1
|
Li S, Qiu J, Guo Z, Gao Q, Huang CY, Hao Y, Hu Y, Liang T, Zhai M, Zhang Y, Nie B, Chang WJ, Wang W, Xi R, Wei R. Formation and culture of cell spheroids by using magnetic nanostructures resembling a crown of thorns. Biofabrication 2024; 16:045018. [PMID: 39053493 DOI: 10.1088/1758-5090/ad6794] [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: 09/07/2023] [Accepted: 07/25/2024] [Indexed: 07/27/2024]
Abstract
In contrast to traditional two-dimensional cell-culture conditions, three-dimensional (3D) cell-culture models closely mimic complexin vivoconditions. However, constructing 3D cell culture models still faces challenges. In this paper, by using micro/nano fabrication method, including lithography, deposition, etching, and lift-off, we designed magnetic nanostructures resembling a crown of thorns. This magnetic crown of thorns (MCT) nanostructure enables the isolation of cells that have endocytosed magnetic particles. To assess the utility of this nanostructure, we used high-flux acquisition of Jurkat cells, an acute-leukemia cell line exhibiting the native phenotype, as an example. The novel structure enabled Jurkat cells to form spheroids within just 30 min by leveraging mild magnetic forces to bring together endocytosed magnetic particles. The size, volume, and arrangement of these spheroids were precisely regulated by the dimensions of the MCT nanostructure and the array configuration. The resulting magnetic cell clusters were uniform in size and reached saturation after 1400 s. Notably, these cell clusters could be easily separated from the MCT nanostructure through enzymatic digestion while maintaining their integrity. These clusters displayed a strong proliferation rate and survival capabilities, lasting for an impressive 96 h. Compared with existing 3D cell-culture models, the approach presented in this study offers the advantage of rapid formation of uniform spheroids that can mimicin vivomicroenvironments. These findings underscore the high potential of the MCT in cell-culture models and magnetic tissue enginerring.
Collapse
Affiliation(s)
- Shijiao Li
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Jingjiang Qiu
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
- Industrial Technology Research Institute, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Zhongwei Guo
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Qiulei Gao
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Chen-Yu Huang
- Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD, United States of America
| | - Yilin Hao
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Yifan Hu
- Industrial Technology Research Institute, Zhengzhou University, Zhengzhou 450001, People's Republic of China
- Henan Spring Biotechnology Ltd Company, Zhengzhou 450001, People's Republic of China
- Division of Logistics, Weistron Co., Ltd, Zhengzhou 450001, People's Republic of China
| | - Tianshui Liang
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Ming Zhai
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Yudong Zhang
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Bangbang Nie
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Wei-Jen Chang
- Department of Biology, Hamilton College, Clinton, NY, United States of America
| | - Wen Wang
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Rui Xi
- School of Mechanical Engineering, North China University of Water Resources and Electric Power, Zhengzhou 450046, People's Republic of China
| | - Ronghan Wei
- Engineering Technology Research Center of Henan Province for MEMS Manufacturing and Applications, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, People's Republic of China
- Industrial Technology Research Institute, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| |
Collapse
|
2
|
Meng J, Xu X, Jiang C, Xia P, Xu P, Tian L, Xu Y, Li D, Tan Y, Ji B. Tensile force field plays a crucial role in local invasion of tumor cells through a mechano-chemical coupling mechanism. SOFT MATTER 2024. [PMID: 39027971 DOI: 10.1039/d4sm00335g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cancer metastasis starts from early local invasion, during which tumor cells detach from the primary tumor, penetrate the extracellular matrix (ECM), and then invade neighboring tissues. However, the cellular mechanics in the detaching and penetrating processes have not been fully understood, and the underlying mechanisms that influence cell polarization and migration in the 3D matrix during tumor invasion remain largely unknown. In this study, we employed a dual tumor-spheroid model to investigate the cellular mechanisms of the tumor invasion. Our results revealed that the tensional force field developed by the active contraction of cells and tissues played a pivotal role in tumor invasion, acting as the driving force for remodeling the collagen fibers during the invasion process. The remodeled collagen fibers promoted cell polarization and migration because of the stiffening of the fiber matrix. The aligned fibers facilitated tumor cell invasion and directed migration from one spheroid to the other. Inhibiting/shielding the cellular contractility abolished matrix remodeling and re-alignment and significantly decreased tumor cell invasion. By developing a coarse-grained cell model that considers the mutual interaction between cells and fibers, we predicted the tensional force field in the fiber network and the associated cell polarization and cell-matrix interaction during cell invasion, which revealed a mechano-chemical coupling mechanism at the cellular level of the tumor invasion process. Our study highlights the roles of cellular mechanics at the early stage of tumor metastasis and may provide new therapeutic strategies for cancer therapy.
Collapse
Affiliation(s)
- Jianfeng Meng
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xiangyu Xu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chaohui Jiang
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Peng Xia
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Science Institute, Zhejiang University, Hangzhou 310058, China
| | - Pengfei Xu
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Liangfei Tian
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yingke Xu
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Dechang Li
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
| | - Youhua Tan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Baohua Ji
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
3
|
Jahin I, Phillips T, Marcotti S, Gorey MA, Cox S, Parsons M. Extracellular matrix stiffness activates mechanosensitive signals but limits breast cancer cell spheroid proliferation and invasion. Front Cell Dev Biol 2023; 11:1292775. [PMID: 38125873 PMCID: PMC10731024 DOI: 10.3389/fcell.2023.1292775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Breast cancer is characterized by physical changes that occur in the tumor microenvironment throughout growth and metastasis of tumors. Extracellular matrix stiffness increases as tumors develop and spread, with stiffer environments thought to correlate with poorer disease prognosis. Changes in extracellular stiffness and other physical characteristics are sensed by integrins which integrate these extracellular cues to intracellular signaling, resulting in modulation of proliferation and invasion. However, the co-ordination of mechano-sensitive signaling with functional changes to groups of tumor cells within 3-dimensional environments remains poorly understood. Here we provide evidence that increasing the stiffness of collagen scaffolds results in increased activation of ERK1/2 and YAP in human breast cancer cell spheroids. We also show that ERK1/2 acts upstream of YAP activation in this context. We further demonstrate that YAP, matrix metalloproteinases and actomyosin contractility are required for collagen remodeling, proliferation and invasion in lower stiffness scaffolds. However, the increased activation of these proteins in higher stiffness 3-dimensional collagen gels is correlated with reduced proliferation and reduced invasion of cancer cell spheroids. Our data collectively provide evidence that higher stiffness 3-dimensional environments induce mechano-signaling but contrary to evidence from 2-dimensional studies, this is not sufficient to promote pro-tumorigenic effects in breast cancer cell spheroids.
