Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.570
Filter
1.
Sci Rep ; 14(1): 17861, 2024 08 01.
Article in English | MEDLINE | ID: mdl-39090238

ABSTRACT

The development of non-destructive, tomographic imaging systems is a current topic of research in biomedical technologies. One of these technologies is Scanning Laser Optical Tomography (SLOT), which features a highly modular setup with various contrast mechanisms. Extending this technology with new acquisition mechanisms allows us to investigate untreated and non-stained biological samples, leaving their natural biological physiology intact. To enhance the development of SLOT, we aimed to extend the density of information with a significant increase of acquisition channels. This should allow us to investigate samples with unknown emission spectra and even allow for label-fee cell identification. We developed and integrated a hyperspectral module into an existing SLOT system. The adaptations allow for the acquisition of three-dimensional datasets containing a highly increased information density. For validation, artificial test objects were made from fluorescent acrylic and acquired with the new hyperspectral setup. In addition, measurements were made on two different human cell spheroids with an unknown spectra, to test the possibilities of label-free cell identification. The validation measurements of the artificial test target show the expected results. Furthermore, the measurements of the biological cell spheroids show small variations in their tomographic spectrum that allow for label-free cell type differentiation. The results of the biological sample demonstrate the potential of label-free cell identification of the newly developed setup.


Subject(s)
Tomography, Optical , Tomography, Optical/methods , Tomography, Optical/instrumentation , Humans , Lasers , Spheroids, Cellular/cytology , Imaging, Three-Dimensional/methods
2.
Nat Commun ; 15(1): 5894, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003281

ABSTRACT

Remarkable advances in protocol development have been achieved to manufacture insulin-secreting islets from human pluripotent stem cells (hPSCs). Distinct from current approaches, we devised a tunable strategy to generate islet spheroids enriched for major islet cell types by incorporating PDX1+ cell budding morphogenesis into staged differentiation. In this process that appears to mimic normal islet morphogenesis, the differentiating islet spheroids organize with endocrine cells that are intermingled or arranged in a core-mantle architecture, accompanied with functional heterogeneity. Through in vitro modelling of human pancreas development, we illustrate the importance of PDX1 and the requirement for EphB3/4 signaling in eliciting cell budding morphogenesis. Using this new approach, we model Mitchell-Riley syndrome with RFX6 knockout hPSCs illustrating unexpected morphogenesis defects in the differentiation towards islet cells. The tunable differentiation system and stem cell-derived islet models described in this work may facilitate addressing fundamental questions in islet biology and probing human pancreas diseases.


Subject(s)
Cell Differentiation , Homeodomain Proteins , Islets of Langerhans , Morphogenesis , Pluripotent Stem Cells , Spheroids, Cellular , Trans-Activators , Humans , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Receptors, Eph Family/metabolism , Receptors, Eph Family/genetics
3.
Biofabrication ; 16(4)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39053497

ABSTRACT

Three-dimensional cell spheroids show promise for the reconstruction of native tissues. Herein, we report a sophisticated, uniform, and highly reproducible spheroid culture system for tissue reconstruction. A mesh-integrated culture system was designed to precisely control the uniformity and reproducibility of spheroid formation. Furthermore, we synthesized hexanoyl glycol chitosan, a material with ultralow cell adhesion properties, to further improve spheroid formation efficiency and biological function. Our results demonstrate improved biological function in various types of cells and ability to generate spheroids with complex structures composed of multiple cell types. In conclusion, our spheroid culture system offers a highly effective and widely applicable approach to generating customized spheroids with desired structural and biological features for a variety of biomedical applications.


