To see the other types of publications on this topic, follow the link: 3D organotypic cell structures.

Journal articles on the topic '3D organotypic cell structures'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic '3D organotypic cell structures.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Zubareva, Ekaterina V., Sergey V. Nadezhdin, Natalia A. Nadezhdina, et al. "3D organotypic cell structures for drug development and Microorganism-Host interaction research." Research Results in Pharmacology 7, no. 1 (2021): 47–64. http://dx.doi.org/10.3897/rrpharmacology.7.62118.

Full text
Abstract:
Introduction: The article describes a new method of tissue engineering, which is based on the use of three-dimensional multicellular constructs consisting of stem cells that mimic the native tissue in vivo – organoids. 3D cell cultures: The currently existing model systems of three-dimensional cultures are described. Characteristics of organoids and strategies for their culturing: The main approaches to the fabrication of 3D cell constructs using pluripotent (embryonic and induced) stem cells or adult stem cells are described. Brain organoids (Cerebral organoids): Organoids of the brain, which are used to study the development of the human brain, are characterized, with the description of biology of generating region-specific cerebral organoids. Lung organoids: Approaches to the generation of lung organoids are described, by means of pluripotent stem cells and lung tissue cell lines. Liver organoids: The features of differentiation of stem cells into hepatocyte-like cells and the creation of 3D hepatic organoids are characterized. Intestinal organoids: The formation of small intestine organoids from stem cells is described. Osteochondral organoids: Fabrication of osteochondral organoids is characterised. Use of organoids as test systems for drugs screening: The information on drug screening using organoids is provided. Using organoids to model infectious diseases and study adaptive responses of microorganisms when interacting with the host: The use of organoids for modeling infectious diseases and studying the adaptive responses of microorganisms when interacting with the host organism is described. Conclusion: The creation of three-dimensional cell structures that reproduce the structural and functional characteristics of tissue in vivo, makes it possible to study the biology of the body’s development, the features of intercellular interactions, screening drugs and co-cultivating with viruses, bacteria and parasites.
APA, Harvard, Vancouver, ISO, and other styles
2

Zubareva, Ekaterina V., Sergey V. Nadezhdin, Natalia A. Nadezhdina, et al. "3D organotypic cell structures for drug development and Microorganism-Host interaction research." Research Results in Pharmacology 7, no. (1) (2021): 47–64. https://doi.org/10.3897/rrpharmacology.7.62118.

Full text
Abstract:
Introduction: The article describes a new method of tissue engineering, which is based on the use of three-dimensional multicellular constructs consisting of stem cells that mimic the native tissue in vivo – organoids. 3D cell cultures: The currently existing model systems of three-dimensional cultures are described. Characteristics of organoids and strategies for their culturing: The main approaches to the fabrication of 3D cell constructs using pluripotent (embryonic and induced) stem cells or adult stem cells are described. Brain organoids (Cerebral organoids): Organoids of the brain, which are used to study the development of the human brain, are characterized, with the description of biology of generating region-specific cerebral organoids. Lung organoids: Approaches to the generation of lung organoids are described, by means of pluripotent stem cells and lung tissue cell lines. Liver organoids: The features of differentiation of stem cells into hepatocyte-like cells and the creation of 3D hepatic organoids are characterized. Intestinal organoids: The formation of small intestine organoids from stem cells is described. Osteochondral organoids: Fabrication of osteochondral organoids is characterised. Use of organoids as test systems for drugs screening: The information on drug screening using organoids is provided. Using organoids to model infectious diseases and study adaptive responses of microorganisms when interacting with the host: The use of organoids for modeling infectious diseases and studying the adaptive responses of microorganisms when interacting with the host organism is described. Conclusion: The creation of three-dimensional cell structures that reproduce the structural and functional characteristics of tissue in vivo, makes it possible to study the biology of the body's development, the features of intercellular interactions, screening drugs and co-cultivating with viruses, bacteria and parasites.
APA, Harvard, Vancouver, ISO, and other styles
3

Salinas-Vera, Yarely M., Jesús Valdés, Alfredo Hidalgo-Miranda, et al. "Three-Dimensional Organotypic Cultures Reshape the microRNAs Transcriptional Program in Breast Cancer Cells." Cancers 14, no. 10 (2022): 2490. http://dx.doi.org/10.3390/cancers14102490.

Full text
Abstract:
The 3D organotypic cultures, which depend on the growth of cells over the extracellular matrix (ECM) used as a scaffold, can better mimic several characteristics of solid cancers that influence tumor biology and the response to drug therapies. Most of our current knowledge on cancer is derived from studies in 2D cultures, which lack the ECM-mediated microenvironment. Moreover, the role of miRNAs that is critical for fine-tuning of gene expression is poorly understood in 3D cultures. The aim of this study was to compare the miRNA expression profiles of breast cancer cells grown in 2D and 3D conditions. On an on-top 3D cell culture model using a basement membrane matrix enriched with laminin, collagen IV, entactin, and heparin-sulfate proteoglycans, the basal B (Hs578T) and luminal (T47D) breast cancer cells formed 3D spheroid-like stellate and rounded mass structures, respectively. Morphological changes in 3D cultures were observed as cell stretching, cell–cell, and cell–ECM interactions associated with a loss of polarity and reorganization on bulk structures. Interestingly, we found prolongations of the cytoplasmic membrane of Hs578T cells similar to tunneled nanotubes contacting between neighboring cells, suggesting the existence of cellular intercommunication processes and the possibility of fusion between spheroids. Expression profiling data revealed that 354 miRNAs were differentially expressed in 3D relative to 2D cultures in Hs578T cells. Downregulated miRNAs may contribute to a positive regulation of genes involved in hypoxia, catabolic processes, and focal adhesion, whereas overexpressed miRNAs modulate genes involved in negative regulation of the cell cycle. Target genes of the top ten modulated miRNAs were selected to construct miRNA/mRNA coregulation networks. Around 502 interactions were identified for downregulated miRNAs, including miR-935/HIF1A and miR-5189-3p/AKT that could contribute to cell migration and the response to hypoxia. Furthermore, the expression levels of miR-935 and its target HIF1A correlated with the expression found in clinical tumors and predicted poor outcomes. On the other hand, 416 interactions were identified for overexpressed miRNAs, including miR-6780b-5p/ANKRD45 and miR-7641/CDK4 that may result in cell proliferation inhibition and cell cycle arrest in quiescent layers of 3D cultures. In conclusion, 3D cultures could represent a suitable model that better resembles the miRNA transcriptional programs operating in tumors, with implications not only in the understanding of basic cancer biology in 3D microenvironments, but also in the identification of novel biomarkers of disease and potential targets for personalized therapies in cancer.
APA, Harvard, Vancouver, ISO, and other styles
4

Shved, Natalia, Anna Egorova, Natalia Osinovskaya, and Anton Kiselev. "Development of Primary Monolayer Cell Model and Organotypic Model of Uterine Leiomyoma." Methods and Protocols 5, no. 1 (2022): 16. http://dx.doi.org/10.3390/mps5010016.

Full text
Abstract:
Cellular technologies are one of the most promising areas of biomedicine, which is based on the isolation of cells of various types, followed by their cultivation and use, or the use of their metabolic products, for medical purposes. Today, a significant part of biomedical research is carried out in vitro. On the other hand, organotypic culture can be used as a powerful model system and can complement cell culture and in vivo studies in different biomedical applications. Uterine leiomyoma (UL) is a very common benign tumor and often leads to many reproductive complications. Herein we describe a fast and reliable method of isolation and UL primary cells culturing along with the development of a UL organotypic model. We propose the usage of UL primary cells in experimental work at a first passage to prevent loss of driver mutations in MED12 and HMGA2 genes. New optimized conditions for the growth and maintenance of 2D and 3D models of uterine leiomyoma in vitro are suggested.
APA, Harvard, Vancouver, ISO, and other styles
5

Walsh, C. L., P. Tafforeau, W. L. Wagner, et al. "Imaging intact human organs with local resolution of cellular structures using hierarchical phase-contrast tomography." Nature Methods 18, no. 12 (2021): 1532–41. http://dx.doi.org/10.1038/s41592-021-01317-x.

Full text
Abstract:
AbstractImaging intact human organs from the organ to the cellular scale in three dimensions is a goal of biomedical imaging. To meet this challenge, we developed hierarchical phase-contrast tomography (HiP-CT), an X-ray phase propagation technique using the European Synchrotron Radiation Facility (ESRF)’s Extremely Brilliant Source (EBS). The spatial coherence of the ESRF-EBS combined with our beamline equipment, sample preparation and scanning developments enabled us to perform non-destructive, three-dimensional (3D) scans with hierarchically increasing resolution at any location in whole human organs. We applied HiP-CT to image five intact human organ types: brain, lung, heart, kidney and spleen. HiP-CT provided a structural overview of each whole organ followed by multiple higher-resolution volumes of interest, capturing organotypic functional units and certain individual specialized cells within intact human organs. We demonstrate the potential applications of HiP-CT through quantification and morphometry of glomeruli in an intact human kidney and identification of regional changes in the tissue architecture in a lung from a deceased donor with coronavirus disease 2019 (COVID-19).
APA, Harvard, Vancouver, ISO, and other styles
6

Liu, Sai, and Pan Jin. "Advances and Challenges in 3D Bioprinted Cancer Models: Opportunities for Personalized Medicine and Tissue Engineering." Polymers 17, no. 7 (2025): 948. https://doi.org/10.3390/polym17070948.

Full text
Abstract:
Cancer is the second leading cause of death worldwide, after cardiovascular disease, claiming not only a staggering number of lives but also causing considerable health and economic devastation, particularly in less-developed countries. Therapeutic interventions are impeded by differences in patient-to-patient responses to anti-cancer drugs. A personalized medicine approach is crucial for treating specific patient groups and includes using molecular and genetic screens to find appropriate stratifications of patients who will respond (and those who will not) to treatment regimens. However, information on which risk stratification method can be used to hone in on cancer types and patients who will be likely responders to a specific anti-cancer agent remains elusive for most cancers. Novel developments in 3D bioprinting technology have been widely applied to recreate relevant bioengineered tumor organotypic structures capable of mimicking the human tissue and microenvironment or adequate drug responses in high-throughput screening settings. Parts are autogenously printed in the form of 3D bioengineered tissues using a computer-aided design concept where multiple layers include different cell types and compatible biomaterials to build specific configurations. Patient-derived cancer and stromal cells, together with genetic material, extracellular matrix proteins, and growth factors, are used to create bioprinted cancer models that provide a possible platform for the screening of new personalized therapies in advance. Both natural and synthetic biopolymers have been used to encourage the growth of cells and biological materials in personalized tumor models/implants. These models may facilitate physiologically relevant cell–cell and cell–matrix interactions with 3D heterogeneity resembling real tumors.
APA, Harvard, Vancouver, ISO, and other styles
7

Das, Ridhima, Lisa Harper, Kayoko Kitajima, et al. "Embryonic Stem Cells Can Generate Oral Epithelia under Matrix Instruction." International Journal of Molecular Sciences 24, no. 9 (2023): 7694. http://dx.doi.org/10.3390/ijms24097694.

