101
|
Cox CR, Lynch S, Goldring C, Sharma P. Current Perspective: 3D Spheroid Models Utilizing Human-Based Cells for Investigating Metabolism-Dependent Drug-Induced Liver Injury. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:611913. [PMID: 35047893 PMCID: PMC8757888 DOI: 10.3389/fmedt.2020.611913] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) remains a leading cause for the withdrawal of approved drugs. This has significant financial implications for pharmaceutical companies, places increasing strain on global health services, and causes harm to patients. For these reasons, it is essential that in-vitro liver models are capable of detecting DILI-positive compounds and their underlying mechanisms, prior to their approval and administration to patients or volunteers in clinical trials. Metabolism-dependent DILI is an important mechanism of drug-induced toxicity, which often involves the CYP450 family of enzymes, and is associated with the production of a chemically reactive metabolite and/or inefficient removal and accumulation of potentially toxic compounds. Unfortunately, many of the traditional in-vitro liver models fall short of their in-vivo counterparts, failing to recapitulate the mature hepatocyte phenotype, becoming metabolically incompetent, and lacking the longevity to investigate and detect metabolism-dependent DILI and those associated with chronic and repeat dosing regimens. Nevertheless, evidence is gathering to indicate that growing cells in 3D formats can increase the complexity of these models, promoting a more mature-hepatocyte phenotype and increasing their longevity, in vitro. This review will discuss the use of 3D in vitro models, namely spheroids, organoids, and perfusion-based systems to establish suitable liver models to investigate metabolism-dependent DILI.
Collapse
Affiliation(s)
- Christopher R. Cox
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: Christopher R. Cox
| | - Stephen Lynch
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Goldring
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Parveen Sharma
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| |
Collapse
|
102
|
Karges J, Kuang S, Ong YC, Chao H, Gasser G. One‐ and Two‐Photon Phototherapeutic Effects of Ru
II
Polypyridine Complexes in the Hypoxic Centre of Large Multicellular Tumor Spheroids and Tumor‐Bearing Mice**. Chemistry 2020; 27:362-370. [DOI: 10.1002/chem.202003486] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Johannes Karges
- Chimie ParisTech PSL University CNRS Institute of Chemistry for Life and Health Sciences Laboratory for Inorganic Chemical Biology 75005 Paris France
| | - Shi Kuang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University 510275 Guangzhou People's Republic of China
| | - Yih Ching Ong
- Chimie ParisTech PSL University CNRS Institute of Chemistry for Life and Health Sciences Laboratory for Inorganic Chemical Biology 75005 Paris France
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University 510275 Guangzhou People's Republic of China
| | - Gilles Gasser
- Chimie ParisTech PSL University CNRS Institute of Chemistry for Life and Health Sciences Laboratory for Inorganic Chemical Biology 75005 Paris France
| |
Collapse
|
103
|
Emami F, Pathak S, Nguyen TT, Shrestha P, Maharjan S, Kim JO, Jeong JH, Yook S. Photoimmunotherapy with cetuximab-conjugated gold nanorods reduces drug resistance in triple negative breast cancer spheroids with enhanced infiltration of tumor-associated macrophages. J Control Release 2020; 329:645-664. [PMID: 33022330 DOI: 10.1016/j.jconrel.2020.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Tumor-associated macrophages (TAM) constitute up to 50-80% of stromal cells in breast cancer (BC), and are correlated with poor prognosis. As epidermal growth factor receptor (EGFR) is overexpressed in 60-80% of patients with triple negative breast cancer (TNBC), photoimmunotherapy (PIT) with cetuximab-targeted gold nanorods (CTX-AuNR) is an attractive therapeutic strategy for TNBC. The 3D cell culture model can mimic drug resistance conferred by the tumor microenvironment and its 3D organization; therefore, TAM and non-TAM embedded TNBC spheroids were constructed to evaluate the therapeutic efficacy of CTX-AuNR plus near infrared (NIR) irradiation. Cytotoxicity, reactive oxygen species (ROS) generation, and protein expression were compared in TNBC (± TAM) spheroids. The IC50 values of doxorubicin (DOX) in TAM-embedded TNBC spheroids were significantly higher than those in TNBC spheroids, demonstrating drug resistance, which could be explained by activation of IL-10/IL-10 receptor/STAT3/Bcl-2 signaling. However, 3D in vitro and in vivo results demonstrated that the efficacy of CTX-AuNR plus NIR irradiation was not significantly different in (± TAM) embedded TNBC cells. By enhancing ROS generation, CTX-AuNR plus NIR irradiation reprogrammed TAM polarization to the M1 anti-tumor phenotype, as indicated by macrophage mannose receptor (MMR) downregulation. Thus, CTX-AuNR plus NIR can serve as a potent PIT strategy for treating EGFR-overexpressing TNBC cells.
Collapse
Affiliation(s)
- Fakhrossadat Emami
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Shiva Pathak
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Tiep Tien Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Prakash Shrestha
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Srijan Maharjan
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea.
| |
Collapse
|
104
|
Premaratne ID, Toyoda Y, Celie KB, Brown KA, Spector JA. Tissue Engineering Models for the Study of Breast Neoplastic Disease and the Tumor Microenvironment. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:423-442. [DOI: 10.1089/ten.teb.2019.0347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ishani D. Premaratne
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Yoshiko Toyoda
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Karel-Bart Celie
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jason A. Spector
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
105
|
Doctor A, Seifert V, Ullrich M, Hauser S, Pietzsch J. Three-Dimensional Cell Culture Systems in Radiopharmaceutical Cancer Research. Cancers (Basel) 2020; 12:cancers12102765. [PMID: 32993034 PMCID: PMC7600608 DOI: 10.3390/cancers12102765] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
In preclinical cancer research, three-dimensional (3D) cell culture systems such as multicellular spheroids and organoids are becoming increasingly important. They provide valuable information before studies on animal models begin and, in some cases, are even suitable for reducing or replacing animal experiments. Furthermore, they recapitulate microtumors, metastases, and the tumor microenvironment much better than monolayer culture systems could. Three-dimensional models show higher structural complexity and diverse cell interactions while reflecting (patho)physiological phenomena such as oxygen and nutrient gradients in the course of their growth or development. These interactions and properties are of great importance for understanding the pathophysiological importance of stromal cells and the extracellular matrix for tumor progression, treatment response, or resistance mechanisms of solid tumors. Special emphasis is placed on co-cultivation with tumor-associated cells, which further increases the predictive value of 3D models, e.g., for drug development. The aim of this overview is to shed light on selected 3D models and their advantages and disadvantages, especially from the radiopharmacist's point of view with focus on the suitability of 3D models for the radiopharmacological characterization of novel radiotracers and radiotherapeutics. Special attention is paid to pancreatic ductal adenocarcinoma (PDAC) as a predestined target for the development of new radionuclide-based theranostics.
