Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.104
Filter
1.
Methods Mol Biol ; 2833: 11-21, 2024.
Article in English | MEDLINE | ID: mdl-38949696

ABSTRACT

In vitro biofilm models have allowed researchers to investigate the role biofilms play in the pathogenesis, virulence, and antimicrobial drug susceptibility of a wide range of bacterial pathogens. Rotary cell culture systems create three-dimensional cellular structures, primarily applied to eukaryotic organoids, that better capture characteristics of the cells in vivo. Here, we describe how to apply a low-shear, detergent-free rotary cell culture system to generate biofilms of Mycobacterium bovis BCG. The three-dimensional biofilm model forms mycobacterial cell aggregates in suspension as surface-detached biomass, without severe nutrient starvation or environmental stress, that can be harvested for downstream experiments. Mycobacterium bovis BCG derived from cell clusters display antimicrobial drug tolerance, presence of an extracellular matrix, and evidence of cell wall remodeling, all features of biofilm-associated bacteria that may be relevant to the treatment of tuberculosis.


Subject(s)
Biofilms , Mycobacterium bovis , Biofilms/drug effects , Biofilms/growth & development , Mycobacterium bovis/growth & development , Mycobacterium bovis/drug effects , Mycobacterium bovis/physiology , Cell Culture Techniques/methods , Cell Culture Techniques, Three Dimensional/methods
2.
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
3.
J Vis Exp ; (208)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38975788

ABSTRACT

Tendons and ligaments (T/L) are strong hierarchically organized structures uniting the musculoskeletal system. These tissues have a strictly arranged collagen type I-rich extracellular matrix (ECM) and T/L-lineage cells mainly positioned in parallel rows. After injury, T/L require a long time for rehabilitation with high failure risk and often unsatisfactory repair outcomes. Despite recent advancements in T/L biology research, one of the remaining challenges is that the T/L field still lacks a standardized differentiation protocol that is able to recapitulate T/L formation process in vitro. For example, bone and fat differentiation of mesenchymal precursor cells require just standard two-dimensional (2D) cell culture and the addition of specific stimulation media. For differentiation to cartilage, three-dimensional (3D) pellet culture and supplementation of TGFß is necessary. However, cell differentiation to tendon needs a very orderly 3D culture model, which ideally should also be subjectable to dynamic mechanical stimulation. We have established a 3-step (expansion, stimulation, and maturation) organoid model to form a 3D rod-like structure out of a self-assembled cell sheet, which delivers a natural microenvironment with its own ECM, autocrine, and paracrine factors. These rod-like organoids have a multi-layered cellular architecture within rich ECM and can be handled quite easily for exposure to static mechanical strain. Here, we demonstrated the 3-step protocol by using commercially available dermal fibroblasts. We could show that this cell type forms robust and ECM-abundant organoids. The described procedure can be further optimized in terms of culture media and optimized toward dynamic axial mechanical stimulation. In the same way, alternative cell sources can be tested for their potential to form T/L organoids and thus undergo T/L differentiation. In sum, the established 3D T/L organoid approach can be used as a model for tendon basic research and even for scaffold-free T/L engineering.


Subject(s)
Cell Culture Techniques , Fibroblasts , Ligaments , Organoids , Tendons , Humans , Tendons/cytology , Fibroblasts/cytology , Organoids/cytology , Ligaments/cytology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Dermis/cytology
4.
J Vis Exp ; (208)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38975789

ABSTRACT

Lens epithelial cells (LECs) play multiple important roles in maintaining the homeostasis and normal function of the lens. LECs determine lens growth, development, size, and transparency. Conversely, dysfunctional LECs can lead to cataract formation and posterior capsule opacification (PCO). Consequently, establishing a robust primary LEC culture system is important to researchers engaged in lens development, biochemistry, cataract therapeutics, and PCO prevention. However, cultivating primary LECs has long presented challenges due to their limited availability, slow proliferation rate, and delicate nature. This study addresses these hurdles by presenting a comprehensive protocol for primary LEC culture. The protocol encompasses essential steps such as the formulation of an optimized culture medium, precise isolation of lens capsules, trypsinization techniques, subculture procedures, harvest protocols, and guidelines for storage and shipment. Throughout the culture process, cell morphology was monitored using phase-contrast microscopy. To confirm the authenticity of the cultured LECs, immunofluorescence assays were conducted to detect the presence and subcellular distribution of critical lens proteins, namely αA- and γ-crystallins. This detailed protocol equips researchers with a valuable resource for cultivating and characterizing primary LECs, enabling advancements in our comprehension of lens biology and the development of therapeutic strategies for lens-related disorders.


