1
|
Taghizadeh-Tabarsi R, Akbari-Birgani S, Amjadi M, Mohammadi S, Nikfarjam N, Kusamori K. Aptamer-guided graphene oxide quantum dots for targeted suicide gene therapy in an organoid model of luminal breast cancer. Sci Rep 2024; 14:24104. [PMID: 39406784 PMCID: PMC11480468 DOI: 10.1038/s41598-024-74312-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Breast cancer is one of the most common cancers in women. One of the best therapeutic methods against breast cancer is gene therapy, while having an appropriate gene carrier is the biggest challenge of gene therapy. Hence, developing carriers with low cytotoxicity and high gene transfection efficiency, and preferentially with the selective function of gene delivery is a critical demand for this method. In the present study, we introduce a novel targeted carrier to deliver the inducible caspase-9 suicide gene (pLVSIN-iC9) into breast cancer cells. The carrier is composed of graphene oxide quantum dots decorated with polyethyleneimine, and S2.2; an aptamer with high affinity to MUC1 (GOQD-PEI/S2.2). Due to the overexpression of MUC1 in breast cancer cells, the designed GOQD-PEI/S2.2/pLVSIN-iC9 can selectively target cancer cells. Moreover, to better mimic solid tumor conditions, and to evaluate the selective effect of the GOQD-PEI/S2.2/pLVSIN-iC9, an organoid model derived from human dermal fibroblasts (HDF) and MCF-7 cells (coculture organoid) was generated and characterized. The results demonstrate that the coculture organoid model adapts the tissue structure of luminal breast cancer, as well. Therefore, the organoids were subjected to treatment with targeted gene therapy using GOQD-PEI/S2.2/pLVSIN-iC9. Our evidence supports the targeted killing effect of iC9 on the breast cancer cells of the organoids and suggests the good potential of the newly introduced carriers in targeted gene delivery.
Collapse
Affiliation(s)
- Reza Taghizadeh-Tabarsi
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 4513766731, Iran
| | - Shiva Akbari-Birgani
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 4513766731, Iran.
- Research Center for Basic Sciences and Modern Technologies (RBST), Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran.
| | - Mehrnaz Amjadi
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 4513766731, Iran
| | - Soheila Mohammadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasser Nikfarjam
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, 29208, USA
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 4513766731, Iran
| | - Kosuke Kusamori
- Laboratory of Cellular Drug Discovery and Development, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
2
|
Yadav R, Mahajan S, Singh H, Mehra NK, Madan J, Doijad N, Singh PK, Guru SK. Emerging In Vitro and In Vivo Models: Hope for the Better Understanding of Cancer Progression and Treatment. Adv Biol (Weinh) 2024; 8:e2300487. [PMID: 38581078 DOI: 10.1002/adbi.202300487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Various cancer models have been developed to aid the understanding of the underlying mechanisms of tumor development and evaluate the effectiveness of various anticancer drugs in preclinical studies. These models accurately reproduce the critical stages of tumor initiation and development to mimic the tumor microenvironment better. Using these models for target validation, tumor response evaluation, resistance modeling, and toxicity comprehension can significantly enhance the drug development process. Herein, various in vivo or animal models are presented, typically consisting of several mice and in vitro models ranging in complexity from transwell models to spheroids and CRISPR-Cas9 technologies. While in vitro models have been used for decades and dominate the early stages of drug development, they are still limited primary to simplistic tests based on testing on a single cell type cultivated in Petri dishes. Recent advancements in developing new cancer therapies necessitate the generation of complicated animal models that accurately mimic the tumor's complexity and microenvironment. Mice make effective tumor models as they are affordable, have a short reproductive cycle, exhibit rapid tumor growth, and are simple to manipulate genetically. Human cancer mouse models are crucial to understanding the neoplastic process and basic and clinical research improvements. The following review summarizes different in vitro and in vivo metastasis models, their advantages and disadvantages, and their ability to serve as a model for cancer research.
Collapse
Affiliation(s)
- Rachana Yadav
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Hoshiyar Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Neelesh Kumar Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Nandkumar Doijad
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
3
|
Northcutt LA, Questell AM, Rhoades J, Rafat M. Development of an alginate-Matrigel hydrogel system to evaluate cancer cell behavior in the stiffness range of the bone marrow. FRONTIERS IN BIOMATERIALS SCIENCE 2023; 2:1140641. [PMID: 38169992 PMCID: PMC10760992 DOI: 10.3389/fbiom.2023.1140641] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Bone metastasis is highly prevalent in breast cancer patients with metastatic disease. These metastatic cells may eventually form osteolytic lesions and affect the integrity of the bone, causing pathological fractures and impairing patient quality of life. Although some mechanisms have been determined in the metastatic cascade to the bone, little is known about how the mechanical cues of the bone marrow microenvironment influence tumor cell growth and invasion once they have homed to the secondary site. The mechanical properties within the bone marrow range from 0.5 kPa in the sinusoidal region to 40 kPa in the endosteal region. Here, we report an alginate-Matrigel hydrogel that can be modulated to the stiffness range of the bone marrow and used to evaluate tumor cell behavior. We fabricated alginate-Matrigel hydrogels with varying calcium sulfate (CaSO4) concentrations to tune stiffness, and we demonstrated that these hydrogels recapitulated the mechanical properties observed in the bone marrow microenvironment (0.7-16 kPa). We encapsulated multiple breast cancer cell lines into these hydrogels to assess growth and invasion. Tumor cells in stiffer hydrogels exhibited increased proliferation and enhanced elongation compared to lower stiffness hydrogels, which suggests that stiffer environments in the bone marrow promote cellular invasive capacity. This work establishes a system that replicates bone marrow mechanical properties to elucidate the physical factors that contribute to metastatic growth.
Collapse
Affiliation(s)
- Logan A. Northcutt
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
| | - Alyssa M. Questell
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Julie Rhoades
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Marjan Rafat
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
4
|
Ingavle G, Das M. Bench to Bedside: New Therapeutic Approaches with Extracellular Vesicles and Engineered Biomaterials for Targeting Therapeutic Resistance of Cancer Stem Cells. ACS Biomater Sci Eng 2022; 8:4673-4696. [PMID: 36194142 DOI: 10.1021/acsbiomaterials.2c00484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer has recently been the second leading cause of death worldwide, trailing only cardiovascular disease. Cancer stem cells (CSCs), represented as tumor-initiating cells (TICs), are mainly liable for chemoresistance and disease relapse due to their self-renewal capability and differentiating capacity into different types of tumor cells. The intricate molecular mechanism is necessary to elucidate CSC's chemoresistance properties and cancer recurrence. Establishing efficient strategies for CSC maintenance and enrichment is essential to elucidate the mechanisms and properties of CSCs and CSC-related therapeutic measures. Current approaches are insufficient to mimic the in vivo chemical and physical conditions for the maintenance and growth of CSC and yield unreliable research results. Biomaterials are now widely used for simulating the bone marrow microenvironment. Biomaterial-based three-dimensional (3D) approaches for the enrichment of CSC provide an excellent promise for future drug discovery and elucidation of molecular mechanisms. In the future, the biomaterial-based model will contribute to a more operative and predictive CSC model for cancer therapy. Design strategies for materials, physicochemical cues, and morphology will offer a new direction for future modification and new methods for studying the CSC microenvironment and its chemoresistance property. This review highlights the critical roles of the microenvironmental cues that regulate CSC function and endow them with drug resistance properties. This review also explores the latest advancement and challenges in biomaterial-based scaffold structure for therapeutic approaches against CSC chemoresistance. Since the recent entry of extracellular vesicles (EVs), cell-derived nanostructures, have opened new avenues of investigation into this field, which, together with other more conventionally studied signaling pathways, play an important role in cell-to-cell communication. Thus, this review further explores the subject of EVs in-depth. This review also discusses possible future biomaterial and biomaterial-EV-based models that could be used to study the tumor microenvironment (TME) and will provide possible therapeutic approaches. Finally, this review concludes with potential perspectives and conclusions in this area.
Collapse
Affiliation(s)
- Ganesh Ingavle
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| | - Madhurima Das
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| |
Collapse
|
5
|
Kyykallio H, Faria AVS, Hartman R, Capra J, Rilla K, Siljander PR. A quick pipeline for the isolation of 3D cell culture-derived extracellular vesicles. J Extracell Vesicles 2022; 11:e12273. [PMID: 36257915 PMCID: PMC9579059 DOI: 10.1002/jev2.12273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 08/10/2021] [Accepted: 09/27/2022] [Indexed: 11/12/2022] Open
Abstract
Recent advances in cell biology research regarding extracellular vesicles have highlighted an increasing demand to obtain 3D cell culture-derived EVs, because they are considered to more accurately represent EVs obtained in vivo. However, there is still a grave need for efficient and tunable methodologies to isolate EVs from 3D cell cultures. Using nanofibrillar cellulose (NFC) scaffold as a 3D cell culture matrix, we developed a pipeline of two different approaches for EV isolation from cancer spheroids. A batch method was created for delivering high EV yield at the end of the culture period, and a harvesting method was created to enable time-dependent collection of EVs to combine EV profiling with spheroid development. Both these methods were easy to set up, quick to perform, and they provided a high EV yield. When compared to scaffold-free 3D spheroid cultures on ultra-low affinity plates, the NFC method resulted in similar EV production/cell, but the NFC method was scalable and easier to perform resulting in high EV yields. In summary, we introduce here an NFC-based, innovative pipeline for acquiring EVs from 3D cancer spheroids, which can be tailored to support the needs of variable EV research objectives.
Collapse
Affiliation(s)
- Heikki Kyykallio
- Institute of BiomedicineUniversity of Eastern FinlandKuopioFinland
| | - Alessandra V. S. Faria
- EV GroupMolecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Rosabella Hartman
- EV GroupMolecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Janne Capra
- Institute of BiomedicineUniversity of Eastern FinlandKuopioFinland
| | - Kirsi Rilla
- Institute of BiomedicineUniversity of Eastern FinlandKuopioFinland
| | - Pia R‐M Siljander
- EV GroupMolecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
6
|
Monti N, Verna R, Piombarolo A, Querqui A, Bizzarri M, Fedeli V. Paradoxical Behavior of Oncogenes Undermines the Somatic Mutation Theory. Biomolecules 2022; 12:662. [PMID: 35625590 PMCID: PMC9138429 DOI: 10.3390/biom12050662] [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: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
The currently accepted theory on the influence of DNA mutations on carcinogenesis (the Somatic Mutation Theory, SMT) is facing an increasing number of controversial results that undermine the explanatory power of mutated genes considered as "causative" factors. Intriguing results have demonstrated that several critical genes may act differently, as oncogenes or tumor suppressors, while phenotypic reversion of cancerous cells/tissues can be achieved by modifying the microenvironment, the mutations they are carrying notwithstanding. Furthermore, a high burden of mutations has been identified in many non-cancerous tissues without any apparent pathological consequence. All things considered, a relevant body of unexplained inconsistencies calls for an in depth rewiring of our theoretical models. Ignoring these paradoxes is no longer sustainable. By avoiding these conundrums, the scientific community will deprive itself of the opportunity to achieve real progress in this important biomedical field. To remedy this situation, we need to embrace new theoretical perspectives, taking the cell-microenvironment interplay as the privileged pathogenetic level of observation, and by assuming new explanatory models based on truly different premises. New theoretical frameworks dawned in the last two decades principally focus on the complex interaction between cells and their microenvironment, which is thought to be the critical level from which carcinogenesis arises. Indeed, both molecular and biophysical components of the stroma can dramatically drive cell fate commitment and cell outcome in opposite directions, even in the presence of the same stimulus. Therefore, such a novel approach can help in solving apparently inextricable paradoxes that are increasingly observed in cancer biology.
