1
|
Fernández-Olivares A, Orellana VP, Llanquinao J, Nuñez G, Pérez-Moreno P, Contreras-Riquelme S, Martin AJ, Mammano F, Alfaro IE, Calderón JF, Stehberg J, Sáez MA, Retamal MA. Connexin46 in the nucleus of cancer cells: a possible role as transcription modulator. Cell Commun Signal 2025; 23:153. [PMID: 40148950 PMCID: PMC11948717 DOI: 10.1186/s12964-025-02151-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Oncogenes drive cancer progression, but few are active exclusively in tumor cells. Connexins (Cxs), traditionally recognized as ion channel proteins, can localize to the nucleus and regulate gene expression, playing key roles in both physiological and pathological processes. Cx46, once thought to be restricted to the eye lens, has been implicated in tumor growth, though its underlying mechanisms remain unclear. This study investigates the nuclear presence of Cx46 in cancer cells and its potential role as a transcriptional modulator. METHODS We employed ChIP-Seq, confocal immunofluorescence, and nuclear protein purification to assess Cx46 localization and DNA interactions. Functional assays were conducted to evaluate its effects on invasion, division, spheroid formation, and mesenchymal marker expression. Single-point mutations and molecular dynamics simulations were used to explore potential Cx46-DNA interactions. RESULTS Cx46 mRNA upregulation was found in a variety of tumors compared to adjacent healthy tissue. In HeLa cells, which do not express Cx46, its transfection promoted proliferation, invasion and self-renewal capacity, cancer stem cell traits and mesenchymal features. Consistently, in Sk-Mel-2, which naturally express Cx46, reduced Cx46 expression led to a decrease in the similar parameters. In HeLa cells, nuclear Cx46 was detected in two forms, full length 46 kDa and a 30 kDa fragment (GJA3-30 k), ChIP-Seq experiments revealed that Cx46 binds to the DNA at intergenic and promoter regions, leading to the activation of oncogenic pathways. Molecular dynamics simulations suggest that GJA3-30 k dimerizes in a RAD50-like structure, forming stable DNA complexes. Cx46 and in some cases GJA3-30 k were detected in the nuclei of multiple cancer cell lines, including prostate, breast and skin cancers. CONCLUSIONS Our findings reveal a novel nuclear role for Cx46 in cancer, demonstrating its function as a transcriptional regulator and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Ainoa Fernández-Olivares
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Viviana P Orellana
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Jesús Llanquinao
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
- Translational Medicine Laboratory, Instituto Oncológico Fundación Arturo López Pérez, Santiago 7500691, Chile
| | - Gonzalo Nuñez
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Pablo Pérez-Moreno
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Sebastián Contreras-Riquelme
- Plant Genome Regulation Lab, Centro de Biotecnología Vegetal, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alberto Jm Martin
- Laboratorio de Redes Biológicas, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile
- Escuela de Ingeniería, Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago, Chile
| | - Fabio Mammano
- Department of Physics and Astronomy "G. Galilei", University of Padova, Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Padova, 35131, Italy
| | - Ivan E Alfaro
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
- Centro Ciencia & Vida, Fundación Ciencia & Vida. Nuñoa, Santiago, Chile
| | - Juan F Calderón
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago, 8370146, Chile
| | - Mauricio A Sáez
- Departamento de Procesos Diagnósticos y Evaluación, Laboratorio de Investigación en Salud de Precisión, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco, Chile.
| | - Mauricio A Retamal
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.