Collapse
Affiliation(s)
| | | | | | | | | | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| |
Collapse
|
4
|
Ren T, Maitusong M, Zhou X, Hong X, Cheng S, Lin Y, Xue J, Xu D, Chen J, Qian Y, Lu Y, Liu X, Zhu Y, Wang J. Programing Cell Assembly via Ink-Free, Label-Free Magneto-Archimedes Based Strategy. ACS NANO 2023; 17:12072-12086. [PMID: 37363813 DOI: 10.1021/acsnano.2c10704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Tissue engineering raised a high requirement to control cell distribution in defined materials and structures. In "ink"-based bioprintings, such as 3D printing and photolithography, cells were associated with inks for spatial orientation; the conditions suitable for one ink are hard to apply on other inks, which increases the obstacle in their universalization. The Magneto-Archimedes effect based (Mag-Arch) strategy can modulate cell locomotion directly without impelling inks. In a paramagnetic medium, cells were repelled from high magnetic strength zones due to their innate diamagnetism, which is independent of substrate properties. However, Mag-Arch has not been developed into a powerful bioprinting strategy as its precision, complexity, and throughput are limited by magnetic field distribution. By controlling the paramagnetic reagent concentration in the medium and the gaps between magnets, which decide the cell repelling scope of magnets, we created simultaneously more than a hundred micrometer scale identical assemblies into designed patterns (such as alphabets) with single/multiple cell types. Cell patterning models for cell migration and immune cell adhesion studies were conveniently created by Mag-Arch. As a proof of concept, we patterned a tumor/endothelial coculture model within a covered microfluidic channel to mimic epithelial-mesenchymal transition (EMT) under shear stress in a cancer pathological environment, which gave a potential solution to pattern multiple cell types in a confined space without any premodification. Overall, our Mag-Arch patterning presents an alternative strategy for the biofabrication and biohybrid assembly of cells with biomaterials featured in controlled distribution and organization, which can be broadly employed in tissue engineering, regenerative medicine, and cell biology research.
Collapse
Affiliation(s)
- Tanchen Ren
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Miribani Maitusong
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Xuhao Zhou
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Xiaoqian Hong
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Si Cheng
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yin Lin
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Junhui Xue
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Dilin Xu
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Jinyong Chen
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yi Qian
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yuwen Lu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China
| | - Xianbao Liu
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yang Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China
| | - Jian'an Wang
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| |
Collapse
|
5
|
Hedrich V, Breitenecker K, Ortmayr G, Pupp F, Huber H, Chen D, Sahoo S, Jolly MK, Mikulits W. PRAME Is a Novel Target of Tumor-Intrinsic Gas6/Axl Activation and Promotes Cancer Cell Invasion in Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:2415. [PMID: 37173882 PMCID: PMC10177160 DOI: 10.3390/cancers15092415] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
(1) Background: Activation of the receptor tyrosine kinase Axl by Gas6 fosters oncogenic effects in hepatocellular carcinoma (HCC), associating with increased mortality of patients. The impact of Gas6/Axl signaling on the induction of individual target genes in HCC and its consequences is an open issue. (2) Methods: RNA-seq analysis of Gas6-stimulated Axl-proficient or Axl-deficient HCC cells was used to identify Gas6/Axl targets. Gain- and loss-of-function studies as well as proteomics were employed to characterize the role of PRAME (preferentially expressed antigen in melanoma). Expression of Axl/PRAME was assessed in publicly available HCC patient datasets and in 133 HCC cases. (3) Results: Exploitation of well-characterized HCC models expressing Axl or devoid of Axl allowed the identification of target genes including PRAME. Intervention with Axl signaling or MAPK/ERK1/2 resulted in reduced PRAME expression. PRAME levels were associated with a mesenchymal-like phenotype augmenting 2D cell migration and 3D cell invasion. Interactions with pro-oncogenic proteins such as CCAR1 suggested further tumor-promoting functions of PRAME in HCC. Moreover, PRAME showed elevated expression in Axl-stratified HCC patients, which correlates with vascular invasion and lowered patient survival. (4) Conclusions: PRAME is a bona fide target of Gas6/Axl/ERK signaling linked to EMT and cancer cell invasion in HCC.
Collapse
Affiliation(s)
- Viola Hedrich
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Kristina Breitenecker
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Gregor Ortmayr
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Franziska Pupp
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Heidemarie Huber
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Doris Chen
- Department of Chromosome Biology, Max Perutz Labs Vienna, University of Vienna, 1030 Vienna, Austria
| | - Sarthak Sahoo
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Wolfgang Mikulits
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| |
Collapse
|
6
|
Kenney RM, Lee MC, Boyce MW, Sitte ZR, Lockett MR. Cellular Invasion Assay for the Real-Time Tracking of Individual Cells in Spheroid or Tumor-like Mimics. Anal Chem 2023; 95:3054-3061. [PMID: 36701161 PMCID: PMC10007898 DOI: 10.1021/acs.analchem.2c05201] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cellular invasion is the gateway to metastasis, with cells moving from a primary tumor into neighboring regions of healthy tissue. Invasion assays provide a tractable experimental platform to quantitatively assess cellular movement in the presence of potential chemokines or inhibitors. Many such assays involve cellular movement from high cell densities to cell-free regions. To improve the physiological relevance of such assays, we developed an assay format to track cellular movement throughout a uniform density of cells. This assay format imparts diffusion-dominated environments along the channel, resulting in oxygen and nutrient gradients found in spheroids or poorly vascularized tumors. By incorporating oxygen- and pH-sensing films, we quantified spatial and temporal changes in the extracellular environment while simultaneously tracking the movement of a subset of cells engineered to express fluorescent proteins constitutively. Our results show the successful invasion into neighboring tissues likely arises from a small population with a highly invasive phenotype. These highly invasive cells continued to move throughout the 48 h experiment, suggesting they have stem-like or persister properties. Surprisingly, the distance these persister cells invaded was unaffected by the density of cells in the channel or the presence or absence of an oxygen gradient. While these datasets cannot determine if the invasive cells are inherent to the population or if diffusion-dominated environments promote them, they highlight the need for further study.
