Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
ACS Biomater Sci Eng ; 10(5): 3478-3488, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38695610

ABSTRACT

Static three-dimensional (3D) cell culture has been demonstrated in ultralow attachment well plates, hanging droplet plates, and microtiter well plates with hydrogels or magnetic nanoparticles. Although it is simple, reproducible, and relatively inexpensive, thus potentially used for high-throughput screening, statically cultured 3D cells often suffer from a necrotic core due to limited nutrient and oxygen diffusion and waste removal and have a limited in vivo-like tissue structure. Here, we overcome these challenges by developing a pillar/perfusion plate platform and demonstrating high-throughput, dynamic 3D cell culture. Cell spheroids were loaded on the pillar plate with hydrogel by simple sandwiching and encapsulation and cultured dynamically in the perfusion plate on a digital rocker. Unlike traditional microfluidic devices, fast flow velocity was maintained within perfusion wells and the pillar plate was separated from the perfusion plate for cell-based assays. It was compatible with common lab equipment and allowed cell culture, testing, staining, and imaging in situ. The pillar/perfusion plate enhanced cell growth by rapid diffusion, reproducibility, assay throughput, and user friendliness in a dynamic 3D cell culture.


Subject(s)
Cell Culture Techniques, Three Dimensional , Cell Proliferation , Cell Culture Techniques, Three Dimensional/methods , Cell Culture Techniques, Three Dimensional/instrumentation , Humans , Reproducibility of Results , Perfusion/instrumentation , Hydrogels/chemistry , Spheroids, Cellular/cytology , Cell Culture Techniques/methods , Cell Culture Techniques/instrumentation
2.
Biomed Microdevices ; 26(2): 26, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38806765

ABSTRACT

Three-dimensional (3D) cell culture models have been extensively utilized in various mechanistic studies as well as for drug development studies as superior in vitro platforms than conventional two-dimensional (2D) cell culture models. This is especially the case in cancer biology, where 3D cancer models, such as spheroids or organoids, have been utilized extensively to understand the mechanisms of cancer development. Recently, many sophisticated 3D models such as organ-on-a-chip models are emerging as advanced in vitro models that can more accurately mimic the in vivo tissue functions. Despite such advancements, spheroids are still considered as a powerful 3D cancer model due to the relatively simple structure and compatibility with existing laboratory instruments, and also can provide orders of magnitude higher throughput than complex in vitro models, an extremely important aspects for drug development. However, creating well-defined spheroids remain challenging, both in terms of throughputs in generation as well as reproducibility in size and shape that can make it challenging for drug testing applications. In the past decades, droplet microfluidics utilizing hydrogels have been highlighted due to their potentials. Importantly, core-shell structured gel droplets can avoid spheroid-to-spheroid adhesion that can cause large variations in assays while also enabling long-term cultivation of spheroids with higher uniformity by protecting the core organoid area from external environment while the outer porous gel layer still allows nutrient exchange. Hence, core-shell gel droplet-based spheroid formation can improve the predictivity and reproducibility of drug screening assays. This review paper will focus on droplet microfluidics-based technologies for cancer spheroid production using various gel materials and structures. In addition, we will discuss emerging technologies that have the potential to advance the production of spheroids, prospects of such technologies, and remaining challenges.


Subject(s)
Hydrogels , Spheroids, Cellular , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Humans , Hydrogels/chemistry , Lab-On-A-Chip Devices , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Culture Techniques, Three Dimensional/instrumentation , Cell Culture Techniques, Three Dimensional/methods , Neoplasms/pathology , Neoplasms/metabolism , Microfluidics/instrumentation , Microfluidics/methods , Animals
3.
ACS Biomater Sci Eng ; 10(5): 3280-3292, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38608136

ABSTRACT

Hydrogels have gained significant popularity as model platforms to study reciprocal interactions between cells and their microenvironment. While hydrogel tools to probe many characteristics of the extracellular space have been developed, fabrication approaches remain challenging and time-consuming, limiting multiplexing or widespread adoption. Thus, we have developed a modular fabrication approach to generate distinct hydrogel microenvironments within the same 96-well plate for increased throughput of fabrication as well as integration with existing high-throughput assay technologies. This approach enables in situ hydrogel mechanical characterization and is used to generate both elastic and viscoelastic hydrogels across a range of stiffnesses. Additionally, this fabrication method enabled a 3-fold reduction in polymer and up to an 8-fold reduction in fabrication time required per hydrogel replicate. The feasibility of this platform for two-dimensional (2D) cell culture applications was demonstrated by measuring both population-level and single-cell-level metrics via microplate reader and high-content imaging. Finally, a 96-well hydrogel array was utilized for three-dimensional (3D) cell culture, demonstrating the ability to support high cell viability. Together, this work demonstrates a versatile and easily adaptable fabrication approach that can support the ever-expanding tool kit of hydrogel technologies for cell culture applications.