Subject(s)
Cell Culture Techniques , Chitosan , Regenerative Medicine , Spheroids, Cellular , Spheroids, Cellular/cytology , Chitosan/chemistry , Humans , Cell Culture Techniques/methods , Tissue Engineering/methods , Animals
4.
Biofabrication ; 16(4)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39019062

ABSTRACT

Three-dimensional (3D) cell culture models capable of emulating the biological functions of natural tissues are pivotal in tissue engineering and regenerative medicine. Despite progress, the fabrication ofin vitroheterocellular models that mimic the intricate structures of natural tissues remains a significant challenge. In this study, we introduce a novel, scaffold-free approach leveraging the inertial focusing effect in rotating hanging droplets for the reliable production of heterocellular spheroids with controllable core-shell structures. Our method offers precise control over the core-shell spheroid's size and geometry by adjusting the cell suspension density and droplet morphology. We successfully applied this technique to create hair follicle organoids, integrating dermal papilla cells within the core and epidermal cells in the shell, thereby achieving markedly enhanced hair inducibility compared to mixed-structure models. Furthermore, we have developed melanoma tumor spheroids that accurately mimic the dynamic interactions between tumor and stromal cells, showing increased invasion capabilities and altered expressions of cellular adhesion molecules and proteolytic enzymes. These findings underscore the critical role of cellular spatial organization in replicating tissue functionalityin vitro. Our method represents a significant advancement towards generating heterocellular spheroids with well-defined architectures, offering broad implications for biological research and applications in tissue engineering.


Subject(s)
Cell Culture Techniques, Three Dimensional , Spheroids, Cellular , Spheroids, Cellular/cytology , Cell Culture Techniques, Three Dimensional/methods , Humans , Tissue Engineering/methods , Organoids/cytology , Hair Follicle/cytology , Animals , Cell Line, Tumor , Tissue Scaffolds/chemistry , Cell Culture Techniques/methods , Cell Culture Techniques/instrumentation
5.
ACS Biomater Sci Eng ; 10(8): 5080-5093, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39038278

ABSTRACT

Ensuring good definition of scaffolds used for 3D cell culture is a prominent challenge that hampers the development of tissue engineering platforms. Since dextran repels cell adhesion, using dextran-based materials biofunctionalized through a bottom-up approach allows for precise control over material definition. Here, we report the design of dextran hydrogels displaying a fully interconnected macropore network for the culture of vascular spheroids in vitro. We biofunctionalized the hydrogels with the RGD peptide sequence to promote cell adhesion. We used an affinity peptide pair, the E/K coiled coil, to load the gels with epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF). Dual functionalization with adhesive and proliferative cues allows vascular spheroids to colonize naturally cell-repellant dextran. In supplement-depleted medium, we report improved colonization of the macropores compared to that of unmodified dextran. Altogether, we propose a well-defined and highly versatile platform for tissue engineering and tissue vascularization applications.


Subject(s)
Dextrans , Hydrogels , Dextrans/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Tissue Engineering/methods , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Cell Adhesion/drug effects , Porosity , Human Umbilical Vein Endothelial Cells/drug effects , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/metabolism , Neovascularization, Physiologic/drug effects , Tissue Scaffolds/chemistry , Intercellular Signaling Peptides and Proteins/pharmacology , Peptides/chemistry , Peptides/pharmacology
6.
Biofabrication ; 16(4)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39053493

ABSTRACT

In contrast to traditional two-dimensional cell-culture conditions, three-dimensional (3D) cell-culture models closely mimic complexin vivoconditions. However, constructing 3D cell culture models still faces challenges. In this paper, by using micro/nano fabrication method, including lithography, deposition, etching, and lift-off, we designed magnetic nanostructures resembling a crown of thorns. This magnetic crown of thorns (MCT) nanostructure enables the isolation of cells that have endocytosed magnetic particles. To assess the utility of this nanostructure, we used high-flux acquisition of Jurkat cells, an acute-leukemia cell line exhibiting the native phenotype, as an example. The novel structure enabled Jurkat cells to form spheroids within just 30 min by leveraging mild magnetic forces to bring together endocytosed magnetic particles. The size, volume, and arrangement of these spheroids were precisely regulated by the dimensions of the MCT nanostructure and the array configuration. The resulting magnetic cell clusters were uniform in size and reached saturation after 1400 s. Notably, these cell clusters could be easily separated from the MCT nanostructure through enzymatic digestion while maintaining their integrity. These clusters displayed a strong proliferation rate and survival capabilities, lasting for an impressive 96 h. Compared with existing 3D cell-culture models, the approach presented in this study offers the advantage of rapid formation of uniform spheroids that can mimicin vivomicroenvironments. These findings underscore the high potential of the MCT in cell-culture models and magnetic tissue enginerring.


