Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 382
Filter
1.
Methods Mol Biol ; 2805: 19-30, 2024.
Article in English | MEDLINE | ID: mdl-39008172

ABSTRACT

Transformed lung organoids have extensive applications in lung cancer modeling and drug screening. Traditional two-dimensional (2D) cultures fail to propagate a large subpopulation of murine primary tumors in vitro. However, three-dimensional (3D) air-liquid interface (ALI) cultures, which are employed to grow normal lung organoids, can be used to efficiently culture cancerous lung tumor cells. Here, we detail a procedure for cultivating genetically modified lung organoids in 3D-ALI cultures. This protocol contains two parts. The first part describes how to transduce lung epithelial cells, which are either freshly sorted from lungs or from actively growing murine organoids, with virus in order to modify gene expression. The target lung cells are incubated with virus for 1-2 h for transduction. Then, the transduced cells are thoroughly washed and mixed with stromal support cells and Matrigel and are loaded into transwell inserts for culture and validated for genetic modifications through downstream assays. The second part describes how to isolate tumor cells growing orthotopically in genetically engineered mouse models to produce organoid cell lines that can be used for ex vivo drug discovery assays. For this protocol, tumors are isolated from lungs of mice, finely chopped and washed. Then, tumor chunks are mixed with Matrigel for 3D-ALI culture. Finally, organoids budding from tumor chunks are trypsinized and passaged to establish an organoid line. Together these two protocols provide a promising platform to study the genesis, progression, and treatment of lung cancer.


Subject(s)
Lung Neoplasms , Lung , Organoids , Organoids/cytology , Animals , Mice , Lung/cytology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Cell Culture Techniques, Three Dimensional/methods , Humans , Cell Culture Techniques/methods , Epithelial Cells/cytology , Transduction, Genetic/methods
2.
Methods Mol Biol ; 2805: 31-50, 2024.
Article in English | MEDLINE | ID: mdl-39008173

ABSTRACT

Cell patterning for 3D culture has increased our understanding of how cells interact among themselves and with their environment during tissue morphogenesis. Building cell communities from the bottom up with size and compositional control is invaluable for studies of morphological transitions. Here, we detail Photolithographic DNA-programmed Assembly of Cells (pDPAC). pDPAC uses a photoactive polyacrylamide gel substrate to capture single-stranded DNA on a 2D surface in large-scale, highly resolved patterns using the photomask technology. Cells are then functionalized with a complementary DNA strand, enabling cells to be temporarily adhered to distinct locations only where their complementary strand is patterned. These temporary 2D patterns can be transferred to extracellular matrix hydrogels for 3D culture of cells in biomimetic microenvironments. Use of a polyacrylamide substrate has advantages, including a simpler photolithography workflow, lower non-specific cell adhesion, and lower stiction to ECM hydrogels during release of patterned hydrogels. The protocol is equally applicable to large (cm)-scale patterns and repetitive arrays of smaller-scale cell interaction or migration experiments.


Subject(s)
Hydrogels , Tissue Engineering , Hydrogels/chemistry , Humans , Tissue Engineering/methods , Acrylic Resins/chemistry , Cell Adhesion , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Cell Culture Techniques/methods , Animals , Cell Culture Techniques, Three Dimensional/methods
3.
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
4.
Biofabrication ; 16(4)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38955197

ABSTRACT

Plasma cells (PCs) in bone marrow (BM) play an important role in both protective and pathogenic humoral immune responses, e.g. in various malignant and non-malignant diseases such as multiple myeloma, primary and secondary immunodeficiencies and autoimmune diseases. Dedicated microenvironmental niches in the BM provide PCs with biomechanical and soluble factors that support their long-term survival. There is a high need for appropriate and robust model systems to better understand PCs biology, to develop new therapeutic strategies for PCs-related diseases and perform targeted preclinical studies with high predictive value. Most preclinical data have been derived fromin vivostudies in mice, asin vitrostudies of human PCs are limited due to restricted survival and functionality in conventional 2D cultures that do not reflect the unique niche architecture of the BM. We have developed a microphysiological, dynamic 3D BM culture system (BM-MPS) based on human primary tissue (femoral biopsies), mechanically supported by a hydrogel scaffold casing. While a bioinert agarose casing did not support PCs survival, a photo-crosslinked collagen-hyaluronic acid (Col-HA) hydrogel preserved the native BM niche architecture and allowed PCs survivalin vitrofor up to 2 weeks. Further, the Col-HA hydrogel was permissive to lymphocyte migration into the microphysiological system´s circulation. Long-term PCs survival was related to the stable presence in the culture of soluble factors, as APRIL, BAFF, and IL-6. Increasing immunoglobulins concentrations in the medium confirm their functionality over culture time. To the best of our knowledge, this study is the first report of successful long-term maintenance of primary-derived non-malignant PCsin vitro. Our innovative model system is suitable for in-depthin vitrostudies of human PCs regulation and exploration of targeted therapeutic approaches such as CAR-T cell therapy or biologics.