Subject(s)
Epithelial Cells , Lens, Crystalline , Trypsin , Epithelial Cells/cytology , Lens, Crystalline/cytology , Animals , Mice , Trypsin/chemistry , Trypsin/metabolism , Cell Culture Techniques/methods , Primary Cell Culture/methods
5.
Stem Cell Res Ther ; 15(1): 191, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956608

ABSTRACT

BACKGROUND: Stem cell-derived therapies hold the potential for treatment of regenerative clinical indications. Static culture has a limited ability to scale up thus restricting its use. Suspension culturing can be used to produce target cells in large quantities, but also presents challenges related to stress and aggregation stability. METHODS: Utilizing a design of experiments (DoE) approach in vertical wheel bioreactors, we evaluated media additives that have versatile properties. The additives evaluated are Heparin sodium salt (HS), polyethylene glycol (PEG), poly (vinyl alcohol) (PVA), Pluronic F68 and dextran sulfate (DS). Multiple response variables were chosen to assess cell growth, pluripotency maintenance and aggregate stability in response to the additive inputs, and mathematical models were generated and tuned for maximal predictive power. RESULTS: Expansion of iPSCs using 100 ml vertical wheel bioreactor assay for 4 days on 19 different media combinations resulted in models that can optimize pluripotency, stability, and expansion. The expansion optimization resulted in the combination of PA, PVA and PEG with E8. This mixture resulted in an expansion doubling time that was 40% shorter than that of E8 alone. Pluripotency optimizer highlighted the importance of adding 1% PEG to the E8 medium. Aggregate stability optimization that minimizes aggregate fusion in 3D culture indicated that the interaction of both Heparin and PEG can limit aggregation as well as increase the maintenance capacity and expansion of hiPSCs, suggesting that controlling fusion is a critical parameter for expansion and maintenance. Validation of optimized solution on two cell lines in bioreactors with decreased speed of 40 RPM, showed consistency and prolonged control over aggregates that have high frequency of pluripotency markers of OCT4 and SOX2 (> 90%). A doubling time of around 1-1.4 days was maintained after passaging as clumps in the optimized medium. Controlling aggregate fusion allowed for a decrease in bioreactor speed and therefore shear stress exerted on the cells in a large-scale expansion. CONCLUSION: This study resulted in a control of aggregate size within suspension cultures, while informing about concomitant state control of the iPSC state. Wider application of this approach can address media optimization complexity and bioreactor scale-up challenges.


Subject(s)
Bioreactors , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Cell Culture Techniques/methods , Cell Proliferation , Cell Aggregation/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Cell Differentiation
6.
J Transl Med ; 22(1): 614, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956643