Subject(s)
Hydrogels , Hydrogels/chemistry , Humans , Cell Culture Techniques/methods , Cell Culture Techniques/instrumentation , Cell Survival , Cell Culture Techniques, Three Dimensional/methods , Cell Culture Techniques, Three Dimensional/instrumentation , Elasticity , Viscosity
4.
J Neurosci Methods ; 407: 110143, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670536

ABSTRACT

BACKGROUND: Silicon-based micro-pillar substrates (MPS), as three-dimensional cell culture platforms with vertically aligned micro-patterned scaffolding structures, are known to facilitate high-quality growth and morphology of dorsal root ganglion (DRG) sensory neurons, promote neurite outgrowth and enhance neurite alignment. However, the electrophysiological aspects of DRG neurons cultured on silicon MPSs have not been thoroughly investigated, which is of greatest importance to ensure that such substrates do not disrupt neuronal homeostasis and function before their widespread adoption in diverse biomedical applications. NEW METHOD: We conducted whole-cell patch-clamp recordings to explore the electrophysiological properties of DRG neurons cultured on MPS arrays, utilizing a custom-made upright patch-clamp setup. RESULTS: Our findings revealed that DRG neurons exhibited similar electrophysiological responses on patterned MPS samples when compared to the control planar glass surfaces. Notably, there were no significant differences observed in the action potential parameters or firing patterns of action potentials between neurons grown on either substrate. COMPARISON WITH EXISTING METHODS: In the current study we for the first time confirmed that successful electrophysiological recordings can be obtained from the cells grown on MPS. CONCLUSION: Our results imply that, despite the potential alterations caused by the cumulative trauma of tissue harvest and cell dissociation, essential functional cell properties of DRG neurons appear to be relatively maintained on MPS surfaces. Therefore, vertically aligned silicon MPSs could be considered as a potentially effective three-dimensional system for supporting a controlled cellular environment in culture.


Subject(s)
Ganglia, Spinal , Patch-Clamp Techniques , Silicon , Ganglia, Spinal/physiology , Ganglia, Spinal/cytology , Animals , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Cells, Cultured , Action Potentials/physiology , Neurons/physiology , Neurons/cytology , Rats, Sprague-Dawley , Rats , Cell Culture Techniques, Three Dimensional/methods , Cell Culture Techniques, Three Dimensional/instrumentation , Electrophysiological Phenomena/physiology
5.
Pharm Res ; 39(2): 329-340, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35166994

ABSTRACT

Although two-dimensional (2D) cell cultures are the standard in cell research, one pivotal disadvantage is the lack of cell-cell and cell-extracellular matrix (ECM) signaling in the culture milieu. However, such signals occur in three-dimensional (3D) in vivo environments and are essential for cell differentiation, proliferation, and a range of cellular functions. In this study, we developed a microfluidic device to proliferate and differentiate functional adipose tissue and adipocytes by utilizing 3D cell culture technology. This device was used to generate a tissue-specific 3D microenvironment to differentiate 3T3-L1 preadipocytes into either visceral white adipocytes using visceral adipose tissue (VAT) or subcutaneous white adipose tissue (SAT). The microchip has been tested and validated by functional assessments including cell morphology, inflammatory response to a lipopolysaccharide (LPS) challenge, GLUT4 tracking, and gene expression analyses. The biomimetic microfluidic chip is expected to mimic functional adipose tissues that can replace 2D cell cultures and allow for more accurate analysis of adipose tissue physiology.


Subject(s)
Adipocytes, White/physiology , Adipogenesis , Biomimetic Materials , Cell Culture Techniques, Three Dimensional/instrumentation , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , 3T3-L1 Cells , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Animals , Cell Proliferation , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Glucose Transporter Type 4/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL
6.
Sci Rep ; 12(1): 317, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013350