Subject(s)
Hydrogels , Plasma Cells , Humans , Plasma Cells/cytology , Plasma Cells/metabolism , Hydrogels/chemistry , Cell Survival/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Bone Marrow Cells/cytology , Collagen/chemistry , Bone Marrow/metabolism , Cells, Cultured , Cell Culture Techniques, Three Dimensional , Models, Biological , Tissue Scaffolds/chemistry , Sepharose/chemistry
5.
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
6.
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
7.
Sci Rep ; 14(1): 15556, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969656

ABSTRACT

Previously, we reported successful cellular expansion of a murine colorectal carcinoma cell line (CT-26) using a three-dimensional (3D) engineered extracellular matrix (EECM) fibrillar scaffold structure. CCL-247 were grown over a limited time period of 8 days on 3D EECM or tissue culture polystyrene (TCPS). Cells were then assayed for growth, electroporation efficiency and Vigil manufacturing release criteria. Using EECM scaffolds, we report an expansion of CCL-247 (HCT116), a colorectal carcinoma cell line, from a starting concentration of 2.45 × 105 cells to 1.9 × 106 cells per scaffold. Following expansion, 3D EECM-derived cells were assessed based on clinical release criteria of the Vigil manufacturing process utilized for Phase IIb trial operation with the FDA. 3D EECM-derived cells passed all Vigil manufacturing release criteria including cytokine expression. Here, we demonstrate successful Vigil product manufacture achieving the specifications necessary for the clinical trial product release of Vigil treatment. Our results confirm that 3D EECM can be utilized for the expansion of human cancer cell CCL-247, justifying further clinical development involving human tissue sample manufacturing including core needle biopsy and minimal ascites samples.


Subject(s)
Extracellular Matrix , Immunotherapy , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Immunotherapy/methods , Tissue Engineering/methods , HCT116 Cells , Colorectal Neoplasms/pathology , Animals , Mice , Cell Proliferation , Cell Line, Tumor , Cell Culture Techniques, Three Dimensional/methods
8.
Biotechnol J ; 19(6): e2400159, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38896414

ABSTRACT

The liver is one of the most important organs in the human body. It performs many important functions, including being responsible for the metabolism of most drugs, which is often associated with its drug-induced damage. Currently, there are no ideal pharmacological models that would allow the evaluation of the effect of newly tested drugs on the liver in preclinical studies. Moreover, the influence of hepatic metabolism on the effectiveness of the tested drugs is rarely evaluated. Therefore, in this work we present an advanced model of the liver, which reflects most of the morphologically and metabolically important features of the liver in vivo, namely: three-dimensionality, cellular composition, presence of extracellular matrix, distribution of individual cell types in the structure of the liver model, high urea and albumin synthesis efficiency, high cytochrome p450 activity. In addition, the work, based on the example of commonly used anticancer drugs, shows how important it is to take into account hepatic metabolism in the effective assessment of their impact on the target organ, in this case cancer. In our research, we have shown that the most similar to liver in vivo are 3D cellular aggregates composed of three important liver cells, namely hepatocytes (HepG2), hepatic stellate cells (HSCs), and hepatic sinusoidal endothelial cells (HSECs). Moreover, we showed that the cells in 3D aggregate structure need time (cell-cell interactions) to improve proper liver characteristic. The triculture model additionally showed the greatest ability to metabolize selected anticancer drugs.