Full text
Abstract:
We aimed to investigate whether molecular clues from the extracellular matrix (ECM) can induce oral epithelial differentiation of pluripotent stem cells. Mouse embryonic stem cells (ESC) of the feeder-independent cell line E14 were used as a model for pluripotent stem cells. They were first grown in 2D on various matrices in media containing vitamin C and without leukemia inhibitory factor (LIF). Matrices investigated were gelatin, laminin, and extracellular matrices (ECM) synthesized by primary normal oral fibroblasts and keratinocytes in culture. Differentiation into epithelial lineages was assessed by light microscopy, immunocytochemistry, and flow cytometry for cytokeratins and stem cell markers. ESC grown in 2D on various matrices were afterwards grown in 3D organotypic cultures with or without oral fibroblasts in the collagen matrix and examined histologically and by immunohistochemistry for epithelial (keratin pairs 1/10 and 4/13 to distinguish epidermal from oral epithelia and keratins 8,18,19 to phenotype simple epithelia) and mesenchymal (vimentin) phenotypes. ECM synthesized by either oral fibroblasts or keratinocytes was able to induce, in 2D cultures, the expression of cytokeratins of the stratified epithelial phenotype. When grown in 3D, all ESC developed into two morphologically distinct cell populations on collagen gels: (i) epithelial-like cells organized in islands with occasional cyst- or duct-like structures and (ii) spindle-shaped cells suggestive of mesenchymal differentiation. The 3D culture on oral fibroblast-populated collagen matrices was necessary for further differentiation into oral epithelia. Only ESC initially grown on 2D keratinocyte or fibroblast-synthesized matrices reached full epithelial maturation. In conclusion, ESC can generate oral epithelia under matrix instruction.
APA, Harvard, Vancouver, ISO, and other styles
8

Kuehlbach, Claudia, Sabine Hensler, and Margareta M. Mueller. "Recapitulating the Angiogenic Switch in a Hydrogel-Based 3D In Vitro Tumor-Stroma Model." Bioengineering 8, no. 11 (2021): 186. http://dx.doi.org/10.3390/bioengineering8110186.

Full text
Abstract:
To ensure nutrient and oxygen supply, tumors beyond a size of 1–2 mm3 need a connection to the vascular system. Thus, tumor cells modify physiological tissue homeostasis by secreting inflammatory and angiogenic cytokines. This leads to the activation of the tumor microenvironment and the turning of the angiogenic switch, resulting in tumor vascularization and growth. To inhibit tumor growth by developing efficient anti-angiogenic therapies, an in depth understanding of the molecular mechanism initiating angiogenesis is essential. Yet so far, predominantly 2D cell cultures or animal models have been used to clarify the interactions within the tumor stroma, resulting in poor transferability of the data obtained to the in vivo situation. Consequently, there is an abundant need for complex, humanized, 3D models in vitro. We established a dextran-hydrogel-based 3D organotypic in vitro model containing microtumor spheroids, macrophages, neutrophils, fibroblasts and endothelial cells, allowing for the analysis of tumor–stroma interactions in a controlled and modifiable environment. During the cultivation period of 21 days, the microtumor spheroids in the model grew in size and endothelial cells formed elongated tubular structures resembling capillary vessels, that appeared to extend towards the tumor spheroids. The tubular structures exhibited complex bifurcations and expanded without adding external angiogenic factors such as VEGF to the culture. To allow high-throughput screening of therapeutic candidates, the 3D cell culture model was successfully miniaturized to a 96-well format, while still maintaining the same level of tumor spheroid growth and vascular sprouting. The quantification of VEGF in the conditioned medium of these cultures showed a continuous increase during the cultivation period, suggesting the contribution of endogenous VEGF to the induction of the angiogenic switch and vascular sprouting. Thus, this model is highly suitable as a testing platform for novel anticancer therapeutics targeting the tumor as well as the vascular compartment.
APA, Harvard, Vancouver, ISO, and other styles
9

Aizawa, Yuka, Kenta Haga, Nagako Yoshiba, et al. "Development and Characterization of a Three-Dimensional Organotypic In Vitro Oral Cancer Model with Four Co-Cultured Cell Types, Including Patient-Derived Cancer-Associated Fibroblasts." Biomedicines 12, no. 10 (2024): 2373. http://dx.doi.org/10.3390/biomedicines12102373.

Full text
Abstract:
Background/Objectives: Cancer organoids have emerged as a valuable tool of three-dimensional (3D) cell cultures to investigate tumor heterogeneity and predict tumor behavior and treatment response. We developed a 3D organotypic culture model of oral squamous cell carcinoma (OSCC) to recapitulate the tumor–stromal interface by co-culturing four cell types, including patient-derived cancer-associated fibroblasts (PD-CAFs). Methods: A stainless-steel ring was used twice to create the horizontal positioning of the cancer stroma (adjoining normal oral mucosa connective tissue) and the OSCC layer (surrounding normal oral mucosa epithelial layer). Combined with a structured bi-layered model of the epithelial component and the underlying stroma, this protocol enabled us to construct four distinct portions mimicking the oral cancer tissue arising in the oral mucosa. Results: In this model, α-smooth muscle actin-positive PD-CAFs were localized in close proximity to the OSCC layer, suggesting a crosstalk between them. Furthermore, a linear laminin-γ2 expression was lacking at the interface between the OSCC layer and the underlying stromal layer, indicating the loss of the basement membrane-like structure. Conclusions: Since the specific 3D architecture and polarity mimicking oral cancer in vivo provides a more accurate milieu of the tumor microenvironment (TME), it could be crucial in elucidating oral cancer TME.
APA, Harvard, Vancouver, ISO, and other styles
10

Monteiro, R. F., S. M. Bakht, M. Gomez-Florit, R. L. Reis, M. E. Gomes, and R. M. A. Domingues. "3D WRITING OF MULTICELLULAR TENDON-ON-CNC-CHIP MODELS." Orthopaedic Proceedings 106-B, SUPP_2 (2024): 15. http://dx.doi.org/10.1302/1358-992x.2024.2.015.

Full text
Abstract:
Relevant in vitro models emulating tendinopathies are highly needed to study these diseases and develop better treatments. We have recently proposed a new strategy that allows the automated 3D writing of microphysiological systems (MPS) embedded into its own biomimetic fibrillar support platform based on the self-assembling of cellulose nanocrystals (CNCs). Here, we explored this CNC platform for writing humanized in vitro tendon models using tendon decellularized extracellular matrix (dECM)-based bioinks to closely recapitulate the biophysical and biochemical cues of tendon cell niche and self-induce the tenogenic differentiation of stem cells. The proposed concept was further explored to study the crosstalk between the tendon core and vascular compartment.Porcine flexor tendons were decellularized to produce the dECM bioink hydrogel. hASCs were used as cell source and the bioink was directly printed within the CNC fluid gel. Tendon constructs were co-printed with compartmentalized microvascular structures to evaluate the cellular crosstalk with endothelial cells. The tendon-on-chip models showed high cell viability and proliferation during culture up to 21 days, and the synergy between dECM cues and printed patterns induced anisotropic cell organization similar to tendon tissues. Gene and protein analysis showed upregulation of the most important tendon related markers on tendon constructs, demonstrating that the biophysical and biochemical cues of dECM induced hASCs commitment toward tenogenic phenotype. In co-culture system, chemotaxis induced endothelial cells migration toward the tendon compartment, but without significant infiltration. Gene and protein expression results suggest that the cellular crosstalk established in this MPS with endothelial cells boosted hASCs tenogenesis, emulating tendon development stages.Overall, the proposed system might be promising for the automated fabrication of organotypic tendon-on-chip models that will be a valuable new tool to study tendon physiology, pathology, or the effect of drugs for the treatment of tendinopathy.Acknowledgments: EU H2020 for ERC-2017-CoG-772817; ERC-PoC-BioCHIPs-101069302; FCT/MCTES for 2022.05526.PTDC, 2020.03410.CEECIND, and PD/BD/129403/2017
APA, Harvard, Vancouver, ISO, and other styles
11

Ahmed, Adeel, Nathan W. Hendrikse, Marcos A. Lares, et al. "Abstract B023: Patient-specific, organotypic head and neck cancer model for personalized medicine." Cancer Research 85, no. 5_Supplement (2025): B023. https://doi.org/10.1158/1538-7445.genfunc25-b023.