| |
Collapse
|
2
|
Nalla LV, Kanukolanu A, Yeduvaka M, Gajula SNR. Advancements in Single-Cell Proteomics and Mass Spectrometry-Based Techniques for Unmasking Cellular Diversity in Triple Negative Breast Cancer. Proteomics Clin Appl 2025; 19:e202400101. [PMID: 39568435 PMCID: PMC11726282 DOI: 10.1002/prca.202400101] [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: 10/11/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive and complex subtype of breast cancer characterized by a lack of targeted treatment options. Intratumoral heterogeneity significantly drives disease progression and complicates therapeutic responses, necessitating advanced analytical approaches to understand its underlying biology. This review aims to explore the advancements in single-cell proteomics and their application in uncovering cellular diversity in TNBC. It highlights innovations in sample preparation, mass spectrometry-based techniques, and the potential for integrating proteomics into multi-omics platforms. METHODS The review discusses the combination of improved sample preparation methods and cutting-edge mass spectrometry techniques in single-cell proteomics. It emphasizes the challenges associated with protein analysis, such as the inability to amplify proteins akin to transcripts, and examines strategies to overcome these limitations. RESULTS Single-cell proteomics provides a direct link to phenotype and cell behavior, complementing transcriptomic approaches and offering new insights into the mechanisms driving TNBC. The integration of advanced techniques has enabled deeper exploration of cellular heterogeneity and disease mechanisms. CONCLUSION Despite the challenges, single-cell proteomics holds immense potential to evolve into a high-throughput and scalable multi-omics platform. Addressing existing hurdles will enable deeper biological insights, ultimately enhancing the diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Lakshmi Vineela Nalla
- Department of Pharmacology, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Aarika Kanukolanu
- Department of Pharmaceutical Analysis, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Madhuri Yeduvaka
- Department of Pharmacology, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| |
Collapse
|
3
|
Porter JM, Yitayew M, Tabrizian M. Renewable Human Cell Model for Type 1 Diabetes Research: EndoC- βH5/HUVEC Coculture Spheroids. J Diabetes Res 2023; 2023:6610007. [PMID: 38162632 PMCID: PMC10757655 DOI: 10.1155/2023/6610007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/20/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024] Open
Abstract
In vitro drug screening for type 1 diabetes therapies has largely been conducted on human organ donor islets for proof of efficacy. While native islets are the ultimate target of these drugs (either in situ or for transplantation), significant benefit can be difficult to ascertain due to the highly heterogeneous nature of individual donors and the overall scarcity of human islets for research. We present an in vitro coculture model based on immortalized insulin-producing beta-cell lines with human endothelial cells in 3D spheroids that aims to recapitulate the islet morphology in an effort towards developing a standardized cell model for in vitro diabetes research. Human insulin-producing immortalized EndoC-βH5 cells are cocultured with human endothelial cells in varying ratios to evaluate 3D cell culture models for type 1 diabetes research. Insulin secretion, metabolic activity, live cell fluorescence staining, and gene expression assays were used to compare the viability and functionality of spheroids composed of 100% beta-cells, 1 : 1 beta-cell/endothelial, and 1 : 3 beta-cell/endothelial. Monoculture and βH5/HUVEC cocultures formed compact spheroids within 7 days, with average diameter ~140 μm. This pilot study indicated that stimulated insulin release from 0 to 20 mM glucose increased from ~8-fold for monoculture and 1 : 1 coculture spheroids to over 20-fold for 1 : 3 EndoC-βH5/HUVEC spheroids. Metabolic activity was also ~12% higher in the 1 : 3 EndoC-βH5/HUVEC group compared to other groups. Stimulating monoculture beta-cell spheroids with 20 mM glucose +1 μg/mL glycine-modified INGAP-P increased the insulin stimulation index ~2-fold compared to glucose alone. Considering their availability and consistent phenotype, EndoC-βH5-based spheroids present a useful 3D cell model for in vitro testing and drug screening applications.
Collapse
Affiliation(s)
- James M. Porter
- Department of Biological and Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada H3A 0G4
| | - Michael Yitayew
- Department of Biological and Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada H3A 0G4
| | - Maryam Tabrizian
- Department of Biological and Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada H3A 0G4
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada H3A 1G1
| |
Collapse
|
4
|
Zhao H, Han R, Wang Z, Xian J, Bai X. Colorectal Cancer Stem Cells and Targeted Agents. Pharmaceutics 2023; 15:2763. [PMID: 38140103 PMCID: PMC10748092 DOI: 10.3390/pharmaceutics15122763] [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: 10/13/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Since their discovery, cancer stem cells have become a hot topic in cancer therapy research. These cells possess stem cell-like self-renewal and differentiation capacities and are important factors that dominate cancer metastasis, therapy-resistance and recurrence. Worse, their inherent characteristics make them difficult to eliminate. Colorectal cancer is the third-most common cancer and the second leading cause of cancer death worldwide. Targeting colorectal cancer stem cells (CR-CSCs) can inhibit colorectal cancer metastasis, enhance therapeutic efficacy and reduce recurrence. Here, we introduced the origin, biomarker proteins, identification, cultivation and research techniques of CR-CSCs, and we summarized the signaling pathways that regulate the stemness of CR-CSCs, such as Wnt, JAK/STAT3, Notch and Hh signaling pathway. In addition to these, we also reviewed recent anti-CR-CSC drugs targeting signaling pathways, biomarkers and other regulators. These will help researchers gain insight into the current agents targeting to CR-CSCs, explore new cancer drugs and propose potential therapies.
Collapse
Affiliation(s)
- Haobin Zhao
- Department of General Practice, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China; (H.Z.); (J.X.)