ABSTRACT

Mesenchymal stem and stromal cells (MSCs) hold potential to treat a broad range of clinical indications, but clinical translation has been limited to date due in part to challenges with batch-to-batch reproducibility of potential critical quality attributes (pCQAs) that can predict potency/efficacy. Here, we designed and implemented a microcarrier-microbioreactor approach to cell therapy manufacturing, specific to anchorage-dependent cells such as MSCs. We sought to assess whether increased control of the biochemical and biophysical environment had the potential to create product with consistent presentation and elevated expression of pCQAs relative to established manufacturing approaches in tissue culture polystyrene (TCPS) flasks. First, we evaluated total cell yield harvested from dissolvable, gelatin microcarriers within a microbioreactor cassette (Mobius Breez) or a flask control with matched initial cell seeding density and culture duration. Next, we identified 24 genes implicated in a therapeutic role for a specific motivating indication, acute respiratory distress syndrome (ARDS); expression of these genes served as our pCQAs for initial in vitro evaluation of product potency. We evaluated mRNA expression for three distinct donors to assess inter-donor repeatability, as well as for one donor in three distinct batches to assess within-donor, inter-batch variability. Finally, we assessed gene expression at the protein level for a subset of the panel to confirm successful translation. Our results indicated that MSCs expanded with this microcarrier-microbioreactor approach exhibited reasonable donor-to-donor repeatability and reliable batch-to-batch reproducibility of pCQAs. Interestingly, the baseline conditions of this microcarrier-microbioreactor approach also significantly improved expression of several key pCQAs at the gene and protein expression levels and reduced total media consumption relative to TCPS culture. This proof-of-concept study illustrates key benefits of this approach to therapeutic cell process development for MSCs and other anchorage-dependent cells that are candidates for cell therapies.


Subject(s)
Bioreactors , Mesenchymal Stem Cells , Respiratory Distress Syndrome , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Humans , Respiratory Distress Syndrome/therapy , Gene Expression Regulation , Cell Culture Techniques/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
7.
Sci Rep ; 14(1): 15592, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971841

ABSTRACT

The production of cultured red blood cells (cRBC) for transfusion purposes requires large scale cultures and downstream processes to purify enucleated cRBC. The membrane composition, and cholesterol content in particular, are important during proliferation of (pro)erythroblasts and for cRBC quality. Therefore, we tested the requirement for cholesterol in the culture medium during expansion and differentiation of erythroid cultures with respect to proliferation, enucleation and purification by filtration. The low cholesterol level (22 µg/dl) in serum free medium was sufficient to expand (pro)erythroblast cultures. Addition of 2.0 or 5.0 mg/dL of free cholesterol at the start of differentiation induction inhibited enucleation compared to the default condition containing 3.3 mg/dl total cholesterol derived from the addition of Omniplasma to serum free medium. Addition of 5.0 mg/dl cholesterol at day 5 of differentiation did not affect the enucleation process but significantly increased recovery of enucleated cRBC following filtration over leukodepletion filters. The addition of cholesterol at day 5 increased the osmotic resistance of cRBC. In conclusion, cholesterol supplementation after the onset of enucleation improved the robustness of cRBC and increased the yield of enucleated cRBC in the purification process.


Subject(s)
Cholesterol , Culture Media , Erythrocytes , Cholesterol/metabolism , Humans , Erythrocytes/metabolism , Culture Media/chemistry , Cells, Cultured , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Culture Techniques/methods , Erythroblasts/metabolism , Erythroblasts/cytology , Culture Media, Serum-Free
8.
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
9.
Methods Mol Biol ; 2829: 3-11, 2024.
Article in English | MEDLINE | ID: mdl-38951323

ABSTRACT

Healthy insect cell cultures are critical for any method described in this book, including making productive baculovirus banks, protein or AAV expression, and determining viral titers. This chapter describes cell maintenance in shake flasks using serum-free conditions and the expansion of virus stocks from a single plaque purified virus. Insect cells can be passaged over multiple generations, but as the cells may undergo changes over multiple passages, limiting the use of your cells to a defined number of passages such as 50 passages is recommendable. Baculovirus stocks once created using serum-free media are not very stable at 4-8 °C. This chapter also includes a simple method to store cells from an early cell passage and your virus stock in liquid nitrogen.


Subject(s)
Baculoviridae , Cell Culture Techniques , Animals , Baculoviridae/genetics , Cell Culture Techniques/methods , Insecta/virology , Insecta/cytology , Cell Line
10.
Methods Mol Biol ; 2829: 49-66, 2024.
Article in English | MEDLINE | ID: mdl-38951326

ABSTRACT

This chapter outlines the workflow using the ExpiSf™ Expression System designed for high-density infection of suspension ExpiSf9™ cells. The system utilizes a chemically defined, serum-free, protein-free, and animal origin free medium, making it suitable for recombinant protein expression experiments. The ExpiSf™ chemically defined medium allows efficient transfection and baculovirus production directly within the same culture medium. The ExpiSf™ Expression System Starter Kit provides all necessary components, including cells, culture medium, and reagents needed to infect one (1) liter of cell culture. The system's versatility and animal origin free nature make it a valuable tool for various protein expression studies and biotechnological applications.