Collapse
Affiliation(s)
| | | | | | | | | | - Valeria Fedeli
- Systems Biology Group Lab, Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (N.M.); (R.V.); (A.P.); (A.Q.); (M.B.)
| |
Collapse
|
7
|
Fedeli V, Cucina A, Dinicola S, Fabrizi G, Catizone A, Gesualdi L, Ceccarelli S, Harrath AH, Alwasel SH, Ricci G, Pedata P, Bizzarri M, Monti N. Microgravity Modifies the Phenotype of Fibroblast and Promotes Remodeling of the Fibroblast-Keratinocyte Interaction in a 3D Co-Culture Model. Int J Mol Sci 2022; 23:ijms23042163. [PMID: 35216279 PMCID: PMC8879576 DOI: 10.3390/ijms23042163] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Microgravity impairs tissue organization and critical pathways involved in the cell–microenvironment interplay, where fibroblasts have a critical role. We exposed dermal fibroblasts to simulated microgravity by means of a Random Positioning Machine (RPM), a device that reproduces conditions of weightlessness. Molecular and structural changes were analyzed and compared to control samples growing in a normal gravity field. Simulated microgravity impairs fibroblast conversion into myofibroblast and inhibits their migratory properties. Consequently, the normal interplay between fibroblasts and keratinocytes were remarkably altered in 3D co-culture experiments, giving rise to several ultra-structural abnormalities. Such phenotypic changes are associated with down-regulation of α-SMA that translocate in the nucleoplasm, altogether with the concomitant modification of the actin-vinculin apparatus. Noticeably, the stress associated with weightlessness induced oxidative damage, which seemed to concur with such modifications. These findings disclose new opportunities to establish antioxidant strategies that counteract the microgravity-induced disruptive effects on fibroblasts and tissue organization.
Collapse
Affiliation(s)
- Valeria Fedeli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
| | - Alessandra Cucina
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via Antonio Scarpa 14, 00161 Rome, Italy;
- Policlinico Umberto I, Viale del Policlinico 155, 00161 Rome, Italy
| | - Simona Dinicola
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via Antonio Scarpa 14, 00161 Rome, Italy;
| | - Gianmarco Fabrizi
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via Antonio Scarpa 14, 00161 Rome, Italy;
| | - Angela Catizone
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (A.C.); (L.G.)
| | - Luisa Gesualdi
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (A.C.); (L.G.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.H.); (S.H.A.)
| | - Saleh H. Alwasel
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.H.); (S.H.A.)
| | - Giulia Ricci
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Paola Pedata
- Department of Medicine, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy;
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
| | - Noemi Monti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
- Correspondence:
| |
Collapse
|
8
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int J Mol Sci 2021; 22:12200. [PMID: 34830082 PMCID: PMC8618305 DOI: 10.3390/ijms222212200] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 01/09/2023] Open
Abstract
The traditional two-dimensional (2D) in vitro cell culture system (on a flat support) has long been used in cancer research. However, this system cannot be fully translated into clinical trials to ideally represent physiological conditions. This culture cannot mimic the natural tumor microenvironment due to the lack of cellular communication (cell-cell) and interaction (cell-cell and cell-matrix). To overcome these limitations, three-dimensional (3D) culture systems are increasingly developed in research and have become essential for tumor research, tissue engineering, and basic biology research. 3D culture has received much attention in the field of biomedicine due to its ability to mimic tissue structure and function. The 3D matrix presents a highly dynamic framework where its components are deposited, degraded, or modified to delineate functions and provide a platform where cells attach to perform their specific functions, including adhesion, proliferation, communication, and apoptosis. So far, various types of models belong to this culture: either the culture based on natural or synthetic adherent matrices used to design 3D scaffolds as biomaterials to form a 3D matrix or based on non-adherent and/or matrix-free matrices to form the spheroids. In this review, we first summarize a comparison between 2D and 3D cultures. Then, we focus on the different components of the natural extracellular matrix that can be used as supports in 3D culture. Then we detail different types of natural supports such as matrigel, hydrogels, hard supports, and different synthetic strategies of 3D matrices such as lyophilization, electrospiding, stereolithography, microfluid by citing the advantages and disadvantages of each of them. Finally, we summarize the different methods of generating normal and tumor spheroids, citing their respective advantages and disadvantages in order to obtain an ideal 3D model (matrix) that retains the following characteristics: better biocompatibility, good mechanical properties corresponding to the tumor tissue, degradability, controllable microstructure and chemical components like the tumor tissue, favorable nutrient exchange and easy separation of the cells from the matrix.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
9
|
Tissue Imaging and Quantification Relying on Endogenous Contrast. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 3233:257-288. [PMID: 34053031 DOI: 10.1007/978-981-15-7627-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Cell-matrix interactions play an important role in regulating a variety of essential processes in multicellular organisms, and are closely associated with numerous diseases. Modified interactions have major effects upon key features of both cells and extracellular matrix (ECM), and a thorough understanding of changes in these features can lead to critically important insights of diseases as well as the identification of effective therapeutic targets. Here, we summarize recent advances in quantitative, optical imaging of cellular metabolism and ECM spatial organization using endogenous sources of contrast. Specifically, we focus on the two-photon excited fluorescence (TPEF) imaging of autofluorescent cellular coenzymes, NAD(P)H and FAD, for the extraction of metabolic information described by optical biomarkers including cellular redox state, NAD(P)H fluorescence lifetime, and mitochondrial clustering. We show representative applications in assessing adipose tissue function and detecting malignant lesions in human skin, and further demonstrate that a combination of these optical metrics can provide complementary insights into the underlying biological mechanisms. In addition, we review the development of quantitative analysis methods to extract spatial orientation and organization metrics of collagen fibers, a major ECM component, and demonstrate applications of these approaches in two and three dimensions in several diseases, including would healing, osteoarthritis and cancer, as well as assessments of matrix remodeling in hormone-regulated engineered breast tissues. Finally, we summarize this chapter and discuss important research directions that we expect will evolve in the near future.
Collapse
|
10
|
Tripathi A, Kashyap A, Tripathi G, Yadav J, Bibban R, Aggarwal N, Thakur K, Chhokar A, Jadli M, Sah AK, Verma Y, Zayed H, Husain A, Bharti AC, Kashyap MK. Tumor reversion: a dream or a reality. Biomark Res 2021; 9:31. [PMID: 33958005 PMCID: PMC8101112 DOI: 10.1186/s40364-021-00280-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Reversion of tumor to a normal differentiated cell once considered a dream is now at the brink of becoming a reality. Different layers of molecules/events such as microRNAs, transcription factors, alternative RNA splicing, post-transcriptional, post-translational modifications, availability of proteomics, genomics editing tools, and chemical biology approaches gave hope to manipulation of cancer cells reversion to a normal cell phenotype as evidences are subtle but definitive. Regardless of the advancement, there is a long way to go, as customized techniques are required to be fine-tuned with precision to attain more insights into tumor reversion. Tumor regression models using available genome-editing methods, followed by in vitro and in vivo proteomics profiling techniques show early evidence. This review summarizes tumor reversion developments, present issues, and unaddressed challenges that remained in the uncharted territory to modulate cellular machinery for tumor reversion towards therapeutic purposes successfully. Ongoing research reaffirms the potential promises of understanding the mechanism of tumor reversion and required refinement that is warranted in vitro and in vivo models of tumor reversion, and the potential translation of these into cancer therapy. Furthermore, therapeutic compounds were reported to induce phenotypic changes in cancer cells into normal cells, which will contribute in understanding the mechanism of tumor reversion. Altogether, the efforts collectively suggest that tumor reversion will likely reveal a new wave of therapeutic discoveries that will significantly impact clinical practice in cancer therapy.
Collapse
Affiliation(s)
- Avantika Tripathi
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon, Haryana, Manesar (Gurugram), -122413, India
| | - Anjali Kashyap
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, India
| | - Greesham Tripathi
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon, Haryana, Manesar (Gurugram), -122413, India
| | - Joni Yadav
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India
| | - Rakhi Bibban
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India
| | - Nikita Aggarwal
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India
| | - Kulbhushan Thakur
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India
| | - Arun Chhokar
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India
| | - Mohit Jadli
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India
| | - Ashok Kumar Sah
- Department of Medical Laboratory Technology, Amity Medical School, Amity University Haryana, Panchgaon, Haryana, Manesar (Gurugram), India
- Department of Pathology and Laboratory Medicine, Medanta-The Medicity, Haryana, Gurugram, India
| | - Yeshvandra Verma
- Department of Toxicology, C C S University, Meerut, UP, 250004, India
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Amjad Husain
- Centre for Science & Society, Indian Institute of Science Education and Research (IISER), Bhopal, India
- Innovation and Incubation Centre for Entrepreneurship (IICE), Indian Institute of Science Education and Research (IISER), Bhopal, India
| | - Alok Chandra Bharti
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India.
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon, Haryana, Manesar (Gurugram), -122413, India.
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), New Delhi, 110007, India.
| |
Collapse
|
11
|
Abreu TR, Biscaia M, Gonçalves N, Fonseca NA, Moreira JN. In Vitro and In Vivo Tumor Models for the Evaluation of Anticancer Nanoparticles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:271-299. [PMID: 33543464 DOI: 10.1007/978-3-030-58174-9_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multiple studies about tumor biology have revealed the determinant role of the tumor microenvironment in cancer progression, resulting from the dynamic interactions between tumor cells and surrounding stromal cells within the extracellular matrix. This malignant microenvironment highly impacts the efficacy of anticancer nanoparticles by displaying drug resistance mechanisms, as well as intrinsic physical and biochemical barriers, which hamper their intratumoral accumulation and biological activity.Currently, two-dimensional cell cultures are used as the initial screening method in vitro for testing cytotoxic nanocarriers. However, this fails to mimic the tumor heterogeneity, as well as the three-dimensional tumor architecture and pathophysiological barriers, leading to an inaccurate pharmacological evaluation.Biomimetic 3D in vitro tumor models, on the other hand, are emerging as promising tools for more accurately assessing nanoparticle activity, owing to their ability to recapitulate certain features of the tumor microenvironment and thus provide mechanistic insights into nanocarrier intratumoral penetration and diffusion rates.Notwithstanding, in vivo validation of nanomedicines remains irreplaceable at the preclinical stage, and a vast variety of more advanced in vivo tumor models is currently available. Such complex animal models (e.g., genetically engineered mice and patient-derived xenografts) are capable of better predicting nanocarrier clinical efficiency, as they closely resemble the heterogeneity of the human tumor microenvironment.Herein, the development of physiologically more relevant in vitro and in vivo tumor models for the preclinical evaluation of anticancer nanoparticles will be discussed, as well as the current limitations and future challenges in clinical translation.
Collapse
Affiliation(s)
- Teresa R Abreu
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal.,UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Mariana Biscaia
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal
| | - Nélio Gonçalves
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal
| | - Nuno A Fonseca
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal.,TREAT U, SA, Parque Industrial de Taveiro, Lote 44, Coimbra, Portugal
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal. .,UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal.
| |
Collapse
|
12
|
Environmental control of mammary carcinoma cell expansion by acidification and spheroid formation in vitro. Sci Rep 2020; 10:21959. [PMID: 33319820 PMCID: PMC7738540 DOI: 10.1038/s41598-020-78989-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the leading cause of cancer death among women worldwide. Like other cancers, mammary carcinoma progression involves acidification of the tumor microenvironment, which is an important factor for cancer detection and treatment strategies. However, the effects of acidity on mammary carcinoma cell morphology and phenotype have not been thoroughly characterized. Here, we evaluated fundamental effects of environmental acidification on mammary carcinoma cells in standard two-dimensional cultures and three-dimensional spheroids. Acidification decreased overall mammary carcinoma cell viability, while increasing their resistance to the anthracycline doxorubicin. Environmental acidification also increased extracellular vesicle production by mammary carcinoma cells. Conditioned media containing these vesicles appeared to increase fibroblast motility. Acidification also increased mammary carcinoma cell motility when cultured with fibroblasts in spheroids. Taken together, results from this study suggest that environmental acidification induces drug resistance and extracellular vesicle production by mammary carcinoma cells that promote tumor expansion.