- Endocrinology Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China
| | - Ruining Han
- Obstetric Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China;
| | - Zhankun Wang
- Emergency Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China;
| | - Junfang Xian
- Department of General Practice, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China; (H.Z.); (J.X.)
| | - Xiaosu Bai
- Endocrinology Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China
| |
Collapse
|
5
|
Pei H, Han Z, Wang Y, Xu C, Li Y, Fan Y, Li L, Tang B. Retraction of "Label-Free Isolation of Low-Adhesion Cells with Stem Properties for Cancer Stem Cell-Specific Drug Evaluation". Anal Chem 2023; 95:6191. [PMID: 36122350 DOI: 10.1021/acs.analchem.2c00444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Habanjar O, Maurin AC, Vituret C, Vachias C, Longechamp L, Garnier C, Decombat C, Bourgne C, Diab-Assaf M, Caldefie-Chezet F, Delort L. A bicellular fluorescent ductal carcinoma in situ (DCIS)-like tumoroid to study the progression of carcinoma: practical approaches and optimization. Biomater Sci 2023; 11:3308-3320. [PMID: 36946175 DOI: 10.1039/d2bm01470j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Recently, many types of 3D culture systems have been developed to preserve the physicochemical environment and biological characteristics of the original tumors better than the conventional 2D monolayer culture system. There are various types of models belonging to this culture, such as the culture based on non-adherent and/or scaffold-free matrices to form the tumors. Agarose mold has been widely used to facilitate tissue spheroid assembly, as it is essentially non-biodegradable, bio-inert, biocompatible, low-cost, and low-attachment material that can promote cell spheroidization. As no studies have been carried out on the development of a fluorescent bicellular tumoroid mimicking ductal carcinoma in situ (DCIS) using human cell lines, our objective was to detail the practical approaches developed to generate this model, consisting of a continuous layer of myoepithelial cells (MECs) around a previously formed in situ breast tumor. The practical approaches developed to generate a bi-cellular tumoroid mimicking ductal carcinoma in situ (DCIS), consisting of a continuous layer of myoepithelial cells (MECs) around a previously formed in situ breast tumoroid. Firstly, the optimal steps and conditions of spheroids generation using a non-adherent agarose gel were described, in particular, the appropriate medium, seeding density of each cell type and incubation period. Next, a lentiviral transduction approach to achieve stable fluorescent protein expression (integrative system) was used to characterize the different cell lines and to track tumoroid generation through immunofluorescence, the organization of the two cell types was validated, specific merits and drawbacks were compared to lentiviral transduction. Two lentiviral vectors expressing either EGFP (Enhanced Green Fluorescent Protein) or m-Cherry (Red Fluorescent Protein) were used. Various rates of a multiplicity of infection (MOI) and multiple types of antibodies (anti-p63, anti-CK8, anti-Maspin, anti-Calponin) for immunofluorescence analysis were tested to determine the optimal conditions for each cell line. At MOI 40 (GFP) and MOI 5 (m-Cherry), the signals were almost homogeneously distributed in the cells which could then be used to generate the DCIS-like tumoroids. Images of the tumoroids in agarose molds were captured with a confocal microscope Micro Zeiss Cell Observer Spinning Disk or with IncuCyte® to follow the progress of the generation. Measurement of protumoral cytokines such as IL-6, IL8 and leptin confirmed their secretion in the supernatants, indicating that the properties of our cells were not altered. Finally the advantages and disadvantages of each fluorescent approach were discussed. This model could also be used for other solid malignancies to study the complex relationship between different cells such as tumor and myoepithelial cells in various microenvironments (inflammatory, adipose and tumor, obesity, etc.). Although, this new model is well established to monitor drug screening applications and perform pharmacokinetic and pharmacodynamic analyses.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, 63000 Clermont-Ferrand, France.
| | | | - Cyrielle Vituret
- Université Clermont-Auvergne, INRAE, UNH, 63000 Clermont-Ferrand, France.
| | - Caroline Vachias
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne, UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Lucie Longechamp
- Université Clermont-Auvergne, INRAE, UNH, 63000 Clermont-Ferrand, France.
| | - Cécile Garnier
- Université Clermont-Auvergne, INRAE, UNH, 63000 Clermont-Ferrand, France.
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH, 63000 Clermont-Ferrand, France.
| | - Céline Bourgne
- Plate-forme CMF, Service d'Hématologie biologique, CHU de Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie moléculaire et anticancéreuse, Faculté des Sciences II, Université libanaise Fanar, Beirut, Lebanon
| | | | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, 63000 Clermont-Ferrand, France.
| |
Collapse
|
7
|
The Use of Biomaterials in Three-Dimensional Culturing of Cancer Cells. Curr Issues Mol Biol 2023; 45:1100-1112. [PMID: 36826018 PMCID: PMC9954970 DOI: 10.3390/cimb45020073] [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: 12/05/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Cell culture is an important tool in biological research. Most studies use 2D cell culture, but cells grown in 2D cell culture have drawbacks, including limited cell and cell-extracellular matrix interactions, which make it inaccurate to model conditions in vivo. Anticancer drug screening is an important research and development process for developing new drugs. As an experiment to mimic the cancer environment in vivo, several studies have been carried out on 3-dimensional (3D) cell cultures with added biomaterials. The use of hydrogel in 3D culture cells is currently developing. The type of hydrogel used might influence cell morphology, viability, and drug screening outcome. Therefore, this review discusses 3D cell culture research regarding the addition of biomaterials.
