patterns as observed in cancer. CONCLUSION: The establishment of a broad ranging and highly effective method to generate FOSs from different organs was successful in terms of the formation of healthy, proliferating 3D organ spheroids that exhibited organ-like characteristics. Potential applications of chick FOSs are their use in studies of cell-to-cell contact, cell fusion and tumor invasion under defined conditions. Future studies will reveal whether chick FOSs also can be applicable in scientific areas such as viral infections, drug screening, cancer diagnostics and/or tissue engineering.


Subject(s)
Cell Culture Techniques, Three Dimensional , Models, Biological , Neoplasm Invasiveness/pathology , Organoids/cytology , Spheroids, Cellular/cytology , Animals , Cell Communication , Cell Line, Tumor , Chick Embryo , Chickens , Humans , Organoids/ultrastructure , Spheroids, Cellular/ultrastructure , Tissue Culture Techniques
5.
Artif Organs ; 45(10): 1208-1218, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34036603

ABSTRACT

As an alternative to the classical tissue engineering approach, bottom-up tissue engineering emerges using building blocks in bioassembly technologies. Spheroids can be used as building blocks to reach a highly complex ordered tissue by their fusion (bioassembly), representing the foundation of biofabrication. In this study, we analyzed the biomechanical properties and the fusion capacity of human adipose stem/stromal cell (ASC) we spheroids during an in vitro model of hypertrophic cartilage established by our research group. Hypertrophic induced-ASC spheroids showed a statistically significant higher Young's modulus at weeks 2 (P < .001) and 3 (P < .0005) compared with non-induced. After fusion, non-induced and induced-ASC spheroids increased the contact area and decreased their pairs' total length. At weeks 3 and 5, induced-ASC spheroids did not fuse completely, and the cells migrate preferentially in the fusion contact region. Alizarin red O staining showed the highest intensity of staining in the fused induced-ASC spheroids at week 5, together with intense staining for collagen type I and osteocalcin. Transmission electron microscopy and element content analysis (X-ray Energy Dispersive Spectroscopy) revealed in the fused quartet at week 3 a crystal-like structure. Hypertrophic induction interferes with the intrinsic capacity of spheroids to fuse. The measurements of contact between spheroids during the fusion process, together with the change in viscoelastic profile to the plastic, will impact the establishment of bioassembly protocols using hypertrophic induced-ASC spheroids as building blocks in biofabrication.


Subject(s)
Adipose Tissue/cytology , Cartilage/growth & development , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Adipose Tissue/physiology , Biomechanical Phenomena , Cartilage/cytology , Cartilage/ultrastructure , Cells, Cultured , Humans , Hypertrophy , Mesenchymal Stem Cells/physiology , Microscopy, Electron, Transmission , Spheroids, Cellular/physiology , Spheroids, Cellular/ultrastructure , Stromal Cells/physiology
6.
J Vis Exp ; (168)2021 02 27.
Article in English | MEDLINE | ID: mdl-33720122

ABSTRACT

Magnetic nanoparticles, made of iron oxide, present a peculiar interest for a wide range of biomedical applications for which they are often internalized in cells and then left within. One challenge is to assess their fate in the intracellular environment with reliable and precise methodologies. Herein, we introduce the use of the vibrating sample magnetometer (VSM) to precisely quantify the integrity of magnetic nanoparticles within cells by measuring their magnetic moment. Stem cells are first labeled with two types of magnetic nanoparticles; the nanoparticles have the same core produced via a fast and efficient microwave-based nonaqueous sol gel synthesis and differ in their coating: the commonly used citric acid molecule is compared to polyacrylic acid. The formation of 3D cell-spheroids is then achieved via centrifugation and the magnetic moment of these spheroids is measured at different times with the VSM. The obtained moment is a direct fingerprint of the nanoparticles' integrity, with decreasing values indicative of a nanoparticle degradation. For both nanoparticles, the magnetic moment decreases over culture time revealing their biodegradation. A protective effect of the polyacrylic acid coating is also shown, when compared to citric acid.