Collapse
|
13
|
Wei D, Li C, Ye J, Xiang F, Liu J. Extracellular Collagen Mediates Osteosarcoma Progression Through an Integrin α2β1/JAK/STAT3 Signaling Pathway. Cancer Manag Res 2020; 12:12067-12075. [PMID: 33262655 PMCID: PMC7699996 DOI: 10.2147/cmar.s273466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background Osteosarcoma development is a complex set which is determined by various factors. Many patients suffered from sustained osteosarcoma growth and revealed poor response to clinical interventions. However, the underlying mechanisms of osteosarcoma development still remain unclear. Methods In our study, we isolated osteosarcoma tissues from clinical patients, which were divided into high degree group (stage G1~G2) and low degree group (stage G0). The expression of type I collagen, integrin and STAT3 in tumor tissues were analyzed by immunohistochemistry or immunofluorescence. The collagen-induced cells proliferation was detected by CCK8 and colony formation analysis. The activation of JAK/STAT3 signal was examined by Western blotting and immunofluorescence. The anticancer effects of integrin α2β1 peptide were analyzed by Sao-2-bearing mice model. Results Our results implicated that type I collagen could facilitate malignant osteosarcoma development in patients. In vitro, 2D collagen culture also efficiently mediated the stemness up-regulation of osteosarcoma cells, resulting in the strengthened capability of cells proliferation and tumorigenesis. In mechanism, we found that type I collagen could facilitate the activation of JAK/STAT3 signals through integrin α2β1, which elicited tumor-sustained growth and cancer relapse. In tumor-bearing mice model, integrin α2β1 signals inhibitor significantly suppressed the osteosarcoma cells proliferation and their tumorigenic ability, which improved the outcome of chemotherapy/radiotherapy. Conclusion Our study demonstrated that type I collagen could mediate osteosarcoma development through an integrin α2β1/JAK/STAT3 signaling pathway. Blockade of integrin α2β1 by α2β1 inhibitor efficiently improved outcome of chemotherapy/radiotherapy, which provided new insights for eradicating tumors in clinic.
Collapse
Affiliation(s)
- Daiqing Wei
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, Sichuan, People's Republic of China
| | - Cui Li
- Department of Nosocomial Infection Control, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Junwu Ye
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, Sichuan, People's Republic of China
| | - Feifan Xiang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, Sichuan, People's Republic of China
| | - Juncai Liu
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, Sichuan, People's Republic of China
| |
Collapse
|
14
|
Girigoswami K, Saini D, Girigoswami A. Extracellular Matrix Remodeling and Development of Cancer. Stem Cell Rev Rep 2020; 17:739-747. [PMID: 33128168 DOI: 10.1007/s12015-020-10070-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2020] [Indexed: 12/21/2022]
Abstract
The importance of stem cell growth and its fate is highly essential for the use of stem cells in therapy and regeneration. There are conflicting evidences regarding the actual role of stem cells when injected into a patient towards damage recovery and its lifespan inside the body. Tumor microenvironment differs from that of normal cells and may have a role in the growth of stem cells when associated with them. In cancer, the uncontrolled growth of cells remodels the extracellular matrix (ECM). The ECM alteration occurs as the mutated fibroblast cells release growth factors into the ECM which further alters the ECM directly or changes the epithelial cells and then alters the ECM. In this review we will discuss about the components and functions of ECM and how does it differ in cancer cells compared to normal cells. Abnormal dynamics of the ECM and its role in cancer progression will also be discussed. Graphical abstract.
Collapse
Affiliation(s)
- Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India.
| | - Devender Saini
- Tissue Engineering and Regenerative Medicine, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, 603103, Tamilnadu, India
| |
Collapse
|
15
|
Proietti S, Cucina A, Pensotti A, Fuso A, Marchese C, Nicolini A, Bizzarri M. Tumor reversion and embryo morphogenetic factors. Semin Cancer Biol 2020; 79:83-90. [DOI: 10.1016/j.semcancer.2020.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 07/09/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
|
16
|
PIAS1 and TIF1γ collaborate to promote SnoN SUMOylation and suppression of epithelial-mesenchymal transition. Cell Death Differ 2020; 28:267-282. [PMID: 32770107 DOI: 10.1038/s41418-020-0599-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023] Open
Abstract
SUMO E3 ligases specify protein substrates for SUMOylation. The SUMO E3 ligases PIAS1 and TIF1γ target the transcriptional regulator SnoN for SUMOylation leading to suppression of epithelial-mesenchymal transition (EMT). Whether and how TIF1γ and PIAS1 might coordinate SnoN SUMOylation and regulation of EMT remained unknown. Here, we reveal that SnoN associates simultaneously with both TIF1γ and PIAS1, leading to a trimeric protein complex. Hence, PIAS1 and TIF1γ collaborate to promote the SUMOylation of SnoN. Importantly, loss of function studies of PIAS1 and TIF1γ suggest that these E3 ligases act in an interdependent manner to suppress EMT of breast cell-derived tissue organoids. Collectively, our findings unveil a novel mechanism by which SUMO E3 ligases coordinate substrate SUMOylation with biological implications.
Collapse
|
17
|
Integrating the Tumor Microenvironment into Cancer Therapy. Cancers (Basel) 2020; 12:cancers12061677. [PMID: 32599891 PMCID: PMC7352326 DOI: 10.3390/cancers12061677] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor progression is mediated by reciprocal interaction between tumor cells and their surrounding tumor microenvironment (TME), which among other factors encompasses the extracellular milieu, immune cells, fibroblasts, and the vascular system. However, the complexity of cancer goes beyond the local interaction of tumor cells with their microenvironment. We are on the path to understanding cancer from a systemic viewpoint where the host macroenvironment also plays a crucial role in determining tumor progression. Indeed, growing evidence is emerging on the impact of the gut microbiota, metabolism, biomechanics, and the neuroimmunological axis on cancer. Thus, external factors capable of influencing the entire body system, such as emotional stress, surgery, or psychosocial factors, must be taken into consideration for enhanced management and treatment of cancer patients. In this article, we review prognostic and predictive biomarkers, as well as their potential evaluation and quantitative analysis. Our overarching aim is to open up new fields of study and intervention possibilities, within the framework of an integral vision of cancer as a functional tissue with the capacity to respond to different non-cytotoxic factors, hormonal, immunological, and mechanical forces, and others inducing stroma and tumor reprogramming.
Collapse
|
18
|
3D-microenvironments initiate TCF4 expression rescuing nuclear β-catenin activity in MCF-7 breast cancer cells. Acta Biomater 2020; 103:153-164. [PMID: 31843716 DOI: 10.1016/j.actbio.2019.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/20/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
Mechanical cues sensed by tumor cells in their microenvironment can influence important mechanisms including adhesion, invasion and proliferation. However, a common mechanosensitive protein and/or pathway can be regulated in different ways among diverse types of tumors. Of particular interest are human breast epithelial cancers, which markedly exhibit a heterogeneous pattern of nuclear β-catenin localization, a protein known to be involved in both mechanotransduction and tumorigenesis. β-catenin can be aberrantly accumulated in the nucleus wherein it binds to and activates lymphoid enhancer factor/T cell factor (LEF/TCF) transcription factors. At present, little is known about how mechanical cues are integrated into breast cancer cells harboring impaired mechanisms of β-catenin's nuclear uptake and/or retention. This prompted us to investigate the influence of mechanical cues on MCF-7 human breast cancer cells which are known to fail in relocating β-catenin into the nucleus due to very low baseline levels of LEF/TCFs. Exploiting three-dimensional (3D) microscaffolds realized by two-photon lithography, we show that surrounding MCF-7 cells have not only a nuclear pool of β-catenin, but also rescue from their defective expression of TCF4 and boost invasiveness. Together with heightened amounts of vimentin, a β-catenin/TCF-target gene regulator of proliferation and invasiveness, such 3D-elicited changes indicate an epithelial-to-mesenchymal phenotypic switch of MCF-7 cells. This is also consistent with an increased in situ MCF-7 cell proliferation that can be abrogated by blocking β-catenin/TCF-transcription activity. Collectively, these data suggest that 3D microenvironments are per se sufficient to prime a TCF4-dependent rescuing of β-catenin nuclear activity in MCF-7 cells. The employed methodology could, therefore, provide a mechanism-based rationale to dissect further aspects of mechanotranscription in breast cancerogenesis, somewhat independent of β-catenin's nuclear accumulation. More importantly, by considering the heterogeneity of β-catenin signaling pathway in breast cancer patients, these data may open alternative avenues for personalized disease management and prevention. STATEMENT OF SIGNIFICANCE: Mechanical cues play a critical role in cancer pathogenesis. Little is known about their influence in breast cancer cells harboring impaired mechanisms of β-catenin's nuclear uptake and/or retention, involved in both mechanotransduction and tumorigenesis. We engineered 3D scaffold, by two-photon lithography, to study the influence of mechanical cues on MCF-7 cells which are known to fail in relocating β-catenin into the nucleus. We found that 3D microenvironments are per se sufficient to prime a TCF4-dependent rescuing of β-catenin nuclear activity that boost cell proliferation and invasiveness. Thus, let us suggest that our system could provide a mechanism-based rationale to further dissect key aspects of mechanotranscription in breast cancerogenesis and progression, somewhat independent of β-catenin's nuclear accumulation.
Collapse
|
19
|
Thong T, Forté CA, Hill EM, Colacino JA. Environmental exposures, stem cells, and cancer. Pharmacol Ther 2019; 204:107398. [PMID: 31376432 PMCID: PMC6881547 DOI: 10.1016/j.pharmthera.2019.107398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022]
Abstract
An estimated 70-90% of all cancers are linked to exposure to environmental risk factors. In parallel, the number of stem cells in a tissue has been shown to be a strong predictor of risk of developing cancer in that tissue. Tumors themselves are characterized by an acquisition of "stem cell" characteristics, and a growing body of evidence points to tumors themselves being sustained and propagated by a stem cell-like population. Here, we review our understanding of the interplay between environmental exposures, stem cell biology, and cancer. We provide an overview of the role of stem cells in development, tissue homeostasis, and wound repair. We discuss the pathways and mechanisms governing stem cell plasticity and regulation of the stem cell state, and describe experimental methods for assessment of stem cells. We then review the current understanding of how environmental exposures impact stem cell function relevant to carcinogenesis and cancer prevention, with a focus on environmental and occupational exposures to chemical, physical, and biological hazards. We also highlight key areas for future research in this area, including defining whether the biological basis for cancer disparities is related to effects of complex exposure mixtures on stem cell biology.
