1
|
Shah S, D'Souza GGM. Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis. Cells 2025; 14:732. [PMID: 40422235 DOI: 10.3390/cells14100732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor-stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models.
Collapse
Affiliation(s)
- Shrey Shah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
- Atom Bioworks Inc., Cary, NC 27513, USA
| | - Gerard G M D'Souza
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| |
Collapse
|
2
|
Madsen NH, Nielsen BS, Skandorff I, Rodriguez-Pardo C, Hadrup SR, Ormhøj M, Holmstrøm K, Larsen J, Gad M. Novel approaches to 3D cancer heterospheroid culture and assay development for immunotherapy screening. Exp Cell Res 2025; 449:114604. [PMID: 40379236 DOI: 10.1016/j.yexcr.2025.114604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/14/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Advanced 3D heterospheroids, composed of cancer, fibroblast, and immune cells, serve as more physiologically relevant models for anticancer drug screening and immunotherapy research compared to traditional 2D cultures. This study aimed to optimize the culturing, dissociation, and analysis of heterospheroids, addressing limitations that restrict their broader use in immunotherapy research. Our study revealed significant effects of Human Plasma-Like culture medium on cell viability, necrotic core formation, and the spatial organization of cancer and fibroblast cells within heterospheroids compared to DMEM and RPMI media. In HT-29 heterospheroids, cell viability decreased from 75 % in DMEM to 20 % in HPLM, which was accompanied by increased necrotic core formation and elevated PD-L1 expression. TrypLE™ effectively dissociated heterospheroids but compromised immune cell viability and surface marker detection. In comparison, Accutase™ significantly reduced cell yield, while collagenase I preserved immune cell markers but affected those on cancer cells. Furthermore, we developed a luciferase-based assay to measure immune-mediated cancer cell killing in heterospheroids, excluding signals from non-target cells, such as dying fibroblasts and immune cells, without requiring spheroid lysis or dissociation. Our findings highlight the importance of tailoring experimental conditions to reflect specific tumor characteristics, thus enhancing the utility of heterospheroids in drug discovery and immunotherapy research.
Collapse
Affiliation(s)
- Natasha Helleberg Madsen
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark.
| | - Boye Schnack Nielsen
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Isabella Skandorff
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | | | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Maria Ormhøj
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Kim Holmstrøm
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Jesper Larsen
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Monika Gad
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| |
Collapse
|
3
|
Joshi P, Nascimento HSD, Kang SY, Lee M, Vanga MG, Lee SH, Ku B, Miranda MDS, Lee MY. Dynamic Culture of Bioprinted Liver Tumor Spheroids in a Pillar/Perfusion Plate for Predictive Screening of Anticancer Drugs. Biotechnol Bioeng 2025; 122:995-1009. [PMID: 39821523 DOI: 10.1002/bit.28924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/13/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025]
Abstract
Recent advancements in three-dimensional (3D) cell culture technologies, such as cell spheroids, organoids, and 3D bioprinted tissue constructs, have significantly improved the physiological relevance of in vitro models. These models better mimic tissue structure and function, closely emulating in vivo characteristics and enhancing phenotypic analysis, critical for basic research and drug screening in personalized cancer therapy. Despite their potential, current 3D cell culture platforms face technical challenges, which include user-unfriendliness in long-term dynamic cell culture, incompatibility with rapid cell encapsulation in biomimetic hydrogels, and low throughput for compound screening. To address these issues, we developed a 144-pillar plate with sidewalls and slits (144PillarPlate) and a complementary 144-perfusion plate with perfusion wells and reservoirs (144PerfusionPlate) for dynamic 3D cell culture and predictive compound screening. To accelerate biomimetic tissue formation, small Hep3B liver tumor spheroids suspended in alginate were printed and encapsulated on the 144PillarPlate rapidly by using microsolenoid valve-driven 3D bioprinting technology. The microarray bioprinting technology enabled precise and rapid loading of small spheroids in alginate on the pillar plate, facilitating reproducible and scalable formation of large tumor spheroids with minimal manual intervention. The bioprinted Hep3B spheroids on the 144PillarPlate were dynamically cultured in the 144PerfusionPlate and tested with anticancer drugs to measure drug effectiveness and determine the concentration required to inhibit 50% of the cell viability (IC50 value). The perfusion plate enabled the convenient dynamic culture of tumor spheroids and facilitated the dynamic testing of anticancer drugs with increased sensitivity. It is envisioned that the integration of microarray bioprinting of tumor spheroids onto the pillar plate, along with dynamic 3D cell culture in the perfusion plate, could more accurately replicate tumor microenvironments. This advancement has the potential to enhance the predictive drug screening process in personalized cancer therapy significantly.
Collapse
Affiliation(s)
- Pranav Joshi
- Bioprinting Laboratories Inc., Dallas, Texas, USA
| | - Hamilton Silva do Nascimento
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
- Institute of Veterinary Medicine, Federal University of Para, Castanhal, Brazil
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | - Minseong Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | | | | | - Bosung Ku
- MBD Co. Ltd., Suwon, Republic of Korea
| | | | - Moo-Yeal Lee
- Bioprinting Laboratories Inc., Dallas, Texas, USA
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| |
Collapse
|
4
|
Nugue G, Martins M, Vitória G, Guimaraes BLDML, Quiñones-Vega M, Rehen S, Guimarães MZ, Junqueira M. Optimized pipeline for personalized neurobiological insights from single patient-derived Neurospheres. J Proteomics 2025; 313:105368. [PMID: 39657900 DOI: 10.1016/j.jprot.2024.105368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024]
Abstract
This pipeline presents a refined approach for deriving personalized neurobiological insights from iPSC-derived neurospheres. By employing Tandem Mass Tag (TMT) labeling, we optimized sample pooling and multiplexing for robust comparative analysis across experimental conditions, maximizing data yield per sample. Through single-patient-derived neurospheres-composed of neural progenitor cells, early neurons, and radial glia-this study explores proteomic profiling to mirror the cellular complexity of neurodevelopment more accurately than traditional 2D cultures. Given their enhanced relevance, these 3D neurospheres serve as a valuable model for elucidating neurogenesis, differentiation, and neuropathological mechanisms, contributing to the advancement of in vitro neural models and reducing dependency on animal models. SIGNIFICANCE: This study evaluates ten protein extraction protocols using TMT 10-plex labeling to optimize proteomic analysis from single neurospheres. It compares cost, protein yield, and the ability to detect differentially expressed proteins, identifying methods like SPEED and S-Trap as efficient for high-throughput studies, while FASP excels in peptide yield. TMT labeling enhances protein identification, particularly for low-abundance proteins, and allows pre-fractionation to maximize analysis from limited samples. However, challenges such as limited PTM analysis and the potential loss of minor proteins highlight the importance of selecting protocols based on specific research goals. This work contributes to optimizing proteomic workflows for in vitro neural models, advancing single-cell analysis with minimal reliance on animal models.
Collapse
Affiliation(s)
- Guillaume Nugue
- Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Michele Martins
- Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Gabriela Vitória
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | | | - Mauricio Quiñones-Vega
- Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; Laboratory of Proteomics (LabProt), LADETEC, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil; Precision Medicine Research Center, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Stevens Rehen
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil; Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Brazil
| | - Marilia Z Guimarães
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Magno Junqueira
- Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil.
| |
Collapse
|
5
|
Lin CH, Leng ZC, Yu CH, Deori Bharali LK, Lin CL, Mao BH, Tu TY. Microscopic-based analysis of nuclei in spheroids via SUNSHINE: An on-chip workflow integrating optical clearing, fluorescence calibration and supervoxel segmentation. Comput Biol Med 2025; 187:109761. [PMID: 39923590 DOI: 10.1016/j.compbiomed.2025.109761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
Multicellular spheroids (MCSs) are increasingly employed as 3D cell culture models in biomedical research due to their ability to effectively replicate in vivo cell interactions, making them suitable for high-throughput drug screening. Accurate cell counting is critical for data normalization, therapeutic evaluation, and exploration of culture conditions; however, affordable software solutions for 3D cell counting using microscopic images are limited. To fill this gap, we created SUNSHINE, an innovative on-chip analytical workflow that uniquely merges optical clearing, histogram matching (HM)-assisted fluorescence calibration, and simple linear iterative clustering (SLIC) supervoxel segmentation. This tool offers an efficient method for analyzing the characteristics and counts of fluorescence-labeled nuclei within MCSs. While optical clearing improves the penetration depth of microscopic imaging, deeper regions of thicker samples often yield faint fluorescence signals. SUNSHINE resolves this issue through the HM image post-processing algorithm. Moreover, SLIC is an effective alternative to traditional contour-wise segmentation, enabling the identification of irregularly shaped fluorescent nuclei. We found that SUNSHINE generated results comparable to commercial software like Imaris and machine learning (ML)-based tools, such as StarDist and Cellpose, in our analysis of the effects of seeding density and cell type on spheroid growth. We also used it to measure the volume and spatial distribution of nuclei, focusing on the hypoxic and peripheral regions of spheroids. Overall, this study finds that SUNSHINE serves as a valuable and economical approach for characterizing cellular activity and interactions in 3D, diminishing the reliance on costly proprietary software.
Collapse
Affiliation(s)
- Chia-Hsiang Lin
- Department of Electrical Engineering, National Cheng Kung University, Tainan City, 70101, Taiwan; Miin Wu School of Computing, National Cheng Kung University, Tainan City, 70101, Taiwan; Meta-nanoPhotonics Center, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Zi-Chao Leng
- Institute of Computer and Communication Engineering, Department of Electrical Engineering, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Chien-Hsin Yu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Lui Kirtan Deori Bharali
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Cheng-Li Lin
- Department of Orthopedic Surgery, National Cheng Kung University Hospital, Tainan City, 704, Taiwan; Skeleton Materials and Biocompatibility Core Lab, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan City, 704, Taiwan; Medical Device Innovation Center (MDIC), National Cheng Kung University Hospital, Tainan City, 704, Taiwan; Musculoskeletal Research Center, Innovative Headquarter, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Bin-Hsu Mao
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan City, 70101, Taiwan.
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan City, 70101, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan City, 70101, Taiwan.
| |
Collapse
|
6
|
Noh S, Park Y, Kim B, Mun JY. Structural Analysis of Cerebral Organoids Using Confocal Microscopy and Transmission/Scanning Electron Microscopy. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2025; 31:ozae119. [PMID: 39999189 DOI: 10.1093/mam/ozae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/14/2024] [Accepted: 11/10/2024] [Indexed: 02/27/2025]
Abstract
Cerebral organoid cultures from human-induced pluripotent stem cells are widely used to study complex human brain development; however, there is still limited ultrastructural information regarding the development. In this study, we examined the structural details of cerebral organoids using various microscopy techniques. Two protocols were chosen as representative methods for the development of brain organoids: the classic whole-cerebral organoid (Whole-CO) culture technique, and the air-liquid interface-cerebral organoid (ALI-CO) culture technique. Immunostained confocal laser scanning microscopy (CLSM) revealed the formation of the CTIP2- and TBR1-positive cortical deep layer on days 90 and 150, depending on the developmental progress of both methods. Furthermore, the presence of astrocytes and oligodendrocytes was verified through immunostained CLSM utilizing two-dimensional and three-dimensional reconstruction images after a 150-day period. Transmission electron microscopy analysis revealed nanometer-resolution details of the cellular organelles and neuron-specific structures including synapses and myelin. Large-area scanning electron microscopy confirmed the well-developed neuronal connectivity from each culture method on day 150. Using those microscopy techniques, we clearly showed significant details within two representative culture protocols, the Whole-CO and ALI-CO culture methods. These multi-level images provide ultrastructural insight into the features of cerebral organoids depending on the developmental stage.
Collapse
Affiliation(s)
- Seulgi Noh
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Yurim Park
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Beomsue Kim
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| | - Ji Young Mun
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| |
Collapse
|
7
|
Vilinski-Mazur K, Kirillov B, Rogozin O, Kolomenskiy D. Numerical modeling of oxygen diffusion in tissue spheroids undergoing fusion using function representation and finite volumes. Sci Rep 2025; 15:5054. [PMID: 39934150 PMCID: PMC11814134 DOI: 10.1038/s41598-025-86805-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
A three-dimensional cell culture called a spheroid serves as a foundational entity in a wide variety of modern tissue engineering applications, including 3D-bioprinting and preclinical drug testing. Lack of oxygen within tissue spheroids hinders metabolism of cells and eventually leads to cell death. Prevention of necrosis is crucial to success of tissue engineering methods and such prevention requires estimation of cell viability in the spheroid. We propose a novel approach for numerical modeling of diffusion in tissue spheroids during their fusion. The approach is based on numerical solutions of partial differential equations and the application of Function Representation (FRep) framework for geometric modeling. We present modeling of oxygen diffusion based on meshes derived from the geometry of fusing spheroids, a method for selecting optimal spheroid size, and several statistics for estimating cellular viability. Our findings provide insights into oxygen diffusion in three-dimensional cell cultures thus improving the robustness of biotechnological methods that employ tissue spheroids.