Collapse
Affiliation(s)
- Alina Doctor
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Verena Seifert
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Martin Ullrich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
- Correspondence: ; Tel.: +49-351-260-2622
| |
Collapse
|
106
|
Nilforoushzadeh MA, Khodadadi Yazdi M, Baradaran Ghavami S, Farokhimanesh S, Mohammadi Amirabad L, Zarrintaj P, Saeb MR, Hamblin MR, Zare M, Mozafari M. Mesenchymal Stem Cell Spheroids Embedded in an Injectable Thermosensitive Hydrogel: An In Situ Drug Formation Platform for Accelerated Wound Healing. ACS Biomater Sci Eng 2020; 6:5096-5109. [PMID: 33455261 DOI: 10.1021/acsbiomaterials.0c00988] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | | | - Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samila Farokhimanesh
- Department of Biotechnology, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, Oklahoma 74078, United States
| | - Mohammad Reza Saeb
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Dermatology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Mehrak Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
107
|
Kang Y, Datta P, Shanmughapriya S, Ozbolat IT. 3D Bioprinting of Tumor Models for Cancer Research. ACS APPLIED BIO MATERIALS 2020; 3:5552-5573. [DOI: 10.1021/acsabm.0c00791] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Youngnam Kang
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
| | - Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, West Bengal 711103, India
| | - Santhanam Shanmughapriya
- Department of Medicine, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Department of Cellular and Molecular Physiology, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Heart and Vascular Institute, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Ibrahim T. Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
- Biomedical Engineering Department, Penn State University, University Park, Pennsylvania 16802, United States
- Materials Research Institute, Penn State University, University Park, Pennsylvania 16802, United States
- Department of Neurosurgery, Penn State University, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
108
|
Hegde RS, Roychoudhury K, Pandey RN. The multi-functional eyes absent proteins. Crit Rev Biochem Mol Biol 2020; 55:372-385. [PMID: 32727223 PMCID: PMC7727457 DOI: 10.1080/10409238.2020.1796922] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/18/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022]
Abstract
The Eyes Absent (EYA) proteins are the only known instance of a single polypeptide housing the following three separable biochemical activities: tyrosine phosphatase, threonine phosphatase, and transactivation. This uniquely positions the EYAs to participate in both transcriptional regulation and signal transduction pathways. But it also complicates the assignment of biological roles to individual biochemical activities through standard loss-of-function experiments. Nevertheless, there is an emerging literature linking developmental and pathological functions with the various EYA activities, and a growing list of disease states that might benefit from EYA-targeted therapeutics. There also remain multiple unresolved issues with significant implications for our understanding of how the EYAs might impact such ubiquitous signaling cascades as the MYC and Notch pathways. This review will describe the unique juxtaposition of biochemical activities in the EYAs, their interaction with signaling pathways and cellular processes, emerging evidence of roles in disease states, and the feasibility of therapeutic targeting of individual EYA activities. We will focus on the phosphatase activities of the vertebrate EYA proteins and will examine the current state of knowledge regarding: • substrates and signaling pathways affected by the EYA tyrosine phosphatase activity; • modes of regulation of the EYA tyrosine phosphatase activity; • signaling pathways that implicate the threonine phosphatase activity of the EYAs including a potential interaction with PP2A-B55α; • the interplay between the two phosphatase activities and the transactivation function of the EYAs; • disease states associated with the EYAs and the current state of development of EYA-targeted therapeutics.
Collapse
Affiliation(s)
- Rashmi S. Hegde
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Department of Pediatrics, University of Cincinnati School of Medicine, 3333 Burnet Avenue, Cincinnati OH 45229
| | - Kaushik Roychoudhury
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Department of Pediatrics, University of Cincinnati School of Medicine, 3333 Burnet Avenue, Cincinnati OH 45229
| | - Ram Naresh Pandey
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Department of Pediatrics, University of Cincinnati School of Medicine, 3333 Burnet Avenue, Cincinnati OH 45229
| |
Collapse
|
109
|
Wang HF, Liu Y, Wang T, Yang G, Zeng B, Zhao CX. Tumor-Microenvironment-on-a-Chip for Evaluating Nanoparticle-Loaded Macrophages for Drug Delivery. ACS Biomater Sci Eng 2020; 6:5040-5050. [DOI: 10.1021/acsbiomaterials.0c00650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hao-Fei Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Yun Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Tong Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Bijun Zeng
- Diamantina Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
110
|
Steinmetz J, Senkowski W, Lengqvist J, Rubin J, Ossipova E, Herman S, Larsson R, Jakobsson PJ, Fryknäs M, Kultima K. Descriptive Proteome Analysis to Investigate Context-Dependent Treatment Responses to OXPHOS Inhibition in Colon Carcinoma Cells Grown as Monolayer and Multicellular Tumor Spheroids. ACS OMEGA 2020; 5:17242-17254. [PMID: 32715210 PMCID: PMC7376893 DOI: 10.1021/acsomega.0c01419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/22/2020] [Indexed: 06/11/2023]
Abstract
We have previously identified selective upregulation of the mevalonate pathway genes upon inhibition of oxidative phosphorylation (OXPHOS) in quiescent cancer cells. Using mass spectrometry-based proteomics, we here investigated whether these responses are corroborated on the protein level and whether proteomics could yield unique insights into context-dependent biology. HCT116 colon carcinoma cells were cultured as monolayer cultures, proliferative multicellular tumor spheroids (P-MCTS), or quiescent (Q-MCTS) multicellular tumor spheroids and exposed to OXPHOS inhibitors: nitazoxanide, FCCP, oligomycin, and salinomycin or the HMG-CoA-reductase inhibitor simvastatin at two different doses for 6 and 24 h. Samples were processed using an in-depth bottom-up proteomics workflow resulting in a total of 9286 identified protein groups. Gene set enrichment analysis showed profound differences between the three cell systems and confirmed differential enrichment of hypoxia, OXPHOS, and cell cycle progression-related protein responses in P-MCTS and Q-MCTS. Treatment experiments showed that the observed drug-induced alterations in gene expression of metabolically challenged cells are not translated directly to the protein level, but the results reaffirmed OXPHOS as a selective vulnerability of quiescent cancer cells. This work provides rationale for the use of deep proteome profiling to identify context-dependent treatment responses and encourages further studies investigating metabolic processes that could be co-targeted together with OXPHOS to eradicate quiescent cancer cells.