Collapse
Affiliation(s)
- Rachael M. Kenney
- Department of Chemistry, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, NC 27599-3290
| | - Maggie C. Lee
- Department of Chemistry, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, NC 27599-3290
| | - Matthew W. Boyce
- Department of Chemistry, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, NC 27599-3290
| | - Zachary R. Sitte
- Department of Chemistry, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, NC 27599-3290
| | - Matthew R. Lockett
- Department of Chemistry, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, NC 27599-3290
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, NC 27599-7295
| |
Collapse
|
7
|
Tutty MA, Prina-Mello A. Three-Dimensional Spheroids for Cancer Research. Methods Mol Biol 2023; 2645:65-103. [PMID: 37202612 DOI: 10.1007/978-1-0716-3056-3_3] [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: 05/20/2023]
Abstract
In vitro cell culture is one of the most widely used tools used today for increasing our understanding of various things such as protein production, mechanisms of drug action, tissue engineering, and overall cellular biology. For the past decades, however, cancer researchers have relied heavily on conventional two-dimensional (2D) monolayer culture techniques to test a variety of aspects of cancer research ranging from the cytotoxic effects of antitumor drugs to the toxicity of diagnostic dyes and contact tracers. However, many promising cancer therapies have either weak or no efficacy in real-life conditions, therefore delaying or stopping altogether their translating to the clinic. This is, in part, due to the reductionist 2D cultures used to test these materials, which lack appropriate cell-cell contacts, have altered signaling, do not represent the natural tumor microenvironment, and have different drug responses, due to their reduced malignant phenotype when compared to real in vivo tumors. With the most recent advances, cancer research has moved into 3D biological investigation. Three-dimensional (3D) cultures of cancer cells not only recapitulate the in vivo environment better than their 2D counterparts, but they have, in recent years, emerged as a relatively low-cost and scientifically accurate methodology for studying cancer. In this chapter, we highlight the importance of 3D culture, specifically 3D spheroid culture, reviewing some key methodologies for forming 3D spheroids, discussing the experimental tools that can be used in conjunction with 3D spheroids and finally their applications in cancer research.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Moissoglu K, Lockett SJ, Mili S. Visualizing and Quantifying mRNA Localization at the Invasive Front of 3D Cancer Spheroids. Methods Mol Biol 2023; 2608:263-280. [PMID: 36653713 PMCID: PMC10411857 DOI: 10.1007/978-1-0716-2887-4_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Localization of mRNAs at the front of migrating cells is a widely used mechanism that functionally supports efficient cell movement. It is observed in single cells on two-dimensional surfaces, as well as in multicellular three-dimensional (3D) structures and in tissue in vivo. 3D multicellular cultures can reveal how the topology of the extracellular matrix and cell-cell contacts influence subcellular mRNA distributions. Here we describe a method for mRNA imaging in an inducible system of collective cancer cell invasion. MDA-MB-231 cancer cell spheroids are embedded in Matrigel, induced to invade, and processed to image mRNAs with single-molecule sensitivity. An analysis algorithm is used to quantify and compare mRNA distributions at the front of invasive leader cells. The approach can be easily adapted and applied to analyze RNA distributions in additional settings where cells polarize along a linear axis.
Collapse
Affiliation(s)
- Konstadinos Moissoglu
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, NIH, Frederick, MD, USA
| | - Stavroula Mili
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
9
|
Qureshi R, Picon-Ruiz M, Sho M, Van Booven D, Nunes de Paiva V, Diaz-Ruano AB, Ince TA, Slingerland J. Estrone, the major postmenopausal estrogen, binds ERa to induce SNAI2, epithelial-to-mesenchymal transition, and ER+ breast cancer metastasis. Cell Rep 2022; 41:111672. [PMID: 36384125 PMCID: PMC9798480 DOI: 10.1016/j.celrep.2022.111672] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 09/22/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Recent work showed that the dominant post-menopausal estrogen, estrone, cooperates with nuclear factor κB (NF-κB) to stimulate inflammation, while pre-menopausal 17β-estradiol opposes NF-κB. Here, we show that post-menopausal estrone, but not 17β-estradiol, activates epithelial-to-mesenchymal transition (EMT) genes to stimulate breast cancer metastasis. HSD17B14, which converts 17β-estradiol to estrone, is higher in cancer than normal breast tissue and in metastatic than primary cancers and associates with earlier metastasis. Treatment with estrone, but not 17β-estradiol, and HSD17B14 overexpression both stimulate an EMT, matrigel invasion, and lung, bone, and liver metastasis in estrogen-receptor-positive (ER+) breast cancer models, while HSD17B14 knockdown reverses the EMT. Estrone:ERα recruits CBP/p300 to the SNAI2 promoter to induce SNAI2 and stimulate an EMT, while 17β-estradiol:ERα recruits co-repressors HDAC1 and NCOR1 to this site. Present work reveals novel differences in gene regulation by these estrogens and the importance of estrone to ER+ breast cancer progression. Upon loss of 17β-estradiol at menopause, estrone-liganded ERα would promote ER+ breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Rehana Qureshi
- Breast Cancer Program, Lombardi Comprehensive Cancer Centre, Department of Oncology, Georgetown University, Washington, DC 20007, USA; Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA; John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Manuel Picon-Ruiz
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18071 Granada, Spain; Biosanitary Institute of Granada (ibs. GRANADA), University of Granada, 18071 Granada, Spain
| | - Maiko Sho
- Breast Cancer Program, Lombardi Comprehensive Cancer Centre, Department of Oncology, Georgetown University, Washington, DC 20007, USA
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vanessa Nunes de Paiva
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anna B Diaz-Ruano
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Tan A Ince
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Joyce Slingerland
- Breast Cancer Program, Lombardi Comprehensive Cancer Centre, Department of Oncology, Georgetown University, Washington, DC 20007, USA; Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
10
|
Oh JM, Gangadaran P, Rajendran RL, Hong CM, Lee J, Ahn BC. Different Expression of Thyroid-Specific Proteins in Thyroid Cancer Cells between 2-Dimensional (2D) and 3-Dimensional (3D) Culture Environment. Cells 2022; 11:cells11223559. [PMID: 36428988 PMCID: PMC9688357 DOI: 10.3390/cells11223559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
The two-dimensional (2D) monolayer culture as a conventional method has been widely applied in molecular biology fields, but it has limited capability to recapitulate real cell environments, being prone to misinterpretation with poor prediction of in vivo behavior. Recently, the three-dimensional (3D) spheroid culture has been studied extensively. Spheroids are self-assembled cell aggregates that have biomimicry capabilities. The behavior of thyroid cancer under the 3D spheroid culture environment has been studied; however, there are no reports regarding differences in the degree of thyroid cancer cell differentiation under 2D and 3D culture conditions. This study investigated the expression of thyroid differentiation proteins related to iodide-metabolizing mechanisms in thyroid cancer cells under different culture conditions. Four thyroid cancer cell lines and one thyroid follicular epithelial cell line were grown in adherent 2D cell culture and 3D spheroid culture with agarose-coated plates. We observed changes in proliferation, hypoxia, extracellular matrix (ECM), cytoskeleton, thyroid-specific proteins, and thyroid transcription factors. All cell lines were successfully established in the spheroid following cell aggregation. Proliferation considerably decreased, while hypoxia-inducible factor 1-α(HIF1-α) was promoted in 3D spheroids; moreover, 3D spheroids with thyroid cancers showed diminished thyroid differentiation markers, but thyroid follicular epithelial cells revealed either a maintenance or weak decline of protein expression. We verified that the 3D spheroid culture environment can be similar to in vivo conditions because of its alterations in numerous cellular and functional activities, including morphology, cellular proliferation, viability, hypoxia, ECM, cytoskeleton, and thyroid differentiation, compared to the conventional 2D monolayer culture environment. An in vitro experimental study using 3D spheroid culture is ideal for the faster discovery of new drugs.