Subject(s)
Baculoviridae , Recombinant Proteins , Workflow , Animals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Baculoviridae/genetics , Transfection/methods , Culture Media/chemistry , Cell Culture Techniques/methods , Cell Line , Gene Expression
11.
Methods Mol Biol ; 2829: 79-90, 2024.
Article in English | MEDLINE | ID: mdl-38951328

ABSTRACT

Adaptive laboratory evolution (ALE) is a powerful tool for enhancing the fitness of cell lines in specific applications, including recombinant protein production. Through adaptation to nonstandard culture conditions, cells can develop specific traits that make them high producers. Despite being widely used for microorganisms and, to lesser extent, for mammalian cells, ALE has been poorly leveraged for insect cells. Here, we describe a method for adapting insect High Five and Sf9 cells to nonstandard culture conditions via an ALE approach. Aiming to demonstrate the potential of ALE to improve productivity of insect cells, two case studies are demonstrated. In the first, we adapted insect High Five cells from their standard pH (6.2) to neutral pH (7.0); this adaptation allowed to improve production of influenza virus-like particles (VLPs) by threefold, using the transient baculovirus expression vector system. In the second, we adapted insect Sf9 cells from their standard culture temperature (27 °C) to hypothermic growth (22 °C); this adaptation allowed to improve production of influenza VLPs by sixfold, using stable cell lines. These examples demonstrate the potential of ALE for enhancing productivity within distinct insect cell hosts and expression systems by manipulating different culture conditions.


Subject(s)
Recombinant Proteins , Animals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Cell Line , Sf9 Cells , Baculoviridae/genetics , Cell Culture Techniques/methods , Insecta/genetics , Insecta/cytology , Directed Molecular Evolution/methods , Hydrogen-Ion Concentration , Temperature
12.
Methods Mol Biol ; 2829: 277-286, 2024.
Article in English | MEDLINE | ID: mdl-38951344

ABSTRACT

Quantitative immunoassays, such as the traditional enzyme-linked immunosorbent assay (ELISA), are used to determine concentrations of an antigen in a matrix of unknown antigen concentration. Magnetic immunoassays, such as the Luminex xMAP technology, allow for the simultaneous detection of multiple analytes and offer heightened sensitivity, specificity, low sample volume requirements, and high-throughput capabilities. Here, we describe a quantitative immunoassay using the Luminex MAGPIX® System to determine the antigen concentration from liquid samples with unknown concentrations. In detail, we describe a newly developed assay for determining production yields of Drosophila S2-produced Marburg virus (MARV) glycoprotein in insect-cell-culture-derived supernatant. The potential applications of this assay could extend to the quantification of viral antigens in fluids derived from both in vitro and in vivo models infected with live MARV, thereby providing additional applications for virological research.


Subject(s)
Antigens, Viral , Microspheres , Animals , Immunoassay/methods , Antigens, Viral/immunology , Antigens, Viral/analysis , Marburgvirus/immunology , Marburgvirus/isolation & purification , Drosophila , Cell Culture Techniques/methods , Cell Line , Enzyme-Linked Immunosorbent Assay/methods
13.
Methods Mol Biol ; 2829: 329-339, 2024.
Article in English | MEDLINE | ID: mdl-38951347

ABSTRACT

Mammalian cell lines are one of the best options when it comes to the production of complex proteins requiring specific glycosylation patterns. Plasmid DNA transfection and stable cell lines are frequently used for recombinant protein production, but they are expensive at large scale or can become time-consuming, respectively. The BacMam baculovirus (BV) is a safe and cost-effective platform to produce recombinant proteins in mammalian cells. The process of generating BacMam BVs is straightforward and similar to the generation of "insect" BVs, with different commercially available platforms. Although there are several protocols that describe recombinant protein expression with the BacMam BV in adherent cell lines, limited information is available on suspension cells. Therefore, it is of relevance to define the conditions to produce recombinant proteins in suspension cell cultures with BacMam BVs that facilitate bioprocess transfer to larger volumes. Here, we describe a method to generate a high titer BacMam BV stock and produce recombinant proteins in suspension HEK293 cells.