Collapse
|
8
|
Liu Y, Chen X, Chen J, Luo Y, Chen Z, Lin D, Zhang J, Liu D. Gel-Free Single-Cell Culture Arrays on a Microfluidic Chip for Highly Efficient Expansion and Recovery of Colon Cancer Stem Cells. ACS Biomater Sci Eng 2022; 8:3623-3632. [PMID: 35786837 DOI: 10.1021/acsbiomaterials.2c00378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The microgel single-cell culture approach we developed to expand tumor stem cells (TSCs) is associated with limited TSC production, which can be attributable to cell viability loss in microgel formation and tumorsphere expansion limitation caused by hydrogel stiffness. In this work, we developed a gel-free single-cell culture array on a microfluidic chip to overcome these issues. The microfluidic chip used in the study has a 16,000 hydrophilic microchamber array, which can capture ∼2000 single cells at a time. After cell capturing, the cell culture chambers were enclosed by forming a chitosan layer through interactions between chitosan and alginate, thus preventing cell loss in the gel-free culture. The hydrophilic coating prevented cell adhesion, so only TSCs with anti-apoptosis and self-renewal properties can survive the harsh culture and form tumorspheres. After a 7 day culture, 19.04% of the HCT116 colon cancer cells formed single-cell-derived tumorspheres with an average size of 46.59 ± 10.58 μm. Compared with the microgel single-cell culture, sphere-forming rate and TSC expansion efficiency were significantly improved by using this gel-free single-cell culture array. After cell culture, the chitosan layer could be destabilized easily, thus allowing recovery of the tumorspheres from the microchip by applying a reverse flow. Approximately 13,600 cells could be obtained in a single culture, which can be used for off-chip cell assays. Flow cytometry analysis indicated high proportions of LGR5(+) and SOX2(+) cells within the single-cell-derived tumorspheres. Moreover, the differentiation experiments confirmed the multi-lineage differentiation potential of single-cell-derived tumorspheres. The gel-free single-cell culture offers a label-free approach to obtain sufficient amounts of TSCs, which is valuable for tumor biology research and the development of TSC-specific therapeutic strategies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xiao Chen
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jueming Chen
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yanzhang Luo
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Zihe Chen
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Dongguo Lin
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China.,Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China.,Clinical Molecular Medicine and Molecular Diagnosis Key Laboratory of Guangdong Province, Guangzhou 510180, China
| | - Jianye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Dayu Liu
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China.,Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China.,Clinical Molecular Medicine and Molecular Diagnosis Key Laboratory of Guangdong Province, Guangzhou 510180, China
| |
Collapse
|
9
|
Ronaldson-Bouchard K, Baldassarri I, Tavakol DN, Graney PL, Samaritano M, Cimetta E, Vunjak-Novakovic G. Engineering complexity in human tissue models of cancer. Adv Drug Deliv Rev 2022; 184:114181. [PMID: 35278521 PMCID: PMC9035134 DOI: 10.1016/j.addr.2022.114181] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/15/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023]
Abstract
Major progress in the understanding and treatment of cancer have tremendously improved our knowledge of this complex disease and improved the length and quality of patients' lives. Still, major challenges remain, in particular with respect to cancer metastasis which still escapes effective treatment and remains responsible for 90% of cancer related deaths. In recent years, the advances in cancer cell biology, oncology and tissue engineering converged into the engineered human tissue models of cancer that are increasingly recapitulating many aspects of cancer progression and response to drugs, in a patient-specific context. The complexity and biological fidelity of these models, as well as the specific questions they aim to investigate, vary in a very broad range. When selecting and designing these experimental models, the fundamental question is "how simple is complex enough" to accomplish a specific goal of cancer research. Here we review the state of the art in developing and using the human tissue models in cancer research and developmental drug screening. We describe the main classes of models providing different levels of biological fidelity and complexity, discuss their advantages and limitations, and propose a framework for designing an appropriate model for a given study. We close by outlining some of the current needs, opportunities and challenges in this rapidly evolving field.