Collapse
Affiliation(s)
- Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea
- Correspondence: ; Tel.: +82-53-420-5583; Fax: +82-53-200-6447
| |
Collapse
|
11
|
CDK14 inhibition reduces mammary stem cell activity and suppresses triple negative breast cancer progression. Cell Rep 2022; 40:111331. [PMID: 36103813 DOI: 10.1016/j.celrep.2022.111331] [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: 02/03/2022] [Revised: 06/09/2022] [Accepted: 08/18/2022] [Indexed: 11/23/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays an important role in regulating mammary organogenesis and oncogenesis. However, therapeutic methods targeting the Wnt pathway against breast cancer have been limited. To address this challenge, we investigate the function of cyclin-dependent kinase 14 (CDK14), a member of the Wnt signaling pathway, in mammary development and breast cancer progression. We show that CDK14 is expressed in the mammary basal layer and elevated in triple negative breast cancer (TNBC). CDK14 knockdown reduces the colony-formation ability and regeneration capacity of mammary basal cells and inhibits the progression of murine MMTV-Wnt-1 basal-like mammary tumor. CDK14 knockdown or pharmacological inhibition by FMF-04-159-2 suppresses the progression and metastasis of TNBC. Mechanistically, CDK14 inhibition inhibits mammary regeneration and TNBC progression by attenuating Wnt/β-catenin signaling. These findings highlight the significance of CDK14 in mammary development and TNBC progression, shedding light on CDK14 as a promising therapeutic target for TNBC.
Collapse
|
12
|
Wu ST, Chen PC, Tseng YH, Chen TH, Wang YJ, Tsai ZL, Lin EC. Assessment of cellular responses in three-dimensional cell cultures through chemical exchange saturation transfer and 1 H MRS. NMR IN BIOMEDICINE 2022; 35:e4757. [PMID: 35510307 DOI: 10.1002/nbm.4757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 06/14/2023]
Abstract
Metabolic responses to physiological changes have been detected using chemical exchange saturation transfer (CEST) imaging in clinical settings. Similarly to other MRI techniques, the CEST technique was based originally on phantoms from buffer solutions and was then further developed through animal experiments. However, CEST imaging can capture certain dynamics of metabolism that solution phantoms cannot model. Cell culture phantoms can fill the gap between buffer phantoms and animal models. In this study, we used 1 H NMR and CEST in a B0 field of 9.4 T to investigate HEK293T cells from two-dimensional (2D) cultures, three-dimensional (3D) cultures, and 3D cultures seeded with cell spheroids. Two CEST dips were observed: the magnitude of the amine dip at 2.8 ppm increased during the incubation period, whereas the hydroxyl dip at 1.2 ppm remained approximately the same or modestly increased. We also observed a CEST dip at 2.8 ppm from the 2D culture responding dramatically to doxorubicin treatment. By cross-validating with pH values and the concentrations of amine and hydroxyl protons extracted through 1 H NMR, we observed that they did not correspond to an increase in the amine pool. We believe that the denaturation or degradation of proteins from the fetal bovine serum increased the size of the amine pool. Although 3D culture conditions can be further improved, our study suggests that 3D cultures have the potential to bridge studies of solution phantoms and those on animals.
Collapse
Affiliation(s)
- Ssu-Ting Wu
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Pin-Chen Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Yu-Hsien Tseng
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Ting-Hao Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Yi-Jiun Wang
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Zong-Lin Tsai
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Eugene C Lin
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
- Center for Nano Bio-detection, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
13
|
Delaunay S, Pascual G, Feng B, Klann K, Behm M, Hotz-Wagenblatt A, Richter K, Zaoui K, Herpel E, Münch C, Dietmann S, Hess J, Benitah SA, Frye M. Mitochondrial RNA modifications shape metabolic plasticity in metastasis. Nature 2022; 607:593-603. [PMID: 35768510 PMCID: PMC9300468 DOI: 10.1038/s41586-022-04898-5] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/24/2022] [Indexed: 12/13/2022]
Abstract
Aggressive and metastatic cancers show enhanced metabolic plasticity1, but the precise underlying mechanisms of this remain unclear. Here we show how two NOP2/Sun RNA methyltransferase 3 (NSUN3)-dependent RNA modifications—5-methylcytosine (m5C) and its derivative 5-formylcytosine (f5C) (refs.2–4)—drive the translation of mitochondrial mRNA to power metastasis. Translation of mitochondrially encoded subunits of the oxidative phosphorylation complex depends on the formation of m5C at position 34 in mitochondrial tRNAMet. m5C-deficient human oral cancer cells exhibit increased levels of glycolysis and changes in their mitochondrial function that do not affect cell viability or primary tumour growth in vivo; however, metabolic plasticity is severely impaired as mitochondrial m5C-deficient tumours do not metastasize efficiently. We discovered that CD36-dependent non-dividing, metastasis-initiating tumour cells require mitochondrial m5C to activate invasion and dissemination. Moreover, a mitochondria-driven gene signature in patients with head and neck cancer is predictive for metastasis and disease progression. Finally, we confirm that this metabolic switch that allows the metastasis of tumour cells can be pharmacologically targeted through the inhibition of mitochondrial mRNA translation in vivo. Together, our results reveal that site-specific mitochondrial RNA modifications could be therapeutic targets to combat metastasis.
Collapse
Affiliation(s)
- Sylvain Delaunay
- German Cancer Research Center - Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Gloria Pascual
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Bohai Feng
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany.,Department of Otorhinolaryngology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kevin Klann
- Institute of Biochemistry II, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mikaela Behm
- German Cancer Research Center - Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Agnes Hotz-Wagenblatt
- German Cancer Research Center - Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Karsten Richter
- German Cancer Research Center - Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Karim Zaoui
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,NCT Tissue Bank, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Christian Münch
- Institute of Biochemistry II, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sabine Dietmann
- Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jochen Hess
- German Cancer Research Center - Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany.,Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Michaela Frye
- German Cancer Research Center - Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany.