Collapse
Affiliation(s)
| | - Bogdan Kirillov
- Center for Materials Technologies, Skolkovo Institute of Science and Technology, Moscow, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Oleg Rogozin
- Center for Materials Technologies, Skolkovo Institute of Science and Technology, Moscow, Russia
- Federal Research Center "Computer Science and Control", Russian Academy of Sciences, Moscow, Russia
| | - Dmitry Kolomenskiy
- Center for Materials Technologies, Skolkovo Institute of Science and Technology, Moscow, Russia
| |
Collapse
|
8
|
Brennan E, Pressete CG, Mohammadi N, Antunes LMG, Shang Q, Chen J, Bennett J, Franchin M, Granato D. Ultrasound-assisted recovery of blackcurrant press cake anthocyanins: Antioxidant and anti-inflammatory properties, bioaccessibility, and application in functional gummies. Food Chem X 2025; 26:102285. [PMID: 40071141 PMCID: PMC11894337 DOI: 10.1016/j.fochx.2025.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Blackcurrant press cake (BPC) anthocyanins were recovered using ultrasound-assisted extraction, and the optimal BPC extract was tested for its antioxidant capacity using chemical and biological assays and applied in a functional food model. Extraction at 400 W for 10 min followed by freeze-drying rendered an extract rich in polyphenols (47.83 mg GAE/g), where delphinidin-3-rutinoside, delphinidin-3-glucoside, cyanidin-3-rutinoside, and cyanidin-3-glucoside accounted for 75 % of total phenolics. When the reverse process of protonation/deprotonation was performed (pH 10 to pH 2), anthocyanins exhibited 79.3 % reversibility. The BPC extract inhibited human plasma oxidation (5052 mg AAE/g) and decreased intracellular reactive oxygen species generation by 54 % in erythrocytes and 45 % in LPS-stimulated THP-1 macrophage-like cells. BPC extract exhibited antiproliferative activity and cytostatic effect on HepG2 cells in monoculture at 100-250 μg/mL. Gummies added with BPC extract had a sensory acceptability of 75 %, but bioactive compounds had a low bioaccessibility.
Collapse
Affiliation(s)
- Emma Brennan
- Bioactivity & Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, V94 T9PX Limerick, Ireland
| | - Carolina Girotto Pressete
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Nima Mohammadi
- Bioactivity & Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, V94 T9PX Limerick, Ireland
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Qixiang Shang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Jihang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Jason Bennett
- Bioactivity & Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, V94 T9PX Limerick, Ireland
| | - Marcelo Franchin
- Bioactivity & Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, V94 T9PX Limerick, Ireland
| | - Daniel Granato
- Bioactivity & Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, V94 T9PX Limerick, Ireland
| |
Collapse
|
9
|
Gao Y, Bissoyi A, Guo Q, Gibson MI. Induced Extracellular Ice Nucleation Protects Cocultured Spheroid Interior and Exterior during Cryopreservation. ACS Biomater Sci Eng 2025; 11:208-212. [PMID: 39315639 PMCID: PMC11733914 DOI: 10.1021/acsbiomaterials.4c00958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
Spheroids and other 3D cellular models more accurately recapitulate physiological responses when compared to 2D models and represent potential alternatives to animal testing. The cryopreservation of spheroids remains challenging, limiting their wider use. Standard DMSO-only cryopreservation results in supercooling to low subzero temperatures, reducing viability, shedding surface cells, and perforating spheroid interiors. Here, cocultured spheroids with differentially labeled outer cell layers allow spatial evaluation of the protective effect of macromolecular ice nucleators by microscopy and histology. Extracellular nucleation is shown to reduce damage to both interior and exterior regions of the spheroids, which will support the development of "off-the-shelf" 3D models.
Collapse
Affiliation(s)
- Yanan Gao
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Akalabya Bissoyi
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess
Street, Manchester M1 7DN, United Kingdom
| | - Qiongyu Guo
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Matthew I. Gibson
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
- Division
of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, United
Kingdom
- Department
of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess
Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
10
|
Streller M, Michlíková S, Ciecior W, Lönnecke K, Kunz-Schughart LA, Lange S, Voss-Böhme A. Image segmentation of treated and untreated tumor spheroids by fully convolutional networks. Gigascience 2025; 14:giaf027. [PMID: 40331344 PMCID: PMC12056507 DOI: 10.1093/gigascience/giaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/07/2025] [Accepted: 02/27/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Multicellular tumor spheroids (MCTS) are advanced cell culture systems for assessing the impact of combinatorial radio(chemo)therapy as they exhibit therapeutically relevant in vivo-like characteristics from 3-dimensional cell-cell and cell-matrix interactions to radial pathophysiological gradients. State-of-the-art assays quantify long-term curative endpoints based on collected brightfield image time series from large treated spheroid populations per irradiation dose and treatment arm. This analyses require laborious spheroid segmentation of up to 100,000 images per treatment arm to extract relevant structural information from the images (e.g., diameter, area, volume, and circularity). While several image analysis algorithms are available for spheroid segmentation, they all focus on compact MCTS with a clearly distinguishable outer rim throughout growth. However, they often fail for the common case of treated MCTS, which may partly be detached and destroyed and are usually obscured by dead cell debris. RESULTS To address these issues, we successfully train 2 fully convolutional networks, UNet and HRNet, and optimize their hyperparameters to develop an automatic segmentation for both untreated and treated MCTS. We extensively test the automatic segmentation on larger, independent datasets and observe high accuracy for most images with Jaccard indices around 90%. For cases with lower accuracy, we demonstrate that the deviation is comparable to the interobserver variability. We also test against previously published datasets and spheroid segmentations. CONCLUSIONS The developed automatic segmentation can not only be used directly but also integrated into existing spheroid analysis pipelines and tools. This facilitates the analysis of 3-dimensional spheroid assay experiments and contributes to the reproducibility and standardization of this preclinical in vitro model.
Collapse
Affiliation(s)
- Matthias Streller
- DataMedAssist Group, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
- Faculty of Informatics/Mathematics, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
| | - Soňa Michlíková
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology–OncoRay, 01328 Dresden, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
| | - Willy Ciecior
- DataMedAssist Group, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
- Faculty of Informatics/Mathematics, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
| | - Katharina Lönnecke
- DataMedAssist Group, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
- Faculty of Informatics/Mathematics, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
| | - Leoni A Kunz-Schughart
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), NCT/UCC Dresden, 69192 Heidelberg, Germany
| | - Steffen Lange
- DataMedAssist Group, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
| | - Anja Voss-Böhme
- DataMedAssist Group, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
- Faculty of Informatics/Mathematics, HTW Dresden–University of Applied Sciences, 01069 Dresden, Germany
| |
Collapse
|
11
|
Xu J, Fang W, Zhou H, Jiang R, Chen Z, Wang X. Application and progress of 3D tumor models in breast cancer. Biotechnol Bioeng 2025; 122:30-43. [PMID: 39402769 DOI: 10.1002/bit.28860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 07/09/2024] [Accepted: 10/01/2024] [Indexed: 12/12/2024]
Abstract
Due to its high heterogeneity and significant impact on women's health globally, breast cancer necessitates robust preclinical models to understand tumor biology and guide personalized treatment strategies. Three-dimensional (3D) in vitro tumor models hold immense promise in this regard. These tumor models not only mimic the spatial structure and growth environment of tumors in vivo, but also retain the pathological and genetic characteristics of solid tumors. This fidelity makes them powerful tools for accelerating advancements in fundamental research and translational medicine. The diversity, modularity, and efficacy of 3D tumor models are driving a biotechnological revolution. As these technologies become increasingly sophisticated, 3D tumor models are poised to become powerful weapons in the fight against breast cancer. This article expounds on the progress made in utilizing 3D tumor models for breast cancer research.
Collapse
Affiliation(s)
- Jiaojiao Xu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wanxia Fang
- The Department of Colorectal Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Huanhuan Zhou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruiyuan Jiang
- The Department of Breast Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhanhong Chen
- The Department of Breast Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiaojia Wang
- The Department of Breast Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
12
|
Sekeroglu ZA, Sekeroglu V. A Review on Patient-derived 3D Micro Cancer Approach for Drug Screen in Personalized Cancer Medicine. Curr Cancer Drug Targets 2025; 25:118-130. [PMID: 38445692 DOI: 10.2174/0115680096285910240206044830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 03/07/2024]
Abstract
Precision medicine in oncology aims to identify an individualized treatment plan based on genomic alterations in a patient's tumor. It helps to select the most beneficial therapy for an individual patient. As it is now known that no patient's cancer is the same, and therefore, different patients may respond differently to conventional treatments, precision medicine, which replaces the one-size-fits-all approach, supports the development of tailored treatments for specific cancers of different patients. Patient-specific organoid or spheroid models as 3D cell culture models are very promising for predicting resistance to anti-cancer drugs and for identifying the most effective cancer therapy for high-throughput drug screening combined with genomic analysis in personalized medicine. Because tumor spheroids incorporate many features of solid tumors and reflect resistance to drugs and radiation, as in human cancers, they are widely used in drug screening studies. Testing patient-derived 3D cancer spheroids with some anticancer drugs based on information from molecular profiling can reveal the sensitivity of tumor cells to drugs and provide the right compounds to be effective against resistant cells. Given that many patients do not respond to standard treatments, patient-specific treatments will be more effective, less toxic. They will affect survival better compared to the standard approach used for all patients.
Collapse
Affiliation(s)
- Zulal Atlı Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| | - Vedat Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| |
Collapse
|
13
|
Olvera-Valencia M, Garcia-Castillo V, Ramos-Payan R, Aguilar-Medina M, Trujano-Camacho S, López-Saavedra A, Marchat LA, López-Camarillo C, Sumagin R, Pérez-Yepez E, Pérez-Plasencia C. Development of a reliable method for human triple-negative breast cancer organotypic culture: Improving imaging and genomic studies in 3D cultures. J Tissue Eng 2025; 16:20417314251326668. [PMID: 40342587 PMCID: PMC12059422 DOI: 10.1177/20417314251326668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/25/2025] [Indexed: 05/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and lacks targeted therapies, posing a major challenge in oncology. Traditional two-dimensional (2D) cell cultures fail to capture the tumor microenvironment's complexity, whereas three-dimensional (3D) cultures provide a more accurate model of tumor biology. We developed an advanced 3D culture system for TNBC cell lines BT-20 and MDA-MB-231, enhancing the hanging-drop method with Matrigel to restore essential extracellular matrix interactions. Confocal imaging showed MDA-MB-231 cells forming clusters typical of aggressive carcinoma, while BT-20 cells organized into duct-like structures. Molecular analysis of PI3K and β-catenin target genes revealed distinct expression patterns, with PI3K overexpressed and β-catenin downregulated in 3D cultures. Moreover, β-catenin distribution in the 3D cell culture closely resembles its pattern in tissue. These findings underscore the value of 3D models in understanding TNBC progression and in supporting the exploration of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mercedes Olvera-Valencia
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Ticoman, CDMX, Mexico
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
| | - Verónica Garcia-Castillo
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México, Mexico
| | - Rosalío Ramos-Payan
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacan, Sinaloa, Mexico
| | - Maribel Aguilar-Medina
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacan, Sinaloa, Mexico
| | - Samuel Trujano-Camacho
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
- Experimental Biology PhD Program, DCBS, Universidad Autónoma Metropolitana- Iztapalapa, Iztapalapa, Mexico
| | - Alejandro López-Saavedra
- Advanced Microscopy Applications Unit (ADMIRA)-Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Laurence A. Marchat
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Ticoman, CDMX, Mexico
| | - Cesar López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Benito Juarez, CDMX, Mexico
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eloy Pérez-Yepez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México, Mexico
| |
Collapse
|
14
|
Berry MA, Bland AR, Major GS, Ashton JC. Development of an ALK-positive Non-Small-Cell Lung Cancer in Vitro Tumor 3D Culture Model for Therapeutic Screening. J Histochem Cytochem 2025; 73:63-79. [PMID: 39991927 PMCID: PMC11851580 DOI: 10.1369/00221554251318435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 01/21/2025] [Indexed: 02/25/2025] Open
Abstract
Cancer cell monolayers are commonly used for preclinical drug screening. However, monolayers do not begin to mimic the complexity of the tumor microenvironment, including hypoxia and nutrient gradients within the tumor. To more accurately mimic solid tumors, we developed and drug-tested an anaplastic lymphoma kinase (ALK)-positive (H3122) non-small-cell lung cancer 3D (three-dimensional) culture model using light-activated gelatin methacryloyl hydrogels. We previously demonstrated that the combination of alectinib, an ALK inhibitor, and SHP099, an SHP2 inhibitor, had synergistic efficacy in ALK-positive cell monolayers. We aimed to test this drug combination in our novel ALK-positive 3D cancer model. We first validated the 3D cultures by comparing the distribution of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in the 3D cultures with sections from time-matched mouse xenografts, finding a comparable percentage of TUNEL-positive cells in the 3D culture and xenograft inner cores at each time point. When we investigated the effect of the combination of alectinib and SHP099 in these novel 3D cultures, we found a comparable cellular response compared with our two-dimensional experiments especially with the drugs in combination. We suggest that 3D cultures be used as preclinical screening platforms to ensure that only the most efficacious drug candidates move on to in vivo testing.