Subject(s)
Baculoviridae , Recombinant Proteins , Baculoviridae/genetics , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/biosynthesis , HEK293 Cells , Animals , Transfection/methods , Genetic Vectors/genetics , Cell Culture Techniques/methods , Gene Expression , Glycosylation
14.
Biol Pharm Bull ; 47(7): 1241-1247, 2024.
Article in English | MEDLINE | ID: mdl-38945897

ABSTRACT

Primary hepatocytes are valuable for studying liver diseases, drug-induced liver injury, and drug metabolism. However, when cultured in a two-dimensional (2D) environment, primary hepatocytes undergo rapid dedifferentiation via an epithelial-mesenchymal transition (EMT) and lose their liver-specific functions. On the other hand, a three-dimensional (3D) culture of primary hepatocyte organoids presents challenges for analyzing cellular functions and molecular behaviors due to strong cell-cell adhesion among heterogeneous cells. In this study, we developed a novel dispersion culture method of hepatocytes within a dome-shaped collagen matrix, overcoming conventional limitations. The expression levels of EMT-related genes were lower in rat primary hepatocytes cultured using this method for 4 d than in cells cultured using the 2D method. Furthermore, albumin production, a marker of liver function, declined sharply in rat primary hepatocytes cultured in two dimensions from 6.40 µg/mL/48 h on day 4 to 1.35 µg/mL/48 h on day 8, and declined gradually from 4.92 µg/mL/48 h on day 8 to 3.89 µg/mL/48 h on day 14 in rat primary hepatocytes cultured using our new method. These findings indicate that the newly developed culture method can suppress EMT and maintain liver functions for 14 d in rat primary hepatocytes, potentially expanding the utility of primary hepatocyte cultured by using conventional 3D methods.


Subject(s)
Collagen , Epithelial-Mesenchymal Transition , Hepatocytes , Liver , Animals , Hepatocytes/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Cells, Cultured , Collagen/metabolism , Male , Liver/metabolism , Liver/cytology , Rats , Cell Culture Techniques/methods , Rats, Sprague-Dawley , Albumins/metabolism
15.
J Biotechnol ; 391: 33-39, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38838744

ABSTRACT

3D printing has become widespread for the manufacture of parts in various industries and enabled radically new designs. This trend has not spread to bioprocess development yet, due to a lack of material suitable for the current workflow, including sterilization by autoclaving. This work demonstrates that commercially available heat temperature stable poly-lactic acid (PLA) can be used to easily manufacture novel bioreactor vessels with included features like harvest tubes and 3D printed spargers. Temperature responsiveness was tested for PLA, temperature stable PLA (PLA-HP) and glass for temperatures relevant for insect and mammalian cell culture, including temperature shifts within the process. Stability at 27 °C and 37 °C as well as temperature shifts to 22 °C and 32 °C showed acceptable performance with slightly higher temperature overshoot for 3D printed vessels. A stable temperature is reached after 2 h for PLA, 3 h for PLA-HP and 1 h for glass reactors. Temperature can be maintained with a fluctuation of 0.1 °C for all materials. A 3D printed sparger design directly integrated into the vessel wall and bottom was tested under three different conditions (0.3 SLPH and 27 °C, 3 SLPH and 37 °C and 13 SLPH and 37 °C). The 3D printed sparger showed a better kLa than the L-Sparger with more pronounced differences for higher flowrates. An insect cell culture run in the novel vessel exhibited the same growth behavior as that in standard glass vessels, reaching the same maximum cell concentration. Being 3D printed from biodegradable materials, these bioreactors offer design flexibility for novel bioreactor formats. Additionally, their autoclavability allows seamless integration into standard workflows.