| |
Collapse
|
14
|
Wang S, Wang Y, Xiong J, Bao W, Li Y, Qin J, Han G, Hu S, Lei J, Yang Z, Qian Y, Dong S, Dong Z. Novel Brain-Stiffness-Mimicking Matrix Gel Enables Comprehensive Invasion Analysis of 3D Cultured GBM Cells. Front Mol Biosci 2022; 9:885806. [PMID: 35755807 PMCID: PMC9218788 DOI: 10.3389/fmolb.2022.885806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in adults, which is fast growing and tends to invade surrounding normal brain tissues. Uncovering the molecular and cellular mechanisms of GBM high invasion potential is of great importance for the treatment and prognostic prediction. However, the commonly used two-dimensional (2D) cell culture and analysis system suffers from lack of the heterogeneity and in vivo property of brain tissues. Here, we established a three-dimensional (3D) cell culture-based analysis system that could better recapitulate the heterogeneity of GBM and mimic the in vivo conditions in the brain. The GBM cell lines, DBTRG and U251, were cultured by hanging drop culture into the GBM multicellular spheroids, which were embedded in the optimized 3D brain-stiffness-mimicking matrix gel (0.5 mg/ml Collagen Ⅰ + 3 mg/ml Matrigel+ 3.3 mg/ml Hyaluronic Acid (HA)). The biochemical composition of the optimized matrix gel is similar to that of the brain microenvironment, and the elastic modulus is close to that of the brain tissue. The dynamics of the GBM spheroids was examined using high-content imaging for 60 h, and four metrics including invasion distance, invasion area, single-cell invasion velocity, and directionality were employed to quantify the invasion capacity. The result showed that DBTRG cells possess higher invasion capacity than U251 cells, which was consistent with the results of the classic transwell test. Transcriptome analysis of both cell lines was performed to explore the underlying molecular mechanisms. Our novel brain-stiffness-mimicking matrix gel enables comprehensive invasion analysis of the 3D cultured GBM cells and provides a model basis for in-depth exploration of the mechanisms regulating GBM invasion including the interaction between GBM cells and brain stroma.
Collapse
Affiliation(s)
- Shuowen Wang
- Brain Research Institute, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yiqi Wang
- Brain Research Institute, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jin Xiong
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wendai Bao
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yaqi Li
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jun Qin
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Guang Han
- Department of Radiation Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Hu
- Department of Thoracic Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Junrong Lei
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Zehao Yang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yu Qian
- Department of Thoracic Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Dong
- Department of Thoracic Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Dong
- Brain Research Institute, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,Central Laboratory, Hubei Cancer Hospital, Wuhan, China
| |
Collapse
|
15
|
The Antitumoral/Antimetastatic Action of the Flavonoid Brachydin A in Metastatic Prostate Tumor Spheroids In Vitro Is Mediated by (Parthanatos) PARP-Related Cell Death. Pharmaceutics 2022; 14:pharmaceutics14050963. [PMID: 35631550 PMCID: PMC9147598 DOI: 10.3390/pharmaceutics14050963] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 12/13/2022] Open
Abstract
Metastatic prostate cancer (mPCa) is resistant to several chemotherapeutic agents. Brachydin A (BrA), a glycosylated flavonoid extracted from Fridericia platyphylla, displays a remarkable antitumoral effect against in vitro mPCa cells cultured as bidimensional (2D) monolayers. Considering that three-dimensional (3D) cell cultures provide a more accurate response to chemotherapeutic agents, this study investigated the antiproliferative/antimetastatic effects of BrA and the molecular mechanisms underlying its action in mPCa spheroids (DU145) in vitro. BrA at 60–100 μM was cytotoxic, altered spheroid morphology/volume, and suppressed cell migration and tumor invasiveness. High-content analysis revealed that BrA (60–100 µM) reduced mitochondrial membrane potential and increased apoptosis and necrosis markers, indicating that it triggered cell death mechanisms. Molecular analysis showed that (i) 24-h treatment with BrA (80–100 µM) increased the protein levels of DNA disruption markers (cleaved-PARP and p-γ-H2AX) as well as decreased the protein levels of anti/pro-apoptotic (BCL-2, BAD, and RIP3K) and cell survival markers (p-AKT1 and p-44/42 MAPK); (ii) 72-h treatment with BrA increased the protein levels of effector caspases (CASP3, CASP7, and CASP8) and inflammation markers (NF-kB and TNF-α). Altogether, our results suggest that PARP-mediated cell death (parthanatos) is a potential mechanism of action. In conclusion, BrA confirms its potential as a candidate drug for preclinical studies against mPCa.
Collapse
|
16
|
Mukherjee A, Park A, Davies KP. PROL1 is essential for xenograft tumor development in mice injected with the human prostate cancer cell-line, LNCaP, and modulates cell migration and invasion. JOURNAL OF MEN'S HEALTH 2022; 18:044. [PMID: 35547856 PMCID: PMC9089447 DOI: 10.31083/jomh.2021.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background and objective A growing body of literature suggests modulated expression of members of the opiorphin family of genes (PROL1, SMR3A and SMR3B) is associated with cancer. Recently, overexpression of PROL1 was shown to be associated with prostate cancer, with evidence of a role in overcoming the hypoxic barrier that develops as tumors grow. The primary goal of the present studies was to support and expand evidence for a role of PROL1 in the development and progression of prostate cancer. Material and methods We engineered knock-out of the opiorphin gene, PROL1, in LNCaP, an androgen-sensitive, human prostate cancer derived, cell-line. Using xenograft assays, we compared the ability of injected LNCaP PROL1 knock-out cell-lines to develop tumors in both castrated and intact male mice with the parental LNCaP and LNCaP PROL1 overexpressing cell-lines. We used RNAseq to compare global gene expression between the parental and LNCaP PROL1 knock-out cell-lines. Wound closure and 3D spheroid invasion assays were used to compare cell motility and migration between parental LNCaP cells and LNCaP cells overexpressing of PROL1. Results The present studies demonstrate that LNCaP cell-lines with consisitutive knock-out of PROL1 fail to develop tumors when injected into both castrated and intact male mice. Using RNAseq to compare global gene expression between the parental and LNCaP PROL1 knock-out cell-lines, we confirmed a role for PROL1 in regulating molecular pathways associated with angiogenesis and tumor blood supply, and also identified a potential role in pathways related to cell motility and migration. Through the use of wound closure and 3D spheroid invasion assays, we confirmed that overexpression of PROL1 in LNCaP cells leads to greater cell motility and migration compared to parental cells, suggesting that PROL1 overexpression results in a more invasive phenotype. Conclusion Overall, our studies add to the growing body of evidence that opiorphin-encoding genes play a role in cancer development and progression. PROL1 is essential for establishment and growth of tumors in mice injected with LNCaP cells, and we provide evidence that PROL1 has a possible role in progression towards a more invasive, metastatic and castration resistant prostate cancer (PrCa).