Full text
Abstract:
Abstract Background: Head and neck cancer (HNC) treatment outcomes are highly variable among patients, and lack of actionable data on individual treatment responses limits effective clinical decision-making. Microphysiological systems (MPS) are advanced in vitro models that replicate the biological, physical, and functional properties of human tissues, offering a promising approach to testing treatment responses in vitro. Despite their potential, MPS adoption for evaluating standard clinical treatments—such as radiation and chemotherapy—has been hindered by the challenge of recreating the complexity of the tumor microenvironment. To address this, we developed an HNC MPS that mimics patient-specific tumor microenvironments using primary cells from patient tumors embedded in a 3D matrix with vascular components. This model was used to evaluate individualized responses to radiation and chemotherapy through cell-scale readouts, including DNA damage, proliferation, and migration. Methods: The HNC MPS comprises a microscale chamber containing two removable rods. A collagen-fibronectin matrix incorporating patient-derived tumor spheroids, fibroblasts, and immune cells was cast in the chamber. The matrix polymerized around the removable rods, which were subsequently extracted to create molded luminal structures. These lumens were lined with vascular and lymphatic endothelial cells to recreate a vascularized tumor microenvironment. An optimized media formulation supported the growth of all cell types within the MPS. The model was treated with radiation, cisplatin, or both to simulate the current standard of care. Cellular responses, including viability, proliferation, migration, and DNA damage, were assessed using immunofluorescence and cytokine secretion. A custom image processing pipeline analyzed multiple orthogonal readouts to reveal treatment responses at the cellular level with metrics to compare to actual patient outcomes under development. The model was further validated by comparing it with tumor tissue using single-cell RNA sequencing (scRNA-seq). Experiments were conducted with samples from multiple patients. Results: We successfully developed a patient-specific model of the HNC microenvironment using primary cells isolated from patient tumor tissue. This model captured key patient-specific features, such as angiogenesis and cell migration, and provided insights into treatment responses across different modalities. Cellular responses, such as proliferation and migration, were quantified, and scRNA-seq demonstrated close concordance between the MPS and patient tumor tissue. Our model presents a valuable functional assay for predicting patient-specific responses to therapy and validating biomarkers for treatment stratification. Conclusion: The HNC MPS is a robust in vitro platform that recapitulates patient-specific tumor microenvironments and treatment responses. It offers a promising approach for personalized cancer therapy, enabling functional assays to guide clinical decision-making and improve treatment outcomes for patients with HNC. Citation Format: Adeel Ahmed, Nathan W Hendrikse, Marcos A Lares, Fauzan Ahmed, Adam R Burr, Paul M Harari, David J Beebe, Sheena C Kerr. Patient-specific, organotypic head and neck cancer model for personalized medicine [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Functional and Genomic Precision Medicine in Cancer: Different Perspectives, Common Goals; 2025 Mar 11-13; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2025;85(5 Suppl):Abstract nr B023.
APA, Harvard, Vancouver, ISO, and other styles
12

Schaeffer, Alexander, Ewelina Czlonka, Irene Tirado-González, et al. "Development and Exploitation of a Fully Human and Modular Organotypic Bone Marrow Niche Model to Study the Role of Stroma-Produced Factors in Human MDS." Blood 136, Supplement 1 (2020): 23. http://dx.doi.org/10.1182/blood-2020-142782.

Full text
Abstract:
Background: Myelodysplastic syndromes (MDS) are a heterogenous group of stem cell driven disorders primarily affecting the elderly and characterized by inefficient production of mature blood cells and a high risk (30%) of evolution to secondary acute myeloid leukemia. Despite tremendous progress in the past decade, treatment options for MDS patients remain limited, and primarily address disease symptoms, rather than altering disease course. This points to the urgent need to better understand the pathogenesis of this heterogenous group of syndromes to develop new therapies that address disease vulnerabilities. However, this effort has been largely hampered by the limited availability of model systems that allow the exploration of MDS biology in a fully humanized setting. In recent years, studies from our lab and others, have highlighted the crucial role niche cells play in human MDS, hence reinforcing the notion that MDS is a disease of a tissue rather than hematopoietic cells alone. Therefore, exploration of MDS biology requires the further development of fully human MDS models in which both constituents of the disease, namely hematopoietic and niche cells, are present. Methods: To address this issue we successfully isolated endothelial cells (ECs) and mesenchymal stromal cells (MSC) from bone marrow biopsies obtained from MDS patients or healthy age matched controls, and subsequently utilized them to develop fully human 2D and 3D organotypic niche models, which were successfully used to support normal and MDS HSPCs expansion ex-vivo. The 3D system makes use of a collagen scaffold, as this protein makes up for 90% of the matrix proteins in the bone. Importantly, MSC and EC cultures could be successfully established from several independent donors and immortalized to generate primary cell lines that can be used to reproducibly establish these ex-vivo systems in a robust manner. Moreover, we could show that these niche cells were easily amenable to genetic editing using CRISPR-Cas9 technology as well as modified to carry fluorescent reporter proteins for tracking cellular interactions using live cell imaging and confocal microscopy. Results: In this work, we successfully isolated human mesenchymal and endothelial cells, from primary bone marrow biopsies (MDS and healthy) and established fully human 2D and 3D organotypic co-cultures ex-vivo. Of note, although bone marrow ECs represent an essential component of the hematopoietic niche, they have so far been omitted in previously described human bone marrow niche models, owing to the notorious difficulties in isolating and expanding this cell type from primary bone marrow biopsies. Therefore, we established immortalized EC lines (iECs) that faithfully recapitulate the morphological, phenotypic and functional features of primary bone marrow ECs. When cultured at defined ratios and under defined conditions, MSCs instructed ECs and iECs to form of vessel-like structures that mimic the meshwork observed in vivo and are typically escheated by aSMA positive cells that stabilize the structures. Genetic manipulation of the cellular components of the niche also allowed to explore the functional relevance of a specific ECM protein, which we previously identified to be significantly upregulated in MSCs isolated from MDS patients, namely the Secreted Protein Acidic and Rich in Cysteine (SPARC). SPARC ablation triggered enhanced proliferation of MDS derived HSPCs and sensitized them treatment with 5-Azacytidine, a standard of care hypomethylating agent used for the treatment of MDS patients. Additional studies are underway to further understand the underlying molecular mechanisms and define a potential druggable target that could sensitize MDS cells to standard of care treatment. Besides gene targeting studies, these organotypic models are also being used to evaluate the relative fitness of MDS and healthy stem/progenitor cells in healthy versus patient derived niches, to explore the contribution of niche components to the establishment of the progressive clonal dominance observed in MDS. Disclosures Bönig: Terumo BCT: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Kiadis: Honoraria; Bayer: Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Fresenius: Honoraria; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Uniqure: Research Funding; medac: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties; Genzyme: Consultancy, Membership on an entity's Board of Directors or advisory committees; Healthineers: Current equity holder in publicly-traded company; Chugai: Honoraria, Research Funding; Erydel: Research Funding; Miltenyi: Honoraria, Research Funding; Polyphor: Research Funding; Sandor-Hexal: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Stage: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Platzbecker:Amgen: Honoraria, Research Funding; Geron: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding. Götze:Celgene: Research Funding. Medyouf:Bergenbio: Consultancy, Research Funding.
APA, Harvard, Vancouver, ISO, and other styles
13

Hammer, S., A. Weltin, Y. Kaminski, F. Noor, J. Kieninger, and G. A. Urban. "Lactate Monitoring in Organotypic 3D Cell Cultures." Procedia Engineering 120 (2015): 961–64. http://dx.doi.org/10.1016/j.proeng.2015.08.825.

Full text
APA, Harvard, Vancouver, ISO, and other styles
14

Lu, Emily Ming-Chieh. "Three-Dimensional Organotypic Systems for Modelling and Understanding Molecular Regulation of Oral Dentogingival Tissues." International Journal of Molecular Sciences 25, no. 21 (2024): 11552. http://dx.doi.org/10.3390/ijms252111552.

Full text
Abstract:
Three-dimensional organotypic models benefit from the ability to mimic physiological cell–cell or cell–matrix interactions and therefore offer superior models for studying pathological or physiological conditions compared to 2D cultures. Organotypic models consisting of keratinocytes supported by fibroblasts embedded in collagen matrices have been utilised for the study of oral conditions. However, the provision of a suitable model for investigating the pathogenesis of periodontitis has been more challenging. Part of the complexity relates to the different regional epithelial specificities and connective tissue phenotypes. Recently, it was confirmed, using 3D organotypic models, that distinct fibroblast populations were implicated in the provision of specific inductive and directive influences on the overlying epithelia. This paper presents the organotypic model of the dentogingival junction (DGJ) constructed to demonstrate the differential fibroblast influences on the maintenance of regionally specific epithelial phenotypes. Therefore, the review aims are (1) to provide the biological basis underlying 3D organotypic cultures and (2) to comprehensively detail the experimental protocol for the construction of the organotypic cultures and the unique setup for the DGJ model. The latter is the first organotypic culture model used for the reconstruction of the DGJ and is recommended as a useful tool for future periodontal research.
APA, Harvard, Vancouver, ISO, and other styles
15

Brulin, Bénédicte, John C. Nolan, Tecla Marangon, et al. "Evaluation of the Chemotherapy Drug Response Using Organotypic Cultures of Osteosarcoma Tumours from Mice Models and Canine Patients." Cancers 13, no. 19 (2021): 4890. http://dx.doi.org/10.3390/cancers13194890.

Full text
Abstract:
Improvements in the clinical outcome of osteosarcoma have plateaued in recent decades with poor translation between preclinical testing and clinical efficacy. Organotypic cultures retain key features of patient tumours, such as a myriad of cell types organized within an extracellular matrix, thereby presenting a more realistic and personalised screening of chemotherapeutic agents ex vivo. To test this concept for the first time in osteosarcoma, murine and canine osteosarcoma organotypic models were maintained for up to 21 days and in-depth analysis identified proportions of immune and stromal cells present at levels comparable to that reported in vivo in the literature. Cytotoxicity testing of a range of chemotherapeutic drugs (mafosfamide, cisplatin, methotrexate, etoposide, and doxorubicin) on murine organotypic culture ex vivo found limited response to treatment, with immune and stromal cells demonstrating enhanced survival over the global tumour cell population. Furthermore, significantly decreased sensitivity to a range of chemotherapeutics in 3D organotypic culture relative to 2D monolayer was observed, with subsequent investigation confirming reduced sensitivity in 3D than in 2D, even at equivalent levels of drug uptake. Finally, as proof of concept for the application of this model to personalised drug screening, chemotherapy testing with doxorubicin was performed on biopsies obtained from canine osteosarcoma patients. Together, this study highlights the importance of recapitulating the 3D tumour multicellular microenvironment to better predict drug response and provides evidence for the utility and possibilities of organotypic culture for enhanced preclinical selection and evaluation of chemotherapeutics targeting osteosarcoma.
APA, Harvard, Vancouver, ISO, and other styles
16

Metelmann, Isabella B., Sebastian Kraemer, Matthias Steinert, Stefan Langer, Peggy Stock, and Olga Kurow. "Novel 3D organotypic co-culture model of pleura." PLOS ONE 17, no. 12 (2022): e0276978. http://dx.doi.org/10.1371/journal.pone.0276978.