Subject(s)
Nanostructures , Spheroids, Cellular , Humans , Spheroids, Cellular/cytology , Jurkat Cells , Nanostructures/chemistry , Cell Culture Techniques/methods
7.
Front Immunol ; 15: 1356397, 2024.
Article in English | MEDLINE | ID: mdl-38975341

ABSTRACT

Introduction: Within adipose tissue (AT), different macrophage subsets have been described, which played pivotal and specific roles in upholding tissue homeostasis under both physiological and pathological conditions. Nonetheless, studying resident macrophages in-vitro poses challenges, as the isolation process and the culture for extended periods can alter their inherent properties. Methods: Stroma-vascular cells isolated from murine subcutaneous AT were seeded on ultra-low adherent plates in the presence of macrophage colony-stimulating factor. After 4 days of culture, the cells spontaneously aggregate to form spheroids. A week later, macrophages begin to spread out of the spheroid and adhere to the culture plate. Results: This innovative three-dimensional (3D) culture method enables the generation of functional mature macrophages that present distinct genic and phenotypic characteristics compared to bone marrow-derived macrophages. They also show specific metabolic activity and polarization in response to stimulation, but similar phagocytic capacity. Additionally, based on single-cell analysis, AT-macrophages generated in 3D culture mirror the phenotypic and functional traits of in-vivo AT resident macrophages. Discussion: Our study describes a 3D in-vitro system for generating and culturing functional AT-resident macrophages, without the need for cell sorting. This system thus stands as a valuable resource for exploring the differentiation and function of AT-macrophages in vitro in diverse physiological and pathological contexts.


Subject(s)
Adipose Tissue , Cell Culture Techniques, Three Dimensional , Cell Differentiation , Macrophages , Animals , Macrophages/immunology , Macrophages/metabolism , Mice , Adipose Tissue/cytology , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Phagocytosis , Mice, Inbred C57BL , Spheroids, Cellular/cytology , Cell Culture Techniques/methods , Phenotype
8.
Bull Exp Biol Med ; 177(1): 115-123, 2024 May.
Article in English | MEDLINE | ID: mdl-38963596

ABSTRACT

The cardiac perivascular niche is a cellular microenvironment of a blood vessel. The principles of niche regulation are still poorly understood. We studied the effect of TGFß1 on cells forming the cardiac perivascular niche using 3D cell culture (cardiospheres). Cardiospheres contained progenitor (c-Kit), endothelial (CD31), and mural (αSMA) cells, basement membrane proteins (laminin) and extracellular matrix proteins (collagen I, fibronectin). TGFß1 treatment decreased the length of CD31+ microvasculature, VE cadherin protein level, and proportion of NG2+ cells, and increased proportion of αSMA+ cells and transgelin/SM22α protein level. We supposed that this effect is related to the stabilizing function of TGFß1 on vascular cells: decreased endothelial cell proliferation, as shown for HUVEC, and activation of mural cell differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Cell Differentiation/drug effects , Humans , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Animals , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Cadherins/metabolism , Laminin/metabolism , Laminin/pharmacology , Muscle Proteins/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/cytology , Fibronectins/metabolism , Fibronectins/pharmacology , Antigens, CD/metabolism , Myocardium/metabolism , Myocardium/cytology , Stem Cell Niche/drug effects , Stem Cell Niche/physiology , Collagen Type I/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Cell Culture Techniques, Three Dimensional/methods
9.
Proc Natl Acad Sci U S A ; 121(28): e2404210121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38954541