Subject(s)
Magnetic Iron Oxide Nanoparticles/chemistry , Magnetometry , Mesenchymal Stem Cells/metabolism , Endocytosis , Humans , Magnetic Iron Oxide Nanoparticles/ultrastructure , Mesenchymal Stem Cells/ultrastructure , Microwaves , Solutions , Spheroids, Cellular/metabolism , Spheroids, Cellular/ultrastructure
7.
Theranostics ; 11(7): 3331-3347, 2021.
Article in English | MEDLINE | ID: mdl-33537090

ABSTRACT

A spheroid is an aggregation of single cells with structural and functional characteristics similar to those of 3D native tissues, and it has been utilized as one of the typical in vitro three-dimensional (3D) cell models. Scaffold-free spheroids provide outstanding reflection of tissue complexity in a 3D in vivo-like environment, but they can neither fabricate realistic macroscale 3D complex structures without avoiding necrosis nor receive direct external stimuli (i.e., stimuli from mechanical or topographical cues). Here, we propose a spheroid-laden electrospinning process to obtain in vitro model achieved using the synergistic effect of the unique bioactive components provided by the spheroids and stimulating effects provided by the aligned nanofibers. Methods: To show the functional activity of the spheroid-laden structures, we used myoblast-spheroids to obtain skeletal muscle, comprising highly aligned myotubes, utilizing an uniaxially arranged topographical cue. The spheroid-electrospinning was used to align spheroids directly by embedding them in aligned alginate nanofibers, which were controlled with various materials and processing parameters. Results: The spheroids laden in the alginate nanofibers showed high cell viability (>90%) and was compared with that of a cell-laden alginate nanofiber that was electrospun with single cells. Consequently, the spheroids laden in the aligned nanofibers showed a significantly higher degree of myotube formation and maturation. Conclusion: Results suggested that the in vitro model using electrospun spheroids could potentially be employed to understand myogenic responses for various in vitro drug tests.


Subject(s)
Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Myoblasts/drug effects , Spheroids, Cellular/drug effects , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Alginates/chemistry , Alginates/pharmacology , Animals , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Electrochemical Techniques , Mice , Models, Biological , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/ultrastructure , Myoblasts/physiology , Myoblasts/ultrastructure , Spheroids, Cellular/physiology , Spheroids, Cellular/ultrastructure
8.
Macromol Biosci ; 20(12): e2000173, 2020 12.
Article in English | MEDLINE | ID: mdl-32881380

ABSTRACT

This work focuses on the development of ultrasound contrast vesicles for ultrasound-mediated enhanced transfection of nucleic acids in the cancer cells and projects its application as a tool for diagnostic imaging. The ultrasound contrast vesicles are stable, anionic, nanoscaled vesicles with ultrasound contrast equivalent to the commercially available SonoVue. These anionic lipid vesicles establish electrostatic interaction with cationic polyplexes based on linear polyethylenimine (22kDa) forming lipopolyplexes with ultrasound contrast. The lipopolyplexes are characterized regarding shape, size, and zeta potential. When exposed to low frequency ultrasound, these carriers show elevated transfection efficiency and reduced cytotoxicity. The effect of post-transfection ultrasound on cellular uptake of lipopolyplexes is also evaluated. An analogous transfection is also observed in the tumor mimicking multicellular 3D spheroid culture of ovarian cancer cells. The emergence of tumor imaging and enhanced gene delivery by medical ultrasound, a noninvasive imaging modality, is considered paving the way for efficient theranostic gene therapy.


Subject(s)
Contrast Media/pharmacology , Lipids/pharmacology , Neoplasms/diagnostic imaging , Ultrasonography , Anions/chemistry , Anions/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Contrast Media/chemistry , Gene Transfer Techniques/trends , Humans , Lipids/chemistry , Liposomes/chemistry , Liposomes/pharmacology , Spheroids, Cellular/ultrastructure
9.
Int J Mol Sci ; 21(15)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32751344