Collapse
Affiliation(s)
- Julia Steinmetz
- Division
of Rheumatology, Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Wojciech Senkowski
- Department
of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala SE-751 05, Sweden
| | - Johan Lengqvist
- Department
of Oncology-Pathology, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Jenny Rubin
- Department
of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala SE-751 05, Sweden
| | - Elena Ossipova
- Division
of Rheumatology, Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Stephanie Herman
- Department
of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala SE-751 85, Sweden
| | - Rolf Larsson
- Department
of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala SE-751 05, Sweden
| | - Per-Johan Jakobsson
- Division
of Rheumatology, Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Mårten Fryknäs
- Department
of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala SE-751 05, Sweden
| | - Kim Kultima
- Department
of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala SE-751 85, Sweden
| |
Collapse
|
111
|
Head and Neck Cancer Stem Cell-Enriched Spheroid Model for Anticancer Compound Screening. Cells 2020; 9:cells9071707. [PMID: 32708734 PMCID: PMC7408407 DOI: 10.3390/cells9071707] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs), a rare cell population in tumors, are resistant to conventional chemotherapy and thus responsible for tumor recurrence. To screen for active compounds targeting CSCs, a good CSC-enriched model compatible with high-throughput screening (HTS) is needed. Here, we describe a new head and neck cancer stem cell-enriched spheroid model (SCESM) suitable for HTS analyses of anti-CSC compounds. We used FaDu cells, round-bottom ultra-low adherent (ULA) microplates, and stem medium. The formed spheroids displayed increased expression of all stem markers tested (qRT-PCR and protein analysis) in comparison to the FaDu cells grown in a standard adherent culture or in a well-known HTS-compatible multi-cellular tumor spheroid model (MCTS). Consistent with increased stemness of the cells in the spheroid, confocal microscopy detected fast proliferating cells only at the outer rim of the SCESM spheroids, with poorly/non-proliferating cells deeper in. To confirm the sensitivity of our model, we used ATRA treatment, which strongly reduced the expression of selected stem markers. Altogether, we developed a CSC-enriched spheroid model with a simple protocol, a microplate format compatible with multimodal detection systems, and a high detection signal, making it suitable for anti-CSC compounds' HTS.
Collapse
|
112
|
Deynoux M, Sunter N, Ducrocq E, Dakik H, Guibon R, Burlaud-Gaillard J, Brisson L, Rouleux-Bonnin F, le Nail LR, Hérault O, Domenech J, Roingeard P, Fromont G, Mazurier F. A comparative study of the capacity of mesenchymal stromal cell lines to form spheroids. PLoS One 2020; 15:e0225485. [PMID: 32484831 PMCID: PMC7266346 DOI: 10.1371/journal.pone.0225485] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 05/17/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC)-spheroid models favor maintenance of stemness, ex vivo expansion and transplantation efficacy. Spheroids may also be considered as useful surrogate models of the hematopoietic niche. However, accessibility to primary cells, from bone marrow (BM) or adipose tissues, may limit their experimental use and the lack of consistency in methods to form spheroids may affect data interpretation. In this study, we aimed to create a simple model by examining the ability of cell lines, from human (HS-27a and HS-5) and murine (MS-5) BM origins, to form spheroids, compared to primary human MSCs (hMSCs). Our protocol efficiently allowed the spheroid formation from all cell types within 24 hours. Whilst hMSC-spheroids began to shrink after 24 hours, the size of spheroids from cell lines remained constant during three weeks. The difference was partially explained by the balance between proliferation and cell death, which could be triggered by hypoxia and induced oxidative stress. Our results demonstrate that, like hMSCs, MSC cell lines make reproductible spheroids that are easily handled. Thus, this model could help in understanding mechanisms involved in MSC functions and may provide a simple model by which to study cell interactions in the BM niche.
Collapse
Affiliation(s)
- Margaux Deynoux
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Nicola Sunter
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Elfi Ducrocq
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Hassan Dakik
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Roseline Guibon
- Anatomie et cytologie pathologique, CHRU de Tours, Tours, France
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Julien Burlaud-Gaillard
- Plateforme IBiSA de Microscopie Electronique, Université et CHRU de Tours, Tours, France
- INSERM U1259 MAVIVH, Université et CHRU de Tours, Tours, France
| | - Lucie Brisson
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | | | | | - Olivier Hérault
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- Service d'hématologie biologique, CHRU de Tours, Tours, France
| | - Jorge Domenech
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- Service d'hématologie biologique, CHRU de Tours, Tours, France
| | - Philippe Roingeard
- Plateforme IBiSA de Microscopie Electronique, Université et CHRU de Tours, Tours, France
- INSERM U1259 MAVIVH, Université et CHRU de Tours, Tours, France
| | - Gaëlle Fromont
- Anatomie et cytologie pathologique, CHRU de Tours, Tours, France
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Frédéric Mazurier
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- * E-mail:
| |
Collapse
|
113
|
Amaral R, Zimmermann M, Ma AH, Zhang H, Swiech K, Pan CX. A Simple Three-Dimensional In Vitro Culture Mimicking the In Vivo-Like Cell Behavior of Bladder Patient-Derived Xenograft Models. Cancers (Basel) 2020; 12:E1304. [PMID: 32455634 PMCID: PMC7281103 DOI: 10.3390/cancers12051304] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022] Open
Abstract
Patient-derived xenograft (PDX) models allow for personalized drug selection and the identification of drug resistance mechanisms in cancer cells. However, PDX models present technical disadvantages, such as long engraftment time, low success rate, and high maintenance cost. On the other hand, tumor spheroids are emerging as an in vitro alternative model that can maintain the phenotype of cancer cells long enough to perform all assays and predict a patient's outcome. The present work aimed to describe a simple, reproducible, and low-cost 3D in vitro culture method to generate bladder tumor spheroids using human cells from PDX mice. Cancer cells from PDX BL0293 and BL0808 models, previously established from advanced bladder cancer, were cultured in 96-well round-bottom ultra-low attachment (ULA) plates with 5% Matrigel and generated regular and round-shaped spheroids (roundness > 0.8) with a diameter larger than 400 μm and a hypoxic core (a feature related to drug resistance in solid tumors). The responses of the tumor spheroids to the antineoplastic drugs cisplatin, gemcitabine, and their combination were similar to tumor responses in in vivo studies with PDX BL0293 and BL0808 mice. Therefore, the in vitro 3D model using PDX tumor spheroids appears as a valuable tool that may predict the outcome of in vivo drug-screening assays and represents a low-cost strategy for such purpose.
Collapse
Affiliation(s)
- Robson Amaral
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, 14040-903 Sao Paulo, Brazil;
| | - Maike Zimmermann
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis Medical School, Sacramento, CA 95817, USA; (M.Z.); (H.Z.); (C.-X.P.)
| | - Ai-Hong Ma
- Department of Urology, UC Davis Comprehensive Cancer Center, University of California Davis Medical School, Sacramento, CA 95817, USA;
| | - Hongyong Zhang
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis Medical School, Sacramento, CA 95817, USA; (M.Z.); (H.Z.); (C.-X.P.)
| | - Kamilla Swiech
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, 14040-903 Sao Paulo, Brazil;
| | - Chong-Xian Pan
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis Medical School, Sacramento, CA 95817, USA; (M.Z.); (H.Z.); (C.-X.P.)