Collapse
Affiliation(s)
- Amarnath Mukherjee
- Department of Urology, Albert Einstein College of Medicine,
Bronx, NY 10461, USA
| | - Augene Park
- Department of Urology, Albert Einstein College of Medicine,
Bronx, NY 10461, USA
| | - Kelvin Paul Davies
- Department of Urology, Albert Einstein College of Medicine,
Bronx, NY 10461, USA
- Department of Physiology and Biophysics, Albert Einstein
College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
17
|
Ono K, Sato K, Nakamura T, Yoshida Y, Murata S, Yoshida K, Kanemoto H, Umemori K, Kawai H, Obata K, Ryumon S, Hasegawa K, Kunisada Y, Okui T, Ibaragi S, Nagatsuka H, Sasaki A. Reproduction of the Antitumor Effect of Cisplatin and Cetuximab Using a Three-dimensional Spheroid Model in Oral Cancer. Int J Med Sci 2022; 19:1320-1333. [PMID: 35928727 PMCID: PMC9346383 DOI: 10.7150/ijms.74109] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/05/2022] [Indexed: 11/06/2022] Open
Abstract
Background/Aim: Cancer research has been conducted using cultured cells as part of drug discovery testing, but conventional two-dimensional culture methods are unable to reflect the complex tumor microenvironment. On the other hand, three-dimensional cultures have recently been attracting attention as in vitro models that more closely resemble the in vivo physiological environment. The purpose of this study was to establish a 3D culture method for oral cancer and to verify its practicality. Materials and Methods: Three-dimensional cultures were performed using several oral cancer cell lines. Western blotting was used for protein expression analysis of the collected cell masses (spheroids), and H-E staining was used for structural observation. The cultures were exposed to cisplatin and cetuximab and the morphological changes of spheroids over time and the expression changes of target proteins were compared. Results: Each cell line formed spheroidal cell aggregates and showed enhancement of cell adhesion molecules over time. H-E staining showed tumor tissue-like structures specific to each cell line. Cisplatin showed concentration-dependent antitumor effects due to loss of cell adhesion and spheroid disruption in each cell line, while cetuximab exhibited antitumor effects that correlated with EGFR expression in each cell line. Conclusion: Spheroids made from oral cancer cell lines appeared to have tumor-like characteristics that may reflect their clinical significance. In the future, it may become possible to produce tumor spheroids from tissue samples of oral cancer patients, and then apply them to drug screening and to develop individualized diagnostic and treatment methods.
Collapse
Affiliation(s)
- Kisho Ono
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Kohei Sato
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Tomoya Nakamura
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Yume Yoshida
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Shogo Murata
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Kunihiro Yoshida
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Hideka Kanemoto
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Koki Umemori
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Hotaka Kawai
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Kyoichi Obata
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Shoji Ryumon
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Kazuaki Hasegawa
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Yuki Kunisada
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Tatsuo Okui
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan
| | - Soichiro Ibaragi
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Hitoshi Nagatsuka
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Akira Sasaki
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| |
Collapse
|
18
|
Wang Y, Li N, Zheng Y, Wang A, Yu C, Song Z, Wang S, Sun Y, Zheng L, Wang G, Liu L, Yi J, Huang Y, Zhang M, Bao Y, Sun L. KIAA1217 Promotes Epithelial-Mesenchymal Transition and Hepatocellular Carcinoma Metastasis by Interacting with and Activating STAT3. Int J Mol Sci 2021; 23:104. [PMID: 35008530 PMCID: PMC8745027 DOI: 10.3390/ijms23010104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/01/2022] Open
Abstract
The survival and prognosis of hepatocellular carcinoma (HCC) are poor, mainly due to metastasis. Therefore, insights into the molecular mechanisms underlying HCC invasion and metastasis are urgently needed to develop a more effective antimetastatic therapy. Here, we report that KIAA1217, a functionally unknown macromolecular protein, plays a crucial role in HCC metastasis. KIAA1217 expression was frequently upregulated in HCC cell lines and tissues, and high KIAA1217 expression was closely associated with shorter survival of patients with HCC. Overexpression and knockdown experiments revealed that KIAA1217 significantly promoted cell migration and invasion by inducing epithelial-mesenchymal transition (EMT) in vitro. Consistently, HCC cells overexpressing KIAA1217 exhibited markedly enhanced lung metastasis in vivo. Mechanistically, KIAA1217 enhanced EMT and accordingly promoted HCC metastasis by interacting with and activating JAK1/2 and STAT3. Interestingly, KIAA1217-activated p-STAT3 was retained in the cytoplasm instead of translocating into the nucleus, where p-STAT3 subsequently activated the Notch and Wnt/β-catenin pathways to facilitate EMT induction and HCC metastasis. Collectively, KIAA1217 may function as an adaptor protein or scaffold protein in the cytoplasm and coordinate multiple pathways to promote EMT-induced HCC metastasis, indicating its potential as a therapeutic target for curbing HCC metastasis.
Collapse
Affiliation(s)
- Yanhong Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
- NMPA Key Laboratory for Quality of Cell and Gene Therapy Medicinal Products, Northeast Normal University, Changchun 130024, China; (Z.S.); (S.W.); (L.Z.); (G.W.); (L.L.); (J.Y.)
| | - Na Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| | - Yanping Zheng
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| | - Anqing Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| | - Chunlei Yu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| | - Zhenbo Song
- NMPA Key Laboratory for Quality of Cell and Gene Therapy Medicinal Products, Northeast Normal University, Changchun 130024, China; (Z.S.); (S.W.); (L.Z.); (G.W.); (L.L.); (J.Y.)
| | - Shuyue Wang
- NMPA Key Laboratory for Quality of Cell and Gene Therapy Medicinal Products, Northeast Normal University, Changchun 130024, China; (Z.S.); (S.W.); (L.Z.); (G.W.); (L.L.); (J.Y.)
| | - Ying Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| | - Lihua Zheng
- NMPA Key Laboratory for Quality of Cell and Gene Therapy Medicinal Products, Northeast Normal University, Changchun 130024, China; (Z.S.); (S.W.); (L.Z.); (G.W.); (L.L.); (J.Y.)
| | - Guannan Wang
- NMPA Key Laboratory for Quality of Cell and Gene Therapy Medicinal Products, Northeast Normal University, Changchun 130024, China; (Z.S.); (S.W.); (L.Z.); (G.W.); (L.L.); (J.Y.)
| | - Lei Liu
- NMPA Key Laboratory for Quality of Cell and Gene Therapy Medicinal Products, Northeast Normal University, Changchun 130024, China; (Z.S.); (S.W.); (L.Z.); (G.W.); (L.L.); (J.Y.)
| | - Jingwen Yi
- NMPA Key Laboratory for Quality of Cell and Gene Therapy Medicinal Products, Northeast Normal University, Changchun 130024, China; (Z.S.); (S.W.); (L.Z.); (G.W.); (L.L.); (J.Y.)
| | - Yanxin Huang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| | - Muqing Zhang
- School of Molecular and Cellular Biology, University of Illinois Urbana Champaign, Urbana, IL 61801, USA;
| | - Yongli Bao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| | - Luguo Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130024, China; (Y.W.); (N.L.); (Y.Z.); (A.W.); (C.Y.); (Y.S.); (Y.H.); (Y.B.)
| |
Collapse
|
19
|
Nousi A, Søgaard MT, Audoin M, Jauffred L. Single-cell tracking reveals super-spreading brain cancer cells with high persistence. Biochem Biophys Rep 2021; 28:101120. [PMID: 34541340 PMCID: PMC8435994 DOI: 10.1016/j.bbrep.2021.101120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 01/06/2023] Open
Abstract
Cell migration is a fundamental characteristic of vital processes such as tissue morphogenesis, wound healing and immune cell homing to lymph nodes and inflamed or infected sites. Therefore, various brain defect diseases, chronic inflammatory diseases as well as tumor formation and metastasis are associated with aberrant or absent cell migration. We embedded multicellular brain cancer spheroids in Matrigel™ and utilized single-particle tracking to extract the paths of cells migrating away from the spheroids. We found that - in contrast to local invasion - single cell migration is independent of Matrigel™ concentration and is characterized by high directionality and persistence. Furthermore, we identified a subpopulation of super-spreading cells with >200-fold longer persistence times than the majority of cells. These results highlight yet another aspect of cell heterogeneity in tumors.