ABSTRACT

Human breast adenocarcinoma cells (MCF7) grow in three-dimensional culture as spheroids that represent the structural complexity of avascular tumors. Therefore, spheroids offer a powerful tool for studying cancer development, aggressiveness, and drug resistance. Notwithstanding the large amount of data regarding the formation of MCF7 spheroids, a detailed description of the morpho-functional changes during their aggregation and maturation is still lacking. In this study, in addition to the already established role of gap junctions, we show evidence of tunneling nanotube (TNT) formation, amyloid fibril production, and opening of large stable cellular bridges, thus reporting the sequential events leading to MCF7 spheroid formation. The variation in cell phenotypes, sustained by dynamic expression of multiple proteins, leads to complex networking among cells similar to the sequence of morphogenetic steps occurring in embryogenesis/organogenesis. On the basis of the observation that early events in spheroid formation are strictly linked to the redox homeostasis, which in turn regulate amyloidogenesis, we show that the administration of N-acetyl-l-cysteine (NAC), a reactive oxygen species (ROS) scavenger that reduces the capability of cells to produce amyloid fibrils, significantly affects their ability to aggregate. Moreover, cells aggregation events, which exploit the intrinsic adhesiveness of amyloid fibrils, significantly decrease following the administration during the early aggregation phase of neutral endopeptidase (NEP), an amyloid degrading enzyme.


Subject(s)
Acetylcysteine/pharmacology , Amyloid/chemistry , Free Radical Scavengers/pharmacology , Gap Junctions/ultrastructure , Homeostasis/drug effects , Spheroids, Cellular/ultrastructure , Amyloid/drug effects , Amyloid/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Aggregation/drug effects , Connexin 43/genetics , Connexin 43/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Gene Expression , Homeostasis/genetics , Humans , Interleukin-18/genetics , Interleukin-18/metabolism , MCF-7 Cells , Neprilysin/pharmacology , Oxidation-Reduction , Phenotype , Proteolysis , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism
10.
Micron ; 137: 102917, 2020 10.
Article in English | MEDLINE | ID: mdl-32693343

ABSTRACT

Three-dimensional (3D) spheroids mimic important properties of tumors and may soon become a reasonable substitute for animal models and human tissue, eliminating numerous problems related to in vivo and ex vivo experiments and pre-clinical drug trials. Currently, various imaging methods including X-ray microtomography (micro-CT), exist but their spatial resolution is limited. Here, we visualized and provided a morphological analysis of spheroid cell cultures using micro-CT and compared it to that of confocal microscopy. An approach is proposed that can potentially open new diagnostic opportunities to determine the morphology of cancer cells cultured in 3D structures instead of using actual tumors. Spheroids were formed from human melanoma cell lines WM266-4 and WM115 seeded at different cell densities using the hanging drop method. Micro-CT analysis of spheroid showed that spheroid size and shape differed depending on the cell line, initial cell number, and duration of culture. The melanoma cell lines used in this study can successfully be cultured as 3D spheroids and used to substitute human and animal models in pre-clinical studies. The micro-CT allows for high-resolution visualization of the spheroids structure.


Subject(s)
Cell Culture Techniques/methods , Neoplasms/ultrastructure , Spheroids, Cellular/ultrastructure , X-Ray Microtomography/methods , Animals , Cell Line , Cell Line, Tumor , High-Throughput Screening Assays , Humans , Melanoma
11.
Sci Rep ; 10(1): 11583, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665565

ABSTRACT

Multicellular tumor spheroid (MCTS) systems provide an in vitro cell culture model system which mimics many of the complexities of an in vivo solid tumor and tumor microenvironment, and are often used to study cancer cell growth and drug efficacy. Here, we present a coupled experimental-computational framework to estimate phenotypic growth and biophysical tumor microenvironment properties. This novel framework utilizes standard microscopy imaging of MCTS systems to drive a biophysical mathematical model of MCTS growth and mechanical interactions. By extending our previous in vivo mechanically-coupled reaction-diffusion modeling framework we developed a microscopy image processing framework capable of mechanistic characterization of MCTS systems. Using MDA-MB-231 breast cancer MCTS, we estimated biophysical parameters of cellular diffusion, rate of cellular proliferation, and cellular tractions forces. We found significant differences in these model-based biophysical parameters throughout the treatment time course between untreated and treated MCTS systems, whereas traditional size-based morphometric parameters were inconclusive. The proposed experimental-computational framework estimates mechanistic MCTS growth and invasion parameters with significant potential to assist in better and more precise assessment of in vitro drug efficacy through the development of computational analysis methodologies for three-dimensional cell culture systems to improve the development and evaluation of antineoplastic drugs.