Full text
Abstract:
Pleural mesothelial cells are the predominant cell type in the pleural cavity, but their role in the pathogenesis of pleural diseases needs to be further elucidated. 3D organotypic models are an encouraging approach for an in vivo understanding of molecular disease development. The aim of the present study was to develop a 3D organotypic model of the pleural mesothelium. Specimens of human pleura parietalis were obtained from patients undergoing surgery at the University Hospital Leipzig, Germany. 3D co-culture model of pleura was established from human pleural mesothelial cells and fibroblasts. The model was compared to human pleura tissue by phase-contrast and light microscopy, immunochemistry and -fluorescence as well as solute permeation test. Histological assessment of the 3D co-culture model displayed the presence of both cell types mimicking the morphology of the human pleura. Vimentin and Cytokeratin, PHD1 showed a similar expression pattern in pleural biopsies and 3D model. Expression of Ki-67 indicates the presence of proliferating cells. Tight junctional marker ZO-1 was found localized at contact zones between mesothelial cells. Each of these markers were expressed in both the 3D co-culture model and human biopsies. Permeability of 3D organotypic co-culture model of pleura was found to be higher for 70 kDa-Dextran and no significant difference was seen in the permeability for small dextran (4 kDa). In summary, the presented 3D organoid of pleura functions as a robust assay for pleural research serving as a precise reproduction of the in vivo morphology and microenvironment.
APA, Harvard, Vancouver, ISO, and other styles
17

Watters, Karen M., Preety Bajwa, and Hilary A. Kenny. "Organotypic 3D Models of the Ovarian Cancer Tumor Microenvironment." Cancers 10, no. 8 (2018): 265. http://dx.doi.org/10.3390/cancers10080265.

Full text
Abstract:
Ovarian cancer progression involves multifaceted and variable tumor microenvironments (TMEs), from the in situ carcinoma in the fallopian tube or ovary to dissemination into the peritoneal cavity as single cells or spheroids and attachment to the mesothelial-lined surfaces of the omentum, bowel, and abdominal wall. The TME comprises the tumor vasculature and lymphatics (including endothelial cells and pericytes), in addition to mesothelial cells, fibroblasts, immune cells, adipocytes and extracellular matrix (ECM) proteins. When generating 3D models of the ovarian cancer TME, researchers must incorporate the most relevant stromal components depending on the TME in question (e.g., early or late disease). Such complexity cannot be captured by monolayer 2D culture systems. Moreover, immortalized stromal cell lines, such as mesothelial or fibroblast cell lines, do not always behave the same as primary cells whose response in functional assays may vary from donor to donor; 3D models with primary stromal cells may have more physiological relevance than those using stromal cell lines. In the current review, we discuss the latest developments in organotypic 3D models of the ovarian cancer early metastatic microenvironment. Organotypic culture models comprise two or more interacting cell types from a particular tissue. We focus on organotypic 3D models that include at least one type of primary stromal cell type in an ECM background, such as collagen or fibronectin, plus ovarian cancer cells. We provide an overview of the two most comprehensive current models—a 3D model of the omental mesothelium and a microfluidic model. We describe the cellular and non-cellular components of the models, the incorporation of mechanical forces, and how the models have been adapted and utilized in functional assays. Finally, we review a number of 3D models that do not incorporate primary stromal cells and summarize how integration of current models may be the next essential step in tackling the complexity of the different ovarian cancer TMEs.
APA, Harvard, Vancouver, ISO, and other styles
18

Dukes, M. W., B. E. Perez White, and T. J. Meade. "847 Modeling basal cell carcinoma in 3D organotypic raft cultures." Journal of Investigative Dermatology 142, no. 8 (2022): S147. http://dx.doi.org/10.1016/j.jid.2022.05.861.

Full text
APA, Harvard, Vancouver, ISO, and other styles
19

Hatterschide, Joshua, Christopher A. Natale, Todd W. Ridky, and Elizabeth A. White. "Monitoring cell fate in 3D organotypic human squamous epithelial cultures." STAR Protocols 4, no. 1 (2023): 102101. http://dx.doi.org/10.1016/j.xpro.2023.102101.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Sakib, Sadman, Aya Uchida, Paula Valenzuela-Leon, et al. "Formation of organotypic testicular organoids in microwell culture†." Biology of Reproduction 100, no. 6 (2019): 1648–60. http://dx.doi.org/10.1093/biolre/ioz053.

Full text
Abstract:
Abstract Three-dimensional (3D) organoids can serve as an in vitro platform to study cell–cell interactions, tissue development, and toxicology. Development of organoids with tissue architecture similar to testis in vivo has remained a challenge. Here, we present a microwell aggregation approach to establish multicellular 3D testicular organoids from pig, mouse, macaque, and human. The organoids consist of germ cells, Sertoli cells, Leydig cells, and peritubular myoid cells forming a distinct seminiferous epithelium and interstitial compartment separated by a basement membrane. Sertoli cells in the organoids express tight junction proteins claudin 11 and occludin. Germ cells in organoids showed an attenuated response to retinoic acid compared to germ cells in 2D culture indicating that the tissue architecture of the organoid modulates response to retinoic acid similar to in vivo. Germ cells maintaining physiological cell–cell interactions in organoids also had lower levels of autophagy indicating lower levels of cellular stress. When organoids were treated with mono(2-ethylhexyl) phthalate (MEHP), levels of germ cell autophagy increased in a dose-dependent manner, indicating the utility of the organoids for toxicity screening. Ablation of primary cilia on testicular somatic cells inhibited the formation of organoids demonstrating an application to screen for factors affecting testicular morphogenesis. Organoids can be generated from cryopreserved testis cells and preserved by vitrification. Taken together, the testicular organoid system recapitulates the 3D organization of the mammalian testis and provides an in vitro platform for studying germ cell function, testicular development, and drug toxicity in a cellular context representative of the testis in vivo.
APA, Harvard, Vancouver, ISO, and other styles
21

Ritch, Sabrina J., Alicia A. Goyeneche, Abu S. Noman, and Carlos M. Telleria. "Abstract 3456: Discrepancies in the metastatic potential of high-grade serous ovarian cancer cells representing disease progression when comparing two-dimensional assays versus three-dimensional organotypic culture model systems." Cancer Research 82, no. 12_Supplement (2022): 3456. http://dx.doi.org/10.1158/1538-7445.am2022-3456.

Full text
Abstract:
Abstract Due to the lack of early symptoms and valid screening methods, most patients with the most prevalent histotype of ovarian cancer, high-grade serous ovarian cancer (HGSOC), are diagnosed at late stage, at which point metastasis has already occurred. The purpose of this study was to characterize the migratory, invasive, and adhesive abilities of HGSOC cells, representing different stages of disease evolution, in both two-dimensional (2D) and three-dimensional (3D) organotypic assays. Five HGSOC cell lines, collected at different stages of disease progression were used throughout this study; PEO1, PEO4, and PEO6 cells were obtained from a first patient, whereas PEO14 and PEO23 cells were isolated from a second patient. Both wound healing (WH) and Boyden chamber (BC) assays were performed to study the migratory capacity of each cell line, while invasion was assessed by adding a layer of extracellular matrix to the BC assays. Finally, 2D adhesion assays were performed on fibronectin coated plates. On the other hand, 3D organotypic models composed of a mixture of collagen I and fibroblasts, topped with a monolayer of mesothelial cells, were constructed to further study the metastatic potential of all five HGSOC cell lines; HGSOC were plated on top of the mesothelial cells, and their adhesive behavior and capacity to displace mesothelial cells was followed by fluorescence microscopy. In 2D studies, the migratory and invasive capacities of the HGSOC cells decreased along disease evolution in both cell line series. Moreover, a distinct migration pattern was observed; cell lines representing early-stage disease had a tendency to migrate as individual cells, while cells representing late stage disease migrated in clusters. Also in 2D conditions, adhesion rates were found to be similar between all cell lines. Noteworthy, these results obtained in 2D conditions were somewhat contradicted by the results obtained in the 3D organotypic models, as both late-state disease PEO6 and PEO23 cells adhered more rapidly to the mesothelial cell monolayer than their early-stage counterparts. Furthermore, cell lines previously found to be tumorigenic in vivo such as PEO4, PEO6, and PEO14, demonstrated to trigger higher rates of mesothelial cell displacement in the 3D organotypic model system. This mesothelial cell displacement was also observed after exposure of mesothelial cells to conditioned media derived from PEO4, PEO6, or PEO14 cultures. In conclusion, 2D in vitro assays demonstrated lower migration and invasion capacities at late- rather than early-stage disease, while the opposite was observed in the organotypic models. Mesothelial cell displacement was found to be associated with tumorigenicity in vivo and to be independent of physical interactions between cancer cells and mesothelial cells. Citation Format: Sabrina J. Ritch, Alicia A. Goyeneche, Abu S. Noman, Carlos M. Telleria. Discrepancies in the metastatic potential of high-grade serous ovarian cancer cells representing disease progression when comparing two-dimensional assays versus three-dimensional organotypic culture model systems [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3456.
APA, Harvard, Vancouver, ISO, and other styles
22

Baca, Martin, Dana Brauer, Maren Klett, et al. "Automated Analysis of Acetaminophen Toxicity on 3D HepaRG Cell Culture in Microbioreactor." Bioengineering 9, no. 5 (2022): 196. http://dx.doi.org/10.3390/bioengineering9050196.

Full text
Abstract:
Real-time monitoring of bioanalytes in organotypic cell cultivation devices is a major research challenge in establishing stand-alone diagnostic systems. Presently, no general technical facility is available that offers a plug-in system for bioanalytics in diversely available organotypic culture models. Therefore, each analytical device has to be tuned according to the microfluidic and interface environment of the 3D in vitro system. Herein, we report the design and function of a 3D automated culture and analysis device (3D-ACAD) which actively perfuses a custom-made 3D microbioreactor, samples the culture medium and simultaneously performs capillary-based flow ELISA. A microstructured MatriGrid® has been explored as a 3D scaffold for culturing HepaRG cells, with albumin investigated as a bioanalytical marker using flow ELISA. We investigated the effect of acetaminophen (APAP) on the albumin secretion of HepaRG cells over 96 h and compared this with the albumin secretion of 2D monolayer HepaRG cultures. Automated on-line monitoring of albumin secretion in the 3D in vitro mode revealed that the application of hepatotoxic drug-like APAP results in decreased albumin secretion. Furthermore, a higher sensitivity of the HepaRG cell culture in the automated 3D-ACAD system to APAP was observed compared to HepaRG cells cultivated as a monolayer. The results support the use of the 3D-ACAD model as a stand-alone device, working in real time and capable of analyzing the condition of the cell culture by measuring a functional analyte. Information obtained from our system is compared with conventional cell culture and plate ELISA, the results of which are presented herein.
APA, Harvard, Vancouver, ISO, and other styles
23

Neau, Laurent, Charlotte Lorin, Stefan Frentzel, et al. "Optimization of a NovelIn SituHybridization Technology on 3D Organotypic Cell Cultures." Applied In Vitro Toxicology 5, no. 2 (2019): 75–85. http://dx.doi.org/10.1089/aivt.2018.0021.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Chu, Chih-Wen, and Lance A. Davidson. "Chambers for Culturing and Immobilizing Xenopus Embryos and Organotypic Explants for Live Imaging." Cold Spring Harbor Protocols 2022, no. 5 (2021): pdb.prot107649. http://dx.doi.org/10.1101/pdb.prot107649.