Collapse
Affiliation(s)
- Kacey Ronaldson-Bouchard
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Ilaria Baldassarri
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Daniel Naveed Tavakol
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Pamela L Graney
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Maria Samaritano
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Elisa Cimetta
- Department of Industrial Engineering, University of Padua, Via Marzolo 9, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; College of Dental Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA.
| |
Collapse
|
10
|
Germain N, Dhayer M, Dekiouk S, Marchetti P. Current Advances in 3D Bioprinting for Cancer Modeling and Personalized Medicine. Int J Mol Sci 2022; 23:3432. [PMID: 35408789 PMCID: PMC8998835 DOI: 10.3390/ijms23073432] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Tumor cells evolve in a complex and heterogeneous environment composed of different cell types and an extracellular matrix. Current 2D culture methods are very limited in their ability to mimic the cancer cell environment. In recent years, various 3D models of cancer cells have been developed, notably in the form of spheroids/organoids, using scaffold or cancer-on-chip devices. However, these models have the disadvantage of not being able to precisely control the organization of multiple cell types in complex architecture and are sometimes not very reproducible in their production, and this is especially true for spheroids. Three-dimensional bioprinting can produce complex, multi-cellular, and reproducible constructs in which the matrix composition and rigidity can be adapted locally or globally to the tumor model studied. For these reasons, 3D bioprinting seems to be the technique of choice to mimic the tumor microenvironment in vivo as closely as possible. In this review, we discuss different 3D-bioprinting technologies, including bioinks and crosslinkers that can be used for in vitro cancer models and the techniques used to study cells grown in hydrogels; finally, we provide some applications of bioprinted cancer models.
Collapse
Affiliation(s)
- Nicolas Germain
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| | - Melanie Dhayer
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Salim Dekiouk
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Philippe Marchetti
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| |
Collapse
|
11
|
Barbosa MAG, Xavier CPR, Pereira RF, Petrikaitė V, Vasconcelos MH. 3D Cell Culture Models as Recapitulators of the Tumor Microenvironment for the Screening of Anti-Cancer Drugs. Cancers (Basel) 2021; 14:190. [PMID: 35008353 PMCID: PMC8749977 DOI: 10.3390/cancers14010190] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Today, innovative three-dimensional (3D) cell culture models have been proposed as viable and biomimetic alternatives for initial drug screening, allowing the improvement of the efficiency of drug development. These models are gaining popularity, given their ability to reproduce key aspects of the tumor microenvironment, concerning the 3D tumor architecture as well as the interactions of tumor cells with the extracellular matrix and surrounding non-tumor cells. The development of accurate 3D models may become beneficial to decrease the use of laboratory animals in scientific research, in accordance with the European Union's regulation on the 3R rule (Replacement, Reduction, Refinement). This review focuses on the impact of 3D cell culture models on cancer research, discussing their advantages, limitations, and compatibility with high-throughput screenings and automated systems. An insight is also given on the adequacy of the available readouts for the interpretation of the data obtained from the 3D cell culture models. Importantly, we also emphasize the need for the incorporation of additional and complementary microenvironment elements on the design of 3D cell culture models, towards improved predictive value of drug efficacy.
Collapse
Affiliation(s)
- Mélanie A. G. Barbosa
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Biofabrication Group, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickevičiaus g 9, LT-44307 Kaunas, Lithuania;
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
12
|
Deng S, Li L, Xu S, Wang X, Han T. Promotion of gastric tumor initiating cells in a 3D collagen gel culture model via YBX1/SPP1/NF-κB signaling. Cancer Cell Int 2021; 21:599. [PMID: 34758833 PMCID: PMC8579534 DOI: 10.1186/s12935-021-02307-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/29/2021] [Indexed: 01/07/2023] Open
Abstract
Background The high potential for tumor recurrence and chemoresistance is a major challenge of clinical gastric cancer treatment. Increasing evidence suggests that the presence of tumor initiating cells (TICs) is the principal cause of tumor recurrence and chemoresistance. However, the underlying mechanism of TIC development remains controversial. Methods To identify novel molecular pathways in gastric cancer, we screened the genomic expression profile of 155 gastric cancer patients from the TCGA database. We then described an improved 3D collagen I gels and tested the effects of collagen on the TIC phenotype of gastric cells using colony formation assay, transwell assay, and nude mouse models. Additionally, cell apoptosis assay was performed to examine the cytotoxicity of 5-fluorine and paclitaxel on gastric cancer cells cultured in 3D collagen I gels. Results Elevated expression of type I collagen was observed in tumor tissues from high stage patients (stage T3–T4) when compared to the low stage group (n=10, stage T1–T2). Furthermore, tumor cells seeded in a low concentration of collagen gels acquired TIC-like phenotypes and revealed enhanced resistance to chemotherapeutic agents, which was dependent on an integrin β1 (ITGB1)/Y-box Binding Protein 1 (YBX1)/Secreted Phosphoprotein 1 (SPP1)/NF-κB signaling pathway. Importantly, inhibition of ITGB1/NF-κB signaling efficiently reversed the chemoresistance induced by collagen and promoted anticancer effects in vivo. Conclusions Our findings demonstrated that type I collagen promoted TIC-like phenotypes and chemoresistance through ITGB1/YBX1/SPP1/NF-κB pathway, which may provide novel insights into gastric cancer therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02307-x.