Subject(s)
Breast Neoplasms/chemistry , Models, Theoretical , Spheroids, Cellular/chemistry , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/pharmacology , Biophysical Phenomena , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Mammary Neoplasms, Animal/chemistry , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Spheroids, Cellular/drug effects , Spheroids, Cellular/ultrastructure
12.
SLAS Discov ; 25(7): 709-722, 2020 08.
Article in English | MEDLINE | ID: mdl-32484408

ABSTRACT

Three-dimensional (3D) spheroid models are rapidly gaining favor for drug discovery applications due to their improved morphological characteristics, cellular complexity, long lifespan in culture, and higher physiological relevance relative to two-dimensional (2D) cell culture models. High-content imaging (HCI) of 3D spheroid models has the potential to provide valuable information to help researchers untangle disease pathophysiology and assess novel therapies more effectively. The transition from 2D monolayer models to dense 3D spheroids in HCI applications is not trivial, however, and requires 3D-optimized protocols, instrumentation, and resources. Here, we discuss considerations for moving from 2D to 3D models and present a framework for HCI and analysis of 3D spheroid models in a drug discovery setting. We combined scaffold-free, multicellular spheroid models with scalable, automation-compatible plate technology enabling image-based applications ranging from high-throughput screening to more complex, lower-throughput microphysiological systems of organ networks. We used this framework in three case studies: investigation of lipid droplet accumulation in a human liver nonalcoholic steatohepatitis (NASH) model, real-time immune cell interactions in a multicellular 3D lung cancer model, and a high-throughput screening application using a 3D co-culture model of gastric carcinoma to assess dose-dependent drug efficacy and specificity. The results of these proof-of-concept studies demonstrate the potential for high-resolution image-based analysis of 3D spheroid models for drug discovery applications, and confirm that cell-level and temporal-spatial analyses that fully exploit multicellular features of spheroid models are not only possible but soon will be routine practice in drug discovery workflows.


Subject(s)
Drug Discovery , Imaging, Three-Dimensional/trends , Molecular Imaging/trends , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Humans , Lipid Droplets/ultrastructure , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Spheroids, Cellular/drug effects , Spheroids, Cellular/ultrastructure , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology
13.
SLAS Discov ; 25(7): 723-733, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32396489

ABSTRACT

The nonadherent mammosphere assay has been commonly used to investigate cancer stem cell activities in breast cancers that have the ability to form tumorspheres and maintain tumor growth. The sphere formation step is critical, in that it enables the construction of the mammosphere models for downstream assays. The mammosphere assay has also been used to assess the effects of drug treatment on the tumorspheres formed from primary cancer cells or cell lines. Traditionally, the mammosphere formation has been evaluated by standard microscopy systems that required external software for additional analyses. However, this method can be time-consuming and low-throughput, thus impractical for high-throughput characterization of mammosphere models and screening for potential therapeutic cancer drugs. To overcome these challenges, we developed a plate-based high-throughput method to rapidly analyze mammospheres in whole wells using the Celigo Image Cytometer. The method is employed to characterize mammosphere formation and morphology for adherent and nonadherent propagation of four breast cancer cell lines (MCF7, MDA-MB-436, MDA-MB-231, and SKBR3). Next, the dose-dependent effects of four small molecule drugs (doxorubicin, paclitaxel, 8-quinolinol, and salinomycin) are characterized based on sphere formation and viability stained with calcein AM and propidium iodide. We observed growth and morphometric differences between adherent and nonadherent propagation of the four cell lines. Furthermore, drug treatments induced various effects on mammosphere formation, morphology, and viability. The proposed image cytometry method provides a useful tool suitable for high-throughput characterization and analysis of mammospheres, which can improve assay efficiency when investigating the formation capabilities and drug-induced cytotoxicity effects.


Subject(s)
Breast Neoplasms/drug therapy , Image Cytometry , Neoplastic Stem Cells/drug effects , Spheroids, Cellular/drug effects , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/pharmacology , Female , High-Throughput Screening Assays/methods , Humans , MCF-7 Cells , Neoplastic Stem Cells/pathology , Oxyquinoline/pharmacology , Paclitaxel/pharmacology , Pyrans/pharmacology , Spheroids, Cellular/ultrastructure
14.
BMC Cancer ; 20(1): 475, 2020 May 27.
Article in English | MEDLINE | ID: mdl-32460715