Collapse
Affiliation(s)
- Madeleine A. Berry
- Department of Pharmacology and Toxicology, Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Abigail R. Bland
- Department of Pharmacology and Toxicology, Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Gretel S. Major
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering and Nanomedicine, University of Otago, Christchurch, New Zealand
| | - John C. Ashton
- Department of Pharmacology and Toxicology, Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
15
|
Lin BJ, Fujie H, Yamazaki M, Sakamoto N. The dual effect of fiber density and matrix stiffness on A549 tumor multicellular migration. Biochem Biophys Res Commun 2024; 741:151018. [PMID: 39579534 DOI: 10.1016/j.bbrc.2024.151018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
The tumor microenvironment features dynamic biomechanical interactions between extracellular matrix physics and tumor progression. Tumor growth compresses the supportive matrix, and the stiffness-gradient guides tumor invasion. From the mechanical perspective, the complexity of the matrix topology involving durotaxis-driven metastasis remains lacking in a comprehensive description. In this study, A549 adenocarcinoma spheroids were exposed to a stiffness-and fiber-adjusted collagen matrix to examine the influence of collective motility. Centrifugated compression on the collagen constructs was adopted to mimic the matrix deformation in response to solid tumor development. Centrifugated compression physically stiffened and condensed collagen constructs simultaneously. Cultured with A549 spheroids for 7 days, compressed collagen constructs disadvantaged spheroid expansion without the effect of tumor proliferation potency but promoted matrix metalloproteinase activity corresponding to softened rigidity. Results suggested that the fibrous structure may counterbalance the matrix stiffness-induced motility.
Collapse
Affiliation(s)
- Bo-Jiang Lin
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 1920397, Japan.
| | - Hiromichi Fujie
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 1920397, Japan; Research Center for Medicine-Engineering Collaboration, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 1920397, Japan.
| | - Masashi Yamazaki
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 1920397, Japan; Research Center for Medicine-Engineering Collaboration, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 1920397, Japan.
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 1920397, Japan; Research Center for Medicine-Engineering Collaboration, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 1920397, Japan.
| |
Collapse
|
16
|
Mungai RW, Hartman RJ, Jolin GE, Piskorowski KW, Billiar KL. Towards a more objective and high-throughput spheroid invasion assay quantification method. Sci Rep 2024; 14:31007. [PMID: 39730859 DOI: 10.1038/s41598-024-82191-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
Multicellular spheroids embedded in 3D hydrogels are prominent in vitro models for 3D cell invasion. Yet, quantification methods for spheroid cell invasion that are high-throughput, objective and accessible are still lacking. Variations in spheroid sizes and the shapes of the cells within render it difficult to objectively assess invasion extent. The goal of this work is to develop a high-throughput quantification method of cell invasion into 3D matrices that minimizes sensitivity to initial spheroid size and cell spreading and provides precise integrative directionally-dependent metrics of invasion. By analyzing images of fluorescent cell nuclei, invasion metrics are automatically calculated at the pixel level. The initial spheroid boundary is segmented and automated calculations of the nuclear pixel distances from the initial boundary are used to compute common invasion metrics (i.e., the change in invasion area, mean distance) for the same spheroid at a later timepoint. We also introduce the area moment of inertia as an integrative metric of cell invasion that considers the invasion area as well as the pixel distances from the initial spheroid boundary. Further, we show that principal component analysis can be used to quantify the directional influence of a stimuli to invasion (e.g., due to a chemotactic gradient or contact guidance). To demonstrate the power of the analysis for cell types with different invasive potentials and the utility of this method for a variety of biological applications, the method is used to analyze the invasiveness of five different cell types. In all, implementation of this high-throughput quantification method results in consistent and objective analysis of 3D multicellular spheroid invasion. We provide the analysis code in both MATLAB and Python languages as well as a GUI for ease of use for researchers with a range of computer programming skills and for applications in a variety of biological research areas such as wound healing and cancer metastasis.
Collapse
Affiliation(s)
- Rozanne W Mungai
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | | | - Grace E Jolin
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | - Kevin W Piskorowski
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | - Kristen L Billiar
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA.
| |
Collapse
|
17
|
Basso J, Matos AM, Ghavami S, Fortuna A, Vitorino R, Vitorino C. Are we better together? Addressing a combined treatment of pitavastatin and temozolomide for brain cancer. Eur J Pharmacol 2024; 985:177087. [PMID: 39491742 DOI: 10.1016/j.ejphar.2024.177087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Pitavastatin is commonly prescribed to treat hypercholesterolemia through the regulation of cholesterol biosynthesis. Interestingly, it has also demonstrated a great potential for treating brain tumors, although the detailed cytotoxic mechanism, particularly in glioblastoma, remains incompletely understood. This work explores the activity of pitavastatin in 2D and 3D glioblastoma models, in an attempt to provide a more representative and robust overview of its anticancer potential in glioblastoma. The results show that not only is pitavastatin 10-1000 times-fold more effective in reducing tumoral metabolic activity than temozolomide, but also demonstrate a synergistic activity with this alkylating drug. In addition, low micromolar concentrations of this statin strongly impair the growth and the invasion ability of multicellular tumor spheroids. The obtained qRT-PCR and proteomics data highlight the modulation of cell death via apoptosis (BAX/BCL2, CASP9) and autophagy (BECN1, BNIP3, BNIP3L and LC3B), as well as an epithelial to mesenchymal transition blockage (HTRA1, SERPINE1, WNT5A, ALDH3B1 and EPHA2) and remodeling of the extracellular matrix (VCAN, SERPINE1 and TGFBI). Overall, these results lay the foundation for further investigations on the potential combinatory clinical treatment with temozolomide.
Collapse
Affiliation(s)
- João Basso
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Ana Miguel Matos
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Chemical Engineering and Renewable Resources for Sustainability, CERES, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB, R3E 0J9, Canada; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB, R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Ana Fortuna
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal; UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
18
|
Lim MC, Kim TY, Ok G, Kim HJ, Choi YS, Kim YR. Concave Microwell Formation Induced by PDMS Water Vapor Permeability for Spheroid Generation. MICROMACHINES 2024; 15:1496. [PMID: 39770249 PMCID: PMC11679915 DOI: 10.3390/mi15121496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
This study introduces a novel method for the fabrication of concave microwells involving water vapor permeation through polydimethylsiloxane (PDMS). This method leverages the exceptional water vapor permeability of PDMS to enable a scalable and cost-effective fabrication process, addressing the limitations of existing techniques such as photolithography that are resource-intensive and complex. PDMS is more permeable to water vapor than to other gas molecules, resulting in the formation of microwells. Smooth-sloped concave microwells are formed by depositing droplets of 10% ethylene glycol on a PDMS substrate followed by curing at 70 °C and evaporation of water vapor. These microwells exhibit a unique structural gradient that is highly conducive for biological applications. Concave microwells were further used as a platform to generate animal cell spheroids, demonstrating their potential for three-dimensional cell culture. Unlike conventional methods, this approach allows precise control over microwell morphology by simply adjusting droplet size and curing conditions, offering enhanced tunability and reproducibility. The formation yield of these microwells is dependent on the volume of the water droplets, demonstrating the importance of droplet size in controlling microwell morphology. This approach provides a simple and effective method for creating microwells without complex lithographic processes, making it a highly promising tool for a range of biomedical applications, including tissue engineering, cancer research, and high-throughput drug screening.
Collapse
Affiliation(s)
- Min-Cheol Lim
- Research Group of Food Safety and Distribution, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
- Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Tai-Yong Kim
- Research Group of Food Safety and Distribution, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Gyeongsik Ok
- Research Group of Food Safety and Distribution, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Hyun Jung Kim
- Research Group of Food Safety and Distribution, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
- Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yun-Sang Choi
- Research Group of Food Processing, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Young-Rok Kim
- Institute of Life Science and Resources & Department of Food Science and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
19
|
Dilloo S, Whittaker A, Chang X, D’Amen E, Spisni E, Hrelia S, Angeloni C, Malaguti M, Dinelli G, Truzzi F. Administration of Spermidine and Eugenol Demonstrates Anti-Tumorigenic Efficacy on Metastatic SW620 and Primary Caco-2 Colorectal Cancer Spheroids. Int J Mol Sci 2024; 25:13362. [PMID: 39769127 PMCID: PMC11679521 DOI: 10.3390/ijms252413362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The anti-cancer potential of eugenol (EUG) is well recognized, whereas that of spermidine (SPD) is subject to dispute and requires further research. The anti-tumorigenic potential of wheat germ SPD (150 µM) and clove EUG (100 µM), alone, in combination as SPD+EUG (50 µM + 100 µM) and, as a supplement (SUPPL; 0.6 µM SPD + 50 µM EUG), was investigated on both metastatic SW620 and primary Caco-2 colorectal cancer (CRC) spheroids. Compared to untreated controls, all treatments significantly reduced the vitality and spheroid area, increased the necrotic area, and induced apoptosis on both cell-type spheroids after 96 h, with a reduced migration evident in 2D (two-dimensional) cultures after 48 h. The comparable anti-CRC effects of the SPD+EUG and the SUPPL reflected a wide-range dose efficacy of SPD and EUG. It is of note that SPD+EUG induced a synergistic effect on the increased caspase-3 expression and reduced the migration percentage in SW620. In more physiologically relevant intestinal equivalents (healthy enterocytes [NCM460], fibroblasts [L929], and monocytes [U937]) containing embedded SW620/Caco-2 spheroids, SPD+EUG administration significantly reduced the spheroid CEA marker and proliferation, whilst simultaneously increasing occludin, autophagy LC3-II expression, and monocyte differentiation, compared to the control models. Exogenous SPD, alone and in combination with EUG, displayed an anti-CRC potential on tumor growth and metastasis, and warrants further investigation.
Collapse
Affiliation(s)
- Silvia Dilloo
- Department of Agricultural and Food Sciences, Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy; (S.D.); (A.W.); (X.C.); (E.D.); (G.D.)
| | - Anne Whittaker
- Department of Agricultural and Food Sciences, Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy; (S.D.); (A.W.); (X.C.); (E.D.); (G.D.)
| | - Xinyue Chang
- Department of Agricultural and Food Sciences, Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy; (S.D.); (A.W.); (X.C.); (E.D.); (G.D.)
| | - Eros D’Amen
- Department of Agricultural and Food Sciences, Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy; (S.D.); (A.W.); (X.C.); (E.D.); (G.D.)
| | - Enzo Spisni
- Department of Biological, Geological, and Environmental Sciences, Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy;
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, 47921 Rimini, Italy; (S.H.); (C.A.); (M.M.)
| | - Cristina Angeloni
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, 47921 Rimini, Italy; (S.H.); (C.A.); (M.M.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, 47921 Rimini, Italy; (S.H.); (C.A.); (M.M.)
| | - Giovanni Dinelli
- Department of Agricultural and Food Sciences, Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy; (S.D.); (A.W.); (X.C.); (E.D.); (G.D.)
| | - Francesca Truzzi
- Department of Agricultural and Food Sciences, Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy; (S.D.); (A.W.); (X.C.); (E.D.); (G.D.)
| |
Collapse
|
20
|
Louati K, Maalej A, Kolsi F, Kallel R, Gdoura Y, Borni M, Hakim LS, Zribi R, Choura S, Sayadi S, Chamkha M, Mnif B, Khemakhem Z, Boudawara TS, Boudawara MZ, Bouraoui A, Kraiem J, Safta F. A Shotgun Proteomic-Based Approach with a Q-Exactive Hybrid Quadrupole-Orbitrap High-Resolution Mass Spectrometer for the Assessment of Pesticide Mixture-Induced Neurotoxicity on a 3D-Developed Neurospheroid Model from Human Brain Meningiomas: Identification of Trityl-Post-Translational Modification. J Proteome Res 2024; 23:5554-5576. [PMID: 39556108 PMCID: PMC11629387 DOI: 10.1021/acs.jproteome.4c00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024]
Abstract
The widespread use of pesticides, particularly in combinations, has resulted in enhanced hazardous health effects. However, little is known about their molecular mechanism of interactions. The aim of this study was to assess the neurotoxicity effect of pesticides in mixtures by adopting a 3D in vitro developed neurospheroid model, followed by treatment by increased concentrations of pesticides for 24 h and analysis by a shotgun proteomic-based approach with high-resolution tandem mass spectrometry. Three proteins, namely, glyceraldehyde-3-phosphate-dehydrogenase (GAPDH), α-enolase, and phosphoglycerate-kinase-1, were selected as key targets in the metabolic process. Only high doses of pesticides mitigated cell-density proliferation with the occurrence of apoptotic cells, which unlikely makes any neurological alterations in environmental regulatory exposures. The proteomic analysis showed that majority of altered proteins were implicated in cell metabolism. De novo peptide sequencing revealed ion losses and adduct formation, namely, a trityl-post-translational modification in the active site of 201-GAPDH protein. The study also highlights the plausible role of pyrethroids to be implicated in the deleterious effects of pesticides in a mixture. To the best of our knowledge, our finding is the first in toxicoproteomics to deeply elucidate pesticides' molecular interactions and their ability to adduct proteins as a pivotal role in the neurotoxicity mechanism.
Collapse
Affiliation(s)
- Kaouthar Louati
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| | - Amina Maalej
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177 , 3018Sfax, Tunisia
| | - Fatma Kolsi
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Rim Kallel
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Yassine Gdoura
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Mahdi Borni
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Leila Sellami Hakim
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
| | - Rania Zribi
- Faculty
of Letters and Humanities, University of
Sfax, Airport Road, Km
4.5, 3023 Sfax, Tunisia
| | - Sirine Choura
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177 , 3018Sfax, Tunisia
| | - Sami Sayadi
- Biotechnology
Program, Center for Sustainable Development, College of Arts and Sciences, Qatar University, 2713 Doha, Qatar
| | - Mohamed Chamkha
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177 , 3018Sfax, Tunisia
| | - Basma Mnif
- Department
of Bacteriology, Habib Bourguiba University
Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Zouheir Khemakhem
- Legal
Medicine
Department, Habib Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Tahya Sellami Boudawara
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Mohamed Zaher Boudawara
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089Sfax, Tunisia
- Faculty
of Medicine, University of Sfax, Avenue of Majida Boulila, 3029Sfax, Tunisia
| | - Abderrahman Bouraoui
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| | - Jamil Kraiem
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| | - Fathi Safta
- Laboratory
of Chemical, Galenic and Pharmacological Drug Development- LR12ES09, University of Monastir, Road Avicenne , 5000Monastir, Tunisia
| |
Collapse
|
21
|
Shi XC, Zhang T, Li C, Guo CJ, Yang Q, Feng Y, Wang J, Qu CX. Impact of Nuclear Peripheral Chromatin Lamin LMNB1 Gene in the Proliferation and Migration of Glioma Cells. Neurochem Res 2024; 50:46. [PMID: 39636549 DOI: 10.1007/s11064-024-04298-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
The goal of this study is to explore the role of the LMNB1 gene in glioma. A cohort of 160 patients who underwent glioma surgery were randomly selected of this study. The LMNB1 expression was assessed employing immunohistochemical and real-time quantitative polymerase chain reaction methods. Initially, RNA interference technology was applied to suppress gene expression, followed by the evaluation of tumor cell proliferation, apoptosis, cell cycle dynamics, and migration. The underlying molecular mechanisms of LMNB1 function were examined by a human phospho-kinase array and immunoblotting. And we established the xenograft models to determine the effect of tumor growth as well as the degree of invasion in shLMNB1 mice. Elevated LMNB1 expression correlated with unfavorable overall survival and disease-free survival. A substantial inhibition in cell growth was observed subsequent to LMNB1 knockdown in SHG-44 and U251 glioma cells. SHG-44-shLMNB1 cells exhibited a reduction in the S phase population, along with an increase in cells in G1 and G2 phases. Similarly, shLMNB1 U251 cells showed fewer cells in the S phase and an elevation in cells in G1 phase. Notably, increased apoptosis was observed in U251-shLMNB1 cells and SHG-44-shLMNB1 cells. Wound healing and Transwell migration assays demonstrated a significant decrease in the migration rate of both SHG-44-shLMNB1 and U251-shLMNB1 cells. The phosphorylation levels of Akt1/2/3, as well as the expressions of PI3K, AKT, and p-AKT proteins, were reduced in the shLMNB1 group. Downregulation of LMNB1 repressed tumor progress in vivo. The silencing of LMNB1 was found to significantly reduce the proliferation of human glioma cells, induce apoptosis in tumor cells, impede the progression of the cell cycle, and inhibit the migration of tumor cells. Consequently, we hypothesize that LMNB1 promotes glioma cell proliferation through mechanisms involving the inhibition of tumor cell apoptosis, acceleration of the cell cycle, and enhancement of tumor cell migration. We found that LMNB1 exert critical roles in glioma progression may via regulation of PI3K/Akt signaling pathway. These observations suggest that LMNB1 holds clinical potential for diagnostic and prognostic applications in glioma, presenting novel targets for drug development.
Collapse
Affiliation(s)
- Xiang-Cheng Shi
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Ting Zhang
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Cheng Li
- The Pathology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030012, China
| | - Chen-Jia Guo
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Qin Yang
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Yao Feng
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Jie Wang
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Chong-Xiao Qu
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China.
- Department of Pathology, Shanxi Provincial People's Hospital, No. 29 of Shuangtasi Road, Yingze District, Taiyuan, 030012, China.
| |
Collapse
|
22
|
Patel T, Jain N. Multicellular tumor spheroids: A convenient in vitro model for translational cancer research. Life Sci 2024; 358:123184. [PMID: 39490437 DOI: 10.1016/j.lfs.2024.123184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
In the attempts to mitigate uncertainties in the results of monolayer culture for the identification of cancer therapeutic targets and compounds, there has been a growing interest in using 3D cancer spheroid models, which include tumorospheres (TSs), tissue-derived tumor spheres (TDTSs), organotypic multicellular tumor spheroids (OMSs), and multicellular tumor spheroids (MCTSs). The MCTSs, either Mono-MCTSs or Hetero-MCTSs, with or without scaffold, in particular, offer numerous advantages over other spheroid models, including easy cultivation, high reproducibility, accessibility, high throughput, controllable size, well-rounded shape, simplicity of genetic manipulation, economical and availability of various biological methods for their development. In this review, we have attempted to discuss the role of MCTSs concerning various aspects of translational cancer research, such as drug response and penetration, cell-cell interaction, and invasion and metastasis. However, the Mono-MCTSs, either scaffold-free or scaffold-based, may not adequately represent the cellular heterogeneity and complexity of clinical tumors, limiting their utility in translational cancer research. Conversely, Hetero-MCTS models, both scaffold-free and scaffold-based, show better suitability due to the presence of a similar in vivo type tumor microenvironment. Nonetheless, scaffold-based Hetero-MCTS models show batch variability and challenges in performing quantitative assays due to difficulties extracting spheroids and cells from scaffolds. Further, incorporating stromal cells with cancer cells in a more precise ratio to develop Hetero-MCTSs can enhance the model's relevance, yielding more clinically reliable outcomes for drug candidates and improving insights into tumor biology.
Collapse
Affiliation(s)
- Tushar Patel
- P D Patel Institute of Applied Sciences, Charotar University of Science and Technology (CHARUSAT), Changa 388 421, India
| | - Neeraj Jain
- Dr. K C Patel Research and Development Centre, University Research Centre(s), Charotar University of Science and Technology (CHARUSAT), Changa 388 421, India.
| |
Collapse
|
23
|
Noguchi R, Osaki J, Ono T, Adachi Y, Iwata S, Yoshimatsu Y, Sasaki K, Kawai A, Kondo T. Pharmacoproteogenomic approach identifies on-target kinase inhibitors for cancer drug repositioning. In Vitro Cell Dev Biol Anim 2024; 60:1200-1214. [PMID: 39422823 DOI: 10.1007/s11626-024-00983-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/01/2024] [Indexed: 10/19/2024]
Abstract
Drug repositioning of approved drugs offers advantages over de novo drug development for a rare type of cancer. To efficiently identify on-target drugs from clinically successful kinase inhibitors in cancer drug repositioning, drug screening and molecular profiling of cell lines are essential to exclude off-targets. We developed a pharmacoproteogenomic approach to identify on-target kinase inhibitors, combining molecular profiling of genomic features and kinase activity, and drug screening of patient-derived cell lines. This study examined eight patient-derived giant cell tumor of the bone (GCTB) cell lines, all of which harbored a signature mutation of H3-3A but otherwise without recurrent copy number variants and mutations. Kinase activity profiles of 100 tyrosine kinases with a three-dimensional substrate peptide array revealed that nine kinases were highly activated. Pharmacological screening of 60 clinically used kinase inhibitors found that nine drugs directed at 29 kinases strongly suppressed cell viability. We regarded ABL1, EGFR, and LCK as on-target kinases; among the two corresponding on-target kinase inhibitors, osimertinib and ponatinib emerged as on-target drugs whose target kinases were significantly activated. The remaining 26 kinases and seven kinase inhibitors were excluded as off-targets. Our pharmacoproteomic approach enabled the identification of on-target kinase inhibitors that are useful for drug repositioning.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Julia Osaki
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Adachi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shuhei Iwata
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Kazuki Sasaki
- Department of Oncopeptidomics, Tochigi Cancer Center; 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology and Rehabilitation Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
24
|
Sueca-Comes M, Rusu EC, Ashworth JC, Collier P, Probert C, Ritchie A, Meakin M, Mongan NP, Egbuniwe IU, Andersen JB, Bates DO, Grabowska AM. The role of mesenchymal cells in cholangiocarcinoma. Dis Model Mech 2024; 17:dmm050716. [PMID: 39492622 DOI: 10.1242/dmm.050716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
The tumour microenvironment (TME) significantly influences tumour formation and progression through dynamic interactions. Cholangiocarcinoma (CCA), a highly desmoplastic tumour, lacks early diagnostic biomarkers and has limited effective treatments owing to incomplete understanding of its molecular pathogenesis. Investigating the role of the TME in CCA progression could lead to better therapies. RNA sequencing was performed on seven CCA patient-derived xenografts (PDXs) and their corresponding patient samples. Differential expression analysis was conducted, and Qiagen Ingenuity Pathway Analysis was used to predict dysregulated pathways and upstream regulators. PDX- and cell line-derived spheroids, with and without immortalised mesenchymal stem cells, were grown and analysed for morphology, growth and viability. Histological analysis confirmed biliary phenotypes. RNA sequencing indicated upregulation of extracellular matrix-receptor interaction and PI3K-AKT pathways in the presence of mesenchymal cells, with several genes linked to poor survival. Mesenchymal cells restored the activity of inhibited cancer-associated kinases. Thus, adding mesenchymal cells to CCA spheroid models restored key paracrine signalling pathways lost in PDXs, enhancing tumour growth and viability. These findings highlight the importance of including stromal components in cancer models to improve pre-clinical studies.
Collapse
Affiliation(s)
- Mireia Sueca-Comes
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Elena Cristina Rusu
- Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas (CSIC), 46980 Valencia, Spain
| | - Jennifer C Ashworth
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Leicestershire LE12 5RD, UK
| | - Pamela Collier
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Catherine Probert
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Alison Ritchie
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Marian Meakin
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nigel P Mongan
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Leicestershire LE12 5RD, UK
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Isioma U Egbuniwe
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jesper Bøje Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - David O Bates
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Anna M Grabowska
- Translational Medical Science, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
25
|
Liu M, Wang Y, Wang C, Li P, Qiu J, Yang N, Sun M, Han L. A Microfluidic 3D-Tumor-Spheroid Model for the Evaluation of Targeted Therapies from Angiogenesis-Related Cytokines at the Single Spheroid Level. Adv Healthc Mater 2024; 13:e2402321. [PMID: 39126126 DOI: 10.1002/adhm.202402321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Indexed: 08/12/2024]
Abstract
Angiogenesis is a key player in drug resistance to targeted therapies for breast cancer. The average expression of angiogenesis-related cytokines is widely associated with the treatments of target therapies for a population of cells or spheroids, overlooking the distinct responses for individuals. In this work, a highly integrated microfluidic platform is developed for the generation of monodisperse multicellular tumor spheroids (MTSs), drug treatments, and the measurement of cytokines for individual MTSs in a single chip. The platform allows the correlation evaluation between cytokine secretion and drug treatment at the level of individual spheroids. For validation, quantities of six representative proangiogenic cytokines are tested against treatments with four model drugs at varying times and concentrations. By applying a linear regression model, significant correlations are established between cytokine secretion and the treated drug concentration for individual spheroids. The proposed platform provides a high-throughput method for the investigation of the molecular mechanism of the cytokine response to targeted therapies and paves the way for future drug screening using predictive regression models at the single-spheroid level.
Collapse
Affiliation(s)
- Mengqi Liu
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Yihe Wang
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Chao Wang
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Ping Li
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Jiaoyan Qiu
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Ningkai Yang
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Mingyuan Sun
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, 250100, P. R. China
| |
Collapse
|
26
|
Crestani M, Kakogiannos N, Iori S, Iannelli F, Dini T, Maderna C, Giannotta M, Pelicci G, Maiuri P, Monzo P, Gauthier NC. Biomimetic Approach of Brain Vasculature Rapidly Characterizes Inter- and Intra-Patient Migratory Diversity of Glioblastoma. SMALL METHODS 2024; 8:e2400210. [PMID: 38747088 PMCID: PMC11671864 DOI: 10.1002/smtd.202400210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/04/2024] [Indexed: 12/28/2024]
Abstract
Glioblastomas exhibit remarkable heterogeneity at various levels, including motility modes and mechanoproperties that contribute to tumor resistance and recurrence. In a recent study using gridded micropatterns mimicking the brain vasculature, glioblastoma cell motility modes, mechanical properties, formin content, and substrate chemistry are linked. Now is presented, SP2G (SPheroid SPreading on Grids), an analytic platform designed to identify the migratory modes of patient-derived glioblastoma cells and rapidly pinpoint the most invasive sub-populations. Tumorspheres are imaged as they spread on gridded micropatterns and analyzed by this semi-automated, open-source, Fiji macro suite that characterizes migration modes accurately. SP2G can reveal intra-patient motility heterogeneity with molecular correlations to specific integrins and EMT markers. This system presents a versatile and potentially pan-cancer workflow to detect diverse invasive tumor sub-populations in patient-derived specimens and offers a valuable tool for therapeutic evaluations at the individual patient level.
Collapse
Affiliation(s)
- Michele Crestani
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
- Present address:
Laboratory of Applied MechanobiologyDepartment of Health Sciences and TechnologyInstitute of Translational MedicineETH ZurichZurichCH‐8093Switzerland
| | - Nikolaos Kakogiannos
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
- Institute of ImmunologyBiomedical Sciences Research Centre “Alexander Fleming”34 Fleming StreetVari16672Greece
| | - Simone Iori
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
| | - Fabio Iannelli
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
- Department of Experimental OncologyIEOEuropean Institute of Oncology IRCCSMilan20139Italy
| | - Tania Dini
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
| | - Claudio Maderna
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
| | - Monica Giannotta
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
| | - Giuliana Pelicci
- Department of Experimental OncologyIEOEuropean Institute of Oncology IRCCSMilan20139Italy
- Department of Translational MedicinePiemonte Orientale University ‘‘Amedeo Avogadro’’Novara28100Italy
| | - Paolo Maiuri
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi diNapoli Federico IIVia S. Pansini 5Naples80131Italy
| | - Pascale Monzo
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
| | - Nils C. Gauthier
- IFOM ETS – The AIRC Institute of Molecular OncologyVia Adamello 16Milan20139Italy
| |
Collapse
|
27
|
Jo H, Lee S, Kim MH, Park S, Lee SY. Recapitulating Glioma Stem Cell Niches Using 3D Spheroid Models for Glioblastoma Research. BIOSENSORS 2024; 14:539. [PMID: 39589998 PMCID: PMC11592235 DOI: 10.3390/bios14110539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Glioblastoma multiforme (GBM) is among the most aggressive brain cancers, and it contains glioma stem cells (GSCs) that drive tumor initiation, progression, and recurrence. These cells resist conventional therapies, contributing to high recurrence rates in GBM patients. Developing in vitro models that mimic the tumor microenvironment (TME), particularly the GSC niche, is crucial for understanding GBM growth and therapeutic resistance. Three-dimensional (3D) spheroid models provide a more physiologically relevant approach than traditional two-dimensional (2D) cultures, recapitulating key tumor features like hypoxia, cell heterogeneity, and drug resistance. This review examines scaffold-free and scaffold-based methods for generating 3D GBM spheroids, focusing on their applications in studying the cancer stem cell niche. The discussion encompasses methods such as the hanging drop, low-adhesion plates, and magnetic levitation, alongside advancements in embedding spheroids within extracellular matrix-based hydrogels and employing 3D bioprinting to fabricate more intricate tumor models. These 3D culture systems offer substantial potential for enhancing our understanding of GBM biology and devising more effective targeted therapies.
Collapse
Affiliation(s)
- Hyunji Jo
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (H.J.); (S.L.)
| | - Seulgi Lee
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (H.J.); (S.L.)
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea;
| | - Sungsu Park
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (H.J.); (S.L.)
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea;
- Department of Quantum Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Seo-Yeon Lee
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
- Department of Biomedical Science, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| |
Collapse
|
28
|
Guo J, Tan W, Xu B. Enzymatic self-assembly of short peptides for cell spheroid formation. J Mater Chem B 2024; 12:11210-11217. [PMID: 39370899 PMCID: PMC11540748 DOI: 10.1039/d4tb01154f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Cell spheroids, including organoids, serve as a valuable link between in vitro systems and in vivo animal models, offering powerful tools for studying cell biology in a three-dimensional environment. However, existing methods for generating cell spheroids are time consuming or difficult to scale up for large-scale production. Our recent study has revealed that transcytotic peptide assemblies, which transform from nanoparticles to nanofibers by enzymatic reactions, can create an intercellular fibril/gel, accelerating cell spheroid formation from a 2D cell culture or a cell suspension. While this finding presents an alternative approach for generating cell spheroids, the specific structural features required for efficient cell spheroid formation remain unclear. Based on our observation that a phosphotetrapeptide with a biphenyl cap at its N-terminus enables cell spheroid formation, we produced 10 variants of the original peptide. The variants explored modifications to the peptide backbone, length, electronic properties of the biphenyl capping group, and the type of phosphorylated amino acid residue. We then evaluated their ability for inducing cell spheroid formation. Our analysis revealed that, among the tested molecules, peptides with C-terminal phosphotyrosine, low critical micelle concentration, and dephosphorylation-guided nanoparticle to nanofiber morphological transition were the most effective in inducing the formation of cell spheroids. This work represents the first example to correlate the thermodynamic properties (e.g., self-assembling ability) and kinetic behavior (e.g., enzymatic dephosphorylation) of peptides with the efficacy of controlling intercellular interaction, thus offering valuable insights into using enzymatic self-assembly to generate peptide assemblies for biological applications.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA.
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA.
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA.
| |
Collapse
|
29
|
Martínez-Alonso C, Izzo L, Rodríguez-Carrasco Y, Ruiz MJ. Integrated Approach to Cyclopiazonic Acid Cytotoxicity Using In Vitro (2D and 3D Models) and In Silico Methods. Toxins (Basel) 2024; 16:473. [PMID: 39591228 PMCID: PMC11598133 DOI: 10.3390/toxins16110473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Cyclopiazonic acid (CPA) is an indole-tetramic acid neurotoxin produced by Aspergillus and Penicillium genera present mainly in fruit, cereals and nuts. This study compares the cytotoxicity produced by CPA after 24, 48 and 72 h of exposure using both monolayers and 3D spheroids in human neuroblastoma SH-SY5Y cells. Furthermore, CPA toxicokinetics was evaluated using in silico models. Cytotoxicity increased dose- and time-dependently, as shown by the MTT assay. The lowest CPA IC50 values were found in the monolayer study compared to the 3D spheroids at all exposure times (24 h: 864.01 vs. 1132; 48 h: 437 vs. 1069; 72 h: 392 vs. 567 nM). The CPA exposure on SH-SY5Y spheroid organization and morphology was also studied. Morphological changes, including spheroid disaggregation, were observed after mycotoxin exposure. The in silico methods, SwissADME and admetSAR, were used for short and full ADMEt profiles of CPA. The ADMEt predictive profile shows high gastrointestinal absorption and ability to penetrate the blood-brain barrier. Including in silico studies emphasizes the comprehensive approach to understanding mycotoxin toxicity and risk assessment. By combining in vitro 3D spheroid models with computational simulations, this study aims to provide a holistic perspective on the effects of CPA, enhancing the accuracy and relevance of our findings.
Collapse
Affiliation(s)
- Carmen Martínez-Alonso
- Department of Preventive Medicine and Public Health, Food Science, Toxicology and Forensic Medicine, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent A Estelles s/n, Burjassot, 46100 Valencia, Spain; (C.M.-A.); (M.-J.R.)
| | - Luana Izzo
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano, 49, 80131 Naples, Italy;
| | - Yelko Rodríguez-Carrasco
- Department of Preventive Medicine and Public Health, Food Science, Toxicology and Forensic Medicine, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent A Estelles s/n, Burjassot, 46100 Valencia, Spain; (C.M.-A.); (M.-J.R.)
| | - María-José Ruiz
- Department of Preventive Medicine and Public Health, Food Science, Toxicology and Forensic Medicine, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent A Estelles s/n, Burjassot, 46100 Valencia, Spain; (C.M.-A.); (M.-J.R.)
| |
Collapse
|
30
|
Yadav P, Singh S, Jaiswal S, Kumar R. Synthetic and natural polymer hydrogels: A review of 3D spheroids and drug delivery. Int J Biol Macromol 2024; 280:136126. [PMID: 39349080 DOI: 10.1016/j.ijbiomac.2024.136126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
This review centers on the synthesis and characterization of both natural and synthetic hydrogels, highlighting their diverse applications across various fields. We will delve into the evolution of hydrogels, focusing on the importance of polysaccharide-based and synthetic variants, which have been particularly chosen for 3D spheroid development in cancer research and drug delivery. A detailed background on the research and specific methodologies, including the in-situ free radical polymerization used for synthesizing these hydrogels, will be extensively discussed. Additionally, the review will explore various applications of these hydrogels, such as their self-healing properties, swelling ratios, pH responsiveness, and cell viability. A comprehensive literature review will support this investigation. Ultimately, this review aims to clearly outline the objectives and significance of hydrogel synthesis and their applications.
Collapse
Affiliation(s)
- Paramjeet Yadav
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Shiwani Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Sheetal Jaiswal
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Rajesh Kumar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| |
Collapse
|
31
|
Arora S, Singh S, Mittal A, Desai N, Khatri DK, Gugulothu D, Lather V, Pandita D, Vora LK. Spheroids in cancer research: Recent advances and opportunities. J Drug Deliv Sci Technol 2024; 100:106033. [DOI: 10.1016/j.jddst.2024.106033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
|
32
|
Serafini CE, Charles S, Casteleiro Costa P, Niu W, Cheng B, Wen Z, Lu H, Robles FE. Non-invasive label-free imaging analysis pipeline for in situ characterization of 3D brain organoids. Sci Rep 2024; 14:22331. [PMID: 39333572 PMCID: PMC11436713 DOI: 10.1038/s41598-024-72038-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024] Open
Abstract
Brain organoids provide a unique opportunity to model organ development in a system similar to human organogenesis in vivo. Brain organoids thus hold great promise for drug screening and disease modeling. Conventional approaches to organoid characterization predominantly rely on molecular analysis methods, which are expensive, time-consuming, labor-intensive, and involve the destruction of the valuable three-dimensional (3D) architecture of the organoids. This reliance on end-point assays makes it challenging to assess cellular and subcellular events occurring during organoid development in their 3D context. As a result, the long developmental processes are not monitored nor assessed. The ability to perform non-invasive assays is critical for longitudinally assessing features of organoid development during culture. In this paper, we demonstrate a label-free high-content imaging approach for observing changes in organoid morphology and structural changes occurring at the cellular and subcellular level. Enabled by microfluidic-based culture of 3D cell systems and a novel 3D quantitative phase imaging method, we demonstrate the ability to perform non-destructive high-resolution quantitative image analysis of the organoid. The highlighted results demonstrated in this paper provide a new approach to performing live, non-destructive monitoring of organoid systems during culture.
Collapse
Affiliation(s)
- Caroline E Serafini
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30318, USA
| | - Seleipiri Charles
- Georgia Institute of Technology, Interdisciplinary Program in Bioengineering, Atlanta, GA, 30332, USA
| | - Paloma Casteleiro Costa
- Georgia Institute of Technology, School of Electrical and Computer Engineering, Atlanta, GA, 30332, USA
| | - Weibo Niu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Brian Cheng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30318, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
- Departments of Cell Biology and Neurology, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Hang Lu
- Georgia Institute of Technology, Interdisciplinary Program in Bioengineering, Atlanta, GA, 30332, USA
- Georgia Institute of Technology, School of Chemical and Biomolecular Engineering, Atlanta, Georgia, 30332, USA
| | - Francisco E Robles
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30318, USA.
- Georgia Institute of Technology, Interdisciplinary Program in Bioengineering, Atlanta, GA, 30332, USA.
- Georgia Institute of Technology, School of Electrical and Computer Engineering, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30318, USA.
| |
Collapse
|
33
|
Ciufolini G, Zampieri S, Cesaroni S, Pasquale V, Bonanomi M, Gaglio D, Sacco E, Vanoni M, Pastore M, Marra F, Cicero DO, Raggi C, Petrella G. 3D Modeling: Insights into the Metabolic Reprogramming of Cholangiocarcinoma Cells. Cells 2024; 13:1536. [PMID: 39329720 PMCID: PMC11430555 DOI: 10.3390/cells13181536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/28/2024] Open
Abstract
Developing accurate in vitro models that replicate the in vivo tumor environment is essential for advancing cancer research and therapeutic development. Traditional 2D cell cultures often fail to capture the complex structural and functional heterogeneity of tumors, limiting the translational relevance of findings. In contrast, 3D culture systems, such as spheroids, provide a more physiologically relevant context by replicating key aspects of the tumor microenvironment. This study aimed to compare the metabolism of three intrahepatic cholangiocarcinoma cell lines in 2D and 3D cultures to identify metabolic shifts associated with spheroid formation. Cells were cultured in 2D on adhesion plates and in 3D using ultra-low attachment plates. Metabolic exchange rates were measured using NMR, and intracellular metabolites were analyzed using LC-MS. Significant metabolic differences were observed between 2D and 3D cultures, with notable changes in central carbon and glutathione metabolism in 3D spheroids. The results suggest that 3D cultures, which more closely mimic the in vivo environment, may offer a more accurate platform for cancer research and drug testing.
Collapse
Affiliation(s)
- Giorgia Ciufolini
- Department of Chemical Science and Technology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.C.); (S.Z.); (S.C.); (D.O.C.)
| | - Serena Zampieri
- Department of Chemical Science and Technology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.C.); (S.Z.); (S.C.); (D.O.C.)
| | - Simona Cesaroni
- Department of Chemical Science and Technology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.C.); (S.Z.); (S.C.); (D.O.C.)
| | - Valentina Pasquale
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, 20126 Milan, Italy; (V.P.); (E.S.); (M.V.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy; (M.B.); (D.G.)
| | - Marcella Bonanomi
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy; (M.B.); (D.G.)
- Institute of Bioimaging and Complex Biological Systems (IBSBC), 20054 Segrate, Italy
| | - Daniela Gaglio
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy; (M.B.); (D.G.)
- Institute of Bioimaging and Complex Biological Systems (IBSBC), 20054 Segrate, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, 20126 Milan, Italy; (V.P.); (E.S.); (M.V.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy; (M.B.); (D.G.)
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, 20126 Milan, Italy; (V.P.); (E.S.); (M.V.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy; (M.B.); (D.G.)
| | - Mirella Pastore
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (M.P.); (F.M.)
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (M.P.); (F.M.)
| | - Daniel Oscar Cicero
- Department of Chemical Science and Technology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.C.); (S.Z.); (S.C.); (D.O.C.)
| | - Chiara Raggi
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (M.P.); (F.M.)
| | - Greta Petrella
- Department of Chemical Science and Technology, University of Rome “Tor Vergata”, 00133 Rome, Italy; (G.C.); (S.Z.); (S.C.); (D.O.C.)
| |
Collapse
|
34
|
Xhafa S, Di Nicola C, Tombesi A, Pettinari R, Pettinari C, Scarpelli F, Crispini A, La Deda M, Candreva A, Garufi A, D'Orazi G, Galindo A, Marchetti F. Pyrazolone-Based Zn(II) Complexes Display Antitumor Effects in Mutant p53-Carrying Cancer Cells. J Med Chem 2024; 67:15676-15690. [PMID: 39221914 DOI: 10.1021/acs.jmedchem.4c01298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The synthesis and characterization of nine Schiff bases of pyrazolone ligands HLn (n = 1-9) and the corresponding zinc(II) complexes 1-9 of composition [Zn(Ln)2] (n = 1-9) are reported. The molecular structures of complexes 2, 3, 4, 8, and 9 were determined by single-crystal X-ray diffraction analysis, highlighting in all cases a distorted tetrahedral geometry around the Zn(II) ion. Density functional theory studies are performed on both the HLn ligands and the derived complexes. A mechanism of dissociation and hydrolyzation of the coordinated Schiff base ligands is suggested, confirmed experimentally by powder X-ray diffraction study and photophysical studies. Complexes 1-9 were investigated in vitro as anticancer agents, along with mutant p53 (mutp53) protein levels in human cancer cell lines carrying R175H and R273H mutp53 proteins. Only those complexes with the highest Zn(II) ion release via dissociation have shown a significant cytotoxic activity with reduction of mutp53 protein levels.
Collapse
Affiliation(s)
- Sonila Xhafa
- ChIP Research Center, School of Science and Technology, University of Camerino, via Madonna delle Carceri Camerino, 62032 Macerata, Italy
| | - Corrado Di Nicola
- ChIP Research Center, School of Science and Technology, University of Camerino, via Madonna delle Carceri Camerino, 62032 Macerata, Italy
| | - Alessia Tombesi
- ChIP Research Center, School of Pharmacy, University of Camerino, via Madonna delle Carceri Camerino, 62032 Macerata, Italy
| | - Riccardo Pettinari
- ChIP Research Center, School of Pharmacy, University of Camerino, via Madonna delle Carceri Camerino, 62032 Macerata, Italy
| | - Claudio Pettinari
- ChIP Research Center, School of Pharmacy, University of Camerino, via Madonna delle Carceri Camerino, 62032 Macerata, Italy
| | - Francesca Scarpelli
- MAT-InLAB, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Alessandra Crispini
- MAT-InLAB, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Massimo La Deda
- MAT-InLAB, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Angela Candreva
- MAT-InLAB, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Alessia Garufi
- Department of Research and Advanced Technologies, IRCCS Regina Elena, National Cancer Institute, via Elio Chianesi 53, 00144 Rome, Italy
| | - Gabriella D'Orazi
- Department of Research and Advanced Technologies, IRCCS Regina Elena, National Cancer Institute, via Elio Chianesi 53, 00144 Rome, Italy
- Department of Neurosciences, Imaging and Clinical Sciences, University G. D'Annunzio, via dei Vestini 31, 66013 Chieti, Italy
| | - Agustín Galindo
- Departamento de Química Inorgánica, Facultad de Química, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Fabio Marchetti
- ChIP Research Center, School of Science and Technology, University of Camerino, via Madonna delle Carceri Camerino, 62032 Macerata, Italy
| |
Collapse
|
35
|
Wang J, Tran-Huynh AM, Kim BJ, Chan DW, Holt MV, Fandino D, Yu X, Qi X, Wang J, Zhang W, Wu YH, Anurag M, Zhang XHF, Zhang B, Cheng C, Foulds CE, Ellis MJ. Death-associated protein kinase 3 modulates migration and invasion of triple-negative breast cancer cells. PNAS NEXUS 2024; 3:pgae401. [PMID: 39319326 PMCID: PMC11421662 DOI: 10.1093/pnasnexus/pgae401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
Sixteen patient-derived xenografts (PDXs) were analyzed using a mass spectrometry (MS)-based kinase inhibitor pull-down assay (KIPA), leading to the observation that death-associated protein kinase 3 (DAPK3) is significantly and specifically overexpressed in the triple-negative breast cancer (TNBC) models. Validation studies confirmed enrichment of DAPK3 protein, in both TNBC cell lines and tumors, independent of mRNA levels. Genomic knockout of DAPK3 in TNBC cell lines inhibited in vitro migration and invasion, along with down-regulation of an epithelial-mesenchymal transition (EMT) signature, which was confirmed in vivo. The kinase and leucine-zipper domains within DAPK3 were shown by a mutational analysis to be essential for functionality. Notably, DAPK3 was found to inhibit the levels of desmoplakin (DSP), a crucial component of the desmosome complex, thereby explaining the observed migration and invasion effects. Further exploration with immunoprecipitation-mass spectrometry (IP-MS) identified that leucine-zipper protein 1 (LUZP1) is a preferential binding partner of DAPK3. LUZP1 engages in a leucine-zipper domain-mediated interaction that protects DAPK3 from proteasomal degradation. Thus, the DAPK3/LUZP1 heterodimer emerges as a newly discovered regulator of EMT/desmosome components that promote TNBC cell migration.
Collapse
Affiliation(s)
- Junkai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anh M Tran-Huynh
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Doug W Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew V Holt
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Diana Fandino
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xin Yu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaoli Qi
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiang H F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chonghui Cheng
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Charles E Foulds
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
36
|
Nowak-Jary J, Płóciennik A, Machnicka B. Functionalized Magnetic Fe 3O 4 Nanoparticles for Targeted Methotrexate Delivery in Ovarian Cancer Therapy. Int J Mol Sci 2024; 25:9098. [PMID: 39201784 PMCID: PMC11354664 DOI: 10.3390/ijms25169098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Magnetic Fe3O4 nanoparticles (MNPs) functionalized with (3-aminopropylo)trietoksysilan (APTES) or N-carboxymethylchitosan (CMC) were proposed as nanocarriers of methotrexate (MTX) to target ovarian cancer cell lines. The successful functionalization of the obtained nanostructures was confirmed by FT-IR spectroscopy. The nanoparticles were characterized by transmission electron spectroscopy (TEM) and dynamic light scattering (DLS) techniques. Their potential zeta, magnetization, and hyperthermic properties were also explored. MTX was conjugated with the nanocarriers by ionic bonds or by amide bonds. The drug release kinetics were examined at different pH and temperatures. The MTT assay showed no toxicity of the MNPs[APTES] and MNPs[CMC]. Finally, the cytotoxicity of the nanostructures with MTX attached towards the ovarian cancer cells was measured. The sensitivity and resistance to methotrexate was determined in simplistic 2D and spheroid 3D conditions. The cytotoxicity tests of the tested nanostructures showed similar values for inhibiting the proliferation of ovarian cancer cells as methotrexate in its free form. Conjugating MTX with nanoparticles allows the drug to be directed to the target site using an external magnetic field, reducing overall toxicity. Combining this approach with hyperthermia could enhance the therapeutic effect in vivo compared to free MTX, though further research on advanced 3D models is needed.
Collapse
Affiliation(s)
- Julia Nowak-Jary
- Department of Biotechnology, Institute of Biological Sciences, University of Zielona Gora, 65-516 Zielona Gora, Poland;
| | - Artur Płóciennik
- Institute of Experimental Biology, University of Poznan, 61-614 Poznan, Poland;
| | - Beata Machnicka
- Department of Biotechnology, Institute of Biological Sciences, University of Zielona Gora, 65-516 Zielona Gora, Poland;
| |
Collapse
|
37
|
Shin YB, Choi JY, Yoon MS, Yoo MK, Shin DH, Lee JW. Evaluation of Anticancer Efficacy of D-α-Tocopheryl Polyethylene-Glycol Succinate and Soluplus ® Mixed Micelles Loaded with Olaparib and Rapamycin Against Ovarian Cancer. Int J Nanomedicine 2024; 19:7871-7893. [PMID: 39114180 PMCID: PMC11304412 DOI: 10.2147/ijn.s468935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Purpose Ovarian cancer has the highest mortality rate and lowest survival rate among female reproductive system malignancies. There are treatment options of surgery and chemotherapy, but both are limited. In this study, we developed and evaluated micelles composed of D-α-tocopheryl polyethylene-glycol (PEG) 1000 succinate (TPGS) and Soluplus® (SOL) loaded with olaparib (OLA), a poly(ADP-ribose)polymerase (PARP) inhibitor, and rapamycin (RAPA), a mammalian target of rapamycin (mTOR) inhibitor in ovarian cancer. Methods We prepared micelles containing different molar ratios of OLA and RAPA embedded in different weight ratios of TPGS and SOL (OLA/RAPA-TPGS/SOL) were prepared and physicochemical characterized. Furthermore, we performed in vitro cytotoxicity experiments of OLA, RAPA, and OLA/RAPA-TPGS/SOL. In vivo toxicity and antitumor efficacy assays were also performed to assess the efficacy of the mixed micellar system. Results OLA/RAPA-TPGS/SOL containing a 4:1 TPGS:SOL weight ratio and a 2:3 OLA:RAPA molar ratio showed synergistic effects and were optimized. The drug encapsulation efficiency of this formulation was >65%, and the physicochemical properties were sustained for 180 days. Moreover, the formulation had a high cell uptake rate and significantly inhibited cell migration (**p < 0.01). In the in vivo toxicity test, no toxicity was observed, with the exception of the high dose group. Furthermore, OLA/RAPA-TPGS/SOL markedly inhibited tumor spheroid and tumor growth in vivo. Conclusion Compared to the control, OLA/RAPA-TPGS/SOL showed significant tumor inhibition. These findings lay a foundation for the use of TPGS/SOL mixed micelles loaded with OLA and RAPA in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yu Been Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Ju-Yeon Choi
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Moon Sup Yoon
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Myeong Kyun Yoo
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
- Chungbuk National University Hospital, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| |
Collapse
|
38
|
Pirayeshfard L, Luo S, Githaka JM, Saini A, Touret N, Goping IS, Julien O. Comparing the BAD Protein Interactomes in 2D and 3D Cell Culture Using Proximity Labeling. J Proteome Res 2024; 23:3433-3443. [PMID: 38959414 PMCID: PMC11302415 DOI: 10.1021/acs.jproteome.4c00111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Protein-protein interaction studies using proximity labeling techniques, such as biotin ligase-based BioID, have become integral in understanding cellular processes. Most studies utilize conventional 2D cell culture systems, potentially missing important differences in protein behavior found in 3D tissues. In this study, we investigated the protein-protein interactions of a protein, Bcl-2 Agonist of cell death (BAD), and compared conventional 2D culture conditions to a 3D system, wherein cells were embedded within a 3D extracellular matrix (ECM) mimic. Using BAD fused to the engineered biotin ligase miniTurbo (BirA*), we identified both overlapping and distinct BAD interactomes under 2D and 3D conditions. The known BAD binding proteins 14-3-3 isoforms and Bcl-XL interacted with BAD in both 2D and 3D. Of the 131 BAD-interactors identified, 56% were specific to 2D, 14% were specific to 3D, and 30% were common to both conditions. Interaction network analysis demonstrated differential associations between 2D and 3D interactomes, emphasizing the impact of the culture conditions on protein interactions. The 2D-3D overlap interactome encapsulated the apoptotic program, which is a well-known role of BAD. The 3D unique pathways were enriched in ECM signaling, suggestive of hitherto unknown functions for BAD. Thus, exploring protein-protein interactions in 3D provides novel clues into cell behavior. This exciting approach has the potential to bridge the knowledge gap between tractable 2D cell culture and organoid-like 3D systems.
Collapse
Affiliation(s)
- Leila Pirayeshfard
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Shu Luo
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | | - Arashdeep Saini
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Nicolas Touret
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department
of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
39
|
Wu K, Jian S, Han Z, Ding C, Li Y, Wen Y, Nie Y, Zhu J, Li T, Zhang P, Zeng Y, Liu Z. Disintegrin Accutin inhibits A549 cell migration though suppression of EMT and FAK/AKT signaling pathway. Int J Biol Macromol 2024; 275:133593. [PMID: 38971284 DOI: 10.1016/j.ijbiomac.2024.133593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/08/2024]
Abstract
Integrins are heterodimers composed of two subunits, α(120-185kD) and β (90-110kD), which mediate the connection between cells and their external environment, such as extracellular matrix (ECM), and play an important role in the regulation of cell shape, proliferation and migration. Herein, we identified a potent anti-tumor migration peptide Accutin from crude venom of Agkistrodon acutus using an A549 3D tumor sphere model, and simulation tools and RNA sequencing were performed to reveal the mechanism of Accutin. Accutin is a disintegrin and docking, molecular dynamics simulations and ITC assay indicate that the RGD motif in the Accutin sequence can stably bind to integrins α5β1. 9.22 nM Accutin can significantly inhibit the migration and invasion of lung cancer cell lines. Transcriptome analysis indicated that many genes are involved in tumor cell adhesion-related biological processes. Several pathways, like the "mTOR signaling pathway", "TGF-β signaling pathway", and "Focal adhesion" were enriched. Interestingly, pathways involved in "N-Glycan biosynthesis" etc. were significantly inhibited. These transcriptomics data suggested that the molecular basis of Accutin-mediated inhibition of cancer cell migration may be by inhibiting N-glycosylation of integrin, then inhibiting signaling pathways such as PI3K/AKT/mTOR and TGFβ/smad. Western blotting analysis further confirmed that Accutin could suppress migration via down-regulating the phosphorylation of FAK and AKT and inhibiting EMT (epithelial-mesenchymal transition). Taken together, as a disintegrin with high efficiency, Accutin may be a potential precursor of a therapeutic agent for the treatment of lung cancer migration.
Collapse
Affiliation(s)
- Kun Wu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Shandong Jian
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Zhuomin Han
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Changhao Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Yaqi Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Yuhan Wen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Yueqi Nie
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Jiaoyue Zhu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Tingting Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Peng Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China.
| | - Yong Zeng
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China.
| | - Zhonghua Liu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha, Hunan 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
40
|
Wang W, Wang H. Modular formation of in vitro tumor models for oncological research/therapeutic drug screening. Adv Cancer Res 2024; 163:223-250. [PMID: 39271264 DOI: 10.1016/bs.acr.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
In recognition of the lethal nature of cancer, extensive efforts have been made to understand the mechanistic causation while identifying the effective therapy modality in hope to eradicate cancerous cells with minimal damage to healthy cells. In search of such effective therapeutics, establishing pathophysiologically relevant in vitro models would be of importance in empowering our capabilities of truly identifying those potent ones with significantly reduction of the preclinical periods for rapid translation. In this regard, wealthy progresses have been achieved over past decades in establishing various in vitro and in vivo tumor models. Ideally, the tumor models should maximally recapture the key pathophysiological attributes of their native counterparts. Many of the current models have demonstrated their utilities but also showed some noticeable limitations. This book chapter will briefly review some of the mainstream platforms for in vitro tumor models followed by detailed elaboration on the modular strategies to form in vitro tumor models with complex structures and spatial organization of cellular components. Clearly, with the ability to modulate the building modules it becomes a new trend to form in vitro tumor models following a bottom-up approach, which offers a high flexibility to satisfy the needs for pathophysiological study, anticancer drug screening or design of personalized treatment.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, United States; School of Life Sciences, Yantai University, Yantai, Shandong, P.R. China
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, United States; Semcer Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, NJ, United States.
| |
Collapse
|
41
|
Karras F, Kunz M. Patient-derived melanoma models. Pathol Res Pract 2024; 259:155231. [PMID: 38508996 DOI: 10.1016/j.prp.2024.155231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024]
Abstract
Melanoma is a very aggressive, rapidly metastasizing tumor that has been studied intensively in the past regarding the underlying genetic and molecular mechanisms. More recently developed treatment modalities have improved response rates and overall survival of patients. However, the majority of patients suffer from secondary treatment resistance, which requires in depth analyses of the underlying mechanisms. Here, melanoma models based on patients-derived material may play an important role. Consequently, a plethora of different experimental techniques have been developed in the past years. Among these are 3D and 4D culture techniques, organotypic skin reconstructs, melanoma-on-chip models and patient-derived xenografts, Every technique has its own strengths but also weaknesses regarding throughput, reproducibility, and reflection of the human situation. Here, we provide a comprehensive overview of currently used techniques and discuss their use in different experimental settings.
Collapse
Affiliation(s)
- Franziska Karras
- Institute of Pathology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg 39120, Germany.
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University Medical Center Leipzig, Philipp-Rosenthal-Str. 23, Leipzig 04103, Germany
| |
Collapse
|
42
|
Fiocchetti M, Raimondi S, Bastari G, Bartoloni S, Marino M, Acconcia F. Characterization of ERα Signaling to Cell Proliferation Induced by Chronic and Pulsatile E2 Stimulation in 2D and 3D Cell Cultures. J Cell Biochem 2024; 125:e30610. [PMID: 38860517 DOI: 10.1002/jcb.30610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/19/2024] [Accepted: 05/24/2024] [Indexed: 06/12/2024]
Abstract
17β-estradiol is a hormone that plays a vital role in human physiology. It acts through estrogen receptors, specifically estrogen receptor α and estrogen receptor β, and its action is determined by the pulsatile secretion in the bloodstream. 17β-estradiol affects cell proliferation, and dysregulation of 17β-estradiol:estrogen receptor α signaling contribute to the development of breast cancer. Previous research on 17β-estradiol:estrogen receptor α signaling has primarily used two-dimensional cell cultures, which do not fully recapitulate the complexity of tumors that exist in a three-dimensional environment and do not consider the pulsatile nature of this hormone. To address these limitations, we studied 17β-estradiol:estrogen receptor α signaling in cell proliferation using both two-dimensional and three-dimensional breast cancer cell culture models under continuous and pulsatile stimulation conditions. Results revealed that breast cancer cells grown in an alginate-based three-dimensional matrix exhibited similar responsiveness to 17β-estradiol compared with cells grown in conventional two-dimensional culture plates. 17β-estradiol induced the expression of proteins containing estrogen response element in the three-dimensional model. The efficacy of the antiestrogen drugs fulvestrant (ICI182,280) and 4OH-tamoxifen was also demonstrated in the three-dimensional model. These results support the use of the three-dimensional culture model for studying tumor response to drugs and provide a more realistic microenvironment for such studies. Furthermore, the study revealed that a brief 5-min exposure to 17β-estradiol triggered a physiological response comparable with continuous hormone exposure, suggesting that the cellular response to 17β-estradiol is more important than the continuous presence of the hormone. In conclusion, the study demonstrates that the alginate-based three-dimensional culture model is suitable for studying the effects of 17β-estradiol and antiestrogen drugs on breast cancer cells, offering a more realistic representation of tumor-microenvironment interactions. The results also highlight the importance of considering the physiological importance of the temporal dynamics in studying 17β-estradiol signaling and cellular responses.
Collapse
Affiliation(s)
- Marco Fiocchetti
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Rome, Italy
| | - Serena Raimondi
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Rome, Italy
| | - Giovanna Bastari
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Rome, Italy
| | - Stefania Bartoloni
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Rome, Italy
| | - Maria Marino
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Rome, Italy
| | - Filippo Acconcia
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Rome, Italy
| |
Collapse
|
43
|
Song WH, Lim YS, Kim JE, Kang HY, Lee C, Rajbongshi L, Hwang SY, Oh SO, Kim BS, Lee D, Song YJ, Yoon S. A Marine Collagen-Based 3D Scaffold for In Vitro Modeling of Human Prostate Cancer Niche and Anti-Cancer Therapeutic Discovery. Mar Drugs 2024; 22:295. [PMID: 39057404 PMCID: PMC11277582 DOI: 10.3390/md22070295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Recently, the need to develop a robust three-dimensional (3D) cell culture system that serves as a valuable in vitro tumor model has been emphasized. This system should closely mimic the tumor growth behaviors observed in vivo and replicate the key elements and characteristics of human tumors for the effective discovery and development of anti-tumor therapeutics. Therefore, in this study, we developed an effective 3D in vitro model of human prostate cancer (PC) using a marine collagen-based biomimetic 3D scaffold. The model displayed distinctive molecular profiles and cellular properties compared with those of the 2D PC cell culture. This was evidenced by (1) increased cell proliferation, migration, invasion, colony formation, and chemoresistance; (2) upregulated expression of crucial multidrug-resistance- and cancer-stemness-related genes; (3) heightened expression of key molecules associated with malignant progressions, such as epithelial-mesenchymal transition transcription factors, Notch, matrix metalloproteinases, and pluripotency biomarkers; (4) robust enrichment of prostate cancer stem cells (CSCs); and (5) enhanced expression of integrins. These results suggest that our 3D in vitro PC model has the potential to serve as a research platform for studying PC and prostate CSC biology, as well as for screening novel therapies targeting PC and prostate CSCs.
Collapse
Affiliation(s)
- Won Hoon Song
- Department of Urology, Pusan National University Yangsan Hospital and Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Ye Seon Lim
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Ji-Eun Kim
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Hae Yeong Kang
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Changyong Lee
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Lata Rajbongshi
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Seon Yeong Hwang
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea;
| | - Yong Jung Song
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital and Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea
| | - Sik Yoon
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| |
Collapse
|
44
|
Alwahsh M, Al-Doridee A, Jasim S, Awwad O, Hergenröder R, Hamadneh L. Cytotoxic and molecular differences of anticancer agents on 2D and 3D cell culture. Mol Biol Rep 2024; 51:721. [PMID: 38829450 DOI: 10.1007/s11033-024-09669-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Cancer and multidrug resistance are regarded as concerns related to poor health outcomes. It was found that the monolayer of 2D cancer cell cultures lacks many important features compared to Multicellular Tumor Spheroids (MCTS) or 3D cell cultures which instead have the ability to mimic more closely the in vivo tumor microenvironment. This study aimed to produce 3D cell cultures from different cancer cell lines and to examine the cytotoxic activity of anticancer medications on both 2D and 3D systems, as well as to detect alterations in the expression of certain genes levels. METHOD 3D cell culture was produced using 3D microtissue molds. The cytotoxic activities of colchicine, cisplatin, doxorubicin, and paclitaxel were tested on 2D and 3D cell culture systems obtained from different cell lines (A549, H1299, MCF-7, and DU-145). IC50 values were determined by MTT assay. In addition, gene expression levels of PIK3CA, AKT1, and PTEN were evaluated by qPCR. RESULTS Similar cytotoxic activities were observed on both 3D and 2D cell cultures, however, higher concentrations of anticancer medications were needed for the 3D system. For instance, paclitaxel showed an IC50 of 6.234 µM and of 13.87 µM on 2D and 3D H1299 cell cultures, respectively. Gene expression of PIK3CA in H1299 cells also showed a higher fold change in 3D cell culture compared to 2D system upon treatment with doxorubicin. CONCLUSION When compared to 2D cell cultures, the behavior of cells in the 3D system showed to be more resistant to anticancer treatments. Due to their shape, growth pattern, hypoxic core features, interaction between cells, biomarkers synthesis, and resistance to treatment penetration, the MCTS have the advantage of better simulating the in vivo tumor conditions. As a result, it is reasonable to conclude that 3D cell cultures may be a more promising model than the traditional 2D system, offering a better understanding of the in vivo molecular changes in response to different potential treatments and multidrug resistance development.