Collapse
Affiliation(s)
- Shuangya Deng
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lun Li
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shu Xu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaobo Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tong Han
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
13
|
Lin D, Chen X, Liu Y, Lin Z, Luo Y, Fu M, Yang N, Liu D, Cao J. Microgel Single-Cell Culture Arrays on a Microfluidic Chip for Selective Expansion and Recovery of Colorectal Cancer Stem Cells. Anal Chem 2021; 93:12628-12638. [PMID: 34495647 DOI: 10.1021/acs.analchem.1c02335] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer stem cells (CSCs) are rare and lack definite biomarkers, necessitating new methods for a robust expansion. Here, we developed a microfluidic single-cell culture (SCC) approach for expanding and recovering colorectal CSCs from both cell lines and tumor tissues. By incorporating alginate hydrogels with droplet microfluidics, a high-density microgel array can be formed on a microfluidic chip that allows for single-cell encapsulation and nonadhesive culture. The SCC approach takes advantage of the self-renewal property of stem cells, as only the CSCs can survive in the SCC and form tumorspheres. Consecutive imaging confirmed the formation of single-cell-derived tumorspheres, mainly from a population of small-sized cells. Through on-chip decapsulation of the alginate microgel, ∼6000 live cells can be recovered in a single run, which is sufficient for most biological assays. The recovered cells were verified to have the genetic and phenotypic characteristics of CSCs. Furthermore, multiple CSC-specific targets were identified by comparing the transcriptomics of the CSCs with the primary cancer cells. To summarize, the microgel SCC array offers a label-free approach to obtain sufficient quantities of CSCs and thus is potentially useful for understanding cancer biology and developing personalized CSC-targeting therapies.
Collapse
Affiliation(s)
- Dongguo Lin
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.,Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China
| | - Xiao Chen
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yang Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Zhun Lin
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yanzhang Luo
- Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Mingpeng Fu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Dayu Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.,Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China
| | - Jie Cao
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of General Surgery, The Second Affiliated Hospital of South China University of Technology, 1, Panfu Road, Guangzhou 510180, China
| |
Collapse
|
14
|
Three-Dimensional Spheroids as In Vitro Preclinical Models for Cancer Research. Pharmaceutics 2020; 12:pharmaceutics12121186. [PMID: 33291351 PMCID: PMC7762220 DOI: 10.3390/pharmaceutics12121186] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/29/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Most cancer biologists still rely on conventional two-dimensional (2D) monolayer culture techniques to test in vitro anti-tumor drugs prior to in vivo testing. However, the vast majority of promising preclinical drugs have no or weak efficacy in real patients with tumors, thereby delaying the discovery of successful therapeutics. This is because 2D culture lacks cell–cell contacts and natural tumor microenvironment, important in tumor signaling and drug response, thereby resulting in a reduced malignant phenotype compared to the real tumor. In this sense, three-dimensional (3D) cultures of cancer cells that better recapitulate in vivo cell environments emerged as scientifically accurate and low cost cancer models for preclinical screening and testing of new drug candidates before moving to expensive and time-consuming animal models. Here, we provide a comprehensive overview of 3D tumor systems and highlight the strategies for spheroid construction and evaluation tools of targeted therapies, focusing on their applicability in cancer research. Examples of the applicability of 3D culture for the evaluation of the therapeutic efficacy of nanomedicines are discussed.
Collapse
|
15
|
A Marine Collagen-Based Biomimetic Hydrogel Recapitulates Cancer Stem Cell Niche and Enhances Progression and Chemoresistance in Human Ovarian Cancer. Mar Drugs 2020; 18:md18100498. [PMID: 33003514 PMCID: PMC7599646 DOI: 10.3390/md18100498] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Recent attention has focused on the development of an effective three-dimensional (3D) cell culture system enabling the rapid enrichment of cancer stem cells (CSCs) that are resistant to therapies and serving as a useful in vitro tumor model that accurately reflects in vivo behaviors of cancer cells. Presently, an effective 3D in vitro model of ovarian cancer (OC) was developed using a marine collagen-based hydrogel. Advantages of the model include simplicity, efficiency, bioactivity, and low cost. Remarkably, OC cells grown in this hydrogel exhibited biochemical and physiological features, including (1) enhanced cell proliferation, migration and invasion, colony formation, and chemoresistance; (2) suppressed apoptosis with altered expression levels of apoptosis-regulating molecules; (3) upregulated expression of crucial multidrug resistance-related genes; (4) accentuated expression of key molecules associated with malignant progression, such as epithelial–mesenchymal transition transcription factors, Notch, and pluripotency biomarkers; and (5) robust enrichment of ovarian CSCs. The findings indicate the potential of our 3D in vitro OC model as an in vitro research platform to study OC and ovarian CSC biology and to screen novel therapies targeting OC and ovarian CSCs.