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma is a devastating disease with poor outcome, generally characterized by an excessive stroma component. The purpose of this study was to develop a simple and reproducible in vitro 3D-assay employing the main constituents of pancreatic ductal adenocarcinoma, namely pancreatic stellate and cancer cells. METHOD: A spheroid assay, directly co-culturing human pancreatic stellate cells with human pancreatic tumour cells in 3D was established and characterized by electron microscopy, immunohistochemistry and real-time RT-PCR. In order to facilitate the cell type-specific crosstalk analysis by real-time RT-PCR, we developed a novel in vitro 3D co-culture model, where the participating cell types were from different species, human and mouse, respectively. Using species-specific PCR primers, we were able to investigate the crosstalk between stromal and cancer cells without previous cell separation and sorting. RESULTS: We found clear evidence for mutual influence, such as increased proliferation and a shift towards a more mesenchymal phenotype in cancer cells and an activation of pancreatic stellate cells towards the myofibroblast phenotype. Using a heterospecies approach, which we coined virtual sorting, confirmed the findings we made initially in the human-human spheroids. CONCLUSIONS: We developed and characterized different easy to set up 3D models to investigate the crosstalk between cancer and stroma cells for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Coculture Techniques/methods , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Spheroids, Cellular/pathology , Cell Communication , Cell Line, Tumor , Cell Proliferation , Humans , Immunohistochemistry , Microscopy, Electron , Phenotype , Real-Time Polymerase Chain Reaction , Spheroids, Cellular/ultrastructure
15.
Cells ; 9(5)2020 05 01.
Article in English | MEDLINE | ID: mdl-32369990

ABSTRACT

We present a hiPSC-based 3D in vitro system suitable to test neurotoxicity (NT). Human iPSCs-derived 3D neurospheres grown in 96-well plate format were characterized timewise for 6-weeks. Changes in complexity and homogeneity were followed by immunocytochemistry and transmission electron microscopy. Transcriptional activity of major developmental, structural, and cell-type-specific markers was investigated at weekly intervals to present the differentiation of neurons, astrocytes, and oligodendrocytes. Neurospheres were exposed to different well-known toxicants with or without neurotoxic effect (e.g., paraquat, acrylamide, or ibuprofen) and examined at various stages of the differentiation with an ATP-based cell viability assay optimized for 3D-tissues. Concentration responses were investigated after acute (72 h) exposure. Moreover, the compound-specific effect of rotenone was investigated by a panel of ER-stress assay, TUNEL assay, immunocytochemistry, electron microscopy, and in 3D-spheroid based neurite outgrowth assay. The acute exposure to different classes of toxicants revealed distinct susceptibility profiles in a differentiation stage-dependent manner, indicating that hiPSC-based 3D in vitro neurosphere models could be used effectively to evaluate NT, and can be developed further to detect developmental neurotoxicity (DNT) and thus replace or complement the use of animal models in various basic research and pharmaceutical applications.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Neurotoxicity Syndromes/diagnosis , Spheroids, Cellular/cytology , Biomarkers/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Models, Biological , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neuronal Outgrowth/drug effects , Neurons/drug effects , Neurons/ultrastructure , Rotenone/toxicity , Spheroids, Cellular/drug effects , Spheroids, Cellular/ultrastructure
16.
Sci Rep ; 10(1): 3362, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32099027

ABSTRACT

Pancreatic cancer is one of the most complex types of cancers to detect, diagnose, and treat. However, the field of nanomedicine has strong potential to address such challenges. When evaluating the diffusion and penetration of theranostic nanoparticles, the extracellular matrix (ECM) is of crucial importance because it acts as a barrier to the tumor microenvironment. In the present study, the penetration of functionalized, fluorescent gold nanorods into large (>500 µm) multicellular 3D tissue spheroids was studied using a multimodal imaging approach. The spheroids were generated by co-culturing pancreatic cancer cells and pancreatic stellate cells in multiple ratios to mimic variable tumor-stromal compositions and to investigate nanoparticle penetration. Fluorescence live imaging, photothermal, and photoacoustic analysis were utilized to examine nanoparticle behavior in the spheroids. Uniquely, the nanorods are intrinsically photoacoustic and photothermal, enabling multi-imaging detection even when fluorescence tracking is not possible or ideal.