Collapse
Affiliation(s)
- Tasha Thong
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Chanese A Forté
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Michigan Institute for Computational Discovery and Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evan M Hill
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
20
|
Zhang Y, Ouyang M, Ray A, Liu T, Kong J, Bai B, Kim D, Guziak A, Luo Y, Feizi A, Tsai K, Duan Z, Liu X, Kim D, Cheung C, Yalcin S, Ceylan Koydemir H, Garner OB, Di Carlo D, Ozcan A. Computational cytometer based on magnetically modulated coherent imaging and deep learning. LIGHT, SCIENCE & APPLICATIONS 2019; 8:91. [PMID: 31645935 PMCID: PMC6804677 DOI: 10.1038/s41377-019-0203-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 05/08/2023]
Abstract
Detecting rare cells within blood has numerous applications in disease diagnostics. Existing rare cell detection techniques are typically hindered by their high cost and low throughput. Here, we present a computational cytometer based on magnetically modulated lensless speckle imaging, which introduces oscillatory motion to the magnetic-bead-conjugated rare cells of interest through a periodic magnetic force and uses lensless time-resolved holographic speckle imaging to rapidly detect the target cells in three dimensions (3D). In addition to using cell-specific antibodies to magnetically label target cells, detection specificity is further enhanced through a deep-learning-based classifier that is based on a densely connected pseudo-3D convolutional neural network (P3D CNN), which automatically detects rare cells of interest based on their spatio-temporal features under a controlled magnetic force. To demonstrate the performance of this technique, we built a high-throughput, compact and cost-effective prototype for detecting MCF7 cancer cells spiked in whole blood samples. Through serial dilution experiments, we quantified the limit of detection (LoD) as 10 cells per millilitre of whole blood, which could be further improved through multiplexing parallel imaging channels within the same instrument. This compact, cost-effective and high-throughput computational cytometer can potentially be used for rare cell detection and quantification in bodily fluids for a variety of biomedical applications.
Collapse
Affiliation(s)
- Yibo Zhang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Mengxing Ouyang
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Aniruddha Ray
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Department of Physics and Astronomy, University of Toledo, Toledo, OH 43606 USA
| | - Tairan Liu
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Janay Kong
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Bijie Bai
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Donghyuk Kim
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Alexander Guziak
- Department of Physics and Astronomy, University of California, Los Angeles, CA 90095 USA
| | - Yi Luo
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Alborz Feizi
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Yale School of Medicine, New Haven, CT 06510 USA
| | - Katherine Tsai
- Department of Biochemistry, University of California, Los Angeles, CA 90095 USA
| | - Zhuoran Duan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
| | - Xuewei Liu
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
| | - Danny Kim
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Chloe Cheung
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Sener Yalcin
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
| | - Hatice Ceylan Koydemir
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Omai B. Garner
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095 USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095 USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095 USA
| | - Aydogan Ozcan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
21
|
Cytotoxic and Antiproliferative Effects of Preussin, a Hydroxypyrrolidine Derivative from the Marine Sponge-Associated Fungus Aspergillus candidus KUFA 0062, in a Panel of Breast Cancer Cell Lines and Using 2D and 3D Cultures. Mar Drugs 2019; 17:md17080448. [PMID: 31366127 PMCID: PMC6722565 DOI: 10.3390/md17080448] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 07/26/2019] [Accepted: 07/27/2019] [Indexed: 12/28/2022] Open
Abstract
Preussin, a hydroxyl pyrrolidine derivative isolated from the marine sponge-associated fungus Aspergillus candidus KUFA 0062, displayed anticancer effects in some cancer cell lines, including MCF7. Preussin was investigated for its cytotoxic and antiproliferative effects in breast cancer cell lines (MCF7, SKBR3, and MDA-MB-231), representatives of major breast cancers subtypes, and in a non-tumor cell line (MCF12A). Preussin was first tested in 2D (monolayer), and then in 3D (multicellular aggregates), cultures, using a multi-endpoint approach for cytotoxicity (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), resazurin and lactate dehydrogenase (LDH)) and proliferative (5-bromo-2'-deoxyuridine (BrdU)) assays, as well as the analysis of cell morphology by optical/electron microscopy and immunocytochemistry for caspase-3 and ki67. Preussin affected cell viability and proliferation in 2D and 3D cultures in all cell lines tested. The results in the 3D culture showed the same tendency as in the 2D culture, however, cells in the 3D culture were less responsive. The effects were observed at different concentrations of preussin, depending on the cell line and assay method. Morphological study of preussin-exposed cells revealed cell death, which was confirmed by caspase-3 immunostaining. In view of the data, we recommend a multi-endpoint approach, including histological evaluation, in future assays with the tested 3D models. Our data showed cytotoxic and antiproliferative activities of preussin in breast cancer cell lines in 2D and 3D cultures, warranting further studies for its anticancer potential.
Collapse
|
22
|
Desyatnik I, Krasner M, Frolov L, Ronen M, Guy O, Wasserman D, Tzur A, Avrahami D, Barbiro-Michaely E, Gerber D. An Integrated Microfluidics Approach for Personalized Cancer Drug Sensitivity and Resistance Assay. ACTA ACUST UNITED AC 2019; 3:e1900001. [PMID: 32648689 DOI: 10.1002/adbi.201900001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/04/2019] [Indexed: 12/22/2022]
Abstract
Cancer is the second leading cause of death globally. Matching proper treatment and dosage is crucial for a positive outcome. Any given drug may affect patients with similar tumors differently. Personalized medicine aims to address this issue. Unfortunately, most cancer samples cannot be expanded in culture, limiting conventional cell-based testing. Herein, presented is a microfluidic device that combines a drug microarray with cell microscopy. The device can perform 512 experiments to test chemosensitivity and resistance to a drug array. MCF7 and 293T cells are cultured inside the device and their chemosensitivity and resistance to docetaxel, applied at various concentrations, are determined. Cell mortality is determined as a function of drug concentration and exposure time. It is found that both cell types form cluster morphology within the device, not evident in conventional tissue culture under similar conditions. Cells inside the clusters are less sensitive to drugs than dispersed cells. These findings support a heterogenous response of cancer cells to drugs. Then demonstrated is the principle of drug microarrays by testing cell response to four different drugs at four different concentrations. This approach may enable the personalization of treatment to the particular tumor and patient and may eventually improve final patient outcome.
Collapse
Affiliation(s)
- Inna Desyatnik
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Matan Krasner
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Ludmila Frolov
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Maria Ronen
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Ortal Guy
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Danit Wasserman
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Amit Tzur
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Dorit Avrahami
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Efrat Barbiro-Michaely
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Doron Gerber
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, 52900, Israel
| |
Collapse
|
23
|
Proietti S, Cucina A, Pensotti A, Biava PM, Minini M, Monti N, Catizone A, Ricci G, Leonetti E, Harrath AH, Alwasel SH, Bizzarri M. Active Fraction from Embryo Fish Extracts Induces Reversion of the Malignant Invasive Phenotype in Breast Cancer through Down-regulation of TCTP and Modulation of E-cadherin/β-catenin Pathway. Int J Mol Sci 2019; 20:E2151. [PMID: 31052313 PMCID: PMC6539734 DOI: 10.3390/ijms20092151] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 12/01/2022] Open
Abstract
Some yet unidentified factors released by both oocyte and embryonic microenvironments demonstrated to be non-permissive for tumor development and display the remarkable ability to foster cell/tissue reprogramming, thus ultimately reversing the malignant phenotype. In the present study we observed how molecular factors extracted from Zebrafish embryos during specific developmental phases (20 somites) significantly antagonize proliferation of breast cancer cells, while reversing a number of prominent aspects of malignancy. Embryo extracts reduce cell proliferation, enhance apoptosis, and dramatically inhibit both invasiveness and migrating capabilities of cancer cells. Counteracting the invasive phenotype is a relevant issue in controlling tumor spreading and metastasis. Moreover, such effect is not limited to cancerous cells as embryo extracts were also effective in inhibiting migration and invasiveness displayed by normal breast cells undergoing epithelial-mesenchymal transition upon TGF-β1 stimulation. The reversion program involves the modulation of E-cadherin/β-catenin pathway, cytoskeleton remodeling with dramatic reduction in vinculin, as well as downregulation of TCTP and the concomitant increase in p53 levels. Our findings highlight that-contrary to the prevailing current "dogma", which posits that neoplastic cells are irreversibly "committed"-the malignant phenotype can ultimately be "reversed", at least partially, in response to environmental morphogenetic influences.
Collapse
Affiliation(s)
- Sara Proietti
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
| | - Alessandra Cucina
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
- Azienda Policlinico Umberto I, 00161 Rome, Italy.
| | | | - Pier Mario Biava
- Scientific Institute of Research and Health Care (IRCCS) Multimedica, 20099 Milano, Italy.
| | - Mirko Minini
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
- Department of Experimental Medicine, Sapienza University of Rome, Systems Biology Group Lab, 00161 Rome, Italy.
| | - Noemi Monti
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
| | - Angela Catizone
- Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy.
| | - Giulia Ricci
- Department. of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Erica Leonetti
- Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy.
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh 2455, Saudi Arabia.
| | - Saleh H Alwasel
- Department of Zoology, College of Science, King Saud University, Riyadh 2455, Saudi Arabia.
| | - Mariano Bizzarri
- Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy.
| |
Collapse
|
24
|
Weng Q, Zhou L, Xia L, Zheng Y, Zhang X, Li F, Li Q. In vitro evaluation of FL118 and 9-Q20 cytotoxicity and cellular uptake in 2D and 3D different cell models. Cancer Chemother Pharmacol 2019; 84:527-537. [DOI: 10.1007/s00280-019-03846-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/22/2019] [Indexed: 12/29/2022]
|
25
|
Dzobo K, Senthebane DA, Thomford NE, Rowe A, Dandara C, Parker MI. Not Everyone Fits the Mold: Intratumor and Intertumor Heterogeneity and Innovative Cancer Drug Design and Development. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:17-34. [PMID: 29356626 DOI: 10.1089/omi.2017.0174] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Disruptive innovations in medicine are game-changing in nature and bring about radical shifts in the way we understand human diseases, their treatment, and/or prevention. Yet, disruptive innovations in cancer drug design and development are still limited. Therapies that cure all cancer patients are in short supply or do not exist at all. Chief among the causes of this predicament is drug resistance, a mechanism that is much more dynamic than previously understood. Drug resistance has limited the initial success experienced with biomarker-guided targeted therapies as well. A major contributor to drug resistance is intratumor heterogeneity. For example, within solid tumors, there are distinct subclones of cancer cells, presenting profound complexity to cancer treatment. Well-known contributors to intratumor heterogeneity are genomic instability, the microenvironment, cellular genotype, cell plasticity, and stochastic processes. This expert review explains that for oncology drug design and development to be more innovative, we need to take into account intratumor heterogeneity. Initially thought to be the preserve of cancer cells, recent evidence points to the highly heterogeneous nature and diverse locations of stromal cells, such as cancer-associated fibroblasts (CAFs) and cancer-associated macrophages (CAMs). Distinct subpopulations of CAFs and CAMs are now known to be located immediately adjacent and distant from cancer cells, with different subpopulations exerting different effects on cancer cells. Disruptive innovation and precision medicine in clinical oncology do not have to be a distant reality, but can potentially be achieved by targeting these spatially separated and exclusive cancer cell subclones and CAF subtypes. Finally, we emphasize that disruptive innovations in drug discovery and development will likely come from drugs whose effect is not necessarily tumor shrinkage.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Dimakatso Alice Senthebane
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Nicholas Ekow Thomford
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Arielle Rowe
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa
| | - Collet Dandara
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - M Iqbal Parker
- 2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| |
Collapse
|
26
|
Cell-seeded 3D scaffolds as in vitro models for electroporation. Bioelectrochemistry 2019; 125:15-24. [DOI: 10.1016/j.bioelechem.2018.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 07/19/2018] [Accepted: 08/24/2018] [Indexed: 12/12/2022]
|
27
|
Sapudom J, Kalbitzer L, Wu X, Martin S, Kroy K, Pompe T. Fibril bending stiffness of 3D collagen matrices instructs spreading and clustering of invasive and non-invasive breast cancer cells. Biomaterials 2018; 193:47-57. [PMID: 30554026 DOI: 10.1016/j.biomaterials.2018.12.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/07/2018] [Accepted: 12/08/2018] [Indexed: 12/12/2022]
Abstract
Extracellular matrix stiffening of breast tissues has been clinically correlated with malignant transformation and poor prognosis. An increase of collagen fibril diameter and lysyl-oxidase mediated crosslinking has been observed in advanced tumor stages. Many current reports suggest that the local mechanical properties of single fibrillar components dominantly regulate cancer cell behavior. Here, we demonstrate by an independent control of fibril diameter and intrafibrillar crosslinking of three-dimensional (3D) collagen matrices that fibril bending stiffness instructs cell behavior of invasive and non-invasive breast cancer cells. Two types of collagen matrices with fibril diameter of either 650 nm or 800 nm at a similar pore size of 10 μm were reconstituted and further modified with the zero-length crosslinker 1-ethyl-3-(3-dimethyl aminopropyl)-carbodiimide (EDC) at concentrations of 0, 20, 100 and 500 mM. This approach yields two sets of collagen matrices with overlapping variation of matrix elasticity. With these matrices we could prove the common assumption that matrix elasticity of collagen networks is bending dominated with a linear dependence on fibril bending stiffness. We derive that the measured variation of matrix elasticity is directly correlated to the variation of fibril bending stiffness, being independently controlled either by fibril diameter or by intrafibrillar crosslinking. We use these defined matrices to demonstrate that the adjustment of fibril bending stiffness allows to instruct the behavior of two different breast cancer cell lines, invasive MDA-MB-231 (human breast carcinoma) and non-invasive MCF-7 cells (human breast adenocarcinoma). Invasiveness and spreading of invasive MDA-MB-231 cells as well as clustering of non-invasive MCF-7 cells is thereby investigated over a broad parameter range. Our results demonstrate and quantify the direct dependence of cancer cell phenotypes on the matrix mechanical properties on the scale of single fibrils.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany; Department of Dermatology, Venerology and Allergology, University of Leipzig Medical Center, Leipzig, 04103, Germany
| | - Liv Kalbitzer
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany
| | - Xiancheng Wu
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany
| | - Steve Martin
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany
| | - Klaus Kroy
- Institute for Theoretical Physics, Leipzig University, Leipzig, 04009, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig, 04103, Germany.