ABSTRACT

Mesenchymal stem cells (MSCs) are essential in regenerative medicine. However, conventional expansion and harvesting methods often fail to maintain the essential extracellular matrix (ECM) components, which are crucial for their functionality and efficacy in therapeutic applications. Here, we introduce a bone marrow-inspired macroporous hydrogel designed for the large-scale production of MSC-ECM spheroids. Through a soft-templating approach leveraging liquid-liquid phase separation, we engineer macroporous hydrogels with customizable features, including pore size, stiffness, bioactive ligand distribution, and enzyme-responsive degradability. These tailored environments are conducive to optimal MSC proliferation and ease of harvesting. We find that soft hydrogels enhance mechanotransduction in MSCs, establishing a standard for hydrogel-based 3D cell culture. Within these hydrogels, MSCs exist as both cohesive spheroids, preserving their innate vitality, and as migrating entities that actively secrete functional ECM proteins. Additionally, we also introduce a gentle, enzymatic harvesting method that breaks down the hydrogels, allowing MSCs and secreted ECM to naturally form MSC-ECM spheroids. These spheroids display heightened stemness and differentiation capacity, mirroring the benefits of a native ECM milieu. Our research underscores the significance of sophisticated materials design in nurturing distinct MSC subpopulations, facilitating the generation of MSC-ECM spheroids with enhanced therapeutic potential.


Subject(s)
Extracellular Matrix , Hydrogels , Mesenchymal Stem Cells , Spheroids, Cellular , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Hydrogels/chemistry , Extracellular Matrix/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Humans , Cell Differentiation , Cell Culture Techniques/methods , Cell Proliferation , Porosity , Mechanotransduction, Cellular/physiology , Cells, Cultured
10.
Biomaterials ; 311: 122691, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38996673

ABSTRACT

Acoustic holography (AH), a promising approach for cell patterning, emerges as a powerful tool for constructing novel invitro 3D models that mimic organs and cancers features. However, understanding changes in cell function post-AH remains limited. Furthermore, replicating complex physiological and pathological processes solely with cell lines proves challenging. Here, we employed acoustical holographic lattice to assemble primary hepatocytes directly isolated from mice into a cell cluster matrix to construct a liver-shaped tissue sample. For the first time, we evaluated the liver functions of AH-patterned primary hepatocytes. The patterned model exhibited large numbers of self-assembled spheroids and superior multifarious core hepatocyte functions compared to cells in 2D and traditional 3D culture models. AH offers a robust protocol for long-term in vitro culture of primary cells, underscoring its potential for future applications in disease pathogenesis research, drug testing, and organ replacement therapy.


Subject(s)
Hepatocytes , Holography , Liver , Hepatocytes/cytology , Hepatocytes/metabolism , Animals , Liver/cytology , Holography/methods , Mice , Acoustics , Cells, Cultured , Spheroids, Cellular/cytology , Mice, Inbred C57BL
11.
Methods Mol Biol ; 2825: 333-343, 2024.
Article in English | MEDLINE | ID: mdl-38913319

ABSTRACT

Cancer cytogenetic analyses often involve cell culture. However, many cytogeneticists overlook interesting phenotypes associated with cultured cells. Given that cytogeneticists need to focus more on phenotypes to comprehend the genotypes, the biological significance of seemingly trivial cellular variations deserves attention. One example is the formation of cellular tunneling tubes (TTs) in cultured cancer cells, which likely play a role in cell-to-cell communication and material transport. In this chapter, we describe protocols for studying these TTs as well as cellular spheres. In addition to diverse chromosomal variants, these different types of variations should be considered for understanding cancer heterogeneity and dynamics, as they illustrate the importance of various forms of fuzzy inheritance.


Subject(s)
Cell Communication , Spheroids, Cellular , Humans , Spheroids, Cellular/cytology , Cell Culture Techniques/methods , Cell Line, Tumor
12.
Exp Cell Res ; 439(2): 114111, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38823471