ABSTRACT

This manuscript describes a new method for forming basal-in MCF10A organoids using commercial 384-well ultra-low attachment (ULA) microplates and the development of associated live-cell imaging and automated analysis protocols. The use of a commercial 384-well ULA platform makes this method more broadly accessible than previously reported hanging drop systems and enables in-incubator automated imaging. Therefore, time points can be captured on a more frequent basis to improve tracking of early organoid formation and growth. However, one major challenge of live-cell imaging in multi-well plates is the rapid accumulation of large numbers of images. In this paper, an automated MATLAB script to handle the increased image load is developed. This analysis protocol utilizes morphological image processing to identify cellular structures within each image and quantify their circularity and size. Using this script, time-lapse images of aggregating and non-aggregating culture conditions are analyzed to profile early changes in size and circularity. Moreover, this high-throughput platform is applied to widely screen concentration combinations of Matrigel and epidermal growth factor (EGF) or heparin-binding EGF-like growth factor (HB-EGF) for their impact on organoid formation. These results can serve as a practical resource, guiding future research with basal-in MCF10A organoids.


Subject(s)
Cell Culture Techniques, Three Dimensional/instrumentation , Cell Proliferation , High-Throughput Screening Assays , Image Processing, Computer-Assisted , Mammary Glands, Human/physiology , Microscopy, Fluorescence , Organoids , Time-Lapse Imaging , Algorithms , Cell Line , Cell Proliferation/drug effects , Collagen/pharmacology , Drug Combinations , Epidermal Growth Factor/pharmacology , Female , Heparin-binding EGF-like Growth Factor/pharmacology , Humans , Laminin/pharmacology , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Proteoglycans/pharmacology , Time Factors
7.
Nat Commun ; 13(1): 493, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35078979

ABSTRACT

Ultrasonic neuromodulation has the unique potential to provide non-invasive control of neural activity in deep brain regions with high spatial precision and without chemical or genetic modification. However, the biomolecular and cellular mechanisms by which focused ultrasound excites mammalian neurons have remained unclear, posing significant challenges for the use of this technology in research and potential clinical applications. Here, we show that focused ultrasound excites primary murine cortical neurons in culture through a primarily mechanical mechanism mediated by specific calcium-selective mechanosensitive ion channels. The activation of these channels results in a gradual build-up of calcium, which is amplified by calcium- and voltage-gated channels, generating a burst firing response. Cavitation, temperature changes, large-scale deformation, and synaptic transmission are not required for this excitation to occur. Pharmacological and genetic inhibition of specific ion channels leads to reduced responses to ultrasound, while over-expressing these channels results in stronger ultrasonic stimulation. These findings provide a mechanistic explanation for the effect of ultrasound on neurons to facilitate the further development of ultrasonic neuromodulation and sonogenetics as tools for neuroscience research.


Subject(s)
Calcium/metabolism , Cerebral Cortex/cytology , Ion Channels/metabolism , Neurons/physiology , Ultrasonic Waves , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Cell Culture Techniques, Three Dimensional/instrumentation , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Gene Knockout Techniques , Ion Channels/genetics , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Physical Stimulation , Signal Transduction/drug effects , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Tetrodotoxin/pharmacology , Thapsigargin/pharmacology
8.
Front Immunol ; 12: 781337, 2021.
Article in English | MEDLINE | ID: mdl-34925361

ABSTRACT

Existing first-line cancer therapies often fail to cope with the heterogeneity and complexity of cancers, so that new therapeutic approaches are urgently needed. Among novel alternative therapies, adoptive cell therapy (ACT) has emerged as a promising cancer treatment in recent years. The limited clinical applications of ACT, despite its advantages over standard-of-care therapies, can be attributed to (i) time-consuming and cost-intensive procedures to screen for potent anti-tumor immune cells and the corresponding targets, (ii) difficulties to translate in-vitro and animal-derived in-vivo efficacies to clinical efficacy in humans, and (iii) the lack of systemic methods for the safety assessment of ACT. Suitable experimental models and testing platforms have the potential to accelerate the development of ACT. Immunocompetent microphysiological systems (iMPS) are microfluidic platforms that enable complex interactions of advanced tissue models with different immune cell types, bridging the gap between in-vitro and in-vivo studies. Here, we present a proof-of-concept iMPS that supports a triple culture of three-dimensional (3D) colorectal tumor microtissues, 3D cardiac microtissues, and human-derived natural killer (NK) cells in the same microfluidic network. Different aspects of tumor-NK cell interactions were characterized using this iMPS including: (i) direct interaction and NK cell-mediated tumor killing, (ii) the development of an inflammatory milieu through enrichment of soluble pro-inflammatory chemokines and cytokines, and (iii) secondary effects on healthy cardiac microtissues. We found a specific NK cell-mediated tumor-killing activity and elevated levels of tumor- and NK cell-derived chemokines and cytokines, indicating crosstalk and development of an inflammatory milieu. While viability and morphological integrity of cardiac microtissues remained mostly unaffected, we were able to detect alterations in their beating behavior, which shows the potential of iMPS for both, efficacy and early safety testing of new candidate ACTs.