| |
Collapse
|
28
|
Joyce MH, Lu C, James ER, Hegab R, Allen SC, Suggs LJ, Brock A. Phenotypic Basis for Matrix Stiffness-Dependent Chemoresistance of Breast Cancer Cells to Doxorubicin. Front Oncol 2018; 8:337. [PMID: 30234012 PMCID: PMC6134055 DOI: 10.3389/fonc.2018.00337] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
The persistence of drug resistant cell populations following chemotherapeutic treatment is a significant challenge in the clinical management of cancer. Resistant subpopulations arise via both cell intrinsic and extrinsic mechanisms. Extrinsic factors in the microenvironment, including neighboring cells, glycosaminoglycans, and fibrous proteins impact therapy response. Elevated levels of extracellular fibrous proteins are associated with tumor progression and cause the surrounding tissue to stiffen through changes in structure and composition of the extracellular matrix (ECM). We sought to determine how this progressively stiffening microenvironment affects the sensitivity of breast cancer cells to chemotherapeutic treatment. MDA-MB-231 triple negative breast carcinoma cells cultured in a 3D alginate-based hydrogel system displayed a stiffness-dependent response to the chemotherapeutic doxorubicin. MCF7 breast carcinoma cells cultured in the same conditions did not exhibit this stiffness-dependent resistance to the drug. This differential therapeutic response was coordinated with nuclear translocation of YAP, a marker of mesenchymal differentiation. The stiffness-dependent response was lost when cells were transferred from 3D to monolayer cultures, suggesting that endpoint ECM conditions largely govern the response to doxorubicin. To further examine this response, we utilized a platform capable of dynamic ECM stiffness modulation to allow for a change in matrix stiffness over time. We found that MDA-MB-231 cells have a stiffness-dependent resistance to doxorubicin and that duration of exposure to ECM stiffness is sufficient to modulate this response. These results indicate the need for additional tools to integrate mechanical stiffness with therapeutic response and inform decisions for more effective use of chemotherapeutics in the clinic.
Collapse
Affiliation(s)
- M. Hunter Joyce
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Carolyne Lu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Emily R. James
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Rachel Hegab
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA, United States
| | - Shane C. Allen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Laura J. Suggs
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| | - Amy Brock
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
29
|
Liu Z, Speroni L, Quinn KP, Alonzo C, Pouli D, Zhang Y, Stuntz E, Sonnenschein C, Soto AM, Georgakoudi I. 3D organizational mapping of collagen fibers elucidates matrix remodeling in a hormone-sensitive 3D breast tissue model. Biomaterials 2018; 179:96-108. [PMID: 29980078 DOI: 10.1016/j.biomaterials.2018.06.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/08/2018] [Accepted: 06/22/2018] [Indexed: 12/11/2022]
Abstract
Hormones play an important role in normal and diseased breast tissue development. However, they can also disrupt cell-matrix interactions and their role in extracellular matrix reorganization during epithelial morphogenesis remains poorly understood, partly due to a lack of sensitive approaches for matrix characterization. Here, we assess the hormonal regulation of matrix reorganization in a three-dimensional (3D) breast tissue culture model using a novel metric, i.e., 3D directional variance, to characterize the 3D organization of collagen fibers visualized via high-resolution, second harmonic generation imaging. This metric enables resolving and quantifying patterns of spatial organization throughout the matrix surrounding epithelial structures treated with 17β-estradiol (E2) alone, and E2 in combination with either promegestone, a progestogen, or prolactin. Addition of promegestone results in the most disorganized fibers, while the E2 alone treatment leads to the most organized ones. Location-dependent organization mapping indicates that only the prolactin treatment leads to significant heterogeneities in the regional organization of collagen fibers, with higher levels of alignment observed at the end of the elongated epithelial structures. The observed collagen organization patterns for all groups persist for tens of micrometers. In addition, a comparison between 3D directional variance and typical 2D analysis approaches reveals an improved sensitivity of the 3D metric to identify organizational heterogeneities and differences among treatment groups. These results demonstrate that 3D directional variance is sensitive to subtle changes in the extracellular micro-environment and has the potential to elucidate reciprocal cell-matrix interactions in the context of numerous applications involving the study of normal and diseased tissue morphogenesis.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lucia Speroni
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | - Carlo Alonzo
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Dimitra Pouli
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yang Zhang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Emily Stuntz
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Carlos Sonnenschein
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
30
|
Recapitulating spatiotemporal tumor heterogeneity in vitro through engineered breast cancer microtissues. Acta Biomater 2018; 73:236-249. [PMID: 29679778 DOI: 10.1016/j.actbio.2018.04.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 01/14/2023]
Abstract
Tumor and microenvironmental heterogeneity hinders the study of breast cancer biology and the assessment of therapeutic strategies, being associated with high variability and drug resistance. In this context, it is mandatory to develop three-dimensional breast tumor models able to reproduce this heterogeneity and the dynamic interaction occurring between tumor cells and microenvironment. Here we show a new breast cancer microtissue model (T-µTP) uniquely able to present intra-tumor morphological heterogeneity in a dynamic and responsive endogenous matrix. T-µTP consists of adenocarcinoma cells, endothelial cells and stromal fibroblasts. These three kinds of cells are totally embedded into an endogenous matrix which is rich in collagen and hyaluronic acid and it is directly produced by human fibroblasts. In this highly physiologically relevant environment, tumor cells evolve in different cluster morphologies recapitulating tumor spatiotemporal heterogeneity. Moreover they activate the desmoplastic and vascular reaction with affected collagen content, assembly and organization and the presence of aberrant capillary-like structures (CLS). Thus, T-µTP allows to outline main crucial events involved in breast cancer progression into a single model overcoming the limit of artificial extra cellular matrix surrogates. We strongly believe that T-µTP is a suitable model for the study of breast cancer and for drug screening assays following key parameters of clinical interest. STATEMENT OF SIGNIFICANCE Tumor and microenvironmental heterogeneity makes very hurdle to find a way to study and treat breast cancer. Here we develop an innovative 3D tumor microtissue model recapitulating in vitro tumor heterogeneity. Tumor microtissues are characterized by the activation of the stromal and vascular reaction too. We underline the importance to mimic different microenvironmental tumor features in the same time and in a single tissue in order to obtain a model of spatiotemporal tumor genesis and progression, suitable for the study of tumor treatment and resistance.
Collapse
|
31
|
Morgan MM, Livingston MK, Warrick JW, Stanek EM, Alarid ET, Beebe DJ, Johnson BP. Mammary fibroblasts reduce apoptosis and speed estrogen-induced hyperplasia in an organotypic MCF7-derived duct model. Sci Rep 2018; 8:7139. [PMID: 29740030 PMCID: PMC5940820 DOI: 10.1038/s41598-018-25461-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/13/2018] [Indexed: 12/22/2022] Open
Abstract
The estrogen receptor (ER) regulates the survival and growth of breast cancer cells, but it is less clear how components of the tissue microenvironment affect ER-mediated responses. We set out to test how human mammary fibroblasts (HMFs) modulate ER signaling and downstream cellular responses. We exposed an organotypic mammary model consisting of a collagen-embedded duct structure lined with MCF7 cells to 17-β estradiol (E2), with and without HMFs in the surrounding matrix. MCF7 cells grown as ductal structures were polarized and proliferated at rates comparable to in vivo breast tissue. In both culture platforms, exposure to E2 increased ER transactivation, increased proliferation, and induced ductal hyperplasia. When the surrounding matrix contained HMFs, the onset and severity of E2-induced ductal hyperplasia was increased due to decreased apoptosis. The reduced apoptosis may be due to fibroblasts modulating ER signaling in MCF7 cells, as suggested by the increased ER transactivation and reduced ER protein in MCF7 cells grown in co-culture. These findings demonstrate the utility of organotypic platforms when studying stromal:epithelial interactions, and add to existing literature that implicate the mammary microenvironment in ER + breast cancer progression.
Collapse
Affiliation(s)
- Molly M Morgan
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.,Department of Chemistry, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Jay W Warrick
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Eli M Stanek
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Elaine T Alarid
- Department of Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.
| |
Collapse
|
32
|
Sweeney MF, Sonnenschein C, Soto AM. Characterization of MCF-12A cell phenotype, response to estrogens, and growth in 3D. Cancer Cell Int 2018; 18:43. [PMID: 29559854 PMCID: PMC5859508 DOI: 10.1186/s12935-018-0534-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/05/2018] [Indexed: 01/28/2023] Open
Abstract
Background Three-dimensional cultures of mammary epithelial cells allow for biologically-relevant studies of the development of the mammary gland in rodents and humans under normal and pathological conditions, like carcinogenesis. Under these conditions, mammotropic hormones play significant roles in tissue morphogenesis. Therefore, a system that recreates the normal, hormonally responsive epithelium would be a valuable tool to study the normal state and its transition to carcinogenesis. MCF-12A cells have been claimed to be non-tumorigenic mammary epithelial cells with reported sensitivity to estrogens. In this study, we aimed at characterizing MCF-12A cells for use in a hormone-responsive 3D culture system to determine their usefulness as a tool to identify normal and abnormal microenvironmental cues. Methods MCF-12A cells were single-cell cloned in order to investigate their heterogeneous makeup. The parental cells were then treated with estradiol to investigate proliferative and transcriptional responses through the estrogen receptor alpha. Finally, parental cells and epithelial-like cell-derived clones were seeded in rat-tail collagen I to profile the morphogenesis of multicellular 3D structures. The resultant structures were then analyzed using unsupervised morphometric analysis. Results MCF-12A cells consist of epithelial-like colonies which shed elongated, freely growing cells on the colony's edges. The cells express E-cadherin as well as mesenchymal vimentin but do not express markers associated with myoepithelial cells or fibroblasts. Treatment with estradiol does not affect either the proliferation rate or the induction of gene expression in MCF-12A cells. Parental MCF-12A cells form acini, solid spheres and elongated branching ducts when grown in rat-tail collagen type I matrix, the geometries and distribution of which are altered following the removal of fibroblast-like cells. Conclusions MCF-12A cells are a heterogeneous pseudo-epithelial cell line capable of forming a variety of multicellular structures in 3D culture. We found no indication that the cells display estrogen-responsive characteristics, thus refuting previous studies which reported estrogen responsiveness. We report that MCF-12A cells are not suited for use in studies in which differential behaviors of "normal" and "cancerous" estrogen-responsive cells are to be compared.