Subject(s)
Multimodal Imaging , Nanoparticles/chemistry , Pancreatic Neoplasms/diagnostic imaging , Stromal Cells/ultrastructure , Cell Line, Tumor , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Gold/chemistry , Humans , Nanotubes/chemistry , Optical Imaging , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/ultrastructure , Spheroids, Cellular/ultrastructure , Tumor Microenvironment/drug effects
17.
Artif Organs ; 44(7): E288-E299, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31950507

ABSTRACT

Human adipose stem/stromal cell (ASC) spheroids were used as a serum-free in vitro model to recapitulate the molecular events and extracellular matrix organization that orchestrate a hypertrophic cartilage phenotype. Induced-ASC spheroids (ø = 450 µm) showed high cell viability throughout the period of culture. The expression of collagen type X alpha 1 chain (COLXA1) and matrix metallopeptidase 13 (MMP-13) was upregulated at week 2 in induced-ASC spheroids compared with week 5 (P < .001) evaluated by quantitative real-time PCR. In accordance, secreted levels of IL-6 (P < .0001), IL-8 (P < .0001), IL-10 (P < .0001), bFGF (P < .001), VEGF (P < .0001), and RANTES (P < .0001) were the highest at week 2. Strong in situ staining for collagen type X and low staining for TSP-1 was associated with the increase of hypertrophic genes expression at week 2 in induced-ASC spheroids. Collagen type I, osteocalcin, biglycan, and tenascin C were detected at week 5 by in situ staining, in accordance with the highest expression of alkaline phosphatase (ALPL) gene and the presence of calcium deposits as evaluated by Alizarin Red O staining. Induced-ASC spheroids showed a higher force required to compression at week 2 (P < .0001). The human ASC spheroids under serum-free inducer medium and normoxic culture conditions were induced to a hypertrophic cartilage phenotype, opening a new perspective to recapitulate endochondral ossification in vivo.


Subject(s)
Cartilage/growth & development , Chondrogenesis/physiology , Mesenchymal Stem Cells/physiology , Primary Cell Culture/methods , Tissue Engineering/methods , Adipose Tissue/cytology , Cartilage/cytology , Cartilage/ultrastructure , Cell Differentiation/physiology , Cells, Cultured , Collagen Type X/metabolism , Culture Media, Serum-Free , Extracellular Matrix/metabolism , Humans , Hypertrophy , Matrix Metalloproteinase 13/metabolism , Microscopy, Electron, Transmission , Spheroids, Cellular/physiology , Spheroids, Cellular/ultrastructure , Stromal Cells/physiology
18.
Pancreas ; 48(9): 1225-1231, 2019 10.
Article in English | MEDLINE | ID: mdl-31593010

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is the third most common cause of cancer death in the United States. Improved characterized models of PDAC are needed for drug screening. METHODS: We grew 4 established pancreatic cancer cell lines in hanging drop cultures to produce spheroids. We also grew organoids from explanted xenografted PDAC and surgically resected primary PDAC. We performed transmission and scanning electron microscopy and compared findings with those of the normal pancreatic duct. We also performed single-cell cloning to determine the potential options for differentiation. RESULTS: Spheroids contained tight junctions and desmosomes but lacked zymogen granules, as expected. The former features were present in normal pancreatic duct but absent from PDAC cell lines grown in standard 2-dimensional culture. Spheroids functionally excluded macromolecules in whole mounts. Cells on the surface of PDAC spheroids were carpeted by microvilli except for rare cells with prominent stereocilia. Carpets of microvilli were also seen in low passage organoids produced from xenografts and surgically resected human PDAC, in addition to normal human pancreatic duct. We performed single-cell cloning and resulting spheroids produced both cell phenotypes at the same approximate ratios as those from bulk cultures. CONCLUSIONS: Pancreatic cancer spheroids/organoids are capable of biphenotypic differentiation.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Culture Techniques/methods , Organoids/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Spheroids, Cellular/pathology , Animals , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Desmosomes/ultrastructure , Female , Heterografts/pathology , Heterografts/ultrastructure , Humans , Mice, Nude , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Organoids/ultrastructure , Pancreatic Ducts/ultrastructure , Pancreatic Neoplasms/ultrastructure , Spheroids, Cellular/ultrastructure , Tight Junctions/ultrastructure
19.
Acta Biomater ; 100: 158-172, 2019 12.
Article in English | MEDLINE | ID: mdl-31542503