Subject(s)
Biocompatible Materials , Bioreactors , Polyesters , Printing, Three-Dimensional , Polyesters/chemistry , Animals , Biocompatible Materials/chemistry , Sterilization/methods , Temperature , Cell Culture Techniques/methods , Cell Culture Techniques/instrumentation , Cell Line
16.
Cells ; 13(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38920683

ABSTRACT

Over the past decade, the development of three-dimensional (3D) models has increased exponentially, facilitating the unravelling of fundamental and essential cellular mechanisms by which cells communicate with each other, assemble into tissues and organs and respond to biochemical and biophysical stimuli under both physiological and pathological conditions. This section presents a concise overview of the most recent updates on the significant contribution of different types of 3D cell cultures including spheroids, organoids and organ-on-chip and bio-printed tissues in advancing our understanding of cellular and molecular mechanisms. The case studies presented include the 3D cultures of breast cancer (BC), endometriosis, the liver microenvironment and infections. In BC, the establishment of 3D culture models has permitted the visualization of the role of cancer-associated fibroblasts in the delivery of exosomes, as well as the significance of the physical properties of the extracellular matrix in promoting cell proliferation and invasion. This approach has also become a valuable tool in gaining insight into general and specific mechanisms of drug resistance. Given the considerable heterogeneity of endometriosis, 3D models offer a more accurate representation of the in vivo microenvironment, thereby facilitating the identification and translation of novel targeted therapeutic strategies. The advantages provided by 3D models of the hepatic environment, in conjunction with the high throughput characterizing various platforms, have enabled the elucidation of complex molecular mechanisms underlying various threatening hepatic diseases. A limited number of 3D models for gut and skin infections have been developed. However, a more profound comprehension of the spatial and temporal interactions between microbes, the host and their environment may facilitate the advancement of in vitro, ex vivo and in vivo disease models. Additionally, it may pave the way for the development of novel therapeutic approaches in diverse research fields. The interested reader will also find concluding remarks on the challenges and prospects of using 3D cell cultures for discovering cellular and molecular mechanisms in the research areas covered in this review.


Subject(s)
Breast Neoplasms , Cell Culture Techniques, Three Dimensional , Endometriosis , Humans , Endometriosis/pathology , Endometriosis/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Cell Culture Techniques, Three Dimensional/methods , Communicable Diseases/metabolism , Communicable Diseases/pathology , Cell Culture Techniques/methods , Spheroids, Cellular/pathology , Spheroids, Cellular/metabolism , Liver/pathology , Liver/metabolism , Organoids/metabolism , Organoids/pathology , Liver Diseases/pathology , Liver Diseases/metabolism , Animals
17.
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
18.
Int J Nanomedicine ; 19: 6201-6228, 2024.
Article in English | MEDLINE | ID: mdl-38911499

ABSTRACT

Due to their ability to replicate the in vivo microenvironment through cell interaction and induce cells to stimulate cell function, three-dimensional cell culture models can overcome the limitations of two-dimensional models. Organoids are 3D models that demonstrate the ability to replicate the natural structure of an organ. In most organoid tissue cultures, matrigel made of a mouse tumor extracellular matrix protein mixture is an essential ingredient. However, its tumor-derived origin, batch-to-batch variation, high cost, and safety concerns have limited the usefulness of organoid drug development and regenerative medicine. Its clinical application has also been hindered by the fact that organoid generation is dependent on the use of poorly defined matrices. Therefore, matrix optimization is a crucial step in developing organoid culture that introduces alternatives as different materials. Recently, a variety of substitute materials has reportedly replaced matrigel. The purpose of this study is to review the significance of the latest advances in materials for cell culture applications and how they enhance build network systems by generating proper cell behavior. Excellence in cell behavior is evaluated from their cell characteristics, cell proliferation, cell differentiation, and even gene expression. As a result, graphene oxide as a matrix optimization demonstrated high potency in developing organoid models. Graphene oxide can promote good cell behavior and is well known for having good biocompatibility. Hence, advances in matrix optimization of graphene oxide provide opportunities for the future development of advanced organoid models.