ABSTRACT

Skeletal muscle ischemia-reperfusion (IR) injury poses significant challenges due to its local and systemic complications. Traditional studies relying on two-dimensional (2D) cell culture or animal models often fall short of faithfully replicating the human in vivo environment, thereby impeding the translational process from animal research to clinical applications. Three-dimensional (3D) constructs, such as skeletal muscle spheroids with enhanced cell-cell interactions from human pluripotent stem cells (hPSCs) offer a promising alternative by partially mimicking human physiological cellular environment in vivo processes. This study aims to establish an innovative in vitro model, human skeletal muscle spheroids based on sphere differentiation from hPSCs, to investigate human skeletal muscle developmental processes and IR mechanisms within a controlled laboratory setting. By eticulously recapitulating embryonic myogenesis through paraxial mesodermal differentiation of neuro-mesodermal progenitors, we successfully established 3D skeletal muscle spheroids that mirror the dynamic colonization observed during human skeletal muscle development. Co-culturing human skeletal muscle spheroids with spinal cord spheroids facilitated the formation of neuromuscular junctions, providing functional relevance to skeletal muscle spheroids. Furthermore, through oxygen-glucose deprivation/re-oxygenation treatment, 3D skeletal muscle spheroids provide insights into the molecular events and pathogenesis of IR injury. The findings presented in this study significantly contribute to our understanding of skeletal muscle development and offer a robust platform for in vitro studies on skeletal muscle IR injury, holding potential applications in drug testing, therapeutic development, and personalized medicine within the realm of skeletal muscle-related pathologies.


Subject(s)
Cell Differentiation , Muscle, Skeletal , Pluripotent Stem Cells , Reperfusion Injury , Spheroids, Cellular , Humans , Reperfusion Injury/pathology , Reperfusion Injury/metabolism , Muscle, Skeletal/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Spheroids, Cellular/cytology , Muscle Development , Coculture Techniques/methods , Cells, Cultured , Cell Culture Techniques/methods
13.
J Control Release ; 371: 386-405, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844177

ABSTRACT

Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.


Subject(s)
Bone Morphogenetic Protein 4 , Cell Differentiation , Gelatin , Hematopoietic Stem Cells , Induced Pluripotent Stem Cells , Spheroids, Cellular , Stem Cell Factor , Bone Morphogenetic Protein 4/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Stem Cell Factor/metabolism , Gelatin/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Animals , Humans , Mice
14.
Biomaterials ; 311: 122681, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38944968

ABSTRACT

Cell-laden bioprinting is a promising biofabrication strategy for regenerating bioactive transplants to address organ donor shortages. However, there has been little success in reproducing transplantable artificial organs with multiple distinctive cell types and physiologically relevant architecture. In this study, an omnidirectional printing embedded network (OPEN) is presented as a support medium for embedded 3D printing. The medium is state-of-the-art due to its one-step preparation, fast removal, and versatile ink compatibility. To test the feasibility of OPEN, exceptional primary mouse hepatocytes (PMHs) and endothelial cell line-C166, were used to print hepatospheroid-encapsulated-artificial livers (HEALs) with vein structures following predesigned anatomy-based printing paths in OPEN. PMHs self-organized into hepatocyte spheroids within the ink matrix, whereas the entire cross-linked structure remained intact for a minimum of ten days of cultivation. Cultivated HEALs maintained mature hepatic functions and marker gene expression at a higher level than conventional 2D and 3D conditions in vitro. HEALs with C166-laden vein structures promoted endogenous neovascularization in vivo compared with hepatospheroid-only liver prints within two weeks of transplantation. Collectively, the proposed platform enables the manufacture of bioactive tissues or organs resembling anatomical architecture, and has broad implications for liver function replacement in clinical applications.


Subject(s)
Bioprinting , Hepatic Veins , Hepatocytes , Liver , Neovascularization, Physiologic , Printing, Three-Dimensional , Spheroids, Cellular , Animals , Bioprinting/methods , Hepatocytes/cytology , Mice , Spheroids, Cellular/cytology , Liver/cytology , Liver Transplantation , Liver, Artificial , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cell Line , Mice, Inbred C57BL , Male
15.
J Vis Exp ; (207)2024 May 31.
Article in English | MEDLINE | ID: mdl-38884490