Collapse
Affiliation(s)
| | - Maria Tangen Søgaard
- The Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, DK-2100, Copenhagen O, Denmark
| | | | - Liselotte Jauffred
- The Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, DK-2100, Copenhagen O, Denmark
| |
Collapse
|
20
|
Hall MK, Burch AP, Schwalbe RA. Functional analysis of N-acetylglucosaminyltransferase-I knockdown in 2D and 3D neuroblastoma cell cultures. PLoS One 2021; 16:e0259743. [PMID: 34748597 PMCID: PMC8575246 DOI: 10.1371/journal.pone.0259743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor development can be promoted/suppressed by certain N-glycans attached to proteins at the cell surface. Here we examined aberrant neuronal properties in 2D and 3D rat neuroblastoma (NB) cell cultures with different N-glycan populations. Lectin binding studies revealed that the engineered N-glycosylation mutant cell line, NB_1(-Mgat1), expressed solely oligomannose N-glycans, and verified that the parental cell line, NB_1, and a previous engineered N-glycosylation mutant, NB_1(-Mgat2), expressed significant levels of higher order N-glycans, complex and hybrid N-glycans, respectively. NB_1 grew faster than mutant cell lines in monolayer and spheroid cell cultures. A 2-fold difference in growth between NB_1 and mutants occurred much sooner in 2D cultures relative to that observed in 3D cultures. Neurites and spheroid cell sizes were reduced in mutant NB cells of 2D and 3D cultures, respectively. Cell invasiveness was highest in 2D cultures of NB_1 cells compared to that of NB_1(-Mgat1). In contrast, NB_1 spheroid cells were much less invasive relative to NB_1(-Mgat1) spheroid cells while they were more invasive than NB_1(-Mgat2). Gelatinase activities supported the ranking of cell invasiveness in various cell lines. Both palladin and HK2 were more abundant in 3D than 2D cultures. Levels of palladin, vimentin and EGFR were modified in a different manner under 2D and 3D cultures. Thus, our results support variations in the N-glycosylation pathway and in cell culturing to more resemble in vivo tumor environments can impact the aberrant cellular properties, particularly cell invasiveness, of NB.
Collapse
Affiliation(s)
- M. Kristen Hall
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Adam P. Burch
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Ruth A. Schwalbe
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, North Carolina, United States of America
| |
Collapse
|
21
|
Malyarenko OS, Malyarenko TV, Usoltseva RV, Surits VV, Kicha AA, Ivanchina NV, Ermakova SP. Combined Anticancer Effect of Sulfated Laminaran from the Brown Alga Alaria angusta and Polyhydroxysteroid Glycosides from the Starfish Protoreaster lincki on 3D Colorectal Carcinoma HCT 116 Cell Line. Mar Drugs 2021; 19:540. [PMID: 34677439 PMCID: PMC8538801 DOI: 10.3390/md19100540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 12/28/2022] Open
Abstract
Colorectal cancer is one of the most frequent types of malignancy in the world. The search for new approaches of increasing the efficacy of cancer therapy is relevant. This work was aimed to study individual, combined anticancer effects, and molecular mechanism of action of sulfated laminaran AaLs of the brown alga Alaria angusta and protolinckiosides A (PL1), B (PL2), and linckoside L1 (L1) of the starfish Protoreaster lincki using a 3D cell culture model. The 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS), soft agar, 3D spheroids invasion, and Western blotting assays were performed to determine the effect and mechanism of the action of investigated compounds or their combinations on proliferation, colony formation, and the invasion of 3D HCT 116 spheroids. AaLs, PL1, PL2, and L1 individually inhibited viability, colony growth, and the invasion of 3D HCT 116 spheroids in a variable degree with greater activity of linckoside L1. AaLs in combination with L1 exerted synergism of a combined anticancer effect through the inactivation of protein kinase B (AKT) kinase and, consequently, the induction of apoptosis via the regulation of proapoptotic/antiapoptotic proteins balance. The obtained data about the efficacy of the combined anticancer effect of a laminaran derivative of brown algae and polyhydroxysteroid glycosides of starfish open up prospects for the development of new therapeutic approaches for colorectal cancer treatment.
Collapse
Affiliation(s)
- Olesya S. Malyarenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 100-let Vladivostok Ave., 690022 Vladivostok, Russia; (T.V.M.); (R.V.U.); (V.V.S.); (A.A.K.); (N.V.I.); (S.P.E.)
| | | | | | | | | | | | | |
Collapse
|
22
|
Aslan M, Hsu EC, Liu S, Stoyanova T. Quantifying the invasion and migration ability of cancer cells with a 3D Matrigel drop invasion assay. Biol Methods Protoc 2021; 6:bpab014. [PMID: 34377838 PMCID: PMC8346651 DOI: 10.1093/biomethods/bpab014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022] Open
Abstract
Metastasis is the main cause of cancer-associated morbidity which will account for ∼ 600,000 deaths in the USA in 2021. Defining new mechanisms that drive cancer metastasis is vital for developing new therapeutic strategies and improving clinical outcomes for cancer patients. Herein, we describe a recently established 3D Matrigel drop invasion assay to measure cancer cell invasion and migration capability in vitro. This assay is a versatile and simple tool to test the ability of cells to invade and migrate, test the functional role of genes of interest in cell invasion and migration, analyze the localization of the target proteins at the cell invasion edge in situ, and screen drug effects on cancer cell invasion and migration.