Subject(s)
Biological Assay/methods , Cell Culture Techniques, Three Dimensional/methods , Immunotherapy, Adoptive , Killer Cells, Natural/transplantation , Neoplasms/therapy , Biological Assay/instrumentation , Cell Culture Techniques, Three Dimensional/instrumentation , Cell Line , Cell Separation , Female , Fetal Blood , Healthy Volunteers , Humans , Induced Pluripotent Stem Cells , Intravital Microscopy , Killer Cells, Natural/immunology , Lab-On-A-Chip Devices , Male , Myocytes, Cardiac , Neoplasms/immunology , Neoplasms/pathology , Primary Cell Culture , Proof of Concept Study
9.
Sci Rep ; 11(1): 19986, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34620916

ABSTRACT

Microfluidics offers promising methods for aligning cells in physiologically relevant configurations to recapitulate human organ functionality. Specifically, microstructures within microfluidic devices facilitate 3D cell culture by guiding hydrogel precursors containing cells. Conventional approaches utilize capillary forces of hydrogel precursors to guide fluid flow into desired areas of high wettability. These methods, however, require complicated fabrication processes and subtle loading protocols, thus limiting device throughput and experimental yield. Here, we present a swift and robust hydrogel patterning technique for 3D cell culture, where preloaded hydrogel solution in a microfluidic device is aspirated while only leaving a portion of the solution in desired channels. The device is designed such that differing critical capillary pressure conditions are established over the interfaces of the loaded hydrogel solution, which leads to controlled removal of the solution during aspiration. A proposed theoretical model of capillary pressure conditions provides physical insights to inform generalized design rules for device structures. We demonstrate formation of multiple, discontinuous hollow channels with a single aspiration. Then we test vasculogenic capacity of various cell types using a microfluidic device obtained by our technique to illustrate its capabilities as a viable micro-manufacturing scheme for high-throughput cellular co-culture.


Subject(s)
Cell Culture Techniques, Three Dimensional/instrumentation , Hydrogels , Microfluidics/instrumentation , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Coculture Techniques/methods , Fibroblasts , Human Umbilical Vein Endothelial Cells , Humans , Microfluidics/methods
10.
Biotechnol Bioeng ; 118(12): 4771-4785, 2021 12.
Article in English | MEDLINE | ID: mdl-34559409

ABSTRACT

Diamagnetic levitation is an emerging technology for remote manipulation of cells in cell and tissue level applications. Low-cost magnetic levitation configurations using permanent magnets are commonly composed of a culture chamber physically sandwiched between two block magnets that limit working volume and applicability. This work describes a single ring magnet-based magnetic levitation system to eliminate physical limitations for biofabrication. Developed configuration utilizes sample culture volume for construct size manipulation and long-term maintenance. Furthermore, our configuration enables convenient transfer of liquid or solid phases during the levitation. Before biofabrication, we first calibrated/ the platform for levitation with polymeric beads, considering the single cell density range of viable cells. By taking advantage of magnetic focusing and cellular self-assembly, millimeter-sized 3D structures were formed and maintained in the system allowing easy and on-site intervention in cell culture with an open operational space. We demonstrated that the levitation protocol could be adapted for levitation of various cell types (i.e., stem cell, adipocyte and cancer cell) representing cells of different densities by modifying the paramagnetic ion concentration that could be also reduced by manipulating the density of the medium. This technique allowed the manipulation and merging of separately formed 3D biological units, as well as the hybrid biofabrication with biopolymers. In conclusion, we believe that this platform will serve as an important tool in broad fields such as bottom-up tissue engineering, drug discovery and developmental biology.


Subject(s)
Cell Culture Techniques, Three Dimensional , Magnets , Tissue Engineering , Animals , Cell Culture Techniques, Three Dimensional/instrumentation , Cell Culture Techniques, Three Dimensional/methods , Cell Line , Equipment Design , Humans , Mice , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tissue Engineering/instrumentation , Tissue Engineering/methods
11.
Stem Cell Reports ; 16(9): 2159-2168, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34478648

ABSTRACT

Crosstalk between endothelial cells (ECs) and pericytes or vascular smooth muscle cells (VSMCs) is essential for the proper functioning of blood vessels. This balance is disrupted in several vascular diseases but there are few experimental models which recapitulate this vascular cell dialogue in humans. Here, we developed a robust multi-cell type 3D vessel-on-chip (VoC) model based entirely on human induced pluripotent stem cells (hiPSCs). Within a fibrin hydrogel microenvironment, the hiPSC-derived vascular cells self-organized to form stable microvascular networks reproducibly, in which the vessels were lumenized and functional, responding as expected to vasoactive stimulation. Vascular organization and intracellular Ca2+ release kinetics in VSMCs could be quantified using automated image analysis based on open-source software CellProfiler and ImageJ on widefield or confocal images, setting the stage for use of the platform to study vascular (patho)physiology and therapy.