Subject(s)
Antineoplastic Agents , Liver , Humans , Antineoplastic Agents/pharmacology , Liver/metabolism , Liver/drug effects , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Models, Biological , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cell Culture Techniques, Three Dimensional/methods
9.
J Am Heart Assoc ; 13(13): e033155, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38934864

ABSTRACT

BACKGROUND: Current protocols generate highly pure human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in vitro that recapitulate characteristics of mature in vivo cardiomyocytes. Yet, a risk of arrhythmias exists when hiPSC-CMs are injected into large animal models. Thus, understanding hiPSC-CM maturational mechanisms is crucial for clinical translation. Forkhead box (FOX) transcription factors regulate postnatal cardiomyocyte maturation through a balance between FOXO and FOXM1. We also previously demonstrated that p53 activation enhances hiPSC-CM maturation. Here, we investigate whether p53 activation modulates the FOXO/FOXM1 balance to promote hiPSC-CM maturation in 3-dimensional suspension culture. METHODS AND RESULTS: Three-dimensional cultures of hiPSC-CMs were treated with Nutlin-3a (p53 activator, 10 µM), LOM612 (FOXO relocator, 5 µM), AS1842856 (FOXO inhibitor, 1 µM), or RCM-1 (FOXM1 inhibitor, 1 µM), starting 2 days after onset of beating, with dimethyl sulfoxide (0.2% vehicle) as control. P53 activation promoted hiPSC-CM metabolic and electrophysiological maturation alongside FOXO upregulation and FOXM1 downregulation, in n=3 to 6 per group for all assays. FOXO inhibition significantly decreased expression of cardiac-specific markers such as TNNT2. In contrast, FOXO activation or FOXM1 inhibition promoted maturational characteristics such as increased contractility, oxygen consumption, and voltage peak maximum upstroke velocity, in n=3 to 6 per group for all assays. Further, by single-cell RNA sequencing of n=2 LOM612-treated cells compared with dimethyl sulfoxide, LOM612-mediated FOXO activation promoted expression of cardiac maturational pathways. CONCLUSIONS: We show that p53 activation promotes FOXO and suppresses FOXM1 during 3-dimensional hiPSC-CM maturation. These results expand our understanding of hiPSC-CM maturational mechanisms in a clinically-relevant 3-dimensional culture system.


Subject(s)
Cell Differentiation , Forkhead Box Protein M1 , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Tumor Suppressor Protein p53 , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Forkhead Box Protein M1/metabolism , Forkhead Box Protein M1/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Signal Transduction , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics
10.
J Mater Chem B ; 12(25): 6063-6078, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38888153

ABSTRACT

Injectable hydrogels have attracted significant interest in the biomedical field due to their minimal invasiveness and accommodation of intricate scenes. Herein, we developed an injectable polyurethane-based thermogel platform by modulating the hydrophilic-hydrophobic balance of the segmented components with pendant PEG. The thermogelling behavior is achieved by a combination of the bridging from the hydrophilic PEG and the percolated network from the hydrophobic micelle core. Firstly, the thermogelation mechanism of this system was demonstrated by both DPD simulation and experimental investigation. The gelling temperature could be modulated by varying the solid content, the component of soft segments, and the length of the pendant PEG. We further applied 3D printing technology to prepare personalized hydrogel structures. This integration highlights the adaptability of our thermogel for fabricating complex and patient-specific constructs, presenting a significant advance in the field of regenerative medicine and tissue engineering. Subsequently, in vitro cell experiments demonstrated that the thermogel had good cell compatibility and could promote the proliferation and migration of L929 cells. Impressively, A549 cells could be expediently in situ parceled in the thermogel for three-dimensional cultivation and gain lifeful 3D cell spheres after 7 days. Further, in vivo experiments demonstrated that the thermogel could promote wound healing with the regeneration of capillaries and hair follicles. Ultimately, our study demonstrates the potential of hydrogels to prepare personalized hydrogel structures via 3D printing technology, offering innovative solutions for complex biomedical applications. This work not only provides a fresh perspective for the design of injectable thermogels but also offers a promising avenue to develop thermoresponsive waterborne polyurethane for various medical applications.