ABSTRACT

Angiogenesis plays a crucial role in both physiological and pathological processes within the body including tumor growth or neovascular eye disease. A detailed understanding of the underlying molecular mechanisms and reliable screening models are essential for targeting diseases effectively and developing new therapeutic options. Several in vitro assays have been developed to model angiogenesis, capitalizing on the opportunities a controlled environment provides to elucidate angiogenic drivers at a molecular level and screen for therapeutic targets. This study presents workflows for investigating angiogenesis in vitro using human umbilical vein endothelial cells (HUVECs). We detail a scratch wound migration assay utilizing a live cell imaging system measuring endothelial cell migration in a 2D setting and the spheroid sprouting assay assessing endothelial cell sprouting in a 3D setting provided by a collagen matrix. Additionally, we outline strategies for sample preparation to enable further molecular analyses such as transcriptomics, particularly in the 3D setting, including RNA extraction as well as immunocytochemistry. Altogether, this framework offers scientists a reliable and versatile toolset to pursue their scientific inquiries in in vitro angiogenesis assays.


Subject(s)
Cell Movement , Human Umbilical Vein Endothelial Cells , Neovascularization, Physiologic , Humans , Neovascularization, Physiologic/physiology , Cell Movement/physiology , Spheroids, Cellular/cytology , Angiogenesis
16.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891883

ABSTRACT

Articular cartilage damage still remains a major problem in orthopedical surgery. The development of tissue engineering techniques such as autologous chondrocyte implantation is a promising way to improve clinical outcomes. On the other hand, the clinical application of autologous chondrocytes has considerable limitations. Mesenchymal stromal cells (MSCs) from various tissues have been shown to possess chondrogenic differentiation potential, although to different degrees. In the present study, we assessed the alterations in chondrogenesis-related gene transcription rates and extracellular matrix deposition levels before and after the chondrogenic differentiation of MSCs in a 3D spheroid culture. MSCs were obtained from three different tissues: umbilical cord Wharton's jelly (WJMSC-Wharton's jelly mesenchymal stromal cells), adipose tissue (ATMSC-adipose tissue mesenchymal stromal cells), and the dental pulp of deciduous teeth (SHEDs-stem cells from human exfoliated deciduous teeth). Monolayer MSC cultures served as baseline controls. Newly formed 3D spheroids composed of MSCs previously grown in 2D cultures were precultured for 2 days in growth medium, and then, chondrogenic differentiation was induced by maintaining them in the TGF-ß1-containing medium for 21 days. Among the MSC types studied, WJMSCs showed the most similarities with primary chondrocytes in terms of the upregulation of cartilage-specific gene expression. Interestingly, such upregulation occurred to some extent in all 3D spheroids, even prior to the addition of TGF-ß1. These results confirm that the potential of Wharton's jelly is on par with adipose tissue as a valuable cell source for cartilage engineering applications as well as for the treatment of osteoarthritis. The 3D spheroid environment on its own acts as a trigger for the chondrogenic differentiation of MSCs.


Subject(s)
Cell Differentiation , Chondrocytes , Chondrogenesis , Extracellular Matrix , Mesenchymal Stem Cells , Spheroids, Cellular , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Humans , Chondrogenesis/genetics , Extracellular Matrix/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Cells, Cultured , Wharton Jelly/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Cell Culture Techniques/methods , Tissue Engineering/methods , Cartilage/cytology , Cartilage/metabolism , Tooth, Deciduous/cytology , Tooth, Deciduous/metabolism , Dental Pulp/cytology , Dental Pulp/metabolism
17.
Biofabrication ; 16(3)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38865994