- VA Northern California Health Care system, Mather, CA 95655, USA
- (Current address): Department of Faculty of Medicine, Harvard Medical School, West Roxbury, MA 02115, USA
| |
Collapse
|
114
|
Kuriu S, Kadonosono T, Kizaka-Kondoh S, Ishida T. Slicing Spheroids in Microfluidic Devices for Morphological and Immunohistochemical Analysis. MICROMACHINES 2020; 11:mi11050480. [PMID: 32384758 PMCID: PMC7281316 DOI: 10.3390/mi11050480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Microfluidic devices utilizing spheroids play important roles in in vitro experimental systems to closely simulate morphological and biochemical characteristics of the in vivo tumor microenvironment. For the observation and analysis of the inner structure of spheroids, sectioning is an efficient approach. However, conventional microfluidic devices are difficult for sectioning, and therefore, spheroids inside the microfluidic channels have not been sliced well. We proposed a microfluidic device created from embedding resin for sectioning. Spheroids were cultured, embedded by resin, and sectioned in the microfluidic device. Slices of the sectioned spheroids yielded clear images at the cellular level. According to morphological and immunohistochemical analyses of the slices of the spheroid, specific protein distribution was observed.
Collapse
Affiliation(s)
- Satoru Kuriu
- Department of Mechanical Engineering, School of Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan
- Correspondence: (S.K.); (T.I.)
| | - Tetsuya Kadonosono
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa 226-8503, Japan; (T.K.); (S.K.-K.)
| | - Shinae Kizaka-Kondoh
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa 226-8503, Japan; (T.K.); (S.K.-K.)
| | - Tadashi Ishida
- Department of Mechanical Engineering, School of Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan
- Correspondence: (S.K.); (T.I.)
| |
Collapse
|
115
|
Bhattacharya S, Calar K, de la Puente P. Mimicking tumor hypoxia and tumor-immune interactions employing three-dimensional in vitro models. J Exp Clin Cancer Res 2020; 39:75. [PMID: 32357910 PMCID: PMC7195738 DOI: 10.1186/s13046-020-01583-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
The heterogeneous tumor microenvironment (TME) is highly complex and not entirely understood. These complex configurations lead to the generation of oxygen-deprived conditions within the tumor niche, which modulate several intrinsic TME elements to promote immunosuppressive outcomes. Decoding these communications is necessary for designing effective therapeutic strategies that can effectively reduce tumor-associated chemotherapy resistance by employing the inherent potential of the immune system.While classic two-dimensional in vitro research models reveal critical hypoxia-driven biochemical cues, three-dimensional (3D) cell culture models more accurately replicate the TME-immune manifestations. In this study, we review various 3D cell culture models currently being utilized to foster an oxygen-deprived TME, those that assess the dynamics associated with TME-immune cell penetrability within the tumor-like spatial structure, and discuss state of the art 3D systems that attempt recreating hypoxia-driven TME-immune outcomes. We also highlight the importance of integrating various hallmarks, which collectively might influence the functionality of these 3D models.This review strives to supplement perspectives to the quickly-evolving discipline that endeavors to mimic tumor hypoxia and tumor-immune interactions using 3D in vitro models.
Collapse
Affiliation(s)
- Somshuvra Bhattacharya
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60th Street N, Sioux Falls, SD, 57104, USA
| | - Kristin Calar
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60th Street N, Sioux Falls, SD, 57104, USA
| | - Pilar de la Puente
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60th Street N, Sioux Falls, SD, 57104, USA.
- Department of Surgery, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA.
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, USA.
| |
Collapse
|
116
|
Pucci M, Gomes Ferreira I, Orlandani M, Malagolini N, Ferracin M, Dall’Olio F. High Expression of the Sd a Synthase B4GALNT2 Associates with Good Prognosis and Attenuates Stemness in Colon Cancer. Cells 2020; 9:cells9040948. [PMID: 32290493 PMCID: PMC7226961 DOI: 10.3390/cells9040948] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The carbohydrate antigen Sda and its biosynthetic enzyme B4GALNT2 are highly expressed in normal colonic mucosa but are down-regulated to a variable degree in colon cancer tissues. Here, we investigated the clinical and biological importance of B4GALNT2 in colon cancer. METHODS Correlations of B4GALNT2 mRNA with clinical data were obtained from The Cancer Genome Atlas (TCGA) database; the phenotypic and transcriptomic changes induced by B4GALNT2 were studied in LS174T cells transfected with B4GALNT2 cDNA. RESULTS TCGA data indicate that patients with high B4GALNT2 expression in cancer tissues display longer survival than non-expressers. In LS174T cells, expression of B4GALNT2 did not affect the ability to heal a scratch wound or to form colonies in standard growth conditions but markedly reduced the growth in soft agar, the tridimensional (3D) growth as spheroids, and the number of cancer stem cells, indicating a specific effect of B4GALNT2 on the growth in poor adherence and stemness. On the transcriptome, B4GALNT2 induced the down-regulation of the stemness-associated gene SOX2 and modulated gene expression towards an attenuation of the cancer phenotype. CONCLUSIONS The level of B4GALNT2 can be proposed as a marker to identify higher- and lower-risk colorectal cancer patients.
Collapse
|
117
|
Remsik J, Chi Y, Tong X, Sener U, Derderian C, Park A, Saadeh F, Bale T, Boire A. Leptomeningeal metastatic cells adopt two phenotypic states. Cancer Rep (Hoboken) 2020; 5:e1236. [PMID: 33372403 PMCID: PMC7772527 DOI: 10.1002/cnr2.1236] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/30/2019] [Accepted: 12/30/2019] [Indexed: 01/26/2023] Open
Abstract
Background Leptomeningeal metastasis (LM), or spread of cancer cells into the cerebrospinal fluid (CSF), is characterized by a rapid onset of debilitating neurological symptoms and markedly bleak prognosis. The lack of reproducible in vitro and in vivo models has prevented the development of novel, LM‐specific therapies. Although LM allows for longitudinal sampling of floating cancer cells with a spinal tap, attempts to culture patient‐derived leptomeningeal cancer cells have not been successful. Aim We, therefore, employ leptomeningeal derivatives of human breast and lung cancer cell lines that reproduce both floating and adherent phenotypes of human LM in vivo and in vitro. Methods and Results We introduce a trypsin/EDTA‐based fractionation method to reliably separate the two cell subsets and demonstrate that in vitro cultured floating cells have decreased proliferation rate, lower ATP content, and are enriched in distinct metabolic signatures. Long‐term fractionation and transcriptomic analysis suggest high degree plasticity between the two phenotypes in vitro. Floating cells colonize mouse leptomeninges more rapidly and associate with shortened survival. In addition, patients harboring LM diagnosed with CSF disease alone succumbed to the disease earlier than patients with adherent (MRI positive) disease. Conclusion Together, these data support mechanistic evidence of a metabolic adaptation that allows cancer cells to thrive in their natural environment but leads to death in vitro.