Subject(s)
Hydrogels , Micelles , Polyurethanes , Polyurethanes/chemistry , Humans , Animals , Hydrogels/chemistry , Mice , Wound Healing/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/chemical synthesis , Cell Proliferation/drug effects , Temperature , Cell Culture Techniques, Three Dimensional/methods , Injections , Cell Movement/drug effects , Particle Size
11.
PLoS Comput Biol ; 20(6): e1012112, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38861575

ABSTRACT

Cell sedimentation in 3D hydrogel cultures refers to the vertical migration of cells towards the bottom of the space. Understanding this poorly examined phenomenon may allow us to design better protocols to prevent it, as well as provide insights into the mechanobiology of cancer development. We conducted a multiscale experimental and mathematical examination of 3D cancer growth in triple negative breast cancer cells. Migration was examined in the presence and absence of Paclitaxel, in high and low adhesion environments and in the presence of fibroblasts. The observed behaviour was modeled by hypothesizing active migration due to self-generated chemotactic gradients. Our results did not reject this hypothesis, whereby migration was likely to be regulated by the MAPK and TGF-ß pathways. The mathematical model enabled us to describe the experimental data in absence (normalized error<40%) and presence of Paclitaxel (normalized error<10%), suggesting inhibition of random motion and advection in the latter case. Inhibition of sedimentation in low adhesion and co-culture experiments further supported the conclusion that cells actively migrated downwards due to the presence of signals produced by cells already attached to the adhesive glass surface.


Subject(s)
Cell Adhesion , Cell Movement , Paclitaxel , Humans , Cell Adhesion/physiology , Cell Movement/physiology , Paclitaxel/pharmacology , Cell Line, Tumor , Models, Biological , Cell Culture Techniques, Three Dimensional/methods , Triple Negative Breast Neoplasms/pathology , Computational Biology , Fibroblasts/physiology , Chemotaxis/physiology
12.
Anal Chem ; 96(25): 10246-10255, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38858132

ABSTRACT

Hypoxia is a representative tumor characteristic associated with malignant progression in clinical patients. Engineered in vitro models have led to significant advances in cancer research, allowing for the investigation of cells in physiological environments and the study of disease mechanisms and processes with enhanced relevance. In this study, we propose a U-shape pillar strip for a 3D cell-lumped organoid model (3D-COM) to study the effects of hypoxia on lung cancer in a high-throughput manner. We developed a U-pillar strip that facilitates the aggregation of PDCs mixed with an extracellular matrix to make the 3D-COM in 384-plate array form. The response to three hypoxia-activated prodrugs was higher in the 3D-COM than in the 2D culture model. The protein expression of hypoxia-inducible factor 1 alpha (HIF-1α) and HIF-2α, which are markers of hypoxia, was also higher in the 3D-COM than in the 2D culture. The results show that 3D-COM better recapitulated the hypoxic conditions of lung cancer tumors than the 2D culture. Therefore, the U-shape pillar strip for 3D-COM is a good tool to study the effects of hypoxia on lung cancer in a high-throughput manner, which can efficiently develop new drugs targeting hypoxic tumors.


Subject(s)
High-Throughput Screening Assays , Lung Neoplasms , Organoids , Humans , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Organoids/metabolism , Organoids/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Hypoxia , Cell Culture Techniques, Three Dimensional , Basic Helix-Loop-Helix Transcription Factors/metabolism
13.
In Vivo ; 38(4): 1579-1593, 2024.
Article in English | MEDLINE | ID: mdl-38936891

ABSTRACT

BACKGROUND/AIM: Melanoma, a variant of skin cancer, presents the highest mortality rates among all skin cancers. Despite advancements in targeted therapies, immunotherapies, and tissue culture techniques, the absence of an effective early treatment model remains a challenge. This study investigated the impact of dabrafenib on both 2D and 3D cell culture models with distinct molecular profiles. MATERIALS AND METHODS: We developed a high-throughput workflow enabling drug screening on spheroids. Our approach involved cultivating 2D and 3D cultures derived from normal melanocytes and metastatic melanoma cells, treating them with dabrafenib and conducting viability, aggregation, migration, cell cycle, and apoptosis assays. RESULTS: Dabrafenib exerted multifaceted influences, particularly on migration at concentrations of 10 and 25 µM. It induced a decrease in cell viability, impeded cellular adhesion to the matrix, inhibited cellular aggregation and spheroid formation, arrested the cell cycle in the G1 phase, and induced apoptosis. CONCLUSION: These results confirm the therapeutic potential of dabrafenib in treating melanoma with the BRAF V600E mutation and that 3D models are validated models to study the potential of new molecules for therapeutic purposes. Furthermore, our study underscores the relevance of 3D models in simulating physiological in vivo microenvironments, providing insights into varied treatment responses between normal and tumor cells.