Full text
Abstract:
Live imaging of Xenopus embryos and organotypic explants can be challenging because of their large size and slippery nature. This protocol covers the preparation of special chambers for immobilizing Xenopus embryos and embryonic explants for live-cell and tissue imaging. The opaque nature of Xenopus embryonic tissues enables simple bright-field imaging techniques for tracking surface movements across large regions. Such surface imaging of embryos or organotypic explants can directly reveal cell behaviors, obviating the need for complex postprocessing commonly required to extract this data from 3D confocal or light-sheet observations of more transparent embryos. Furthermore, Xenopus embryos may be filled with light-absorbing pigment granules and light-scattering yolk platelets, but these limitations are offset by the utilitarian nature of Xenopus organotypic explants that expose and stabilize large embryonic cells in a nearly native context for high-resolution live-cell imaging. Additionally, whole embryos can be stabilized for long-term bright-field and confocal microscopy. Simple explants can be prepared using a single cell type, and organotypic explants can be prepared in which multiple tissue types are dissected while retaining native tissue–tissue interactions. These preparations enable both in-toto imaging of tissue dynamics and super-resolution imaging of protein dynamics within individual cells. We present detailed protocols for these methods together with references to applications.
APA, Harvard, Vancouver, ISO, and other styles
25

Jeon, Jessie S., Simone Bersini, Mara Gilardi, et al. "Human 3D vascularized organotypic microfluidic assays to study breast cancer cell extravasation." Proceedings of the National Academy of Sciences 112, no. 1 (2014): 214–19. http://dx.doi.org/10.1073/pnas.1417115112.

Full text
Abstract:
A key aspect of cancer metastases is the tendency for specific cancer cells to home to defined subsets of secondary organs. Despite these known tendencies, the underlying mechanisms remain poorly understood. Here we develop a microfluidic 3D in vitro model to analyze organ-specific human breast cancer cell extravasation into bone- and muscle-mimicking microenvironments through a microvascular network concentrically wrapped with mural cells. Extravasation rates and microvasculature permeabilities were significantly different in the bone-mimicking microenvironment compared with unconditioned or myoblast containing matrices. Blocking breast cancer cell A3 adenosine receptors resulted in higher extravasation rates of cancer cells into the myoblast-containing matrices compared with untreated cells, suggesting a role for adenosine in reducing extravasation. These results demonstrate the efficacy of our model as a drug screening platform and a promising tool to investigate specific molecular pathways involved in cancer biology, with potential applications to personalized medicine.
APA, Harvard, Vancouver, ISO, and other styles
26

Desai, Diti, Malin Åkerfelt, Neeraj Prabhakar, et al. "Factors Affecting Intracellular Delivery and Release of Hydrophilic Versus Hydrophobic Cargo from Mesoporous Silica Nanoparticles on 2D and 3D Cell Cultures." Pharmaceutics 10, no. 4 (2018): 237. http://dx.doi.org/10.3390/pharmaceutics10040237.

Full text
Abstract:
Intracellular drug delivery by mesoporous silica nanoparticles (MSNs) carrying hydrophilic and hydrophobic fluorophores as model drug cargo is demonstrated on 2D cellular and 3D tumor organoid level. Two different MSN designs, chosen on the basis of the characteristics of the loaded cargo, were used: MSNs with a surface-grown poly(ethylene imine), PEI, coating only for hydrophobic cargo and MSNs with lipid bilayers covalently coupled to the PEI layer as a diffusion barrier for hydrophilic cargo. First, the effect of hydrophobicity corresponding to loading degree (hydrophobic cargo) as well as surface charge (hydrophilic cargo) on intracellular drug release was studied on the cellular level. All incorporated agents were able to release to varying degrees from the endosomes into the cytoplasm in a loading degree (hydrophobic) or surface charge (hydrophilic) dependent manner as detected by live cell imaging. When administered to organotypic 3D tumor models, the hydrophilic versus hydrophobic cargo-carrying MSNs showed remarkable differences in labeling efficiency, which in this case also corresponds to drug delivery efficacy in 3D. The obtained results could thus indicate design aspects to be taken into account for the development of efficacious intracellular drug delivery systems, especially in the translation from standard 2D culture to more biologically relevant organotypic 3D cultures.
APA, Harvard, Vancouver, ISO, and other styles
27

Millard, Melissa, Aaron Carlson, Danielle Y. Nadeau, et al. "Abstract 6768: Matched XPDX™derivative models as a platform for continuity across preclinical drug development." Cancer Research 84, no. 6_Supplement (2024): 6768. http://dx.doi.org/10.1158/1538-7445.am2024-6768.

Full text
Abstract:
Abstract Drug discovery is a reductive process in which druglike entities are carried through increasingly complex assay formats to tease out the compounds best suited for human studies. In vivo efficacy remains the benchmark for successful preclinical drug development. Cell-based models are a mainstay in early stages but translatability of different models across platforms remains a challenge. In oncology, patient-derived xenografts (PDX) are the gold standard for animal studies because they recapitulate key features of human tumors including the cellular heterogeneity, growth patterns, and drug response of the original patient tumor. Ex vivo, PDX derivatives have the potential to harness these same traits thereby extending the utility of PDX models to earlier stages of preclinical drug development. The KIYA-Predict™ platform is a clinically validated tool for the assessment of ex vivo drug responses across a broad range of drug classes that when applied to XPDX models shows good correlation to in vivo drug responses and excellent intra-sample reproducibility over time. Working with the well-characterized XenoSTART™ XPDX model repository, Kiyatec has derived 2D cell lines and organotypic 3D-expansion cultures to complement the ex vivo predictive value of our KIYA-PREDICT™ XPDX drug response profiling (DRP) platform. The development of syngeneic models applicable to simpler, high-throughput library screening (2D formats) and lower-throughput more complex mechanistic studies (3D organotypic models) could alleviate the burden and pitfalls of extrapolating data across unrelated models. Matched sets of 2D cell lines (2D-XPDX™) and 3D organotypic (XPDXO™) cultures were established from single cell suspensions of digested, mouse-cell depleted XPDX™ tissue samples. Drug response profiling of the XPDX™ in their various ex vivo culture formats showed relative similarities across platforms. Genomic profiling of the derivates confirmed shared tissue origin and retention of genomic and phenotypic signatures to varying degrees across models. Mechanistic evaluation of 3D organotypic culture demonstrated that disease-relevant biomarkers and processes are maintained. Together these results demonstrate the feasibility of this novel drug discovery platform that avoids ambiguity imparted by discordant models. Citation Format: Melissa Millard, Aaron Carlson, Danielle Y. Nadeau, Kimberly J. Burgess, Alyssa Simonson, Armando Diaz, Michael Wick, Teresa DesRochers. Matched XPDX™derivative models as a platform for continuity across preclinical drug development [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 6768.
APA, Harvard, Vancouver, ISO, and other styles
28

Engelmann, Luca, Julia Thierauf, Natalia Koerich Laureano, et al. "Organotypic Co-Cultures as a Novel 3D Model for Head and Neck Squamous Cell Carcinoma." Cancers 12, no. 8 (2020): 2330. http://dx.doi.org/10.3390/cancers12082330.

Full text
Abstract:
Background: Head and neck squamous cell carcinomas (HNSCC) are phenotypically and molecularly heterogeneous and frequently develop therapy resistance. Reliable patient-derived 3D tumor models are urgently needed to further study the complex pathogenesis of these tumors and to overcome treatment failure. Methods: We developed a three-dimensional organotypic co-culture (3D-OTC) model for HNSCC that maintains the architecture and cell composition of the individual tumor. A dermal equivalent (DE), composed of healthy human-derived fibroblasts and viscose fibers, served as a scaffold for the patient sample. DEs were co-cultivated with 13 vital HNSCC explants (non-human papillomavirus (HPV) driven, n = 7; HPV-driven, n = 6). Fractionated irradiation was applied to 5 samples (non-HPV-driven, n = 2; HPV-driven n = 3). To evaluate expression of ki-67, cleaved caspase-3, pan-cytokeratin, p16INK4a, CD45, ∝smooth muscle actin and vimentin over time, immunohistochemistry and immunofluorescence staining were performed Patient checkup data were collected for up to 32 months after first diagnosis. Results: All non-HPV-driven 3D-OTCs encompassed proliferative cancer cells during cultivation for up to 21 days. Proliferation indices of primaries and 3D-OTCs were comparable and consistent over time. Overall, tumor explants displayed heterogeneous growth patterns (i.e., invasive, expansive, silent). Cancer-associated fibroblasts and leukocytes could be detected for up to 21 days. HPV DNA was detectable in both primary and 3D-OTCs (day 14) of HPV-driven tumors. However, p16INK4a expression levels were varying. Morphological alterations and radioresistant tumor cells were detected in 3D-OTC after fractionated irradiation in HPV-driven and non-driven samples. Conclusions: Our 3D-OTC model for HNSCC supports cancer cell survival and proliferation in their original microenvironment. The model enables investigation of invasive cancer growth and might, in the future, serve as a platform to perform sensitivity testing upon treatment to predict therapy response.
APA, Harvard, Vancouver, ISO, and other styles
29

Shin, Hyun-Soo, Hye Jin Hong, Won-Gun Koh, and Jae-Yol Lim. "Organotypic 3D Culture in Nanoscaffold Microwells Supports Salivary Gland Stem-Cell-Based Organization." ACS Biomaterials Science & Engineering 4, no. 12 (2018): 4311–20. http://dx.doi.org/10.1021/acsbiomaterials.8b00894.