Collapse
Affiliation(s)
- Jan Remsik
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA.,Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Yudan Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA.,Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Xinran Tong
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Ugur Sener
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Camille Derderian
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA.,Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Abigail Park
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Fadi Saadeh
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA.,Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Tejus Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Adrienne Boire
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA.,Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA.,Department of Neurology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| |
Collapse
|
118
|
Sakaguchi R, Mori Y. Transient receptor potential (TRP) channels: Biosensors for redox environmental stimuli and cellular status. Free Radic Biol Med 2020; 146:36-44. [PMID: 31682917 DOI: 10.1016/j.freeradbiomed.2019.10.415] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/26/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022]
Abstract
Transient receptor potential (TRP) channels are a family of cation channels that depolarizes the membrane potential and regulates intracellular concentrations of cations such as Ca2+. TRP channels are also known to function as "biosensors" to detect changes of the surrounding environment and cellular status. Lines of evidence have unveiled that numerous proteins are subject to redox modification and subsequent signaling. For example, TRPM2, TRPC5, TRPV1, and TRPA1 are known as redox sensors activated by hydrogen peroxide (H2O2), nitric oxide (NO), and electrophiles. Thus, these channels facilitate the influx of cations which in turn triggers the appropriate cellular responses against environmental redox stimuli and cellular redox status. In this review, we focus on the recent findings regarding the functions of TRP channels in relation to other ion channels, and other proteins which also go through redox modification of cysteine (Cys) residues. We aim to understand the structural and molecular basis of the redox-sensing mechanisms of TRP channels in exerting various functions under physiological conditions as well as pathological conditions such as cancer malignancy. Their future potential as drug targets will also be discussed.
Collapse
Affiliation(s)
- Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan; The World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 615-8510, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan; The World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 615-8510, Japan.
| |
Collapse
|
119
|
Hypoxia-tropic nanozymes as oxygen generators for tumor-favoring theranostics. Biomaterials 2019; 230:119635. [PMID: 31767443 DOI: 10.1016/j.biomaterials.2019.119635] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/22/2019] [Accepted: 11/16/2019] [Indexed: 12/16/2022]
Abstract
Oxygen deficiency is the main obstacle of hypoxia-related theranostics, thus this is a considerable amount of research focusing on the development of methods to supply oxygen by taking advantage of hypoxia-responsive properties of nanoparticles. However, strategies to properly penetrate hypoxic regions by the nanoparticles remains an unmet challenge. In this work, a biomimetic nanozyme capable of possessing catalase-like activity and the efficient direct penetration of hypoxic areas in tumor tissues was developed to supply oxygen based on catalytic tumor microenvironment-responsive reaction, providing substantial tumor hypoxia relief with nearly 3-fold reduction compared to untreated tumor tissues. To demonstrate the advantages of the nanozymes in overcoming hypoxia, a theranostic nanosystem model composed of the core/shell nanozymes and aggregation-induced emission (AIE) molecules was designed. The nanosystem was able to present multi-modal imaging of tumors and modulated the tumor microenvironment for improved photodynamic therapy (PDT) by cascade reactions of therapeutic effector molecules, thereby providing significantly enhanced therapeutic benefits in inhibiting tumor growth and lung metastasis of orthotopic breast cancer. This conceptual study showed the multifaceted features of biomimetic nanozymes as tumor therapeutics and demonstrated the encouraging potential for modulating hypoxia as an application for tumor theranostics.
Collapse
|
120
|
Leedale J, Colley HE, Gaskell H, Williams DP, Bearon RN, Chadwick AE, Murdoch C, Webb SD. In silico-guided optimisation of oxygen gradients in hepatic spheroids. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.comtox.2019.100093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
121
|
Ayuso JM, Virumbrales-Munoz M, McMinn PH, Rehman S, Gomez I, Karim MR, Trusttchel R, Wisinski KB, Beebe DJ, Skala MC. Tumor-on-a-chip: a microfluidic model to study cell response to environmental gradients. LAB ON A CHIP 2019; 19:3461-3471. [PMID: 31506657 PMCID: PMC6785375 DOI: 10.1039/c9lc00270g] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Limited blood supply and rapid tumor metabolism within solid tumors leads to nutrient starvation, waste product accumulation and the generation of pH gradients across the tumor mass. These environmental conditions modify multiple cellular functions, including metabolism, proliferation, and drug response. However, capturing the spatial metabolic and phenotypic heterogeneity of the tumor with classic in vitro models remains challenging. Thus, in this work a microfluidic tumor slice model was developed to study cell behavior under metabolic starvation gradients. The presented microdevice comprises a central chamber where tumor cells were cultured in a 3D collagen hydrogel. A lumen on the flank of the chamber was used to perfuse media, mimicking the vasculature. Under these circumstances, tumor cell metabolism led to the generation of viability, proliferation and pH gradients. The model decoupled the influence of oxygen from other nutrients, revealing that cell necrosis at the core of the model could be explained by nutrient starvation. The microdevice can be disassembled to retrieve the cells from the desired locations to study molecular adaptions due to nutrient starvation. When exposed to these pH gradients and low nutrient conditions, cancer cells showed multiple changes in their gene expression profile depending on their distance from the lumen. Those cells located further from the lumen upregulated several genes related to stress and survival response, whereas genes related to proliferation and DNA repair were downregulated. This model may help to identify new therapeutic opportunities to target the metabolic heterogeneity observed in solid tumors.