Subject(s)
Apoptosis , Cell Movement , Cell Survival , Imidazoles , Melanoma , Oximes , Proto-Oncogene Proteins B-raf , Spheroids, Cellular , Oximes/pharmacology , Humans , Imidazoles/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Cell Line, Tumor , Apoptosis/drug effects , Cell Movement/drug effects , Spheroids, Cellular/drug effects , Cell Survival/drug effects , Cell Culture Techniques , Protein Kinase Inhibitors/pharmacology , Cell Cycle/drug effects , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Culture Techniques, Three Dimensional/methods , Drug Screening Assays, Antitumor/methods
14.
J Cell Biochem ; 125(7): e30610, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38860517

ABSTRACT

17ß-estradiol is a hormone that plays a vital role in human physiology. It acts through estrogen receptors, specifically estrogen receptor α and estrogen receptor ß, and its action is determined by the pulsatile secretion in the bloodstream. 17ß-estradiol affects cell proliferation, and dysregulation of 17ß-estradiol:estrogen receptor α signaling contribute to the development of breast cancer. Previous research on 17ß-estradiol:estrogen receptor α signaling has primarily used two-dimensional cell cultures, which do not fully recapitulate the complexity of tumors that exist in a three-dimensional environment and do not consider the pulsatile nature of this hormone. To address these limitations, we studied 17ß-estradiol:estrogen receptor α signaling in cell proliferation using both two-dimensional and three-dimensional breast cancer cell culture models under continuous and pulsatile stimulation conditions. Results revealed that breast cancer cells grown in an alginate-based three-dimensional matrix exhibited similar responsiveness to 17ß-estradiol compared with cells grown in conventional two-dimensional culture plates. 17ß-estradiol induced the expression of proteins containing estrogen response element in the three-dimensional model. The efficacy of the antiestrogen drugs fulvestrant (ICI182,280) and 4OH-tamoxifen was also demonstrated in the three-dimensional model. These results support the use of the three-dimensional culture model for studying tumor response to drugs and provide a more realistic microenvironment for such studies. Furthermore, the study revealed that a brief 5-min exposure to 17ß-estradiol triggered a physiological response comparable with continuous hormone exposure, suggesting that the cellular response to 17ß-estradiol is more important than the continuous presence of the hormone. In conclusion, the study demonstrates that the alginate-based three-dimensional culture model is suitable for studying the effects of 17ß-estradiol and antiestrogen drugs on breast cancer cells, offering a more realistic representation of tumor-microenvironment interactions. The results also highlight the importance of considering the physiological importance of the temporal dynamics in studying 17ß-estradiol signaling and cellular responses.


Subject(s)
Cell Proliferation , Estradiol , Estrogen Receptor alpha , Signal Transduction , Humans , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Cell Proliferation/drug effects , Signal Transduction/drug effects , Female , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , MCF-7 Cells , Cell Culture Techniques, Three Dimensional/methods , Cell Culture Techniques/methods , Fulvestrant/pharmacology
15.
J Vet Sci ; 25(3): e40, 2024 May.
Article in English | MEDLINE | ID: mdl-38834510