Full text
APA, Harvard, Vancouver, ISO, and other styles
30

Struber, Andreas, Georg Auer, Martin Fischlechner, et al. "Low-Cost Devices for Three-Dimensional Cell Aggregation, Real-Time Monitoring Microscopy, Microfluidic Immunostaining, and Deconvolution Analysis." Bioengineering 9, no. 2 (2022): 60. http://dx.doi.org/10.3390/bioengineering9020060.

Full text
Abstract:
The wide use of 3D-organotypic cell models is imperative for advancing our understanding of basic cell biological mechanisms. For this purpose, easy-to-use enabling technology is required, which should optimally link standardized assessment methods to those used for the formation, cultivation, and evaluation of cell aggregates or primordial tissue. We thus conceived, manufactured, and tested devices which provide the means for cell aggregation and online monitoring within a hanging drop. We then established a workflow for spheroid manipulation and immune phenotyping. This described workflow conserves media and reagent, facilitates the uninterrupted tracking of spheroid formation under various conditions, and enables 3D-marker analysis by means of 3D epifluorescence deconvolution microscopy. We provide a full description of the low-cost manufacturing process for the fluidic devices and microscopic assessment tools, and the detailed blueprints and building instructions are disclosed. Conclusively, the presented compilation of methods and techniques promotes a quick and barrier-free entry into 3D cell biology.
APA, Harvard, Vancouver, ISO, and other styles
31

Hensler, Sabine, Claudia Kühlbach, Jacquelyn Dawn Parente, Knut Moeller, and Margareta M. Mueller. "Establishment and initial characterization of a simple 3D wound healing model." Current Directions in Biomedical Engineering 5, no. 1 (2019): 581–84. http://dx.doi.org/10.1515/cdbme-2019-0146.

Full text
Abstract:
Abstract Poor wound healing as consequence of malfunctions in the regulation of the healthy tissue repair response affects millions of people worldwide. The number of therapies available to successfully treat chronic wound is still very limited and their development is costly and time consuming. Therefore simple to use 3D systems, reflecting the in vivo tissue complexity, are urgently needed. We introduce a novel 3D organotypic model (OTC) containing the major cell components active during wound healing i.e. keratinocytes, fibroblasts and inflammatory cells that allows to determine the effects of different therapeutic approaches on wound closure, cell differentiation and cytokine secretion in chronic wounds. There are first reports on irradiation with visible light of different wave length (Low Level Light Therapy) as a means to enhance wound closure. However the mechanisms underlying this therapy as well as optimized irradiation wavelength and dose are not clear and were therefore analyzed using our 3D organotypic model. In the standardized OTC model we could demonstrate epithelial closure under control conditions as well as differential effects of red and blue light irradiation with respect to stability of the newly formed epithelium and time until epithelial closure. First results show differential cytokine profiles upon different wavelength irradiation e.g. high expression of TGF beta and IL-1 beta in red light irradiated cultures and increased GM-CSF expression in blue light irradiated and control cultures.
APA, Harvard, Vancouver, ISO, and other styles
32

Angelopoulos, Ioannis, Konstantinos Ioannidis, Konstantina Gr Lyroni, et al. "A 3D SVZonChip Model for In Vitro Mimicry of the Subventricular Zone Neural Stem Cell Niche." Bioengineering 12, no. 6 (2025): 562. https://doi.org/10.3390/bioengineering12060562.

Full text
Abstract:
Neural stem cells (NSCs) are crucial components of the nervous system, primarily located in the subventricular zone (SVZ) and subgranular zone (SGZ). The SVZ neural stem cell niche (NSCN) is a specialized microenvironment where growth factors and extracellular matrix (ECM) components collaborate to regulate NSC self-renewal and differentiation. Despite its importance, our understanding of the SVZ remains incomplete due to the inherent challenges of animal research, particularly given the tissue’s dynamic nature. To address these limitations, we developed a proof-of-concept, dynamic, and tissue-specific 3D organotypic SVZ model to reduce reliance on animal models. This static 3D organotypic model integrates a region-specific decellularized ECM derived from the SVZ, mimicking the native NSCN and supporting mouse-derived ependymal cells (ECs), radial glial cells (RGCs), astrocytes, and NSCs. To further improve physiological relevance, we incorporated a dynamic microfluidic culture system (SVZonChip), replicating cerebrospinal fluid (CSF) flow as observed in vivo. The resulting SVZonChip platform, combining region-specific ECM proteins with dynamic culture conditions, provides a sustainable and reproducible tool to minimize animal model use. It holds significant promise for studying SVZ-related diseases, such as congenital hydrocephalus, stroke, and post-stroke neurogenesis, while advancing translational research and enabling personalized medicine protocols.
APA, Harvard, Vancouver, ISO, and other styles
33

Abady, Mariam, Ibrahim A. Zahran, and Yasmin Elmokhtar. "The liver organoid's past, present and future: a personalized medicine strategy." International journal of health sciences 8, S1 (2024): 972–98. http://dx.doi.org/10.53730/ijhs.v8ns1.14980.

Full text
Abstract:
Organoids are three-dimensional (3D) cell culture systems derived from human pluripotent stem cells or organotypic differentiation, replicating the complex interactions and functionalities of actual organs. These systems offer significant advantages for studying human tissue and organ biology, addressing limitations of animal models related to sample accessibility and ethical concerns. Liver organoids, in particular, are advanced models developed to study hepatic phenotypes, encompassing various cell types and enabling detailed investigation of cellular, molecular, and genetic aspects of liver diseases, drug metabolism, and protein secretion. They hold promise for fundamental research, drug discovery, and regenerative medicine applications. Despite their potential, organoids face limitations such as simplicity, lack of high-fidelity cell types, flexibility, and atypical physiology. Enhancements in organotypic liver-like surrogates, incorporating in vivo-like cell interactions and architecture, along with advancements in microfluidic chip technology, are expected to improve models for disease, toxicity, and drug discovery, paving the way for new treatments. This review will provide an overview of the history and development of liver organoids, their current progress, challenges, applications, and future prospects in the field of personalized medicine.
APA, Harvard, Vancouver, ISO, and other styles
34

Ahsan, Aarif, Danny Jeyaraju, Kamlesh Bisht, et al. "Modeling Tumor Microenvironment Interactions of Imid Sensitive and Resistant Cells in 3D Organotypic Culture Models." Blood 132, Supplement 1 (2018): 5622. http://dx.doi.org/10.1182/blood-2018-99-118885.

Full text
Abstract:
Abstract Background: Organotypic culture models developed using 3D conditions recapitulate tissue-specific structural features and cell-cell interactions more accurately than conventional 2D cultures. Our ultimate goal is to optimize culture conditions which promote the survival and proliferation of multiple myeloma (MM) cells and could serve as a platform for molecular mechanistic, clinical biomarker and pharmacodynamic marker studies using immune-modulatory compounds (IMIDs) and other myeloma drugs alone and in combination. Design/Results: Using gas permeable microfluidic devices, we cultured and compared growth/morphologic properties of six multiple myeloma cell lines, MM1.S, MM1.SPR, H929, H929PR, H929-220R and RPMI-8226 in 2D and 3D conditions. Collagen type IV was used as an extra-cellular matrix source to grow these cells. Cell growth and morphology was captured at regular intervals. Ten days post culture, cells were harvested from the device and stained for proliferation (Ki67 staining) index and expression of key MM oncogenic molecules, CD138, CD38 and BCMA. Cell lines grown in 3D conditions had, with some exceptions, higher proliferation index compared to 2D conditions. Thus, Ki67-mean fluorescence intensity (MFI) for 3D vs 2D were: 2038 vs 1130 for MM1.S; 1614 vs 1912 for MM1.PR; 2067 vs 1169 for H929; 2057 vs 1702 for H929PR; 2300 vs 1889 for H929-220R; 2018 vs 1220 for RPMI-8226. Similar trends for higher proliferation under 3D conditions were observed for the CD138, CD38 and BCMA cell subsets. Expression of FOXM1, a potential marker of IMID resistance, was reduced in Pomalidomide sensitive non-synchronous cells compared to resistant cells, although a few clusters with higher FOXM1 expression were observed among sensitive cells. To further study the effects of other components of MM tumor micro-environment on Pomalidomide response, we optimized the culture conditions to co-culture MM cell lines with bone marrow stromal cells. The co-culture of bone marrow stromal cells, HS5 with MM cell line H929 protected Ikaros degradation induced by Pomalidomide. Interestingly, CD44 expression in H929 cells was upregulated in co-culture conditions with stromal cells. Future Directions: These culture conditions are currently being optimized to study the (1) drug effects in MM and immune cells alone and in combination and (2) use the co-culture derived cells for single cell level evaluation of genetic, transcriptomic or proteomic changes associated with drug treatment and (3) ultimately grow primary Myeloma cells in these conditions for ex vivo manipulation and downstream molecular and biological effects. Figure. Figure. Disclosures Ahsan: celgene: Employment, Equity Ownership. Jeyaraju:Celgene Corporation: Employment, Equity Ownership. Bisht:Celgene Corporation: Employment, Equity Ownership. Hagner:Celgene Corporation: Employment, Equity Ownership. Bjorklund:Celgene Corporation: Employment, Equity Ownership. Pierceall:Celgene: Employment, Equity Ownership. Thakurta:Celgene Corporation: Employment, Equity Ownership.
APA, Harvard, Vancouver, ISO, and other styles
35

UMEZU, Shinjiro, Tomohiko AOKI, and Hitoshi OHMORI. "Patterning collagen for 3D cell structures." Journal of Advanced Science 24, no. 1+2 (2012): 11–15. http://dx.doi.org/10.2978/jsas.24.11.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Gastélum-López, María de los Ángeles, Maribel Aguilar-Medina, Cristina García Mata, et al. "Organotypic 3D Cell-Architecture Impacts the Expression Pattern of miRNAs–mRNAs Network in Breast Cancer SKBR3 Cells." Non-Coding RNA 9, no. 6 (2023): 66. http://dx.doi.org/10.3390/ncrna9060066.