Collapse
|
16
|
Granger CJ, Hoyt AK, Moran A, Becker B, Sedani A, Saigh S, Conway SA, Brown J, Galoian K. Cancer stem cells as a therapeutic target in 3D tumor models of human chondrosarcoma: An encouraging future for proline rich polypeptide‑1. Mol Med Rep 2020; 22:3747-3758. [PMID: 32901865 PMCID: PMC7533489 DOI: 10.3892/mmr.2020.11480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
Chondrosarcoma is a malignant bone neoplasm that is refractory to chemotherapy and radiation. With no current biological treatments, mutilating surgical resection is the only effective treatment. Proline rich polypeptide 1 (PRP-1), which is a 15-amino acid inhibitor of mammalian target of rapamycin complex-1 (mTORC1), has been indicated to exert cytostatic and immunomodulatory properties in human chondrosarcoma cells in a monolayer. The aim of the present study was to evaluate the effects of PRP-1 on an in vitro 3D chondrosarcoma tumor model, known as spheroids, and on the cancer stem cells (CSCs) which form spheroids. JJ012 cells were cultured and treated with PRP-1. An ALDEFLUOR™ assay was conducted (with N,N-diethylaminobenzaldehyde as the negative control) to assess aldehyde dehydrogenase (ALDH) activity (a recognized CSC marker), and bulk JJ012, ALDHhigh and PRP-1 treated ALDHlow cells were sorted using flow cytometry. Colony formation and spheroid formation assays of cell fractions, including CSCs, were used to compare the PRP-1-treated groups with the control. CSCs were assessed for early apoptosis and cell death with a modified Annexin V/propidium iodide assay. Western blotting was used to identify mesenchymal stem cell markers (STRO1, CD44 and STAT3), and spheroid self-renewal assays were also conducted. A clonogenic dose-response assay demonstrated that 20 µg/ml PRP-1 was the most effective dose for reducing colony formation capacity. Furthermore, CSC spheroid growth was significantly reduced with increasing doses of PRP-1. Annexin V analysis demonstrated that PRP-1 induced CSC cell death, and that this was not attributed to apoptosis or necrosis. Western blot analysis confirmed the expression of mesenchymal markers, and the spheroid self-renewal assay confirmed the presence of self-renewing CSCs. The results of the present study demonstrate that PRP-1 eliminates anchorage independent CSC growth and spheroid formation, indicating that PRP-1 likely inhibits tumor formation in a murine model. Additionally, a decrease in non-CSC bulk tumor cells indicates an advantageous decline in tumor stromal cells. These findings confirm that PRP-1 inhibits CSC proliferation in a 3D tumor model which mimics the behavior of chondrosarcoma in vivo.
Collapse
Affiliation(s)
- Caroline J Granger
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Aaron K Hoyt
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alexandra Moran
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Beatrice Becker
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anil Sedani
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Shannon Saigh
- Department of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Sheila A Conway
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jeffrey Brown
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Karina Galoian
- RMSB Room 8012 (D27), Department of Orthopedic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
17
|
Head and Neck Cancer Stem Cell-Enriched Spheroid Model for Anticancer Compound Screening. Cells 2020; 9:cells9071707. [PMID: 32708734 PMCID: PMC7408407 DOI: 10.3390/cells9071707] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs), a rare cell population in tumors, are resistant to conventional chemotherapy and thus responsible for tumor recurrence. To screen for active compounds targeting CSCs, a good CSC-enriched model compatible with high-throughput screening (HTS) is needed. Here, we describe a new head and neck cancer stem cell-enriched spheroid model (SCESM) suitable for HTS analyses of anti-CSC compounds. We used FaDu cells, round-bottom ultra-low adherent (ULA) microplates, and stem medium. The formed spheroids displayed increased expression of all stem markers tested (qRT-PCR and protein analysis) in comparison to the FaDu cells grown in a standard adherent culture or in a well-known HTS-compatible multi-cellular tumor spheroid model (MCTS). Consistent with increased stemness of the cells in the spheroid, confocal microscopy detected fast proliferating cells only at the outer rim of the SCESM spheroids, with poorly/non-proliferating cells deeper in. To confirm the sensitivity of our model, we used ATRA treatment, which strongly reduced the expression of selected stem markers. Altogether, we developed a CSC-enriched spheroid model with a simple protocol, a microplate format compatible with multimodal detection systems, and a high detection signal, making it suitable for anti-CSC compounds' HTS.