ABSTRACT

IMPORTANCE: The creation of robust maternal-embryonic interactions and implantation models is important for comprehending the early stages of embryonic development and reproductive disorders. Traditional two-dimensional (2D) cell culture systems often fail to accurately mimic the highly complex in vivo conditions. The employment of three-dimensional (3D) organoids has emerged as a promising strategy to overcome these limitations in recent years. The advancements in the field of organoid technology have opened new avenues for studying the physiology and diseases affecting female reproductive tract. OBSERVATIONS: This review summarizes the current strategies and advancements in the field of 3D organoids to establish maternal-embryonic interaction and implantation models for use in research and personalized medicine in assisted reproductive technology. The concepts of endometrial organoids, menstrual blood flow organoids, placental trophoblast organoids, stem cell-derived blastoids, and in vitro-generated embryo models are discussed in detail. We show the incorportaion of organoid systems and microfluidic technology to enhance tissue performance and precise management of the cellular surroundings. CONCLUSIONS AND RELEVANCE: This review provides insights into the future direction of modeling maternal-embryonic interaction research and its combination with other powerful technologies to interfere with this dialogue either by promoting or hindering it for improving fertility or methods for contraception, respectively. The merging of organoid systems with microfluidics facilitates the creation of sophisticated and functional organoid models, enhancing insights into organ development, disease mechanisms, and personalized medical investigations.


Subject(s)
Organoids , Female , Animals , Pregnancy , Humans , Cell Culture Techniques, Three Dimensional/methods , Embryo Implantation/physiology
16.
SLAS Discov ; 29(4): 100158, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38852983

ABSTRACT

3D in vitro systems offer advantages over the shortcomings of two-dimensional models by simulating the morphological and functional features of in vivo-like environments, such as cell-cell and cell-extracellular matrix interactions, as well as the co-culture of different cell types. Nevertheless, these systems present technical challenges that limit their potential in cancer research requiring cell line- and culture-dependent standardization. This protocol details the use of a magnetic 3D bioprinting method and other associated techniques (cytotoxicity assay and histological analysis) using oral squamous cell carcinoma cell line, HSC3, which offer advantages compared to existing widely used approaches. This protocol is particularly timely, as it validates magnetic bioprinting as a method for the rapid deployment of 3D cultures as a tool for compound screening and development of heterotypic cultures such as co-culture of oral squamous cell carcinoma cells with cancer-associated fibroblasts (HSC3/CAFs).


Subject(s)
Bioprinting , Carcinoma, Squamous Cell , Coculture Techniques , Mouth Neoplasms , Printing, Three-Dimensional , Spheroids, Cellular , Humans , Mouth Neoplasms/pathology , Bioprinting/methods , Cell Line, Tumor , Carcinoma, Squamous Cell/pathology , Coculture Techniques/methods , Spheroids, Cellular/pathology , Cell Culture Techniques, Three Dimensional/methods
17.
Carbohydr Polym ; 339: 122253, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823920

ABSTRACT

In vitro tumor models are essential for understanding tumor behavior and evaluating tumor biological properties. Hydrogels that can mimic the tumor extracellular matrix have become popular for creating 3D in vitro tumor models. However, designing biocompatible hydrogels with appropriate chemical and physical properties for constructing tumor models is still a challenge. In this study, we synthesized a series of ß-cyclodextrin (ß-CD)-crosslinked polyacrylamide hydrogels with different ß-CD densities and mechanical properties and evaluated their potential for use in 3D in vitro tumor model construction, including cell capture and spheroid formation. By utilizing a combination of ß-CD-methacrylate (CD-MA) and a small amount of N,N'-methylene bisacrylamide (BIS) as hydrogel crosslinkers and optimizing the CD-MA/BIS ratio, the hydrogels performed excellently for tumor cell 3D culture and spheroid formation. Notably, when we co-cultured L929 fibroblasts with HeLa tumor cells on the hydrogel surface, co-cultured spheroids were formed, showing that the hydrogel can mimic the complexity of the tumor extracellular matrix. This comprehensive investigation of the relationship between hydrogel mechanical properties and biocompatibility provides important insights for hydrogel-based in vitro tumor modeling and advances our understanding of the mechanisms underlying tumor growth and progression.