Full text
Abstract:
Background. Currently, most of the research on breast cancer has been carried out in conventional two-dimensional (2D) cell cultures due to its practical benefits, however, the three-dimensional (3D) cell culture is becoming the model of choice in cancer research because it allows cell–cell and cell–extracellular matrix (ECM) interactions, mimicking the native microenvironment of tumors in vivo. Methods. In this work, we evaluated the effect of 3D cell organization on the expression pattern of miRNAs (by Small-RNAseq) and mRNAs (by microarrays) in the breast cancer SKBR3 cell line and analyzed the biological processes and signaling pathways regulated by the differentially expressed protein-coding genes (DE-mRNAs) and miRNAs (DE-microRNAs) found in the organoids. Results. We obtained well-defined cell-aggregated organoids with a grape cluster-like morphology with a size up to 9.2 × 105 μm3. The transcriptomic assays showed that cell growth in organoids significantly affected (all p < 0.01) the gene expression patterns of both miRNAs, and mRNAs, finding 20 upregulated and 19 downregulated DE-microRNAs, as well as 49 upregulated and 123 downregulated DE-mRNAs. In silico analysis showed that a subset of 11 upregulated DE-microRNAs target 70 downregulated DE-mRNAs. These genes are involved in 150 gene ontology (GO) biological processes such as regulation of cell morphogenesis, regulation of cell shape, regulation of canonical Wnt signaling pathway, morphogenesis of epithelium, regulation of cytoskeleton organization, as well as in the MAPK and AGE–RAGE signaling KEGG-pathways. Interestingly, hsa-mir-122-5p (Fold Change (FC) = 15.4), hsa-mir-369-3p (FC = 11.4), and hsa-mir-10b-5p (FC = 20.1) regulated up to 81% of the 70 downregulated DE-mRNAs. Conclusion. The organotypic 3D cell-organization architecture of breast cancer SKBR3 cells impacts the expression pattern of the miRNAs–mRNAs network mainly through overexpression of hsa-mir-122-5p, hsa-mir-369-3p, and hsa-mir-10b-5p. All these findings suggest that the interaction between cell–cell and cell–ECM as well as the change in the culture architecture impacts gene expression, and, therefore, support the pertinence of migrating breast cancer research from conventional cultures to 3D models.
APA, Harvard, Vancouver, ISO, and other styles
37

Marescotti, Diego, David Bovard, Moran Morelli, et al. "In Vitro High-Content Imaging-Based Phenotypic Analysis of Bronchial 3D Organotypic Air–Liquid Interface Cultures." SLAS TECHNOLOGY: Translating Life Sciences Innovation 25, no. 3 (2020): 247–52. http://dx.doi.org/10.1177/2472630319895473.

Full text
Abstract:
High-content imaging (HCI) is a powerful method for quantifying biological effects in vitro. Historically, HCI has been applied to adherent cells growing in monolayers. With the advent of confocal versions of HCI devices, researchers now have the option of performing analyses on 3D cell cultures. However, some obstacles remain in integrating the third dimension, such as limited light penetration and less sophisticated image analysis. Here, we report the development of an HCI technique for imaging human bronchial 3D organotypic air–liquid interface (ALI) cultures (hBR-ALI). In this method, we monitored differentiation status through HCI evaluation markers representative of ciliated epithelial cells and goblet cells (Muc5AC [mucin 5AC]). As a second use case for demonstrating the utility of this technique, we induced goblet cell hyperplasia in hBR-ALI by using interleukin (IL)-13. Our results demonstrate the utility of the HCI technique for imaging hBR-ALI grown on Transwell inserts. This technique may be expanded to other cell culture systems, such as skin epithelia and 3D intestinal systems.
APA, Harvard, Vancouver, ISO, and other styles
38

Mai, Patrick, Jörg Hampl, Martin Baca, et al. "MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing." Bioengineering 9, no. 5 (2022): 220. http://dx.doi.org/10.3390/bioengineering9050220.

Full text
Abstract:
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
APA, Harvard, Vancouver, ISO, and other styles
39

Iordachescu, Alexandra, Richard L. Williams, Philippa A. Hulley, and Liam M. Grover. "Organotypic Culture of Bone-Like Structures Using Composite Ceramic-Fibrin Scaffolds." Current Protocols in Stem Cell Biology 48, no. 1 (2019): e79. http://dx.doi.org/10.1002/cpsc.79.

Full text
APA, Harvard, Vancouver, ISO, and other styles
40

Bedard, Mary C., Marion G. Brusadelli, Adrean Carlile, et al. "Patient-Derived Organotypic Epithelial Rafts Model Phenotypes in Juvenile-Onset Recurrent Respiratory Papillomatosis." Viruses 13, no. 1 (2021): 68. http://dx.doi.org/10.3390/v13010068.

Full text
Abstract:
Juvenile-onset recurrent respiratory papillomatosis (JoRRP) is driven by human papillomavirus (HPV) low-risk strains and is associated with significant morbidity. While previous studies of 2D cultures have shed light on disease pathogenesis and demonstrated the utility of personalized medicine approaches, monolayer cultures lack the 3D tissue architecture and physiology of stratified, sequentially differentiated mucosal epithelium important in RRP disease pathogenesis. Herein we describe the establishment of JoRRP-derived primary cell populations that retain HPV genomes and viral gene expression in culture. These were directly compared to cells from matched adjacent non-diseased tissue, given the known RRP patient-to-patient variability. JoRRP papilloma versus control cells displayed decreased growth at subconfluency, with a switch to increased growth after reaching confluency, suggesting relative resistance to cell-cell contact and/or differentiation. The same papilloma cells grown as 3D organotypic rafts harbored hyperproliferation as compared to controls, with increased numbers of proliferating basal cells and inappropriately replicating suprabasal cells, mimicking phenotypes in the patient biopsies from which they were derived. These complementary model systems provide novel opportunities to elucidate disease mechanisms at distinct stages in JoRRP progression and to identify diagnostic, prognostic and therapeutic factors to personalize patient management and treatment.
APA, Harvard, Vancouver, ISO, and other styles
41

Shehzad, Ahmer, Fariza Mukasheva, Muhammad Moazzam, et al. "Dual-Crosslinking of Gelatin-Based Hydrogels: Promising Compositions for a 3D Printed Organotypic Bone Model." Bioengineering 10, no. 6 (2023): 704. http://dx.doi.org/10.3390/bioengineering10060704.

Full text
Abstract:
Gelatin-based hydrogels have emerged as a popular scaffold material for tissue engineering applications. The introduction of variable crosslinking methods has shown promise for fabricating stable cell-laden scaffolds. In this work, we examine promising composite biopolymer-based inks for extrusion-based 3D bioprinting, using a dual crosslinking approach. A combination of carefully selected printable hydrogel ink compositions and the use of photoinduced covalent and ionic crosslinking mechanisms allows for the fabrication of scaffolds of high accuracy and low cytotoxicity, resulting in unimpeded cell proliferation, extracellular matrix deposition, and mineralization. Three selected bioink compositions were characterized and the respective cell-laden scaffolds were bioprinted. Temporal stability, morphology, swelling, and mechanical properties of the scaffolds were thoroughly studied and the biocompatibility of the constructs was assessed using rat mesenchymal stem cells while focusing on osteogenesis. Experimental results showed that the composition of 1% alginate, 4% gelatin, and 5% (w/v) gelatine methacrylate, was found to be optimal among the examined, with shape fidelity of 88%, large cell spreading area and cell viability at around 100% after 14 days. The large pore diameters that exceed 100 µm, and highly interconnected scaffold morphology, make these hydrogels extremely potent in bone tissue engineering and bone organoid fabrication.
APA, Harvard, Vancouver, ISO, and other styles
42

Barua, Nilakshi, Lin Huang, Carmen Li, et al. "Comparative Study of Two-Dimensional (2D) vs. Three-Dimensional (3D) Organotypic Kertatinocyte-Fibroblast Skin Models for Staphylococcus aureus (MRSA) Infection." International Journal of Molecular Sciences 23, no. 1 (2021): 299. http://dx.doi.org/10.3390/ijms23010299.

Full text
Abstract:
The invasion of skin tissue by Staphylococcus aureus is mediated by mechanisms that involve sequential breaching of the different stratified layers of the epidermis. Induction of cell death in keratinocytes is a measure of virulence and plays a crucial role in the infection progression. We established a 3D-organotypic keratinocyte-fibroblast co-culture model to evaluate whether a 3D-skin model is more effective in elucidating the differences in the induction of cell death by Methicillin-resistant Staphylococcus aureus (MRSA) than in comparison to 2D-HaCaT monolayers. We investigated the difference in adhesion, internalization, and the apoptotic index in HaCaT monolayers and our 3D-skin model using six strains of MRSA representing different clonal types, namely, ST8, ST30, ST59, ST22, ST45 and ST239. All the six strains exhibited internalization in HaCaT cells. Due to cell detachment, the invasion study was limited up to two and a half hours. TUNEL assay showed no significant difference in the cell death induced by the six MRSA strains in the HaCaT cells. Our 3D-skin model provided a better insight into the interactions between the MRSA strains and the human skin during the infection establishment as we could study the infection of MRSA in our skin model up to 48 h. Immunohistochemical staining together with TUNEL assay in the 3D-skin model showed co-localization of the bacteria with the apoptotic cells demonstrating the induction of apoptosis by the bacteria and revealed the variation in bacterial transmigration among the MRSA strains. The strain representing ST59 showed maximum internalization in HaCaT cells and the maximum cell death as measured by Apoptotic index in the 3D-skin model. Our results show that 3D-skin model might be more likely to imitate the physiological response of skin to MRSA infection than 2D-HaCaT monolayer keratinocyte cultures and will enhance our understanding of the difference in pathogenesis among different MRSA strains.
APA, Harvard, Vancouver, ISO, and other styles
43

Zhuang, Lizhe, Rahul M. Visalakshan, and Pritinder Kaur. "Dermal Pericytes Exhibit Declined Ability to Promote Human Skin Regeneration with Ageing in 3D Organotypic Culture Models." Cells 10, no. 11 (2021): 3051. http://dx.doi.org/10.3390/cells10113051.