Subject(s)
Cell Culture Techniques, Three Dimensional/methods , Endothelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Lab-On-A-Chip Devices , Myocytes, Smooth Muscle/cytology , Tissue Engineering/methods , Biomarkers , Calcium/metabolism , Cell Culture Techniques, Three Dimensional/instrumentation , Cell Differentiation , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Humans , Induced Pluripotent Stem Cells/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Tissue Engineering/instrumentation
12.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Article in English | MEDLINE | ID: mdl-33941674

ABSTRACT

Tissue-on-chip systems represent promising platforms for monitoring and controlling tissue functions in vitro for various purposes in biomedical research. The two-dimensional (2D) layouts of these constructs constrain the types of interactions that can be studied and limit their relevance to three-dimensional (3D) tissues. The development of 3D electronic scaffolds and microphysiological devices with geometries and functions tailored to realistic 3D tissues has the potential to create important possibilities in advanced sensing and control. This study presents classes of compliant 3D frameworks that incorporate microscale strain sensors for high-sensitivity measurements of contractile forces of engineered optogenetic muscle tissue rings, supported by quantitative simulations. Compared with traditional approaches based on optical microscopy, these 3D mechanical frameworks and sensing systems can measure not only motions but also contractile forces with high accuracy and high temporal resolution. Results of active tension force measurements of engineered muscle rings under different stimulation conditions in long-term monitoring settings for over 5 wk and in response to various chemical and drug doses demonstrate the utility of such platforms in sensing and modulation of muscle and other tissues. Possibilities for applications range from drug screening and disease modeling to biohybrid robotic engineering.


Subject(s)
Cell Culture Techniques, Three Dimensional/methods , Imaging, Three-Dimensional/methods , Muscles/metabolism , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Acetylcholine/pharmacology , Actinin/metabolism , Animals , Caffeine/pharmacology , Cell Culture Techniques, Three Dimensional/instrumentation , Cell Differentiation , Cell Line , Dantrolene/pharmacology , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myosins/metabolism , Tissue Engineering/instrumentation , Vasodilator Agents/pharmacology
13.
ACS Appl Bio Mater ; 4(2): 1319-1329, 2021 02 15.
Article in English | MEDLINE | ID: mdl-35014483

ABSTRACT

Melt electrowriting (MEW) is an emerging technique that precisely fabricates microfibrous scaffolds, ideal for tissue engineering, where biomimetic microarchitectural detail is required. Polycaprolactone (PCL), a synthetic polymer, was selected as the scaffold material due to its biocompatibility, biodegradability, mechanical strength, and melt processability. To increase PCL bioactivity, a natural polymer, chitosan, was added to construct MEW fibrous composite scaffolds. To date, this is the first study of its kind detailing the effects of stem cell behavior on PCL containing chitosan MEW scaffolds. The aim of this study was to melt electrowrite a range of PCL/chitosan tissue-engineered constructs (TECs) and assess their suitability to promote the growth of human bone-marrow-derived mesenchymal stem cells (hBMSCs). In vitro physical and biological characterizations of melt-electrowritten TECs were performed. Physical characterization showed that reproducible, layered micron-range scaffolds could be successfully fabricated. As well, cell migration and proliferation were assessed via an assay to monitor cell infiltration throughout the three-dimensional (3D) melt-electrowritten scaffold structure. A statistically significant increase (∼140%) in hBMSC proliferation in 1 wt % chitosan PCL blends in comparison to PCL-only scaffolds was found when monitored over two weeks. Overall, our study demonstrates the fabrication of melt-electrowritten PCL/chitosan composite scaffolds with controlled microarchitecture and their potential use for regenerative, tissue engineering applications.


Subject(s)
Cell Culture Techniques, Three Dimensional/instrumentation , Chitosan/chemistry , Mesenchymal Stem Cells/physiology , Polyesters/chemistry , Cell Culture Techniques, Three Dimensional/methods , Humans , Tissue Engineering/methods , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...