Collapse
|
18
|
Salati MA, Khazai J, Tahmuri AM, Samadi A, Taghizadeh A, Taghizadeh M, Zarrintaj P, Ramsey JD, Habibzadeh S, Seidi F, Saeb MR, Mozafari M. Agarose-Based Biomaterials: Opportunities and Challenges in Cartilage Tissue Engineering. Polymers (Basel) 2020; 12:1150. [PMID: 32443422 PMCID: PMC7285176 DOI: 10.3390/polym12051150] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 12/17/2022] Open
Abstract
The lack of adequate blood/lymphatic vessels as well as low-potential articular cartilage regeneration underlines the necessity to search for alternative biomaterials. Owing to their unique features, such as reversible thermogelling behavior and tissue-like mechanical behavior, agarose-based biomaterials have played a key role in cartilage tissue repair. Accordingly, the need for fabricating novel highly efficient injectable agarose-based biomaterials as hydrogels for restoration of injured cartilage tissue has been recognized. In this review, the resources and conspicuous properties of the agarose-based biomaterials were reviewed. First, different types of signals together with their functionalities in the maintenance of cartilage homeostasis were explained. Then, various cellular signaling pathways and their significant role in cartilage tissue engineering were overviewed. Next, the molecular structure and its gelling behavior have been discussed. Eventually, the latest advancements, the lingering challenges, and future ahead of agarose derivatives from the cartilage regeneration perspective have been discussed.
Collapse
Affiliation(s)
- Mohammad Amin Salati
- Polymer Engineering Department, Faculty of Engineering, Urmia University, Urmia 5756151818, Iran; (M.A.S.); (J.K.); (A.M.T.); (A.S.)
| | - Javad Khazai
- Polymer Engineering Department, Faculty of Engineering, Urmia University, Urmia 5756151818, Iran; (M.A.S.); (J.K.); (A.M.T.); (A.S.)
| | - Amir Mohammad Tahmuri
- Polymer Engineering Department, Faculty of Engineering, Urmia University, Urmia 5756151818, Iran; (M.A.S.); (J.K.); (A.M.T.); (A.S.)
| | - Ali Samadi
- Polymer Engineering Department, Faculty of Engineering, Urmia University, Urmia 5756151818, Iran; (M.A.S.); (J.K.); (A.M.T.); (A.S.)
| | - Ali Taghizadeh
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran 11155-4563, Iran; (A.T.); (M.T.)
| | - Mohsen Taghizadeh
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran 11155-4563, Iran; (A.T.); (M.T.)
| | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, USA;
| | - Josh D. Ramsey
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, USA;
| | - Sajjad Habibzadeh
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591639675, Iran;
| | - Farzad Seidi
- Provincial Key Lab of Pulp and Paper Science and Technology and Joint International Research Lab of Lignocellulosic Functional Materials, Nanjing Forestry University, Nanjing 210037, China;
| | - Mohammad Reza Saeb
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran P.O. Box 16765-654, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 144961-4535, Iran
| |
Collapse
|
19
|
Modeling the Efficacy of Oncolytic Adenoviruses In Vitro and In Vivo: Current and Future Perspectives. Cancers (Basel) 2020; 12:cancers12030619. [PMID: 32155969 PMCID: PMC7139921 DOI: 10.3390/cancers12030619] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic adenoviruses (OAd) selectively target and lyse tumor cells and enhance anti- tumor immune responses. OAds have been used as promising cancer gene therapies for many years and there are a multitude of encouraging pre-clinical studies. However, translating OAd therapies to the clinic has had limited success, in part due to the lack of realistic pre-clinical models to rigorously test the efficacy of OAds. Solid tumors have a heterogenous and hostile microenvironment that provides many barriers to OAd treatment, including structural and immunosuppressive components that cannot be modeled in two-dimensional tissue culture. To replicate these characteristics and bridge the gap between pre-clinical and clinical success, studies must test OAd therapy in three-dimensional culture and animal models. This review focuses on current methods to test OAd efficacy in vitro and in vivo and the development of new model systems to test both oncolysis and immune stimulatory components of oncolytic adenovirotherapy.
Collapse
|
20
|
Leal-Egaña A, Balland M, Boccaccini AR. Re-engineering Artificial Neoplastic Milieus: Taking Lessons from Mechano- and Topobiology. Trends Biotechnol 2020; 38:142-153. [DOI: 10.1016/j.tibtech.2019.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/30/2022]
|
21
|
Dondossola E, Paindelli C. In vitro three-dimensional modeling for prostate cancer. BIOMATERIALS FOR 3D TUMOR MODELING 2020:251-286. [DOI: 10.1016/b978-0-12-818128-7.00012-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|