Full text
Abstract:
The well documented decline in the regenerative ability of ageing human skin has been attributed to many factors including genomic instability, telomere shortening, poor nutrient sensing, cellular senescence, and stem cell exhaustion. However, a role for the dermal cellular and molecular microenvironment in skin ageing is just emerging. We previously showed that dermal pericytes co-operate with fibroblasts to improve human skin regeneration in an organotypic skin culture model, and even do so in the absence of fibroblasts. Here, we report that the number of dermal cells, particularly pericytes, declines significantly in human skin of donors aged > 50 years. Notably, aged pericytes promoted epidermal regeneration of neonatal keratinocytes in organotypic cultures and the resulting epithelium exhibited a Ki67+/ΔNp63+ basal layer and terminal differentiation. However, the epithelium lacked several features of homeostasis displaying lower levels of ΔNp63 expression, decreased LAMA5 deposition at the dermo-epidermal junction, and the absence of basement membrane and hemi-desmosome assembly. We conclude that a decline in pericyte incidence and function contribute to an impaired epidermal microenvironment and poor skin regeneration with ageing in the human skin.
APA, Harvard, Vancouver, ISO, and other styles
44

Pratsinis, Harris, and Dimitris Kletsas. "Organotypic Cultures of Intervertebral Disc Cells: Responses to Growth Factors and Signaling Pathways Involved." BioMed Research International 2015 (2015): 1–10. http://dx.doi.org/10.1155/2015/427138.

Full text
Abstract:
Intervertebral disc (IVD) degeneration is strongly associated with low back pain, a major cause of disability worldwide. An in-depth understanding of IVD cell physiology is required for the design of novel regenerative therapies. Accordingly, aim of this work was the study of IVD cell responses to mitogenic growth factors in a three-dimensional (3D) organotypic milieu, comprising characteristic molecules of IVD’s extracellular matrix. In particular, annulus fibrosus (AF) cells were cultured inside collagen type-I gels, while nucleus pulposus (NP) cells in chondroitin sulfate A (CSA) supplemented collagen gels, and the effects of Platelet-Derived Growth Factor (PDGF), basic Fibroblast Growth Factor (bFGF), and Insulin-Like Growth Factor-I (IGF-I) were assessed. All three growth factors stimulated DNA synthesis in both AF and NP 3D cell cultures, with potencies similar to those observed previously in monolayers. CSA supplementation inhibited basal DNA synthesis rates, without affecting the response to growth factors. ERK and Akt were found to be phosphorylated following growth factor stimulation. Blockade of these two signaling pathways using pharmacologic inhibitors significantly, though not completely, inhibited growth factor-induced DNA synthesis. The proposed culture systems may prove useful for further in vitro studies aiming at future interventions for IVD regeneration.
APA, Harvard, Vancouver, ISO, and other styles
45

Härmä, Ville, Hannu-Pekka Schukov, Antti Happonen, et al. "Quantification of Dynamic Morphological Drug Responses in 3D Organotypic Cell Cultures by Automated Image Analysis." PLoS ONE 9, no. 5 (2014): e96426. http://dx.doi.org/10.1371/journal.pone.0096426.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Barrila, Jennifer, Andrea L. Radtke, Aurélie Crabbé, et al. "Organotypic 3D cell culture models: using the rotating wall vessel to study host–pathogen interactions." Nature Reviews Microbiology 8, no. 11 (2010): 791–801. http://dx.doi.org/10.1038/nrmicro2423.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Maggs, Luke, Yi Sun, Giulia Cattaneo, et al. "Abstract 4003: TBK1 inhibition enhances CAR-T cell efficacy against patient-derived organotypic tumor spheroids." Cancer Research 84, no. 6_Supplement (2024): 4003. http://dx.doi.org/10.1158/1538-7445.am2024-4003.

Full text
Abstract:
Abstract Novel therapeutic strategies are needed to improve the efficacy of chimeric antigen receptor (CAR)-T cells in the treatment of solid tumors. Here, we set out to evaluate novel therapeutic strategies to improve the efficacy of B7-H3 (CD276) directed (B7-H3.CAR-T) using 3D microfluidic cultures of patient-derived organotypic tumor spheroids (PDOTS). PDOTS are generated from fresh patient tumor tissue and contain tumor cells, autologous immune cells and stromal cells, therefore recapitulating key features of the tumor microenvironment (TME), enabling the study of tumor-immune dynamics. We confirmed activity of B7-H3.CAR-T in PDOTS, although a strong correlation between B7-H3 expression and CAR-T efficacy was not observed. Mechanistic studies subsequently demonstrated dynamic upregulation of co-inhibitory receptors on CAR-T cells following target cell encounter leading to CAR-T cell dysfunction and limiting CAR-T efficacy in B7-H3 expressing tumors. PD-1 blockade restored CAR-T activity in monotypic and organotypic tumor spheroids with improved tumor control and upregulation of effector cytokines. Given the emerging role of TANK-binding kinase 1 (TBK1) as an immune evasion gene, we examined the effect of TBK1 inhibition on CAR-T efficacy. Similar to PD-1 blockade, TBK1 inhibition restored CAR-T activity in monotypic and organotypic tumor spheroids, prevented CAR-T cell dysfunction, and enhanced T cell proliferation. Inhibition or deletion of TBK1 also enhanced sensitivity of cancer cells to immune-mediated killing. Taken together, our results demonstrate the feasibility and utility of ex vivo profiling of CAR-T cells using PDOTS, and suggest that targeting TBK1 is a novel strategy to enhance CAR-T efficacy by overcoming tumor-intrinsic and -extrinsic resistance mechanisms. Citation Format: Luke Maggs, Yi Sun, Giulia Cattaneo, Marco Ventin, Feng Chen, Martin Q. Rasmussen, Xinhui Wang, Cristina R. Ferrone, Soldano Ferrone, Russell W. Jenkins. TBK1 inhibition enhances CAR-T cell efficacy against patient-derived organotypic tumor spheroids [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 4003.
APA, Harvard, Vancouver, ISO, and other styles
48

Tavares-Valente, Diana, Stefania Cannone, Maria Raffaella Greco, et al. "Extracellular Matrix Collagen I Differentially Regulates the Metabolic Plasticity of Pancreatic Ductal Adenocarcinoma Parenchymal Cell and Cancer Stem Cell." Cancers 15, no. 15 (2023): 3868. http://dx.doi.org/10.3390/cancers15153868.

Full text
Abstract:
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of less than 10 percent largely due to the intense fibrotic desmoplastic reaction, characterized by high levels of extracellular matrix (ECM) collagen I that constitutes a niche for a subset of cancer cells, the cancer stem cells (CSCs). Cancer cells undergo a complex metabolic adaptation characterized by changes in metabolic pathways and biosynthetic processes. The use of the 3D organotypic model in this study allowed us to manipulate the ECM constituents and mimic the progression of PDAC from an early tumor to an ever more advanced tumor stage. To understand the role of desmoplasia on the metabolism of PDAC parenchymal (CPC) and CSC populations, we studied their basic metabolic parameters in organotypic cultures of increasing collagen content to mimic in vivo conditions. We further measured the ability of the bioenergetic modulators (BMs), 2-deoxyglucose, dichloroacetate and phenformin, to modify their metabolic dependence and the therapeutic activity of paclitaxel albumin nanoparticles (NAB-PTX). While all the BMs decreased cell viability and increased cell death in all ECM types, a distinct, collagen I-dependent profile was observed in CSCs. As ECM collagen I content increased (e.g., more aggressive conditions), the CSCs switched from glucose to mostly glutamine metabolism. All three BMs synergistically potentiated the cytotoxicity of NAB-PTX in both cell lines, which, in CSCs, was collagen I-dependent and the strongest when treated with phenformin + NAB-PTX. Metabolic disruption in PDAC can be useful both as monotherapy or combined with conventional drugs to more efficiently block tumor growth.
APA, Harvard, Vancouver, ISO, and other styles
49

Jackson, Robert, Esha V. Rajadhyaksha, Reid S. Loeffler, Caitlyn E. Flores, and Koenraad Van Doorslaer. "Characterization of 3D organotypic epithelial tissues reveals tonsil-specific differences in tonic interferon signaling." PLOS ONE 18, no. 10 (2023): e0292368. http://dx.doi.org/10.1371/journal.pone.0292368.

Full text
Abstract:
Three-dimensional (3D) culturing techniques can recapitulate the stratified nature of multicellular epithelial tissues. Organotypic 3D epithelial tissue culture methods have several applications, including the study of tissue development and function, drug discovery and toxicity testing, host-pathogen interactions, and the development of tissue-engineered constructs for use in regenerative medicine. We grew 3D organotypic epithelial tissues from foreskin, cervix, and tonsil-derived primary cells and characterized the transcriptome of these in vitro tissue equivalents. Using the same 3D culturing method, all three tissues yielded stratified squamous epithelium, validated histologically using basal and superficial epithelial cell markers. The goal of this study was to use RNA-seq to compare gene expression patterns in these three types of epithelial tissues to gain a better understanding of the molecular mechanisms underlying their function and identify potential therapeutic targets for various diseases. Functional profiling by over-representation and gene set enrichment analysis revealed tissue-specific differences: i.e., cutaneous homeostasis and lipid metabolism in foreskin, extracellular matrix remodeling in cervix, and baseline innate immune differences in tonsil. Specifically, tonsillar epithelia may play an active role in shaping the immune microenvironment of the tonsil balancing inflammation and immune responses in the face of constant exposure to microbial insults. Overall, these data serve as a resource, with gene sets made available for the research community to explore, and as a foundation for understanding the epithelial heterogeneity and how it may impact their in vitro use. An online resource is available to investigate these data (https://viz.datascience.arizona.edu/3DEpiEx/).
APA, Harvard, Vancouver, ISO, and other styles
50

Cavaliere, Fabio, Monica Benito-Muñoz, and Carlos Matute. "Organotypic Cultures as a Model to Study Adult Neurogenesis in CNS Disorders." Stem Cells International 2016 (2016): 1–6. http://dx.doi.org/10.1155/2016/3540568.

Full text
Abstract:
Neural regeneration resides in certain specific regions of adult CNS. Adult neurogenesis occurs throughout life, especially from the subgranular zone of hippocampus and the subventricular zone, and can be modulated in physiological and pathological conditions. Numerous techniques and animal models have been developed to demonstrate and observe neural regeneration but, in order to study the molecular and cellular mechanisms and to characterize multiple types of cell populations involved in the activation of neurogenesis and gliogenesis, investigators have to turn toin vitromodels. Organotypic cultures best recapitulate the 3D organization of the CNS and can be explored taking advantage of many techniques. Here, we review the use of organotypic cultures as a reliable and well defined method to study the mechanisms of neurogenesis under normal and pathological conditions. As an example, we will focus on the possibilities these cultures offer to study the pathophysiology of diseases like Alzheimer disease, Parkinson’s disease, and cerebral ischemia.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography