Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.771
Filter
1.
J Vis Exp ; (207)2024 May 10.
Article in English | MEDLINE | ID: mdl-38801268

ABSTRACT

Non-small cell lung cancer (NSCLC) is a highly lethal disease with a complex and heterogeneous tumor microenvironment. Currently, common animal models based on subcutaneous inoculation of cancer cell suspensions do not recapitulate the tumor microenvironment in NSCLC. Herein we describe a murine orthotopic lung cancer xenograft model that employs the intrapulmonary inoculation of three-dimensional multicellular spheroids (MCS). Specifically, fluorescent human NSCLC cells (A549-iRFP) were cultured in low-attachment 96-well microplates with collagen for 3 weeks to form MCS, which were then inoculated intercostally into the left lung of athymic nude mice to establish the orthotopic lung cancer model. Compared with the original A549 cell line, MCS of the A549-iRFP cell line responded similarly to anticancer drugs. The long-wavelength fluorescent signal of the A549-iRFP cells correlated strongly with common markers of cancer cell growth, including spheroid volume, cell viability, and cellular protein level, thus allowing dynamic monitoring of the cancer growth in vivo by fluorescent imaging. After inoculation into mice, the A549-iRFP MCS xenograft reliably progressed through phases closely resembling the clinical stages of NSCLC, including the expansion of the primary tumor, the emergence of neighboring secondary tumors, and the metastases of cancer cells to the contralateral right lung and remote organs. Moreover, the model responded to the benchmark antilung cancer drug, cisplatin with the anticipated toxicity and slower cancer progression. Therefore, this murine orthotopic xenograft model of NSCLC would serve as a platform to recapitulate the disease's progression and facilitate the development of potential anticancer drugs.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice, Nude , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Humans , Mice , Xenograft Model Antitumor Assays/methods , Disease Progression , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Disease Models, Animal , A549 Cells , Neoplasm Transplantation
2.
Artif Cells Nanomed Biotechnol ; 52(1): 309-320, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38781462

ABSTRACT

Photodynamic therapy (PDT) holds great potential to overcome limitations associated with common colorectal cancer (CRC) treatment approaches. Targeted photosensitiser (PS) delivery systems using nanoparticles (NPs) with targeting moieties are continually being designed, which are aimed at enhancing PS efficacy in CRC PDT. However, the optimisation of targeted PS delivery systems in most, in vitro PDT studies has been conducted on two dimensional (2D) monolayers cell cultures. In our present study, we developed a nano PS delivery system for in vitro cultured human colorectal three-dimensional multicellular spheroids (3D MCTS). PEGylated gold nanoparticles (PEG-AuNPs) were prepared and attached to ZnPcS4PS and further functionalised with specific CRC targeting anti-Guanylate Cyclase monoclonal antibodies(mAb). The ZnPcS4-AuNP-Anti-GCC Ab (BNC) nanoconjugates were successfully synthesised and their photodynamic effect investigated following exposure to laser irradiation and demonstrated enhanced anticancer effects in Caco-2 cells cultivated as 3D MCTS spheroids. Our findings suggest that targeted BNC nanoconjugates can improve the efficacy of PDT and highlight the potential of 3D MCTS tumour model for evaluating of targeted PDT.


Subject(s)
Colorectal Neoplasms , Gold , Metal Nanoparticles , Photochemotherapy , Spheroids, Cellular , Humans , Gold/chemistry , Gold/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Spheroids, Cellular/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Metal Nanoparticles/chemistry , Caco-2 Cells , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Polyethylene Glycols/chemistry
3.
J Mech Behav Biomed Mater ; 155: 106571, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38744118

ABSTRACT

Cardiac fibrosis refers to the abnormal accumulation of extracellular matrix within the cardiac muscle, leading to increased stiffness and impaired heart function. From a rheological standpoint, knowledge about myocardial behavior is still lacking, partially due to a lack of appropriate techniques to investigate the rheology of in vitro cardiac tissue models. 3D multicellular cardiac spheroids are powerful and versatile platforms for modeling healthy and fibrotic cardiac tissue in vitro and studying how their mechanical properties are modulated. In this study, cardiac spheroids were created by co-culturing neonatal rat ventricular cardiomyocytes and fibroblasts in definite ratios using the hanging-drop method. The rheological characterization of such models was performed by Atomic Force Microscopy-based stress-relaxation measurements on the whole spheroid. After strain application, a viscoelastic bi-exponential relaxation was observed, characterized by a fast relaxation time (τ1) followed by a slower one (τ2). In particular, spheroids with higher fibroblasts density showed reduction for both relaxation times comparing to control, with a more pronounced decrement of τ1 with respect to τ2. Such response was found compatible with the increased production of extracellular matrix within these spheroids, which recapitulates the main feature of the fibrosis pathophysiology. These results demonstrate how the rheological characteristics of cardiac tissue vary as a function of cellular composition and extracellular matrix, confirming the suitability of such system as an in vitro preclinical model of cardiac fibrosis.


Subject(s)
Fibrosis , Myocytes, Cardiac , Rheology , Spheroids, Cellular , Animals , Spheroids, Cellular/cytology , Spheroids, Cellular/pathology , Rats , Myocytes, Cardiac/cytology , Fibroblasts/cytology , Myocardium/cytology , Myocardium/pathology , Myocardium/metabolism , Rats, Wistar , Models, Biological
4.
Biosci Rep ; 44(5)2024 May 29.
Article in English | MEDLINE | ID: mdl-38717250

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.


Subject(s)
DNA-Directed DNA Polymerase , Drug Resistance, Neoplasm , Glioblastoma , Spheroids, Cellular , Temozolomide , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/enzymology , Temozolomide/pharmacology , Drug Resistance, Neoplasm/drug effects , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/enzymology , Antineoplastic Agents, Alkylating/pharmacology
5.
Mol Biol Rep ; 51(1): 603, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698270

ABSTRACT

BACKGROUND: Drug combination studies help to improve new treatment approaches for colon cancer. Tumor spheroids (3D) are better models than traditional 2-dimensional cultures (2D) to evaluate cellular responses to chemotherapy drugs. The cultivation of cancer cells in 2D and 3D cultures affects the apoptotic process, which is a major factor influencing the response of cancer cells to chemotherapeutic drugs. In this study, the antiproliferative effects of 5-fluorouracil (5-FU) and doxorubicin (DOX) were investigated separately and in combination using 2D and 3D cell culture models on two different colon cancer cell lines, HT-29 (apoptosis-resistant cells) and Caco-2 2 (apoptosis-susceptible cells). METHODS: The effect of the drugs on the proliferation of both colon cancer cells was determined by performing an MTT assay in 2D culture. The apoptotic effect of 5-FU and DOX, both as single agents and in combination, was assessed in 2D and 3D cultures through quantitative real-time polymerase chain reaction analysis. The expression of apoptotic genes, such as caspases, p53, Bax, and Bcl-2, was quantified. RESULTS: It was found that the mRNA expression of proapoptotic genes was significantly upregulated, whereas the mRNA expression of the antiapoptotic Bcl-2 gene was significantly downregulated in both colon cancer models treated with 5-FU, DOX, and 5-FU + DOX. CONCLUSION: The results indicated that the 5-FU + DOX combination therapy induces apoptosis and renders 5-FU and DOX more effective at lower concentrations compared to their alone use. This study reveals promising results in reducing the potential side effects of treatment by enabling the use of lower drug doses.


Subject(s)
Apoptosis , Cell Proliferation , Colorectal Neoplasms , Doxorubicin , Fluorouracil , Spheroids, Cellular , Humans , Fluorouracil/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Doxorubicin/pharmacology , Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , HT29 Cells , Cell Proliferation/drug effects , Caco-2 Cells , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Line, Tumor , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics
6.
Cell Death Dis ; 15(5): 306, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693105

ABSTRACT

Colorectal cancers (CRCs) are highly heterogeneous and show a hierarchical organization, with cancer stem cells (CSCs) responsible for tumor development, maintenance, and drug resistance. Our previous studies showed the importance of thyroid hormone-dependent signaling on intestinal tumor development and progression through action on stem cells. These results have a translational value, given that the thyroid hormone nuclear receptor TRα1 is upregulated in human CRCs, including in the molecular subtypes associated with CSC features. We used an established spheroid model generated from the human colon adenocarcinoma cell line Caco2 to study the effects of T3 and TRα1 on spheroid formation, growth, and response to conventional chemotherapies. Our results show that T3 treatment and/or increased TRα1 expression in spheroids impaired the response to FOLFIRI and conferred a survival advantage. This was achieved by stimulating drug detoxification pathways and increasing ALDH1A1-expressing cells, including CSCs, within spheroids. These results suggest that clinical evaluation of the thyroid axis and assessing TRα1 levels in CRCs could help to select optimal therapeutic regimens for patients with CRC. Proposed mechanism of action of T3/TRα1 in colon cancer spheroids. In the control condition, TRα1 participates in maintaining homeostatic cell conditions. The presence of T3 in the culture medium activates TRα1 action on target genes, including the drug efflux pumps ABCG2 and ABCB1. In the case of chemotherapy FOLFIRI, the increased expression of ABC transcripts and proteins induced by T3 treatment is responsible for the augmented efflux of 5-FU and Irinotecan from the cancer cells. Taken together, these mechanisms contribute to the decreased efficacy of the chemotherapy and allow cells to escape the treatment. Created with BioRender.com .


Subject(s)
Camptothecin/analogs & derivatives , Colonic Neoplasms , Fluorouracil , Neoplastic Stem Cells , Spheroids, Cellular , Thyroid Hormone Receptors alpha , Triiodothyronine , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors alpha/genetics , Caco-2 Cells , Colonic Neoplasms/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Triiodothyronine/pharmacology , Leucovorin/pharmacology , Leucovorin/therapeutic use , Camptothecin/pharmacology , Camptothecin/therapeutic use , Phenotype , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Retinal Dehydrogenase/metabolism , Retinal Dehydrogenase/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics
7.
Sci Rep ; 14(1): 11013, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38745039

ABSTRACT

Cancer Stem Cells presumably drive tumor growth and resistance to conventional cancer treatments. From a previous computational model, we inferred that these cells are not uniformly distributed in the bulk of a tumorsphere. To confirm this result, we cultivated tumorspheres enriched in stem cells, and performed immunofluorescent detection of the stemness marker SOX2 using confocal microscopy. In this article, we present an image processing method that reconstructs the amount and location of the Cancer Stem Cells in the spheroids. Its advantage is the use of a statistical criterion to classify the cells in Stem and Differentiated, instead of setting an arbitrary threshold. Moreover, the analysis of the experimental images presented in this work agrees with the results from our computational models, thus enforcing the notion that the distribution of Cancer Stem Cells in a tumorsphere is non-homogeneous. Additionally, the method presented here provides a useful tool for analyzing any image in which different kinds of cells are stained with different markers.


Subject(s)
Neoplastic Stem Cells , Spheroids, Cellular , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Humans , Spheroids, Cellular/pathology , Spheroids, Cellular/metabolism , SOXB1 Transcription Factors/metabolism , Image Processing, Computer-Assisted/methods , Microscopy, Confocal , Cell Line, Tumor
8.
Biofabrication ; 16(3)2024 May 09.
Article in English | MEDLINE | ID: mdl-38663395

ABSTRACT

Three-dimensional (3D) cell culture has been used in many fields of biology because of its unique advantages. As a representative of the 3D systems, 3D spheroids are used as building blocks for tissue construction. Larger tumor aggregates can be assembled by manipulating or stacking the tumor spheroids. The motivation of this study is to investigate the behavior of the cells distributed at different locations of the spheroids in the fusion process and the mechanism behind it. To this aim, spheroids with varying grades of maturity or age were generated for fusion to assemble micro-tumor tissues. The dynamics of the fusion process, the motility of the cells distributed in different heterogeneous architecture sites, and their reactive oxygen species profiles were studied. We found that the larger the spheroid necrotic core, the slower the fusion rate of the spheroid. The cells that move were mainly distributed on the spheroid's surface during fusion. In addition to dense microfilament distribution and low microtubule content, the reactive oxygen content was high in the fusion site, while the non-fusion site was the opposite. Last, multi-spheroids with different maturities were fused to complex micro-tissues to mimic solid tumors and evaluate Doxorubicin's anti-tumor efficacy.


Subject(s)
Doxorubicin , Reactive Oxygen Species , Spheroids, Cellular , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/pathology , Humans , Reactive Oxygen Species/metabolism , Doxorubicin/pharmacology , Cell Fusion , Neoplasms/pathology , Neoplasms/metabolism , Cell Line, Tumor , Cell Culture Techniques, Three Dimensional , Cell Movement , Tissue Engineering
9.
Nat Commun ; 15(1): 3599, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678014

ABSTRACT

Targeting the supportive tumor microenvironment (TME) is an approach of high interest in cancer drug development. However, assessing TME-targeted drug candidates presents a unique set of challenges. We develop a comprehensive screening platform that allows monitoring, quantifying, and ranking drug-induced effects in self-organizing, vascularized tumor spheroids (VTSs). The confrontation of four human-derived cell populations makes it possible to recreate and study complex changes in TME composition and cell-cell interaction. The platform is modular and adaptable for tumor entity or genetic manipulation. Treatment effects are recorded by light sheet fluorescence microscopy and translated by an advanced image analysis routine in processable multi-parametric datasets. The system proved to be robust, with strong interassay reliability. We demonstrate the platform's utility for evaluating TME-targeted antifibrotic and antiangiogenic drugs side-by-side. The platform's output enabled the differential evaluation of even closely related drug candidates according to projected therapeutic needs.


Subject(s)
Breast Neoplasms , Microscopy, Fluorescence , Spheroids, Cellular , Tumor Microenvironment , Humans , Tumor Microenvironment/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Microscopy, Fluorescence/methods , Female , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
10.
Lab Chip ; 24(9): 2561-2574, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38629978

ABSTRACT

Tumor spheroids are now intensively investigated toward preclinical and clinical applications, necessitating the establishment of accessible and cost-effective methods for routine operations. Without losing the advantage of organ-chip technologies, we developed a rocking system for facile formation and culture of tumor spheroids in hydrogel microwells of a suspended membrane under microfluidic conditions. While the rocking is controlled with a step motor, the microfluidic device is made of two plastic plates, allowing plugging directly syringe tubes with Luer connectors. Upon injection of the culture medium into the tubes and subsequent rocking of the chip, the medium flows back and forth in the channel underneath the membrane, ensuring a diffusion-based culture. Our results showed that such a rocking- and diffusion-based culture method significantly improved the quality of the tumor spheroids when compared to the static culture, particularly in terms of growth rate, roundness, junction formation and compactness of the spheroids. Notably, dynamically cultured tumor spheroids showed increased drug resistance, suggesting alternative assay conditions. Overall, the present method is pumpless, connectionless, and user-friendly, thereby facilitating the advancement of tumor-spheroid-based applications.


Subject(s)
Lab-On-A-Chip Devices , Spheroids, Cellular , Spheroids, Cellular/cytology , Spheroids, Cellular/pathology , Humans , Cell Culture Techniques/instrumentation , Diffusion , Microfluidic Analytical Techniques/instrumentation , Hydrogels/chemistry , Cell Line, Tumor , Tumor Cells, Cultured , Equipment Design
11.
J Photochem Photobiol B ; 255: 112910, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663337

ABSTRACT

The prognosis for patients with advanced-stage pancreatic ductal adenocarcinoma (PDAC) remains dismal. It is generally accepted that combination cancer therapies offer the most promise, such as Folforinox, despite their associated high toxicity. This study addresses the issue of chemoresistance by introducing a complementary dual priming approach to attenuate the DNA repair mechanism and to improve the efficacy of a type 1 topoisomerase (Top1) inhibitor. The result is a regimen that integrates drug-repurposing and nanotechnology using 3 clinically relevant FDA-approved agents (1) Top1 inhibitor (irinotecan) at subcytotoxic doses (2) benzoporphyrin derivative (BPD) as a photoactive molecule for photodynamic priming (PDP) to improve the delivery of irinotecan within the cancer cell and (3) minocycline priming (MNP) to modulate DNA repair enzyme Tdp1 (tyrosyl-DNA phosphodiesterase) activity. We demonstrate in heterotypic 3D cancer models that incorporate cancer cells and pancreatic cancer-associated fibroblasts that simultaneous targeting of Tdp1 and Top1 were significantly more effective by employing MNP and photoactivatable multi-inhibitor liposomes encapsulating BPD and irinotecan compared to monotherapies or a cocktail of dual or triple-agents. These data are encouraging and warrant further work in appropriate animal models to evolve improved therapeutic regimens.


Subject(s)
Carcinoma, Pancreatic Ductal , Irinotecan , Minocycline , Pancreatic Neoplasms , Photochemotherapy , Humans , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Cell Line, Tumor , Minocycline/pharmacology , Minocycline/therapeutic use , Irinotecan/pharmacology , Irinotecan/therapeutic use , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Phosphoric Diester Hydrolases/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , Topoisomerase I Inhibitors/chemistry , Liposomes/chemistry
12.
EMBO J ; 43(9): 1770-1798, 2024 May.
Article in English | MEDLINE | ID: mdl-38565950

ABSTRACT

The cancer epigenome has been studied in cells cultured in two-dimensional (2D) monolayers, but recent studies highlight the impact of the extracellular matrix and the three-dimensional (3D) environment on multiple cellular functions. Here, we report the physical, biochemical, and genomic differences between T47D breast cancer cells cultured in 2D and as 3D spheroids. Cells within 3D spheroids exhibit a rounder nucleus with less accessible, more compacted chromatin, as well as altered expression of ~2000 genes, the majority of which become repressed. Hi-C analysis reveals that cells in 3D are enriched for regions belonging to the B compartment, have decreased chromatin-bound CTCF and increased fusion of topologically associating domains (TADs). Upregulation of the Hippo pathway in 3D spheroids results in the activation of the LATS1 kinase, which promotes phosphorylation and displacement of CTCF from DNA, thereby likely causing the observed TAD fusions. 3D cells show higher chromatin binding of progesterone receptor (PR), leading to an increase in the number of hormone-regulated genes. This effect is in part mediated by LATS1 activation, which favors cytoplasmic retention of YAP and CTCF removal.


Subject(s)
Breast Neoplasms , CCCTC-Binding Factor , Chromatin , Protein Serine-Threonine Kinases , Humans , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Chromatin/metabolism , Chromatin/genetics , Female , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Hippo Signaling Pathway
13.
ACS Nano ; 18(17): 11257-11269, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38632933

ABSTRACT

Despite recent advances in the development of scaffold-based three-dimensional (3D) cell models, challenges persist in imaging and monitoring cell behavior within these complex structures due to their heterogeneous cell distribution and geometries. Incorporating sensors into 3D scaffolds provides a potential solution for real-time, in situ sensing and imaging of biological processes such as cell growth and disease development. We introduce a 3D printed hydrogel-based scaffold capable of supporting both surface-enhanced Raman scattering (SERS) biosensing and imaging of 3D breast cancer cell models. The scaffold incorporates plasmonic nanoparticles and SERS tags, for sensing and imaging, respectively. We demonstrate the scaffold's adaptability and modularity in supporting breast cancer spheroids, thereby enabling spatial and temporal monitoring of tumor evolution.


Subject(s)
Spectrum Analysis, Raman , Humans , Spectrum Analysis, Raman/methods , Breast Neoplasms/pathology , Breast Neoplasms/diagnostic imaging , Hydrogels/chemistry , Surface Properties , Cell Line, Tumor , Biosensing Techniques/methods , Tissue Scaffolds/chemistry , Metal Nanoparticles/chemistry , Spheroids, Cellular/pathology
14.
Exp Cell Res ; 438(1): 114033, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38593916

ABSTRACT

Regardless of the clinical response and improved patient survival observed following treatment with BRAFi like Vemurafenib (Vem), rapid development of resistance still remains as a major obstacle in melanoma therapy. In this context, we developed and characterized two acquired Vem-resistant melanoma cell lines, A375V and SK-MEL-28V, and an intrinsically Vem-resistant cell line, RPMI-7951. Altered morphology and growth rate of the resistant cell lines displayed spindle-shaped cells with filopodia formation and enhanced proliferation rate as compared to parental cells. Further in vitro characterization in 2D models confirmed the emergence of a resistant phenotype in melanoma cells. To mimic the in vivo tumor microenvironment, spheroids were developed for both parental and resistant cell lines to recognize materialization of invadopodia structures demonstrating elevated invasiveness and proliferation of resistant cells-based spheroids, especially A375V. Importantly, we validated A375V cell line in vivo to prove its tumorigenicity and drug resistance in tumor xenograft model. Taken together, our established clinically relevant Vem-resistant tumor model could be beneficial to elucidate drug resistance mechanisms, screen and identify novel anticancer therapies to overcome BRAFi resistance in melanoma.


Subject(s)
Cell Proliferation , Drug Resistance, Neoplasm , Melanoma , Proto-Oncogene Proteins B-raf , Vemurafenib , Humans , Melanoma/drug therapy , Melanoma/pathology , Drug Resistance, Neoplasm/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib/pharmacology , Mice , Xenograft Model Antitumor Assays , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment/drug effects , Antineoplastic Agents/pharmacology , Mice, Nude
15.
Cell Death Dis ; 15(4): 303, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684666

ABSTRACT

Scientific literature supports the evidence that cancer stem cells (CSCs) retain inside low reactive oxygen species (ROS) levels and are, therefore, less susceptible to cell death, including ferroptosis, a type of cell death dependent on iron-driven lipid peroxidation. A collection of lung adenocarcinoma (LUAD) primary cell lines derived from malignant pleural effusions (MPEs) of patients was used to obtain 3D spheroids enriched for stem-like properties. We observed that the ferroptosis inducer RSL3 triggered lipid peroxidation and cell death in LUAD cells when grown in 2D conditions; however, when grown in 3D conditions, all cell lines underwent a phenotypic switch, exhibiting substantial resistance to RSL3 and, therefore, protection against ferroptotic cell death. Interestingly, this phenomenon was reversed by disrupting 3D cells and growing them back in adherence, supporting the idea of CSCs plasticity, which holds that cancer cells have the dynamic ability to transition between a CSC state and a non-CSC state. Molecular analyses showed that ferroptosis resistance in 3D spheroids correlated with an increased expression of antioxidant genes and high levels of proteins involved in iron storage and export, indicating protection against oxidative stress and low availability of iron for the initiation of ferroptosis. Moreover, transcriptomic analyses highlighted a novel subset of genes commonly modulated in 3D spheroids and potentially capable of driving ferroptosis protection in LUAD-CSCs, thus allowing to better understand the mechanisms of CSC-mediated drug resistance in tumors.


Subject(s)
Adenocarcinoma of Lung , Ferroptosis , Lung Neoplasms , Neoplastic Stem Cells , Ferroptosis/genetics , Ferroptosis/drug effects , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Spheroids, Cellular/drug effects , Cell Line, Tumor , Lipid Peroxidation , Reactive Oxygen Species/metabolism , Gene Expression Regulation, Neoplastic , Drug Resistance, Neoplasm/genetics , Iron/metabolism
16.
Sci Rep ; 14(1): 9357, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38653823

ABSTRACT

The advent of micro-physiological systems (MPS) in biomedical research has enabled the introduction of more complex and relevant physiological into in vitro models. The recreation of complex morphological features in three-dimensional environments can recapitulate otherwise absent dynamic interactions in conventional models. In this study we developed an advanced in vitro Renal Cell Carcinoma (RCC) that mimics the interplay between healthy and malignant renal tissue. Based on the TissUse Humimic platform our model combines healthy renal proximal tubule epithelial cells (RPTEC) and RCC. Co-culturing reconstructed RPTEC tubules with RCC spheroids in a closed micro-perfused circuit resulted in significant phenotypical changes to the tubules. Expression of immune factors revealed that interleukin-8 (IL-8) and tumor necrosis factor-alfa (TNF-α) were upregulated in the non-malignant cells while neutrophil gelatinase-associated lipocalin (NGAL) was downregulated in both RCC and RPTEC. Metabolic analysis showed that RCC prompted a shift in the energy production of RPTEC tubules, inducing glycolysis, in a metabolic adaptation that likely supports RCC growth and immunogenicity. In contrast, RCC maintained stable metabolic activity, emphasizing their resilience to external factors. RNA-seq and biological process analysis of primary RTPTEC tubules demonstrated that the 3D tubular architecture and MPS conditions reverted cells to a predominant oxidative phosphorylate state, a departure from the glycolytic metabolism observed in 2D culture. This dynamic RCC co-culture model, approximates the physiology of healthy renal tubules to that of RCC, providing new insights into tumor-host interactions. Our approach can show that an RCC-MPS can expand the complexity and scope of pathophysiology and biomarker studies in kidney cancer research.


Subject(s)
Carcinoma, Renal Cell , Coculture Techniques , Epithelial Cells , Kidney Neoplasms , Kidney Tubules, Proximal , Humans , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Cell Line, Tumor , Lipocalin-2/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
17.
Methods Mol Biol ; 2777: 145-161, 2024.
Article in English | MEDLINE | ID: mdl-38478342

ABSTRACT

Cancer stem cells (CSCs) play a significant role in driving several tumor hallmarks. Their behavior and tumor progression are strictly related to the tumor microenvironment (TME). The dynamic interplay between CSCs and TME drives metastasis, chemoresistance, and disease relapse. In this chapter, we describe different techniques and protocols for isolating, culturing, and characterizing CSCs and we explain the methodology for the culture of multicellular spheroids comprising CSCs.


Subject(s)
Neoplasms , Spheroids, Cellular , Humans , Spheroids, Cellular/pathology , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Tumor Microenvironment
18.
Biomaterials ; 307: 122526, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38513434

ABSTRACT

Stem cell therapies have shown great potential for treating myocardial infarction (MI) but are limited by low cell survival and compromised functionality due to the harsh microenvironment at the disease site. Here, we presented a Mesenchymal stem cell (MSC) spheroid-based strategy for MI treatment by introducing a protein/polyphenol self-assembling armor coating on the surface of cell spheroids, which showed significantly enhanced therapeutic efficacy by actively manipulating the hostile pathological MI microenvironment and enabling versatile functionality, including protecting the donor cells from host immune clearance, remodeling the ROS microenvironment and stimulating MSC's pro-healing paracrine secretion. The underlying mechanism was elucidated, wherein the armor protected to prolong MSCs residence at MI site, and triggered paracrine stimulation of MSCs towards immunoregulation and angiogenesis through inducing hypoxia to provoke glycolysis in stem cells. Furthermore, local delivery of coated MSC spheroids in MI rat significantly alleviated local inflammation and subsequent fibrosis via mediation macrophage polarization towards pro-healing M2 phenotype and improved cardiac function. In general, this study provided critical insight into the enhanced therapeutic efficacy of stem cell spheroids coated with a multifunctional armor. It potentially opens up a new avenue for designing immunomodulatory treatment for MI via stem cell therapy empowered by functional biomaterials.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Myocardial Infarction , Rats , Animals , Myocardial Infarction/pathology , Stem Cells/pathology , Spheroids, Cellular/pathology , Wound Healing
19.
Tissue Eng Part C Methods ; 30(5): 193-205, 2024 May.
Article in English | MEDLINE | ID: mdl-38545771

ABSTRACT

Multiple myeloma (MM) clones reside in the bone marrow (BM), which plays a role in its survival and development. The interactions between MM and their neighboring mesenchymal stromal cells (MSCs) have been shown to promote MM growth and drug resistance. However, those interactions are often missing or misrepresented in traditional two-dimensional (2D) culture models. Application of novel three-dimensional (3D) models might recapitulate the BM niche more precisely, which will offer new insights into MM progression and survival. Here, we aimed to establish two 3D models, based on MSC spheroids and collagen droplets incorporating both MM cells and MSCs with the goal of replicating the native myeloma context of the BM niche. This approach revealed that although MSCs can spontaneously assemble spheroids with altered metabolic traits, MSC spheroid culture does not support the integration of MM cells. On the contrary, collagen-droplet culture supported the growth of both cell types. In collagen, MSC proliferation was reduced, with the correlating decrease in ATP production and Ki-67 expression, which might resemble in vivo conditions, rather than 2D abundance of nutrients and space. MSCs and MMs were distributed homogenously throughout the collagen droplet, with an apparent CXCL12 expression in MSCs. In addition, the response of MM cells to bortezomib was substantially reduced in collagen, indicating the importance of 3D culture in the investigation of myeloma cell behavior, as drug resistance is one of the most pertinent issues in cancer therapy.


Subject(s)
Collagen , Mesenchymal Stem Cells , Multiple Myeloma , Spheroids, Cellular , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Humans , Collagen/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Spheroids, Cellular/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Proliferation/drug effects , Cell Line, Tumor , Models, Biological , Cell Culture Techniques/methods
20.
J Vis Exp ; (204)2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38372384

ABSTRACT

In vitro drug sensitivity screens are important tools in the discovery of anti-cancer drug combination therapies. Typically, these in vitro drug screens are performed on cells grown in a monolayer. However, these two-dimensional (2D) models are considered less accurate compared to three-dimensional (3D) spheroid cell models; this is especially true for glioma stem cell lines. Cells grown in spheres activate different signaling pathways and are considered more representative of in vivo models than monolayer cell lines. This protocol describes a method for in vitro drug screening of spheroid lines; mouse and human glioma stem cell lines are used as an example. This protocol describes a 3D spheroid drug sensitivity and synergy assay that can be used to determine if a drug or drug combination induces cell death and if two drugs synergize. Glioma stem cell lines are modified to express RFP. Cells are plated in low attachment round well bottom 96 plates, and spheres are allowed to form overnight. Drugs are added, and the growth is monitored by measuring the RFP signal over time using the Incucyte live imaging system, a fluorescence microscope embedded in the tissue culture incubator. Half maximal inhibitory concentration (IC50), median lethal dose (LD50), and synergy score are subsequently calculated to evaluate sensitivities to drugs alone or in combination. The three-dimensional nature of this assay provides a more accurate reflection of tumor growth, behavior, and drug sensitivities in vivo, thus forming the basis for further preclinical investigation.


Subject(s)
Glioma , Spheroids, Cellular , Humans , Mice , Animals , Drug Evaluation, Preclinical/methods , Cell Line, Tumor , Spheroids, Cellular/pathology , Glioma/pathology , Neoplastic Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...