Subject(s)
Acrylic Resins , Hydrogels , Spheroids, Cellular , beta-Cyclodextrins , Spheroids, Cellular/drug effects , Humans , Acrylic Resins/chemistry , Acrylic Resins/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/chemical synthesis , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacology , HeLa Cells , Animals , Mice , Cross-Linking Reagents/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Culture Techniques, Three Dimensional/methods , Methacrylates/chemistry , Coculture Techniques , Neoplasms/pathology
18.
Mol Biol Rep ; 51(1): 721, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829450

ABSTRACT

BACKGROUND: Cancer and multidrug resistance are regarded as concerns related to poor health outcomes. It was found that the monolayer of 2D cancer cell cultures lacks many important features compared to Multicellular Tumor Spheroids (MCTS) or 3D cell cultures which instead have the ability to mimic more closely the in vivo tumor microenvironment. This study aimed to produce 3D cell cultures from different cancer cell lines and to examine the cytotoxic activity of anticancer medications on both 2D and 3D systems, as well as to detect alterations in the expression of certain genes levels. METHOD: 3D cell culture was produced using 3D microtissue molds. The cytotoxic activities of colchicine, cisplatin, doxorubicin, and paclitaxel were tested on 2D and 3D cell culture systems obtained from different cell lines (A549, H1299, MCF-7, and DU-145). IC50 values were determined by MTT assay. In addition, gene expression levels of PIK3CA, AKT1, and PTEN were evaluated by qPCR. RESULTS: Similar cytotoxic activities were observed on both 3D and 2D cell cultures, however, higher concentrations of anticancer medications were needed for the 3D system. For instance, paclitaxel showed an IC50 of 6.234 µM and of 13.87 µM on 2D and 3D H1299 cell cultures, respectively. Gene expression of PIK3CA in H1299 cells also showed a higher fold change in 3D cell culture compared to 2D system upon treatment with doxorubicin. CONCLUSION: When compared to 2D cell cultures, the behavior of cells in the 3D system showed to be more resistant to anticancer treatments. Due to their shape, growth pattern, hypoxic core features, interaction between cells, biomarkers synthesis, and resistance to treatment penetration, the MCTS have the advantage of better simulating the in vivo tumor conditions. As a result, it is reasonable to conclude that 3D cell cultures may be a more promising model than the traditional 2D system, offering a better understanding of the in vivo molecular changes in response to different potential treatments and multidrug resistance development.


Subject(s)
Antineoplastic Agents , Cell Culture Techniques , Spheroids, Cellular , Humans , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Spheroids, Cellular/drug effects , Cell Culture Techniques/methods , Doxorubicin/pharmacology , Paclitaxel/pharmacology , Cisplatin/pharmacology , Tumor Microenvironment/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Cell Culture Techniques, Three Dimensional/methods , MCF-7 Cells , Gene Expression Regulation, Neoplastic/drug effects , Cell Survival/drug effects
19.
Nat Commun ; 15(1): 5027, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871693

ABSTRACT

Generating 3D bone cell networks in vitro that mimic the dynamic process during early bone formation remains challenging. Here, we report a synthetic biodegradable microporous hydrogel for efficient formation of 3D networks from human primary cells, analysis of cell-secreted extracellular matrix (ECM) and microfluidic integration. Using polymerization-induced phase separation, we demonstrate dynamic in situ formation of microporosity (5-20 µm) within matrix metalloproteinase-degradable polyethylene glycol hydrogels in the presence of living cells. Pore formation is triggered by thiol-Michael-addition crosslinking of a viscous precursor solution supplemented with hyaluronic acid and dextran. The resulting microporous architecture can be fine-tuned by adjusting the concentration and molecular weight of dextran. After encapsulation in microporous hydrogels, human mesenchymal stromal cells and osteoblasts spread rapidly and form 3D networks within 24 hours. We demonstrate that matrix degradability controls cell-matrix remodeling, osteogenic differentiation, and deposition of ECM proteins such as collagen. Finally, we report microfluidic integration and proof-of-concept osteogenic differentiation of 3D cell networks under perfusion on chip. Altogether, this work introduces a synthetic microporous hydrogel to efficiently differentiate 3D human bone cell networks, facilitating future in vitro studies on early bone development.


Subject(s)
Cell Culture Techniques, Three Dimensional , Cell Differentiation , Extracellular Matrix , Hydrogels , Mesenchymal Stem Cells , Osteoblasts , Osteogenesis , Humans , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Cell Differentiation/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Extracellular Matrix/metabolism , Porosity , Cell Culture Techniques, Three Dimensional/methods , Polyethylene Glycols/chemistry , Tissue Engineering/methods , Hyaluronic Acid/chemistry , Cells, Cultured , Tissue Scaffolds/chemistry , Dextrans/chemistry
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...