Subject(s)
Graphite , Organoids , Organoids/drug effects , Organoids/cytology , Animals , Graphite/chemistry , Graphite/pharmacology , Humans , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Drug Combinations , Cell Culture Techniques/methods , Cell Culture Techniques, Three Dimensional/methods , Mice , Laminin/chemistry , Laminin/pharmacology , Collagen , Proteoglycans
19.
Mol Biol Rep ; 51(1): 781, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38913199

ABSTRACT

Mesenchymal Stem Cells, mesodermal origin and multipotent stem cells, have ability to differentiate into vascular endothelial cells. The cells are squamous in morphology, inlining, and protecting blood vessel tissue, as well as maintaining homeostatic conditions. ECs are essential in vascularization and blood vessels formation. The differentiation process, generally carried out in 2D culture systems, were relied on growth factors induction. Therefore, an artificial extracellular matrix with relevant mechanical properties is essential to build 3D culture models. Various 3D fabrication techniques, such as hydrogel-based and fibrous scaffolds, scaffold-free, and co-culture to endothelial cells were reviewed and summarized to gain insights. The obtained MSCs-derived ECs are shown by the expression of endothelial gene markers and tubule-like structure. In order to mimicking relevant vascular tissue, 3D-bioprinting facilitates to form more complex microstructures. In addition, a microfluidic chip with adequate flow rate allows medium perfusion, providing mechanical cues like shear stress to the artificial vascular vessels.


Subject(s)
Cell Culture Techniques, Three Dimensional , Cell Differentiation , Endothelial Cells , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Cell Culture Techniques, Three Dimensional/methods , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Animals , Hydrogels/chemistry , Cell Culture Techniques/methods , Coculture Techniques/methods , Extracellular Matrix/metabolism
20.
Sheng Wu Gong Cheng Xue Bao ; 40(6): 1935-1949, 2024 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-38914502

ABSTRACT

Plant synthetic biology has significant theoretical advantages in exploration and production of plant natural products. However, its contribution to the field of biosynthesis is currently limited due to the lack of efficient chassis systems and related enabling technologies. Synthetic biologists often avoid tobacco as a chassis system because of its long operation cycle, difficulties in genetic and metabolic modification, complex metabolism and purification background, nicotine toxicity, and challenges in accurately controlling for agricultural production. Nevertheless, the tobacco suspension cell chassis system offers a viable solution to these challenges. The objective of this research was to develop a tobacco suspension cell chassis with high scientific and industrial potential. This chassis should exhibit rapid growth, high biomass, excellent dispersion, high transformation efficiency, and minimal nicotine content. Nicotiana benthamiana, which has high applicability in molecular technology, was used to induce suspension cells. The induced suspension cells, named NBS-1, exhibited rapid growth, excellent dispersion, and high biomass, reaching a maximum biomass of 476.39 g/L (fresh weight), which was significantly higher than that of BY-2. The transformation efficiency of the widely utilized pEAQ-HT transient expression system in NBS-1 reached 81%, which was substantially elevated compared to BY-2. The metabolic characteristics and bias of BY-2 and NBS-1 were analyzed using transcriptome data. It was found that the gene expression of pathways related to biosynthesis of flavonoids and their derivatives in NBS-1 was significantly higher, while the pathways related to alkaloid biosynthesis were significantly lower compared to BY-2. These findings were further validated by the total content of flavonoid and alkaloid. In summary, our research demonstrates NBS-1 possesses minimal nicotine content and provides valuable guidance for selecting appropriate chassis for specific products. In conclusion, this study developed NBS-1, a tobacco suspension cell chassis with excellent growth and transformation, high flavonoid content and minimal nicotine content, which has important guiding significance for the development of tobacco suspension cell chassis.


Subject(s)
Nicotiana , Nicotiana/metabolism , Nicotiana/genetics , Synthetic Biology , Plants, Genetically Modified/metabolism , Metabolic Engineering/methods , Cell Culture Techniques/methods , Nicotine/metabolism , Nicotine/biosynthesis , Biomass
SELECTION OF CITATIONS
SEARCH DETAIL
...