ABSTRACT

Cell spheroids as building blocks for engineering micro-tissue should be able to mimic the complex structure of natural tissue. However, control of the distribution of multiple cell populations within cell spheroids is difficult to achieve with current spheroid-harvest methods such as hanging-drop and with the use of microwell plates. In this study, we report the fabrication of core-shell spheroids with the ultimate goal to form 3D complex micro-tissue. We used endothelial cells and two types of stem cells (human turbinate mesenchymal stem cells (hTMSCs)/adipose-derived stem cells (ADSCs)). The stem cells and endothelial cells formed layered micro-sized cell sheets (µCSs) on polydopamine micro-patterned temperature-responsive hydrogel surfaces by a sequential seeding method, and these layered µCSs self-assembled to form core-shell spheroids by expansion of the hydrogels. The co-cultured spheroids formed a core-shell structure irrespective of stem cell type. In addition, the size of the core-shell spheroids was controlled from 90 ± 1 to 144 ± 3 µm by changing pattern sizes (200, 300, and 400 µm). The shell thickness gradually increased from 12 ± 3 to 30 ± 6 µm by adjusting the endothelial cell seeding density. Finally, we fabricated the micro-tissue by fusion of the co-cultured spheroids, and the spheroids with the core-shell structure rapidly induced in vitro vessel-like network in 3 days. Thus, the position of endothelial cells in co-cultured spheroids may be an important factor for the modulation of the vascularization process, which can be useful for the production of 3D complex micro-tissues using spheroids as building blocks. STATEMENT OF SIGNIFICANCE: This manuscript describes our work on the fabrication of core-shell spheroids as building blocks to form 3D complex vascularized micro-tissue. Stem cells (human turbinate mesenchymal stem cells (hTMSCs) or adipose-derived stem cells (ADSCs)) and endothelial cells formed layered micro-sized cell sheets (µCSs) on micro-patterned temperature-responsive hydrogel surfaces by a sequential seeding method, and these layered µCSs self-assembled to form core-shell spheroids (core - stem cells, shell - endothelial cells), irrespective of stem cell type. In addition, the size and shell thickness of the core-shell spheroids were controlled by modifying pattern size and endothelial cell seeding density. We fabricated the vascularized micro-tissue by fusion of the spheroids and demonstrated that the spheroids with a core-shell structure rapidly induced vessel-like network.


Subject(s)
Microtechnology , Neovascularization, Physiologic , Spheroids, Cellular/cytology , Tissue Engineering/methods , Fluorescence , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Spheroids, Cellular/ultrastructure , Temperature
20.
Nanoscale ; 11(35): 16488-16498, 2019 Sep 21.
Article in English | MEDLINE | ID: mdl-31453605

ABSTRACT

Magnetic nanoparticles (MNPs) internalized within stem cells have paved the way for remote magnetic cell manipulation and imaging in regenerative medicine. A full understanding of their interactions with stem cells and of their fate in the intracellular environment is then required, in particular with respect to their surface coatings. Here, we investigated the biological interactions of MNPs composed of an identical magnetic core but coated with different molecules: phosphonoacetic acid, polyethylene glycol phosphonic carboxylic acid, caffeic acid, citric acid, and polyacrylic acid. These coatings vary in the nature of the chelating function, the number of binding sites, and the presence or absence of a polymer. The nanoparticle magnetism was systematically used as an indicator of their internalization within human stem cells and of their structural long-term biodegradation in a 3D stem cell spheroid model. Overall, we evidence that the coating impacts the aggregation status of the nanoparticles and subsequently their uptake within stem cells, but it has little effect on their intracellular degradation. Only a high number of chelating functions (polyacrylic acid) had a significant protective effect. Interestingly, when the nanoparticles aggregated prior to cellular internalization, less degradation was also observed. Finally, for all coatings, a robust dose-dependent intracellular degradation rate was demonstrated, with higher doses of internalized nanoparticles leading to a lower degradation extent.


Subject(s)
Coated Materials, Biocompatible , Magnetite Nanoparticles , Mesenchymal Stem Cells , Spheroids, Cellular , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Humans , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Magnetite Nanoparticles/ultrastructure , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Spheroids, Cellular/metabolism , Spheroids, Cellular/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...