Collapse
Affiliation(s)
- Jose M. Ayuso
- Morgridge Institute for Research, 330 N Orchard street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Maria Virumbrales-Munoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Patrick H. McMinn
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Shujah Rehman
- Morgridge Institute for Research, 330 N Orchard street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Ismael Gomez
- Allergy research group, IdISSC. San Carlos Clinic Hospital, Madrid, Spain
- Materials department, Carlos III University. Leganes, Spain
| | - Mohammad R. Karim
- Morgridge Institute for Research, 330 N Orchard street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Regan Trusttchel
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Kari B. Wisinski
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - David J. Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI,USA
| | - Melissa C. Skala
- Morgridge Institute for Research, 330 N Orchard street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
122
|
Labuschagne CF, Cheung EC, Blagih J, Domart MC, Vousden KH. Cell Clustering Promotes a Metabolic Switch that Supports Metastatic Colonization. Cell Metab 2019; 30:720-734.e5. [PMID: 31447323 PMCID: PMC6863392 DOI: 10.1016/j.cmet.2019.07.014] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/30/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022]
Abstract
Cancer metastasis depends on cell survival following loss of extracellular matrix attachment and dissemination through the circulation. The metastatic spread can be enhanced by the clustering of detached cancer cells and increased antioxidant defense. Here, we link these responses by describing how cell clustering limits reactive oxygen species (ROS). Loss of attachment causes mitochondrial perturbations and increased ROS production. The formation of cell clusters induces a hypoxic environment that drives hypoxia-inducible factor 1-alpha (Hif1α)-mediated mitophagy, clearing damaged mitochondria and limiting ROS. However, hypoxia and reduced mitochondrial capacity promote dependence on glycolysis for ATP production that is supported by cytosolic reductive metabolism. Preventing this metabolic adaptation or disruption of cell clusters results in ROS accumulation, cell death, and a reduction of metastatic capacity in vivo. Our results provide a mechanistic explanation for the role of cell clustering in supporting survival during extracellular matrix detachment and metastatic spread and may point to targetable vulnerabilities.
Collapse
Affiliation(s)
| | - Eric C Cheung
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Julianna Blagih
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | - Karen H Vousden
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
123
|
Moreira MP, Brayner FA, Alves LC, Cassali GD, Silva LM. Phenotypic, structural, and ultrastructural analysis of triple-negative breast cancer cell lines and breast cancer stem cell subpopulation. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2019; 48:673-684. [PMID: 31485678 DOI: 10.1007/s00249-019-01393-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/24/2019] [Accepted: 08/12/2019] [Indexed: 12/24/2022]
Abstract
Triple negative breast cancer (TNBC) is a highly heterogeneous disease, which influences the therapeutic response and makes difficult the discovery of effective targets. This heterogeneity is attributed to the presence of breast cancer stem cells (BCSCs), which determines resistance to chemotherapy and subsequently disease recurrence and metastasis. In this context, this work aimed to evaluate the morphological and phenotypic cellular heterogeneity of two TNBC cell lines cultured in monolayer and tumorsphere (TS) models by fluorescence and electron microscopy and flow cytometry. The BT-549 and Hs 578T analyses demonstrated large phenotypic and morphological heterogeneity between these cell lines, as well as between the cell subpopulations that compose them. BT-549 and Hs 578T are heterogeneous considering the cell surface marker CD44 and CD24 expression, characterizing BCSC mesenchymal-like cells (CD44+/CD24-), epithelial cells (CD44-/CD24+), hybrid cells with mesenchymal and epithelial features (CD44+/CD24+), and CD44-/CD24- cells. BCSC epithelial-like cells (ALDH+) were found in BT-549, BT-549 TS, and Hs 578T TS; however, only BT-549 TS showed a high ALDH activity. Ultrastructural characterization showed the heterogeneity within and among BT-549 and Hs 578T in monolayer and TS models being formed by more than one cellular type. Further, the mesenchymal characteristic of these cells is demonstrated by E-cadherin absence and filopodia. It is well known that tumor cell heterogeneity can influence survival, therapy responses, and the rate of tumor growth. Thus, molecular understanding of this heterogeneity is essential for the identification of potential therapeutic options and vulnerabilities of oncological patients.
Collapse
Affiliation(s)
- Milene Pereira Moreira
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil
- Laboratório de Patologia Comparada, Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Fábio André Brayner
- Laboratório de Imunopatologia Keizo Asami (LIKA), Universidade Federal de Pernambuco (UFPE), Avenida Professor Moraes Rego, s/n, Campus da UFPE, Cidade Universitária, Recife, Pernambuco, 50740-465, Brazil
- Instituto Aggeu Magalhães (IAM), Fundação Oswaldo Cruz (FIOCRUZ), Avenida Professor Moraes Rego, s/n, Campus da UFPE, Cidade Universitária, Recife, Pernambuco, 50670-420, Brazil
| | - Luiz Carlos Alves
- Laboratório de Imunopatologia Keizo Asami (LIKA), Universidade Federal de Pernambuco (UFPE), Avenida Professor Moraes Rego, s/n, Campus da UFPE, Cidade Universitária, Recife, Pernambuco, 50740-465, Brazil
- Instituto Aggeu Magalhães (IAM), Fundação Oswaldo Cruz (FIOCRUZ), Avenida Professor Moraes Rego, s/n, Campus da UFPE, Cidade Universitária, Recife, Pernambuco, 50670-420, Brazil
| | - Geovanni Dantas Cassali
- Laboratório de Patologia Comparada, Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Luciana Maria Silva
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil.
| |
Collapse
|
124
|
Twarock S, Reichert C, Bach K, Reiners O, Kretschmer I, Gorski DJ, Gorges K, Grandoch M, Fischer JW. Inhibition of the hyaluronan matrix enhances metabolic anticancer therapy by dichloroacetate in vitro and in vivo. Br J Pharmacol 2019; 176:4474-4490. [PMID: 31351004 PMCID: PMC6932941 DOI: 10.1111/bph.14808] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022] Open
Abstract
Background and Purpose Aerobic glycolysis is a unique feature of tumour cells that entails several advantages for cancer progression such as resistance to apoptosis. The low MW compound, dichloroacetate, is a pyruvate dehydrogenase kinase inhibitor, which restores oxidative phosphorylation and induces apoptosis in a variety of cancer entities. However, its therapeutic effectiveness is limited by resistance mechanisms. This study aimed to examine the role of the anti‐apoptotic hyaluronan (HA) matrix in this context and to identify a potential add‐on treatment option to overcome this limitation. Experimental Approach The metabolic connection between dichloroacetate treatment and HA matrix augmentation was analysed in vitro by quantitative PCR and affinity cytochemistry. Metabolic pathways were analysed using Seahorse, HPLC, fluorophore‐assisted carbohydrate electrophoresis, colourimetry, immunoblots, and immunochemistry. The effects of combining dichloroacetate with the HA synthesis inhibitor 4‐methylumbelliferone was evaluated in 2D and 3D cell cultures and in a nude mouse tumour xenograft regression model by immunoblot, immunochemistry, and FACS analysis. Key Results Mitochondrial reactivation induced by dichloroacetate metabolically activated HA synthesis by augmenting precursors as well as O‐GlcNAcylation. This process was blocked by 4‐methylumbelliferone, resulting in enhanced anti‐tumour efficacy in 2D and 3D cell culture and in a nude mouse tumour xenograft regression model. Conclusions and Implications The HA rich tumour micro‐environment represents a metabolic factor contributing to chemotherapy resistance. HA synthesis inhibition exhibited pronounced synergistic actions with dichloroacetate treatment on oesophageal tumour cell proliferation and survival in vitro and in vivo suggesting the combination of these two strategies is an effective anticancer therapy.