Collapse
Affiliation(s)
- Michael F Sweeney
- 1Sackler School of Graduate Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Carlos Sonnenschein
- 2Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Ana M Soto
- 2Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| |
Collapse
|
33
|
Thakuri PS, Liu C, Luker GD, Tavana H. Biomaterials-Based Approaches to Tumor Spheroid and Organoid Modeling. Adv Healthc Mater 2018; 7:e1700980. [PMID: 29205942 PMCID: PMC5867257 DOI: 10.1002/adhm.201700980] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/21/2017] [Indexed: 12/22/2022]
Abstract
Evolving understanding of structural and biological complexity of tumors has stimulated development of physiologically relevant tumor models for cancer research and drug discovery. A major motivation for developing new tumor models is to recreate the 3D environment of tumors and context-mediated functional regulation of cancer cells. Such models overcome many limitations of standard monolayer cancer cell cultures. Under defined culture conditions, cancer cells self-assemble into 3D constructs known as spheroids. Additionally, cancer cells may recapitulate steps in embryonic development to self-organize into 3D cultures known as organoids. Importantly, spheroids and organoids reproduce morphology and biologic properties of tumors, providing valuable new tools for research, drug discovery, and precision medicine in cancer. This Progress Report discusses uses of both natural and synthetic biomaterials to culture cancer cells as spheroids or organoids, specifically highlighting studies that demonstrate how these models recapitulate key properties of native tumors. The report concludes with the perspectives on the utility of these models and areas of need for future developments to more closely mimic pathologic events in tumors.
Collapse
Affiliation(s)
- Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Chun Liu
- Departments of Radiology, Biomedical Engineering and Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gary D Luker
- Departments of Radiology, Biomedical Engineering and Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| |
Collapse
|
34
|
Gonzalez-Guerrico AM, Espinoza I, Schroeder B, Park CH, Kvp CM, Khurana A, Corominas-Faja B, Cuyàs E, Alarcón T, Kleer C, Menendez JA, Lupu R. Suppression of endogenous lipogenesis induces reversion of the malignant phenotype and normalized differentiation in breast cancer. Oncotarget 2018; 7:71151-71168. [PMID: 27223424 PMCID: PMC5342069 DOI: 10.18632/oncotarget.9463] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022] Open
Abstract
The correction of specific signaling defects can reverse the oncogenic phenotype of tumor cells by acting in a dominant manner over the cancer genome. Unfortunately, there have been very few successful attempts at identifying the primary cues that could redirect malignant tissues to a normal phenotype. Here we show that suppression of the lipogenic enzyme fatty acid synthase (FASN) leads to stable reversion of the malignant phenotype and normalizes differentiation in a model of breast cancer (BC) progression. FASN knockdown dramatically reduced tumorigenicity of BC cells and restored tissue architecture, which was reminiscent of normal ductal-like structures in the mammary gland. Loss of FASN signaling was sufficient to direct tumors to a reversed phenotype that was near normal when considering the development of polarized growth-arrested acinar-like structure similar to those formed by nonmalignant breast cells in a 3D reconstituted basement membrane in vitro. This process, in vivo, resulted in a low proliferation index, mesenchymal-epithelial transition, and shut-off of the angiogenic switch in FASN-depleted BC cells orthotopically implanted into mammary fat pads. The role of FASN as a negative regulator of correct breast tissue architecture and terminal epithelial cell differentiation was dominant over the malignant phenotype of tumor cells possessing multiple cancer-driving genetic lesions as it remained stable during the course of serial in vivo passage of orthotopic tumor-derived cells. Transient knockdown of FASN suppressed hallmark structural and cytosolic/secretive proteins (vimentin, N-cadherin, fibronectin) in a model of EMT-induced cancer stem cells (CSC). Indirect pharmacological inhibition of FASN promoted a phenotypic switch from basal- to luminal-like tumorsphere architectures with reduced intrasphere heterogeneity. The fact that sole correction of exacerbated lipogenesis can stably reprogram cancer cells back to normal-like tissue architectures might open a new avenue to chronically restrain BC progression by using FASN-based differentiation therapies.
Collapse
Affiliation(s)
- Anatilde M Gonzalez-Guerrico
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ingrid Espinoza
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Schroeder
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA
| | - Cheol Hong Park
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA
| | - Chandra Mohan Kvp
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ashwani Khurana
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA
| | - Bruna Corominas-Faja
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Catalonia, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Elisabet Cuyàs
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Catalonia, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Tomás Alarcón
- Computational and Mathematical Biology Research Group, Centre de Recerca Matemàtica (CRM), Barcelona, Spain.,Departament de Matemàtiques, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,ICREA (Institució Catalana d'Estudis i Recerca Avançats), Barcelona, Spain.,Barcelona Graduate School of Mathematics (BGSMath), Barcelona, Spain
| | - Celina Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Javier A Menendez
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Catalonia, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Ruth Lupu
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Cancer Center, Rochester, MN, USA
| |
Collapse
|
35
|
Differential response to doxorubicin in breast cancer subtypes simulated by a microfluidic tumor model. J Control Release 2017; 266:129-139. [PMID: 28939108 DOI: 10.1016/j.jconrel.2017.09.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 02/08/2023]
Abstract
Successful drug delivery and overcoming drug resistance are the primary clinical challenges for management and treatment of cancer. The ability to rapidly screen drugs and delivery systems within physiologically relevant environments is critically important; yet is currently limited due to lack of appropriate tumor models. To address this problem, we developed the Tumor-microenvironment-on-chip (T-MOC), a new microfluidic tumor model simulating the interstitial flow, plasma clearance, and transport of the drug within the tumor. We demonstrated T-MOC's capabilities by assessing the delivery and efficacy of doxorubicin in small molecular form versus hyaluronic acid nanoparticle (NP) formulation in MCF-7 and MDA-MB-231, two cell lines representative of different molecular subtypes of breast cancer. Doxorubicin accumulated and penetrated similarly in both cell lines while the NP accumulated more in MDA-MB-231 than MCF-7 potentially due to binding of hyaluronic acid to CD44 expressed by MDA-MB-231. However, the penetration of the NP was less than the molecular drug due to its larger size. In addition, both cell lines cultured on the T-MOC showed increased resistance to the drug compared to 2D culture where MDA-MB-231 attained a drug-resistant tumor-initiating phenotype indicated by increased CD44 expression. When grown in immunocompromised mice, both cell lines exhibited cell-type-dependent resistance and phenotypic changes similar to T-MOC, confirming its predictive ability for in vivo drug response. This initial characterization of T-MOC indicates its transformative potential for in vitro testing of drug efficacy towards prediction of in vivo outcomes and investigation of drug resistance mechanisms for advancement of personalized medicine.
Collapse
|
36
|
Liu H, Wang J, Ren G, Zhang Y, Dong F. The relationship between proteoglycan inhibition via xylosyltransferase II silencing and the implantation of salivary pleomorphic adenoma. J Oral Pathol Med 2017; 46:504-512. [PMID: 27732748 DOI: 10.1111/jop.12510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To study the relationship between proteoglycan inhibition and the implantation of salivary pleomorphic adenoma (SPA). METHODS SPA fresh tissue was primitively cultured and identified. The Ad-shRNA-XT-II adenovirus vector was constructed and transfected into SPA cells to silence the XT-II gene. The expression of the XT-II gene and protein was detected using real-time PCR and Western blotting, respectively. The proteoglycan content of the cells was determined 48 h after transfection. The invasion and migration of SPA cells were observed using Matrigel invasion and wound-healing assays. Fibroblasts from the tumour capsule of the same patient were obtained, cultured and seeded onto an acellular dermal matrix (ADM) scaffold. Tumour cells were seeded onto the scaffold with the fibroblasts and then transferred to BALB/C-nu nude mice and allowed to grow in vivo for 3 months. RESULTS The SPA cells cultures were positive for human calponin, S-100 protein, α-SMA and CK. XT-II gene and protein expression was decreased by 42.72% and 34%, respectively. The proteoglycan content was downregulated by 41.15%. XT-II gene silencing decreased the invasion and migration abilities of SPA cells. The assessment of SPA growth in nude mice indicated an absence of tumour growth in the SPA-XT-II group (in which the XT-II gene was silenced), whereas SPA growth was observed in the other two groups (in which the XT-II gene was not silenced), and the tumour tissue was positive for the human S-100 protein, α-SMA and CK8&18. CONCLUSION Proteoglycan inhibition induced via XT-II gene silencing inhibited the implantation of SPA.
Collapse
Affiliation(s)
- Huijuan Liu
- Department of Oral Pathology, College and Hospital of Stomatology, Key Laboratory of Stomatology, Hebei Medical University, Hebei Province, China
| | - Jie Wang
- Department of Oral Pathology, College and Hospital of Stomatology, Key Laboratory of Stomatology, Hebei Medical University, Hebei Province, China
| | - Guiyun Ren
- Department of Oral & Maxillofacial Surgery, College and Hospital of Stomatology, Key Laboratory of Stomatology, Hebei Medical University, Hebei Province, China
| | - Yanning Zhang
- Department of Oral Pathology, College and Hospital of Stomatology, Key Laboratory of Stomatology, Hebei Medical University, Hebei Province, China
| | - Fusheng Dong
- Department of Oral & Maxillofacial Surgery, College and Hospital of Stomatology, Key Laboratory of Stomatology, Hebei Medical University, Hebei Province, China
| |
Collapse
|
37
|
Rianna C, Kumar P, Radmacher M. The role of the microenvironment in the biophysics of cancer. Semin Cell Dev Biol 2017; 73:107-114. [PMID: 28746843 DOI: 10.1016/j.semcdb.2017.07.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 01/23/2023]
Abstract
During the last decades, cell mechanics has been recognized as a quantitative measure to discriminate between many physiological and pathological states of single cells. In the field of biophysics of cancer, a large body of research has been focused on the comparison between normal and cancer mechanics and slowly the hypothesis that cancer cells are softer than their normal counterparts has been accepted, even though in situ tumor tissue is usually stiffer than the surrounding normal tissue. This corroborates the idea that the extra-cellular matrix (ECM) has a critical role in regulating tumor cell properties and behavior. Rearrangements in ECM can lead to changes in cancer cell mechanics and in specific conditions the general assumption about cancer cell softening could be confuted. Here, we highlight the contribution of ECM in cancer cell mechanics and argue that the statement that cancer cells are softer than normal cells should be firmly related to the properties of cell environment and the specific stage of cancer cell progression. In particular, we will discuss that when employing cell mechanics in cancer diagnosis and discrimination, the chemical, the topographical and - last but not least - the mechanical properties of the microenvironment are very important.