Collapse
Affiliation(s)
- Mohammad Alwahsh
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, 17138, Jordan.
| | - Amani Al-Doridee
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, 17138, Jordan
| | - Suhair Jasim
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, 17138, Jordan
| | - Oriana Awwad
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Roland Hergenröder
- Department of Bioanalytics, Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139, Dortmund, Germany
| | - Lama Hamadneh
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
45
|
Dimitriou NM, Flores-Torres S, Kyriakidou M, Kinsella JM, Mitsis GD. Cancer cell sedimentation in 3D cultures reveals active migration regulated by self-generated gradients and adhesion sites. PLoS Comput Biol 2024; 20:e1012112. [PMID: 38861575 PMCID: PMC11195982 DOI: 10.1371/journal.pcbi.1012112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/24/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
Cell sedimentation in 3D hydrogel cultures refers to the vertical migration of cells towards the bottom of the space. Understanding this poorly examined phenomenon may allow us to design better protocols to prevent it, as well as provide insights into the mechanobiology of cancer development. We conducted a multiscale experimental and mathematical examination of 3D cancer growth in triple negative breast cancer cells. Migration was examined in the presence and absence of Paclitaxel, in high and low adhesion environments and in the presence of fibroblasts. The observed behaviour was modeled by hypothesizing active migration due to self-generated chemotactic gradients. Our results did not reject this hypothesis, whereby migration was likely to be regulated by the MAPK and TGF-β pathways. The mathematical model enabled us to describe the experimental data in absence (normalized error<40%) and presence of Paclitaxel (normalized error<10%), suggesting inhibition of random motion and advection in the latter case. Inhibition of sedimentation in low adhesion and co-culture experiments further supported the conclusion that cells actively migrated downwards due to the presence of signals produced by cells already attached to the adhesive glass surface.
Collapse
Affiliation(s)
| | | | - Maria Kyriakidou
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | | | | |
Collapse
|
46
|
Ribeiro D, Latancia M, de Souza I, Ariwoola AB, Mendes D, Rocha CRR, Lengert A, Menck C. Temozolomide resistance mechanisms: unveiling the role of translesion DNA polymerase kappa in glioblastoma spheroids in vitro. Biosci Rep 2024; 44:BSR20230667. [PMID: 38717250 PMCID: PMC11139666 DOI: 10.1042/bsr20230667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Temozolomide (TMZ) is the leading therapeutic agent for combating Glioblastoma Multiforme (GBM). Nonetheless, the persistence of chemotherapy-resistant GBM cells remains an ongoing challenge, attributed to various factors, including the translesion synthesis (TLS) mechanism. TLS enables tumor cells to endure genomic damage by utilizing specialized DNA polymerases to bypass DNA lesions. Specifically, TLS polymerase Kappa (Polκ) has been implicated in facilitating DNA damage tolerance against TMZ-induced damage, contributing to a worse prognosis in GBM patients. To better understand the roles of Polκ in TMZ resistance, we conducted a comprehensive assessment of the cytotoxic, antiproliferative, antimetastatic, and genotoxic effects of TMZ on GBM (U251MG) wild-type (WTE) and TLS Polκ knockout (KO) cells, cultivated as three-dimensional (3D) tumor spheroids in vitro. Initial results revealed that TMZ: (i) induces reductions in GBM spheroid diameter (10-200 µM); (ii) demonstrates significant cytotoxicity (25-200 μM); (iii) exerts antiproliferative effects (≤25 μM) and promotes cell cycle arrest (G2/M phase) in Polκ KO spheroids when compared with WTE counterparts. Furthermore, Polκ KO spheroids exhibit elevated levels of cell death (Caspase 3/7) and display greater genotoxicity (53BP1) than WTE following TMZ exposure. Concerning antimetastatic effects, TMZ impedes invadopodia (3D invasion) more effectively in Polκ KO than in WTE spheroids. Collectively, the results suggest that TLS Polκ plays a vital role in the survival, cell death, genotoxicity, and metastatic potential of GBM spheroids in vitro when subjected to TMZ treatment. While the precise mechanisms underpinning this resistance remain elusive, TLS Polκ emerges as a potential therapeutic target for GBM patients.
Collapse
Affiliation(s)
- Diego Luis Ribeiro
- Departament of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Marcela Teatin Latancia
- Departament of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Izadora de Souza
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Abu-Bakr Adetayo Ariwoola
- Departament of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Davi Mendes
- Departament of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - André Van Helvoort Lengert
- Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | | |
Collapse
|
47
|
Kim S, Lam PY, Jayaraman A, Han A. Uniform sized cancer spheroids production using hydrogel-based droplet microfluidics: a review. Biomed Microdevices 2024; 26:26. [PMID: 38806765 PMCID: PMC11241584 DOI: 10.1007/s10544-024-00712-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2024] [Indexed: 05/30/2024]
Abstract
Three-dimensional (3D) cell culture models have been extensively utilized in various mechanistic studies as well as for drug development studies as superior in vitro platforms than conventional two-dimensional (2D) cell culture models. This is especially the case in cancer biology, where 3D cancer models, such as spheroids or organoids, have been utilized extensively to understand the mechanisms of cancer development. Recently, many sophisticated 3D models such as organ-on-a-chip models are emerging as advanced in vitro models that can more accurately mimic the in vivo tissue functions. Despite such advancements, spheroids are still considered as a powerful 3D cancer model due to the relatively simple structure and compatibility with existing laboratory instruments, and also can provide orders of magnitude higher throughput than complex in vitro models, an extremely important aspects for drug development. However, creating well-defined spheroids remain challenging, both in terms of throughputs in generation as well as reproducibility in size and shape that can make it challenging for drug testing applications. In the past decades, droplet microfluidics utilizing hydrogels have been highlighted due to their potentials. Importantly, core-shell structured gel droplets can avoid spheroid-to-spheroid adhesion that can cause large variations in assays while also enabling long-term cultivation of spheroids with higher uniformity by protecting the core organoid area from external environment while the outer porous gel layer still allows nutrient exchange. Hence, core-shell gel droplet-based spheroid formation can improve the predictivity and reproducibility of drug screening assays. This review paper will focus on droplet microfluidics-based technologies for cancer spheroid production using various gel materials and structures. In addition, we will discuss emerging technologies that have the potential to advance the production of spheroids, prospects of such technologies, and remaining challenges.
Collapse
Affiliation(s)
- Sungjin Kim
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Po Yi Lam
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
48
|
Royo F, Garcia-Vallicrosa C, Azparren-Angulo M, Bordanaba-Florit G, Lopez-Sarrio S, Falcon-Perez JM. Three-Dimensional Hepatocyte Spheroids: Model for Assessing Chemotherapy in Hepatocellular Carcinoma. Biomedicines 2024; 12:1200. [PMID: 38927406 PMCID: PMC11201042 DOI: 10.3390/biomedicines12061200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Three-dimensional cellular models provide a more comprehensive representation of in vivo cell properties, encompassing physiological characteristics and drug susceptibility. METHODS Primary hepatocytes were seeded in ultra-low attachment plates to form spheroids, with or without tumoral cells. Spheroid structure, cell proliferation, and apoptosis were analyzed using histological staining techniques. In addition, extracellular vesicles were isolated from conditioned media by differential ultracentrifugation. Spheroids were exposed to cytotoxic drugs, and both spheroid growth and cell death were measured by microscopic imaging and flow cytometry with vital staining, respectively. RESULTS Concerning spheroid structure, an active outer layer forms a boundary with the media, while the inner core comprises a mass of cell debris. Hepatocyte-formed spheroids release vesicles into the extracellular media, and a decrease in the concentration of vesicles in the culture media can be observed over time. When co-cultured with tumoral cells, a distinct distribution pattern emerges over the primary hepatocytes, resulting in different spheroid conformations. Tumoral cell growth was compromised upon antitumoral drug challenges. CONCLUSIONS Treatment of mixed spheroids with different cytotoxic drugs enables the characterization of drug effects on both hepatocytes and tumoral cells, determining drug specificity effects on these cell types.
Collapse
Affiliation(s)
- Felix Royo
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Clara Garcia-Vallicrosa
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Maria Azparren-Angulo
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Guillermo Bordanaba-Florit
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Silvia Lopez-Sarrio
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Juan Manuel Falcon-Perez
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
49
|
Petersen ME, Brant MG, Lasalle M, Das S, Duan R, Wong J, Ding T, Wu KJ, Siddappa D, Fang C, Zhang W, Wu AML, Hirkala-Schaefer T, Garnett GAE, Fung V, Yang L, Hernandez Rojas A, Lawn SO, Barnscher SD, Rich JR, Colombo R. Design and Evaluation of ZD06519, a Novel Camptothecin Payload for Antibody Drug Conjugates. Mol Cancer Ther 2024; 23:606-618. [PMID: 38354417 PMCID: PMC11063767 DOI: 10.1158/1535-7163.mct-23-0822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/08/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
In recent years, the field of antibody drug conjugates (ADC) has seen a resurgence, largely driven by the clinical benefit observed in patients treated with ADCs incorporating camptothecin-based topoisomerase I inhibitor payloads. Herein, we present the development of a novel camptothecin ZD06519 (FD1), which has been specifically designed for its application as an ADC payload. A panel of camptothecin analogs with different substituents at the C-7 and C-10 positions of the camptothecin core was prepared and tested in vitro. Selected compounds spanning a range of potency and hydrophilicity were elaborated into drug-linkers, conjugated to trastuzumab, and evaluated in vitro and in vivo. ZD06519 was selected on the basis of its favorable properties as a free molecule and as an antibody conjugate, which include moderate free payload potency (∼1 nmol/L), low hydrophobicity, strong bystander activity, robust plasma stability, and high-monomeric ADC content. When conjugated to different antibodies using a clinically validated MC-GGFG-based linker, ZD06519 demonstrated impressive efficacy in multiple cell line-derived xenograft models and noteworthy tolerability in healthy mice, rats, and non-human primates.
Collapse
Affiliation(s)
- Mark E. Petersen
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Michael G. Brant
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Manuel Lasalle
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Samir Das
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Renee Duan
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Jodi Wong
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Tong Ding
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Kaylee J. Wu
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Dayananda Siddappa
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Chen Fang
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Wen Zhang
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Alex M. L. Wu
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | | | - Graham A. E. Garnett
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Vincent Fung
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Luying Yang
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | | | - Samuel O. Lawn
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Stuart D. Barnscher
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Jamie R. Rich
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| | - Raffaele Colombo
- ADC Therapeutic Development, Zymeworks Inc., Vancouver, British Columbia, Canada
| |
Collapse
|
50
|
Cirillo S, Zhang B, Brown S, Zhao X. Antimicrobial peptide A 9K as a gene delivery vector in cancer cells. Eur J Pharm Biopharm 2024; 198:114244. [PMID: 38467336 DOI: 10.1016/j.ejpb.2024.114244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/13/2024]
Abstract
Designed peptides are promising biomaterials for biomedical applications. The amphiphilic cationic antimicrobial peptide (AMP), A9K, can self-assemble into nano-rod structures and has shown cancer cell selectivity and could therefore be a promising candidate for therapeutic delivery into cancer cells. In this paper, we investigate the selectivity of A9K for cancer cell models, examining its effect on two human cancer cell lines, A431 and HCT-116. Little or no activity was observed on the control, human dermal fibroblasts (HDFs). In the cancer cell lines the peptide inhibited cellular growth through changes in mitochondrial morphology and membrane potential while remaining harmless towards HDFs. In addition, the peptide can bind to and protect nucleic acids while transporting them into both 2D cultures and 3D spheroids of cancer cells. A9K showed high efficiency in delivering siRNA molecules into the centre of the spheroids. A9K was also explored in vivo, using a zebrafish (Danio rerio) development toxicity assay, showing that the peptide is safe at low doses. Finally, a high-content imaging screen, using RNA interference (RNAi) targeted towards cellular uptake, in HCT-116 cells was carried out. Our findings suggest that active cellular uptake is involved in peptide internalisation, mediated through clathrin-mediated endocytosis. These new discoveries make A9K attractive for future developments in clinical and biotechnological applications.
Collapse
Affiliation(s)
- Silvia Cirillo
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Bo Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Stephen Brown
- The Sheffield RNAi Screening Facility, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Xiubo Zhao
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK; School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|