Collapse
Affiliation(s)
- Sören Twarock
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Christina Reichert
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Katharina Bach
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Oliver Reiners
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Inga Kretschmer
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Daniel J Gorski
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Katharina Gorges
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Maria Grandoch
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jens W Fischer
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| |
Collapse
|
125
|
Sheth DB, Gratzl M. Electrochemical mapping of oxygenation in the three-dimensional multicellular tumour hemi-spheroid. Proc Math Phys Eng Sci 2019; 475:20180647. [PMID: 31236040 PMCID: PMC6545061 DOI: 10.1098/rspa.2018.0647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/29/2019] [Indexed: 01/09/2023] Open
Abstract
Blood capillaries deliver oxygen and nutrients to surrounding micro-regions of tissue and carry away metabolic waste. In normal tissue, capillaries are close enough to keep all the cells viable. In solid tumours, the capillary system is chaotic and typical inter-capillary distances are larger than in normal tissue. Therefore, hypoxic regions develop. Drug molecules may not reach these areas at concentrations above the lethal level. The combined effect of low drug concentrations and local hypoxia, often exacerbated by acidity, leads to therapy failure. To better understand the interplay between hypoxia and poor drug penetration, oxygenation needs to be assessed in different areas of inter-capillary tissue. The multicellular tumour spheroid is a well-established three-dimensional (3D) in vitro model of the capillary microenvironment. It is used to mimic nascent tumours and micro-metastases as well. In this work, we demonstrate for the first time that dynamic intra-spheroidal oxygen maps can be obtained at the 3D multicellular tumour hemi-spheroid (MCH) using a non-invasive microelectrode array. The same oxygen distributions exist inside the equivalent but less accessible full spheroid. The MCH makes high throughput-high content analysis of spheroids feasible and thus can assist studies on basic cancer biology, drug development and personalized medicine.
Collapse
Affiliation(s)
| | - Miklόs Gratzl
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
126
|
Di Modugno F, Colosi C, Trono P, Antonacci G, Ruocco G, Nisticò P. 3D models in the new era of immune oncology: focus on T cells, CAF and ECM. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:117. [PMID: 30898166 PMCID: PMC6429763 DOI: 10.1186/s13046-019-1086-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitor therapy has changed clinical practice for patients with different cancers, since these agents have demonstrated a significant improvement of overall survival and are effective in many patients. However, an intrinsic or acquired resistance frequently occur and biomarkers predictive of responsiveness should help in patient selection and in defining the adequate treatment options. A deep analysis of the complexity of the tumor microenvironment is likely to further advance the field and hopefully identify more effective combined immunotherapeutic strategies. Here we review the current knowledge on tumor microenvironment, focusing on T cells, cancer associated fibroblasts and extracellular matrix. The use of 3D cell culture models to resemble tumor microenvironment landscape and to screen immunomodulatory drugs is also reviewed.
Collapse
Affiliation(s)
- Francesca Di Modugno
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS-Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy.
| | - Cristina Colosi
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Paola Trono
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS-Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy
| | - Giuseppe Antonacci
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Paola Nisticò
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS-Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
127
|
Badea MA, Balas M, Hermenean A, Ciceu A, Herman H, Ionita D, Dinischiotu A. Influence of Matrigel on Single- and Multiple-Spheroid Cultures in Breast Cancer Research. SLAS DISCOVERY 2019; 24:563-578. [PMID: 30897015 DOI: 10.1177/2472555219834698] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study aimed to develop and compare single and multiple 3D models such as multicellular tumor spheroids and to investigate the influence of Matrigel on their morphological and functional behavior. MDA-MB-231 3D models were generated in the presence and absence of Matrigel and their key biological properties within 6 days of culture were monitored. Our results revealed the formation of well-defined 3D models in the presence of Matrigel, with a uniform morphology, increased diameter, good circularity, and increased expression of a proliferation marker (PCNA). In comparison, 3D models generated without Matrigel were characterized by an irregular border, reduced dimensions and circularity, and a decrease of PCNA expression. Similarities between the single and multiple 3D cultures were found in their viability, Nrf2 expression, and glutathione (GSH) content. The influence of Matrigel on MDA-MB-231 spheroids metabolism under hypoxic conditions was highlighted by released lactate dehydrogenase and nitric oxide, GSH levels and expression of Nrf2 and Hsp70 proteins. Based on the increased expression of PCNA and the development of the hypoxia process in the presence of extracellular matrix, our study showed that the addition of Matrigel improves the growing environment of tumor spheroids, making it closer to that of in vivo tumor conditions.
Collapse
Affiliation(s)
- Madalina Andreea Badea
- 1 Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Mihaela Balas
- 1 Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Anca Hermenean
- 2 Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, Arad, Romania.,3 Department of Experimental and Applied Biology, Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Alina Ciceu
- 3 Department of Experimental and Applied Biology, Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Hildegard Herman
- 3 Department of Experimental and Applied Biology, Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Daniela Ionita
- 4 Department of General Chemistry, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, Bucharest, Romania
| | - Anca Dinischiotu
- 1 Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
128
|
Movia D, Bazou D, Volkov Y, Prina-Mello A. Multilayered Cultures of NSCLC cells grown at the Air-Liquid Interface allow the efficacy testing of inhaled anti-cancer drugs. Sci Rep 2018; 8:12920. [PMID: 30150787 PMCID: PMC6110800 DOI: 10.1038/s41598-018-31332-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/12/2018] [Indexed: 12/11/2022] Open
Abstract
Evidence supports the advantages of inhalation over other drug-administration routes in the treatment of lung diseases, including cancer. Although data obtained from animal models and conventional in vitro cultures are informative, testing the efficacy of inhaled chemotherapeutic agents requires human-relevant preclinical tools. Such tools are currently unavailable. Here, we developed and characterized in vitro models for the efficacy testing of inhaled chemotherapeutic agents against non-small-cell lung cancer (NSCLC). These models recapitulated key elements of both the lung epithelium and the tumour tissue, namely the direct contact with the gas phase and the three-dimensional (3D) architecture. Our in vitro models were formed by growing, for the first time, human adenocarcinoma (A549) cells as multilayered mono-cultures at the Air-Liquid Interface (ALI). The in vitro models were tested for their response to four benchmarking chemotherapeutics, currently in use in clinics, demonstrating an increased resistance to these drugs as compared to sub-confluent monolayered 2D cell cultures. Chemoresistance was comparable to that detected in 3D hypoxic tumour spheroids. Being cultured in ALI conditions, the multilayered monocultures demonstrated to be compatible with testing drugs administered as a liquid aerosol by a clinical nebulizer, offering an advantage over 3D tumour spheroids. In conclusion, we demonstrated that our in vitro models provide new human-relevant tools allowing for the efficacy screening of inhaled anti-cancer drugs.