Collapse
Affiliation(s)
- Carmela Rianna
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany
| | - Prem Kumar
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany
| | - Manfred Radmacher
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany.
| |
Collapse
|
38
|
Pradhan S, Clary JM, Seliktar D, Lipke EA. A three-dimensional spheroidal cancer model based on PEG-fibrinogen hydrogel microspheres. Biomaterials 2016; 115:141-154. [PMID: 27889665 DOI: 10.1016/j.biomaterials.2016.10.052] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/26/2016] [Accepted: 10/29/2016] [Indexed: 12/11/2022]
Abstract
Three-dimensional (3D) in vitro cancer models offer an attractive approach towards the investigation of tumorigenic phenomena and other cancer studies by providing dimensional context and higher degree of physiological relevance than that offered by conventional two-dimensional (2D) models. The multicellular tumor spheroid model, formed by cell aggregation, is considered to be the "gold standard" for 3D cancer models, due to its ease and simplicity of use. Although better than 2D models, tumor spheroids are unable to replicate key features of the native tumor microenvironment, particularly due to a lack of surrounding extracellular matrix components and heterogeneity in shape, size and aggregate forming tendencies. In order to address this issue, we have developed a 3D "tumor microsphere" model, formed by a dual-photoinitiator, aqueous-oil emulsion technique, for the encapsulation of cancer cells within PEG-fibrinogen hydrogel microspheres and for subsequent long-term 3D culture. In comparison to self-aggregated tumor spheroids, the tumor microspheres displayed a higher degree of size and shape homogeneity throughout long-term culture. In sharp contrast to cells in tumor spheroids, cells within tumor microspheres demonstrated significant loss in apico-basal polarity and cellular architecture, cellular and nuclear atypia, increased disorganization, elevated nuclear cytoplasmic ratio and nuclear volume density and reduction in cell-cell junction length, all of which are hallmarks of malignant transformation and tumorigenic progression. Additionally, the tumor microsphere model was extended for the 3D encapsulation and maintenance of a wide range of other cancer cell (metastatic and non-metastatic) types. Taken together, our results reinforce the importance of incorporating a biomimetic matrix in the cellular microenvironment of 3D tumor models and the influential effects of the matrix on the tumorigenic morphology of 3D cultured cells. The tumor microsphere system established in this study has the potential to be used in future investigations of 3D cancer cell-cell and cell-ECM interactions and in drug-testing applications.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Jacob M Clary
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Dror Seliktar
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
39
|
Evolution from genetics to phenotype: reinterpretation of NSCLC plasticity, heterogeneity, and drug resistance. Protein Cell 2016; 8:178-190. [PMID: 27757846 PMCID: PMC5326619 DOI: 10.1007/s13238-016-0330-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Targeted therapy is beneficial in most cases, but the development of drug resistance stands as an obstacle to good prognosis. Multiple mechanisms were explored such as genetic alterations, activation of bypass signaling, and phenotypic transition. These intrinsic and/or extrinsic dynamic regulations facilitate tumor cell survival in meeting the demands of signaling under different stimulus. This review introduces lung cancer plasticity and heterogeneity and their correlation with drug resistance. While cancer plasticity and heterogeneity play an essential role in the development of drug resistance, the manipulation of them may bring some inspirations to cancer prognosis and treatment. That is to say, lung cancer plasticity and heterogeneity present us with not only challenges but also opportunities.
Collapse
|
40
|
Pradhan S, Hassani I, Seeto WJ, Lipke EA. PEG‐fibrinogen hydrogels for three‐dimensional breast cancer cell culture. J Biomed Mater Res A 2016; 105:236-252. [DOI: 10.1002/jbm.a.35899] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/08/2016] [Accepted: 09/07/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Shantanu Pradhan
- Department of Chemical EngineeringAuburn UniversityAuburn Alabama36849
| | - Iman Hassani
- Department of Chemical EngineeringAuburn UniversityAuburn Alabama36849
| | - Wen J. Seeto
- Department of Chemical EngineeringAuburn UniversityAuburn Alabama36849
| | | |
Collapse
|
41
|
Fecher D, Hofmann E, Buck A, Bundschuh R, Nietzer S, Dandekar G, Walles T, Walles H, Lückerath K, Steinke M. Human Organotypic Lung Tumor Models: Suitable For Preclinical 18F-FDG PET-Imaging. PLoS One 2016; 11:e0160282. [PMID: 27501455 PMCID: PMC4976941 DOI: 10.1371/journal.pone.0160282] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/15/2016] [Indexed: 12/20/2022] Open
Abstract
Development of predictable in vitro tumor models is a challenging task due to the enormous complexity of tumors in vivo. The closer the resemblance of these models to human tumor characteristics, the more suitable they are for drug-development and –testing. In the present study, we generated a complex 3D lung tumor test system based on acellular rat lungs. A decellularization protocol was established preserving the architecture, important ECM components and the basement membrane of the lung. Human lung tumor cells cultured on the scaffold formed cluster and exhibited an up-regulation of the carcinoma-associated marker mucin1 as well as a reduced proliferation rate compared to respective 2D culture. Additionally, employing functional imaging with 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography (FDG-PET) these tumor cell cluster could be detected and tracked over time. This approach allowed monitoring of a targeted tyrosine kinase inhibitor treatment in the in vitro lung tumor model non-destructively. Surprisingly, FDG-PET assessment of single tumor cell cluster on the same scaffold exhibited differences in their response to therapy, indicating heterogeneity in the lung tumor model. In conclusion, our complex lung tumor test system features important characteristics of tumors and its microenvironment and allows monitoring of tumor growth and -metabolism in combination with functional imaging. In longitudinal studies, new therapeutic approaches and their long-term effects can be evaluated to adapt treatment regimes in future.
Collapse
Affiliation(s)
- David Fecher
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- * E-mail:
| | - Elisabeth Hofmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ralph Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Sarah Nietzer
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Gudrun Dandekar
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center ´Regenerative Therapies for Oncology and Musculoskeletal Diseases`Wuerzburg, branch of the Fraunhofer Institute Interfacial Engineering and Biotechnology (IGB), Wuerzburg, Germany
| | - Thorsten Walles
- Department of Cardiothoracic Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Heike Walles
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center ´Regenerative Therapies for Oncology and Musculoskeletal Diseases`Wuerzburg, branch of the Fraunhofer Institute Interfacial Engineering and Biotechnology (IGB), Wuerzburg, Germany
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Maria Steinke
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center ´Regenerative Therapies for Oncology and Musculoskeletal Diseases`Wuerzburg, branch of the Fraunhofer Institute Interfacial Engineering and Biotechnology (IGB), Wuerzburg, Germany
| |
Collapse
|
42
|
Collagen Extracted from Persian Gulf Squid Exhibits Anti-Cytotoxic Properties on Apple Pectic Treated Cells: Assessment in an In Vitro Bioassay Model. IRANIAN JOURNAL OF PUBLIC HEALTH 2016; 45:1054-1063. [PMID: 27928532 PMCID: PMC5139963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Collagen-based three-dimensional (3D) in vitro systems have been introduced to study the physiological states of cells. As a biomolecule, collagen is usually extracted from terrestrial animals whilst aquatic animals like squid contain large amounts of collagen. METHODS In order to make effective use of marine organisms, we selected Persian Gulf squid in 2015 to extract the required collagen. Then, a 3D culture system based on the extracted collagen was applied to investigate cellular mechanisms in a native microenvironment. The formed collagen gel was used to investigate the growth of MDA-MB-231 breast cancer cells as well as responses to pectic acid. RESULTS The results revealed that the extracted collagen contained α, ß and γ components with high water holding capacity. This collagen formed a gel-like structure, which could promote the proliferation of MDA-MB-231 breast cancer cells. The MDA-MB-231 cells' viability in presence of pectic acid, demonstrating the cells' behavior in a 3D culture system. CONCLUSION It seems that the collagen extracted from squid skin has type I collagen properties. It might be used as a substrate in 3D cell culture systems.
Collapse
|
43
|
Capturing tumor complexity in vitro: Comparative analysis of 2D and 3D tumor models for drug discovery. Sci Rep 2016; 6:28951. [PMID: 27364600 PMCID: PMC4929472 DOI: 10.1038/srep28951] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/27/2016] [Indexed: 01/14/2023] Open
Abstract
Two-dimensional (2D) cell cultures growing on plastic do not recapitulate the three dimensional (3D) architecture and complexity of human tumors. More representative models are required for drug discovery and validation. Here, 2D culture and 3D mono- and stromal co-culture models of increasing complexity have been established and cross-comparisons made using three standard cell carcinoma lines: MCF7, LNCaP, NCI-H1437. Fluorescence-based growth curves, 3D image analysis, immunohistochemistry and treatment responses showed that end points differed according to cell type, stromal co-culture and culture format. The adaptable methodologies described here should guide the choice of appropriate simple and complex in vitro models.
Collapse
|
44
|
Roberts GC, Morris PG, Moss MA, Maltby SL, Palmer CA, Nash CE, Smart E, Holliday DL, Speirs V. An Evaluation of Matrix-Containing and Humanised Matrix-Free 3-Dimensional Cell Culture Systems for Studying Breast Cancer. PLoS One 2016; 11:e0157004. [PMID: 27300768 PMCID: PMC4907459 DOI: 10.1371/journal.pone.0157004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/23/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND 3D cell cultures are emerging as more physiologically meaningful alternatives to monolayer cultures for many biological applications. They are attractive because they more closely mimic in vivo morphology, especially when co-cultured with stromal fibroblasts. METHODOLOGY/PRINCIPAL FINDINGS We compared the efficacy of 3 different 3D cell culture systems; collagen I, low attachment culture vessels and a modification of Fibrolife®, a specialised humanised cell culture medium devoid of animal-derived components, using breast cancer cell lines representative of the different molecular subtypes of breast cancer, cultured alone or with human mammary fibroblasts with a view to developing matrix-free humanised systems. 3D collagen I culture supported the growth of a range of breast cancer cell lines. By modifying the composition of Fibrolife® to epiFL, matrix-free cell culture was possible. During sequential transfer to epiFL breast cancer cells gradually detached from the flask, growing progressively as spheroids. Phenotype was stable and reversible with cells remaining actively proliferating and easily accessible throughout culture. They could also be revived from frozen stocks. To achieve co-culture with fibroblasts in epiFL required use of low attachment culture vessels instead of standard plastic as fibroblasts remained adherent in epiFL. Here, cancer cell spheroids were allowed to form before adding fibroblasts. Immunohistochemical examination showed fibroblasts scattered throughout the epithelial spheroid, not dissimilar to the relationship of tumour stroma in human breast cancer. CONCLUSIONS Because of its ease of handling, matrix-free 3D cell culture may be a useful model to study the influence of fibroblasts on breast cancer epithelial cells with use of epiFL culture medium taking this a step further towards a fully humanised 3D model. This methodology could be applied to other types of cancer cell lines, making this a versatile technique for cancer researchers wishing to use in vitro systems that better reflect cancer in vivo.
Collapse
Affiliation(s)
- Grace C. Roberts
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Paul G. Morris
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Marcus A. Moss
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Sarah L. Maltby
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Chelsea A. Palmer
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Claire E. Nash
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Emily Smart
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Deborah L. Holliday
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| |
Collapse
|
45
|
Regier MC, Alarid ET, Beebe DJ. Progress towards understanding heterotypic interactions in multi-culture models of breast cancer. Integr Biol (Camb) 2016; 8:684-92. [PMID: 27097801 PMCID: PMC4993016 DOI: 10.1039/c6ib00001k] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microenvironments in primary tumors and metastases include multiple cell types whose dynamic and reciprocal interactions are central to progression of the disease. However, the literature involving breast cancer studied in vitro is dominated by cancer cells in mono-culture or co-cultured with one other cell type. For in vitro studies of breast cancer the inclusion of multiple cell types has led to models that are more representative of in vivo behaviors and functions as compared to more traditional monoculture. Here, we review foundational co-culture techniques and their adaptation to multi-culture (including three or more cell types). Additionally, while macroscale methods involving conditioned media, direct contact, and indirect interactions have been informative, we examined many advances that have been made more recently using microscale systems with increased control over cellular and structural complexity. Throughout this discussion we consider the benefits and limitations of current multi-culture methods and the significant results they have produced.