ABSTRACT

The lack of adequate humanin vitromodels that recapitulate the cellular composition and response of the human liver to injury hampers the development of anti-fibrotic drugs. The goal of this study was to develop a human spheroid culture model to study liver fibrosis by using induced pluripotent stem cell (iPSC)-derived liver cells. iPSCs were independently differentiated towards hepatoblasts (iHepatoblasts), hepatic stellate cells (iHSCs), endothelial cells (iECs) and macrophages (iMΦ), before assembly into free floating spheroids by culturing cells in 96-well U-bottom plates and orbital shaking for up to 21 days to allow further maturation. Through transcriptome analysis, we show further maturation of iECs and iMΦ, the differentiation of the iHepatoblasts towards hepatocyte-like cells (iHeps) and the inactivation of the iHSCs by the end of the 3D culture. Moreover, these cultures display a similar expression of cell-specific marker genes (CYP3A4, PDGFRß, CD31andCD68) and sensitivity to hepatotoxicity as spheroids made using freshly isolated primary human liver cells. Furthermore, we show the functionality of the iHeps and the iHSCs by mimicking liver fibrosis through iHep-induced iHSC activation, using acetaminophen. In conclusion, we have established a reproducible human iPSC-derived liver culture model that can be used to mimic fibrosisin vitroas a replacement of primary human liver derived 3D models. The model can be used to investigate pathways involved in fibrosis development and to identify new targets for chronic liver disease therapy.


Subject(s)
Cell Differentiation , Coculture Techniques , Induced Pluripotent Stem Cells , Liver Cirrhosis , Liver , Spheroids, Cellular , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Spheroids, Cellular/pathology , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Liver/pathology , Liver/cytology , Models, Biological , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/pathology , Cells, Cultured
18.
Adipocyte ; 13(1): 2347215, 2024 12.
Article in English | MEDLINE | ID: mdl-38864486

ABSTRACT

Adipose tissue plays a crucial role in metabolic syndrome, autoimmune diseases, and many cancers. Because of adipose's role in so many aspects of human health, there is a critical need for in vitro models that replicate adipose architecture and function. Traditional monolayer models, despite their convenience, are limited, showing heterogeneity and functional differences compared to 3D models. While monolayer cultures struggle with detachment and inefficient differentiation, healthy adipocytes in 3D culture accumulate large lipid droplets, secrete adiponectin, and produce low levels of inflammatory cytokines. The shift from monolayer models to more complex 3D models aims to better replicate the physiology of healthy adipose tissue in culture. This study introduces a simple and accessible protocol for generating adipose organoids using a scaffold-free spheroid model. The method, utilizing either 96-well spheroid plates or agarose micromolds, demonstrates increased throughput, uniformity, and ease of handling compared to previous techniques. This protocol allows for diverse applications, including drug testing, toxin screening, tissue engineering, and co-culturing. The choice between the two methods depends on the experimental goals, with the 96-well plate providing individualized control and the micromold offering scale advantages. The outlined protocol covers isolation, expansion, and characterization of stromal vascular fraction cells, followed by detailed steps for spheroid formation and optional downstream analyses.


Subject(s)
Adipocytes , Adipose Tissue , Spheroids, Cellular , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Humans , Adipocytes/metabolism , Adipocytes/cytology , Cell Culture Techniques/methods , Animals , Tissue Engineering/methods , Cells, Cultured , Cell Differentiation , Mice
19.
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
20.
ACS Appl Bio Mater ; 7(6): 3766-3776, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38729097

ABSTRACT

Cadherin-mediated tension at adherens junctions (AJs) is fundamental for cell-cell adhesion and maintaining epithelial integrity. Despite the importance of manipulating AJs to dissect cell-cell interactions, existing three-dimensional (3D) multicellular models have not adequately addressed the precise manipulation of these junctions. To fill this gap, we introduce E-cadherin-modified tension gauge tethers (TGTs) at the junctions within spheroids. The system enables both quantification and modulation of junctional tension with specific DNA triggers. Using rupture-induced fluorescence, we successfully measure mechanical forces in 3D spheroids. Furthermore, mechanically strong TGTs can maintain normal E-cadherin-mediated adhesion. Employing toehold-mediated strand displacement allowed us to disrupt E-cadherin-specific cell-cell adhesion, consequently altering intracellular tension within the spheroids. Our methodology offers a robust and precise way to manipulate cell-cell adhesion and intracellular mechanics in spheroid models.


Subject(s)
Cadherins , Cell Adhesion , Spheroids, Cellular , Cadherins/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Adherens Junctions/metabolism , Materials Testing , Particle Size
SELECTION OF CITATIONS
SEARCH DETAIL