Collapse
Affiliation(s)
- Dania Movia
- Department of Clinical Medicine/Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland.
| | - Despina Bazou
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Yuri Volkov
- Department of Clinical Medicine/Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- AMBER Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
- Department of Histology, Cytology and Embryology, First Moscow State Sechenov Medical University, Moskva, Russian Federation
| | - Adriele Prina-Mello
- Department of Clinical Medicine/Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- AMBER Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
129
|
Lukina MM, Dudenkova VV, Ignatova NI, Druzhkova IN, Shimolina LE, Zagaynova EV, Shirmanova MV. Metabolic cofactors NAD(P)H and FAD as potential indicators of cancer cell response to chemotherapy with paclitaxel. Biochim Biophys Acta Gen Subj 2018; 1862:1693-1700. [PMID: 29719197 DOI: 10.1016/j.bbagen.2018.04.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 10/17/2022]
Abstract
Paclitaxel, a widely used antimicrotubular agent, predominantly eliminates rapidly proliferating cancer cells, while slowly proliferating and quiescent cells can survive the treatment, which is one of the main reasons for tumor recurrence and non-responsiveness to the drug. To improve the efficacy of chemotherapy, biomarkers need to be developed to enable monitoring of tumor responses. In this study we considered the auto-fluorescent metabolic cofactors NAD(P)H and FAD as possible indicators of cancer cell response to therapy with paclitaxel. It was found that, among the tested parameters (the fluorescence intensity-based redox ratio FAD/NAD(P)H, and the fluorescence lifetimes of NAD(P)H and FAD), the fluorescence lifetime of NAD(P)H is the most sensitive in tracking the drug response, and is capable of indicating heterogeneous cellular responses both in cell monolayers and in multicellular tumor spheroids. We observed that metabolic reorganization to a more oxidative state preceded the morphological manifestation of cell death and developed faster in cells that were more responsive to the drug. Our results suggest that noninvasive, label-free monitoring of the drug-induced metabolic changes by noting the NAD(P)H fluorescence lifetime is a valuable approach to characterize the responses of cancer cells to anti-cancer treatments and, therefore, to predict the effectiveness of chemotherapy.
Collapse
Affiliation(s)
- Maria M Lukina
- Institute of Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Varvara V Dudenkova
- Institute of Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Nadezhda I Ignatova
- Institute of Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia
| | - Irina N Druzhkova
- Institute of Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia
| | - Lyubov' E Shimolina
- Institute of Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Elena V Zagaynova
- Institute of Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia
| | - Marina V Shirmanova
- Institute of Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia.
| |
Collapse
|
130
|
Carranza-Rosales P, Guzmán-Delgado NE, Carranza-Torres IE, Viveros-Valdez E, Morán-Martínez J. Breast Organotypic Cancer Models. Curr Top Microbiol Immunol 2018:199-223. [PMID: 29556825 DOI: 10.1007/82_2018_86] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most common cancer type diagnosed in women, it represents a critical public health problem worldwide, with 1,671,149 estimated new cases and nearly 571,000 related deaths. Research on breast cancer has mainly been conducted using two-dimensional (2D) cell cultures and animal models. The usefulness of these models is reflected in the vast knowledge accumulated over the past decades. However, considering that animal models are three-dimensional (3D) in nature, the validity of the studies using 2D cell cultures has recently been questioned. Although animal models are important in cancer research, ethical questions arise about their use and usefulness as there is no clear predictivity of human disease outcome and they are very expensive and take too much time to obtain results. The poor performance or failure of most cancer drugs suggests that preclinical research on cancer has been based on an over-dependence on inadequate animal models. For these reasons, in the last few years development of alternative models has been prioritized to study human breast cancer behavior, while maintaining a 3D microenvironment, and to reduce the number of experiments conducted in animals. One way to achieve this is using organotypic cultures, which are being more frequently explored in cancer research because they mimic tissue architecture in vivo. These characteristics make organotypic cultures a valuable tool in cancer research as an alternative to replace animal models and for predicting risk assessment in humans. This chapter describes the cultures of multicellular spheroids, organoids, 3D bioreactors, and tumor slices, which are the most widely used organotypic models in breast cancer research.
Collapse
Affiliation(s)
- Pilar Carranza-Rosales
- Departamento de Biología Celular y Molecular, Instituto Mexicano del Seguro Social. Centro de Investigación Biomédica del Noreste, Monterrey, Nuevo León, Mexico.
| | - Nancy Elena Guzmán-Delgado
- Unidad Médica de Alta Especialidad # 34, División de Investigación, Instituto Mexicano del Seguro Social, Monterrey, Nuevo León, Mexico
| | - Irma Edith Carranza-Torres
- Departamento de Biología Celular y Molecular, Instituto Mexicano del Seguro Social. Centro de Investigación Biomédica del Noreste, Monterrey, Nuevo León, Mexico
| | - Ezequiel Viveros-Valdez
- Departamento de Química Analítica, Ciudad Universitaria, Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, San Nicolás de los Garza, Nuevo León, Mexico
| | - Javier Morán-Martínez
- Departamento de Biología Celular y Ultraestructura, Universidad Autónoma de Coahuila, Facultad de Medicina. Centro de Investigación Biomédica, Torreón, Coahuila, Mexico
| |
Collapse
|
131
|
Abstract
In 2012, cancer affected 14.1 million people worldwide and was responsible for 8.2 million deaths. The disease predominantly affects aged populations and is one of the leading causes of death in most western countries. In tumors, the aggressive growth of the neoplastic cell population and associated overexpression of pro-angiogenic factors lead to the development of disorganized blood vessel networks that are structurally and functionally different from normal vasculature. A disorganized labyrinth of vessels that are immature, tortuous and hyperpermeable typifies tumor vasculature. Functionally, the ability of the tumor vasculature to deliver nutrients and remove waste products is severely diminished. A critical consequence of the inadequate vascular networks in solid tumors is the development of regions of hypoxia [low oxygen tensions typically defined as oxygen tensions (pO2 values) < 10 mm Hg]. Tumor cells existing in such hypoxic environments have long been known to be resistant to anticancer therapy, display an aggressive phenotype, and promote tumor progression and dissemination. This review discusses the physiological basis of hypoxia, methods of detection, and strategies to overcome the resulting therapy resistance.
Collapse
Affiliation(s)
- Veronica S Hughes
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| | - Jennifer M Wiggins
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| | - Dietmar W Siemann
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| |
Collapse
|