Collapse
Affiliation(s)
- Mary C Regier
- Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| | | | | |
Collapse
|
46
|
Andersen AP, Flinck M, Oernbo EK, Pedersen NB, Viuff BM, Pedersen SF. Roles of acid-extruding ion transporters in regulation of breast cancer cell growth in a 3-dimensional microenvironment. Mol Cancer 2016; 15:45. [PMID: 27266704 PMCID: PMC4896021 DOI: 10.1186/s12943-016-0528-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 05/20/2016] [Indexed: 12/20/2022] Open
Abstract
Background The 3-dimensional (3D) microenvironment of breast carcinomas is characterized by profoundly altered pH homeostasis, reflecting increased metabolic acid production and a confined extracellular space characterized by poor diffusion, yet the relative contributions of specific pH-regulatory transporters to 3D growth are poorly understood. The aim of this work was to determine how 3D spheroid growth of breast cancer cells impacts the expression and spatial organization of major acid extruding proteins, and how these proteins in turn are required for spheroid growth. Methods MCF-7 (Luminal-A) and MDA-MB-231 (Triple-negative) human breast cancer cells were grown as ~700-950 μm diameter spheroids, which were subjected to Western blotting for relevant transporters (2- and 3D growth), quantitative immunohistochemical analysis, and spheroid growth assays. Individual transporter contributions were assessed (i) pharmacologically, (ii) by stable shRNA- and transient siRNA-mediated knockdown, and (iii) by CRISPR/Cas9 knockout. Results In MCF-7 spheroids, expression of the lactate-H+ cotransporter MCT1 (SLC16A1) increased from the spheroid periphery to its core, the Na+,HCO3− cotransporter NBCn1 (SLC4A7) was most highly expressed at the periphery, and the Na+/H+ exchanger NHE1 (SLC9A1) and MCT4 (SLC16A3) were evenly distributed. A similar pattern was seen in MDA-MB-231 spheroids, except that these cells do not express MCT1. The relative total expression of NBCn1 and NHE1 was decreased in 3D compared to 2D, while that of MCT1 and MCT4 was unaltered. Inhibition of MCT1 (AR-C155858) attenuated MCF-7 spheroid growth and this was exacerbated by addition of S0859, an inhibitor of Na+,HCO3− cotransporters and MCTs. The pharmacological data was recapitulated by stable knockdown of MCT1 or NBCn1, whereas knockdown of MCT4 had no effect. CRISPR/Cas9 knockout of NHE1, but neither partial NHE1 knockdown nor the NHE1 inhibitor cariporide, inhibited MCF-7 spheroid growth. In contrast, growth of MDA-MB-231 spheroids was inhibited by stable or transient NHE1 knockdown and by NHE1 knockout, but not by knockdown of NBCn1 or MCT4. Conclusions This work demonstrates the distinct expression and localization patterns of four major acid-extruding transporters in 3D spheroids of human breast cancer cells and reveals that 3D growth is dependent on these transporters in a cell type-dependent manner, with potentially important implications for breast cancer therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0528-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Poder Andersen
- Department of Biology, Section for Cell Biology and Physiology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Mette Flinck
- Department of Biology, Section for Cell Biology and Physiology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Eva Kjer Oernbo
- Department of Biology, Section for Cell Biology and Physiology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Nis Borbye Pedersen
- Department of Biology, Section for Cell Biology and Physiology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Birgitte Martine Viuff
- Department of Veterinary Disease Biology, Section for Molecular Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Stine Falsig Pedersen
- Department of Biology, Section for Cell Biology and Physiology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
47
|
Xu W, Qian J, Zhang Y, Suo A, Cui N, Wang J, Yao Y, Wang H. A photo-polymerized poly(N ε-acryloyl l-lysine) hydrogel for 3D culture of MCF-7 breast cancer cells. J Mater Chem B 2016; 4:3339-3350. [PMID: 32263269 DOI: 10.1039/c6tb00511j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The most common in vitro cell culture platform, standard two-dimensional (2D) monolayer cell culture, often fails to mimic the tumor microenvironment, while animal models complicate research on the effect of individual factors on cell behaviors. Both are unsatisfactory in the research of molecular mechanisms of tumor development and progression and the discovery and development of anticancer drugs. In vitro three-dimensional (3D) cell culture can partially simulate in vivo conditions and 3D-cultured cancer cells can recapture many essential features of native tumor tissues. In this study, to mimic the in vivo breast tumor microenvironment, novel reduction-responsive poly(Nε-acryloyl l-lysine) (pLysAAm) hydrogels were synthesized by rapid photo-polymerization of Nε-acryloyl l-lysine and using N,N'-bis(acryloyl)-(l)-cystine as a crosslinker, and their physicochemical properties were characterized systemically. The results showed that the pLysAAm hydrogels were formed within 93 s under UV irradiation and exhibited almost total elastic recovery from compressions as high as 75%. The lyophilized hydrogel samples displayed a highly porous structure with interconnected pores, had an equilibrium swelling ratio of about 20, and were degraded faster in a glutathione-containing solution than in PBS solution. The biological versatility of the pLysAAm hydrogels was demonstrated by both in vitro MCF-7 cell culture and in vivo tumor formation. Compared to cells cultured as 2D monolayers, the 3D-cultured cells presented 3D cell morphology, exhibited better cell viability, expressed higher levels of pro-angiogenic factors, and showed significantly greater migration and invasion abilities. The results from assay of tumorigenicity in nude mice and histologic analysis demonstrated the enhanced tumorigenic and angiogenic capabilities of the MCF-7 cells pre-cultured in pLysAAm hydrogels. These findings suggest that pLysAAm hydrogels may be used to bridge the gap between standard in vitro cell cultures and living tissues, aid breast cancer research, and help researchers to develop novel anticancer therapeutics.
Collapse
Affiliation(s)
- Weijun Xu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Paulose T, Montévil M, Speroni L, Cerruti F, Sonnenschein C, Soto AM. SAMA: A Method for 3D Morphological Analysis. PLoS One 2016; 11:e0153022. [PMID: 27035711 PMCID: PMC4818086 DOI: 10.1371/journal.pone.0153022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/21/2016] [Indexed: 01/04/2023] Open
Abstract
Three-dimensional (3D) culture models are critical tools for understanding tissue morphogenesis. A key requirement for their analysis is the ability to reconstruct the tissue into computational models that allow quantitative evaluation of the formed structures. Here, we present Software for Automated Morphological Analysis (SAMA), a method by which epithelial structures grown in 3D cultures can be imaged, reconstructed and analyzed with minimum human intervention. SAMA allows quantitative analysis of key features of epithelial morphogenesis such as ductal elongation, branching and lumen formation that distinguish different hormonal treatments. SAMA is a user-friendly set of customized macros operated via FIJI (http://fiji.sc/Fiji), an open-source image analysis platform in combination with a set of functions in R (http://www.r-project.org/), an open-source program for statistical analysis. SAMA enables a rapid, exhaustive and quantitative 3D analysis of the shape of a population of structures in a 3D image. SAMA is cross-platform, licensed under the GPLv3 and available at http://montevil.theobio.org/content/sama.
Collapse
Affiliation(s)
- Tessie Paulose
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Maël Montévil
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Lucia Speroni
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Florent Cerruti
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Carlos Sonnenschein
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
49
|
Cardoso TC, Sakamoto SS, Stockmann D, Souza TFB, Ferreira HL, Gameiro R, Vieira FV, Louzada MJQ, Andrade AL, Flores EF. A three-dimensional cell culture system as an in vitro canine mammary carcinoma model for the expression of connective tissue modulators. Vet Comp Oncol 2016; 15:582-593. [PMID: 26991309 DOI: 10.1111/vco.12202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 11/27/2022]
Abstract
In this study, derived complex carcinoma (CC) and simple carcinoma (SC) cell lines were established and cultured under two-dimensional (2D) and three-dimensional (3D) conditions. The 3D was performed in six-well AlgiMatrix™ (LifeTechnologies®, Carlsbad, CA, USA) scaffolds, resulting in spheroids sized 50-125 µm for CC and 175-200 µm for SC. Cell viability was demonstrated up to 14 days for both models. Epidermal growth factor receptor (EGFR) was expressed in CC and SC in both systems. However, higher mRNA and protein levels were observed in SC 2D and 3D systems when compared with CC (P < 0.005). The connective tissue modulators, metalloproteinases-1, -2, -9 and -13 (MMPs), relaxin receptors 1 and 2 (RXR1 and RXR2) and E-cadherin (CDH1) were quantitated. All were upregulated similarly when canine mammary tumour (CMT)-derived cell lines were cultured under 3D AlgiMatrix, except CDH1 that was downregulated (P < 0.005). These results are promising towards the used of 3D system to increase a high throughput in vitro canine tumour model.
Collapse
Affiliation(s)
- T C Cardoso
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil.,Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - S S Sakamoto
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - D Stockmann
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - T F B Souza
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - H L Ferreira
- Department of de Veterinary Medicine, Faculdade Zotecnia e Engenharia de Alimentos-USP, Pirassununga, SP, Brazil
| | - R Gameiro
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil
| | - F V Vieira
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil
| | - M J Q Louzada
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil
| | - A L Andrade
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - E F Flores
- Department of Preventive Veterinary Medicine, Federal University of Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
50
|
Xu W, Qian J, Zhang Y, Suo A, Cui N, Wang J, Yao Y, Wang H. A double-network poly(Nɛ-acryloyl L-lysine)/hyaluronic acid hydrogel as a mimic of the breast tumor microenvironment. Acta Biomater 2016; 33:131-41. [PMID: 26805429 DOI: 10.1016/j.actbio.2016.01.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/19/2015] [Accepted: 01/20/2016] [Indexed: 01/07/2023]
Abstract
To mimic the structure of breast tumor microenvironment, novel double-network poly(Nɛ-acryloyl L-lysine)/hyaluronic acid (pLysAAm/HA) hydrogels were fabricated by a two-step photo-polymerization process for in vitro three-dimensional (3D) cell culture. The morphology, mechanical properties, swelling and degradation behaviors of pLysAAm/HA hydrogels were investigated. The growth behavior and function of MCF-7 cells cultured on the hydrogels and standard 2D culture plates were compared. The results showed that pLysAAm/HA hydrogels had a highly porous microstructure with a double network and that their mechanical properties, swelling ratio and degradation rate depended on the degree of methacrylation of HA. The results of in vitro studies revealed that the pLysAAm/HA hydrogels could support MCF-7 cell adhesion, promote cell proliferation, and induce the diversification of cell morphologies and overexpression of VEGF, IL-8 and bFGF. The MCF-7 cells cultured on 3D hydrogels showed significantly increased migration and invasion abilities as compared to 2D-cultured cells. Preliminary in vivo results confirmed that the 3D culture of MCF-7 cells resulted in greater tumorigenesis than their 2D culture. These results indicate that the pLysAAm/HA hydrogels can provide a 3D microenvironment for MCF-7 cells that is more representative of the in vivo breast cancer. STATEMENT OF SIGNIFICANCE Traditional 2D cell cultures cannot ideally represent their in vivo physiological conditions. In this work, we reported a method for preparing double-network poly(Nɛ-acryloyl L-lysine)/hyaluronic acid hydrogel, and demonstrated its suitability for use in mimicing breast tumor microenvironment. Results showed the prepared hydrogels had controllable mechanical properties, swelling ratio and degradation rate. The MCF-7 cells cultured in hydrogels expressed much higher levels of pro-angiogenic growth factors and displayed significantly enhanced migration and invasion abilities. The tumorigenic capability of MCF-7 cells pre-cultured in 3D hydrogels was enhanced significantly. Therefore, the novel hydrogel may provide a more physiologically relevant 3D in vitro model for breast cancer research. To our knowledge, this is the first report assessing a HA-based double-network hydrogel used as a tumor model.
Collapse
Affiliation(s)
- Weijun Xu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Junmin Qian
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Yaping Zhang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Aili Suo
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Ning Cui
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jinlei Wang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yu Yao
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an 710061, China
| | - Hejing Wang
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|