Collapse
Affiliation(s)
- Merve Aslan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| | - En-Chi Hsu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| | - Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| |
Collapse
|
23
|
Zhang C, Meng X, Gong C, Zhao J, Zhang K, Yang Z. Glutathione-Responsive Biodegradable Nanoplatform with Endogenous Esterase-Triggered Nitric Oxide Release for Gas Therapy and Enhanced Chemotherapy. ACS APPLIED BIO MATERIALS 2021; 4:5212-5221. [DOI: 10.1021/acsabm.1c00384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Chen Zhang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Xiangdan Meng
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Chenchen Gong
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Jianming Zhao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Kai Zhang
- College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zhou Yang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| |
Collapse
|
24
|
Nazari SS. Generation of 3D Tumor Spheroids with Encapsulating Basement Membranes for Invasion Studies. ACTA ACUST UNITED AC 2021; 87:e105. [PMID: 32436628 PMCID: PMC8172047 DOI: 10.1002/cpcb.105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the past, in vitro studies of invasion and tumor progression were performed primarily using cancer cells cultured on a flat, two‐dimensional (2D) surface in a monolayer. In recent years, however, many studies have demonstrated differences in cell signaling and cell migration between 2D and 3D cell cultures. Traditional 2D monolayer cancer cell invasion models do not fully recapitulate 3D cell‐to‐cell and cell−to−extracellular matrix interactions that in vivo models can provide. Moreover, although in vivo animal models are irreplaceable for studying tumor biology and metastasis, they are costly, time‐consuming, and impractical for answering preliminary questions. Thus, emergent and evolving 3D spheroid cell culture models have changed the way we study tumors and their interactions with their surrounding extracellular matrix. In the case of breast cancer, metastasis of breast cancer tumors results in high mortality rates, and thus development of robust cell culture models that are reproducible and practical for studying breast cancer progression is important for ultimately developing preventatives for cancer metastasis. This article provides a set of protocols for generating uniform spheroids with a thin sheet of basement membrane for studying the initial invasion of mammary epithelial cells into a surrounding collagen‐rich extracellular matrix. Details are provided for generating 3D spheroids with a basement membrane, polymerizing collagen I, embedding the spheroids in the 3D collagen gel, and immunostaining the spheroids for invasion studies. Published 2020. U.S. Government. Basic Protocol 1: Growth of uniformly sized tumor spheroids with an encapsulating basement membrane Basic Protocol 2: Polymerization and embedding of tumor spheroids in a 3D type I collagen gel Alternate Protocol: Embedding of tumor spheroids in collagen gels using a sandwich method Basic Protocol 3: Fixing and immunostaining of tumor spheroids embedded in 3D collagen gels
Collapse
Affiliation(s)
- Shayan S Nazari
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
25
|
Cenariu D, Zimta AA, Munteanu R, Onaciu A, Moldovan CS, Jurj A, Raduly L, Moldovan A, Florea A, Budisan L, Pop LA, Magdo L, Albu MT, Tonea RB, Muresan MS, Ionescu C, Petrut B, Buiga R, Irimie A, Gulei D, Berindan-Neagoe I. Hsa-miR-125b Therapeutic Role in Colon Cancer Is Dependent on the Mutation Status of the TP53 Gene. Pharmaceutics 2021; 13:664. [PMID: 34066331 PMCID: PMC8148199 DOI: 10.3390/pharmaceutics13050664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/27/2021] [Accepted: 05/01/2021] [Indexed: 12/13/2022] Open
Abstract
Colon cancer is the third most common cancer type worldwide and is highly dependent on DNA mutations that progressively appear and accumulate in the normal colon epithelium. Mutations in the TP53 gene appear in approximately half of these patients and have significant implications in disease progression and response to therapy. miR-125b-5p is a controversial microRNA with a dual role in cancer that has been reported to target specifically TP53 in colon adenocarcinomas. Our study investigated the differential therapeutic effect of miR-125b-5p replacement in colon cancer based on the TP53 mutation status of colon cancer cell lines. In TP53 mutated models, miR-125b-5p overexpression slows cancer cells' malignant behavior by inhibiting the invasion/migration and colony formation capacity via direct downregulation of mutated TP53. In TP53 wild type cells, the exogenous modulation of miR-125b-5p did not significantly affect the molecular and phenotypic profile. In conclusion, our data show that miR-125b-5p has an anti-cancer effect only in TP53 mutated colon cancer cells, explaining partially the dual behavior of this microRNA in malignant pathologies.
Collapse
Affiliation(s)
- Diana Cenariu
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Alina-Andreea Zimta
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Raluca Munteanu
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Anca Onaciu
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Cristian Silviu Moldovan
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Alin Moldovan
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 6 Louis Pasteur St., 400349 Cluj-Napoca, Romania;
| | - Liviuta Budisan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Laura Ancuta Pop
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
| | - Lorand Magdo
- Faculty of Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (L.M.); (M.T.A.); (R.B.T.)
| | - Mihai Tudor Albu
- Faculty of Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (L.M.); (M.T.A.); (R.B.T.)
| | - Rares Bogdan Tonea
- Faculty of Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (L.M.); (M.T.A.); (R.B.T.)
| | - Mihai-Stefan Muresan
- 5th Surgical Department, Municipal Hospital, 11 Tăbăcarilor Street, 400139 Cluj-Napoca, Romania; (M.-S.M.); (C.I.)
- Surgical and Gynecological Oncology Department, Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii 34–36 Street, 400015 Cluj-Napoca, Romania
- Department of Surgery V, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Calin Ionescu
- 5th Surgical Department, Municipal Hospital, 11 Tăbăcarilor Street, 400139 Cluj-Napoca, Romania; (M.-S.M.); (C.I.)
- Department of Surgery V, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Bogdan Petrut
- Department of Urology, “Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii 34–36 Street, 400015 Cluj-Napoca, Romania;
- Department of Urology, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Rares Buiga
- Department of Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii 34–36 Street, 400015 Cluj-Napoca, Romania;
- Department of Pathology, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Alexandru Irimie
- 11th Department of Surgical Oncology and Gynaecological Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
- Department of Surgery, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Diana Gulei
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, Marinescu 23 Street/Louis Pasteur 4–6 Street, 400337 Cluj-Napoca, Romania; (D.C.); (A.-A.Z.); (R.M.); (A.O.); (C.S.M.); (A.M.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (L.B.); (L.A.P.); (I.B.-N.)
- Department of Functional Genomics and Experimental Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| |
Collapse
|
26
|
Yildiz-Ozturk E, Saglam-Metiner P, Yesil-Celiktas O. Lung carcinoma spheroids embedded in a microfluidic platform. Cytotechnology 2021; 73:457-471. [PMID: 34149177 DOI: 10.1007/s10616-021-00470-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/07/2021] [Indexed: 01/13/2023] Open
Abstract
Three-dimensional (3D) spheroid cell cultures are excellent models used in cancer biology research and drug screening. The objective of this study was to develop a lung carcinoma spheroid based microfluidic platform with perfusion function to mimic lung cancer pathology and investigate the effect of a potential drug molecule, panaxatriol. Spheroids were successfully formed on agar microtissue molds at the end of 10 days, reaching an average diameter of about 317.18 ± 4.05 μm and subsequently transferred to 3D dynamic microfluidic system with perfusion function. While the size of the 3D spheroids embedded in the Matrigel matrix in the platform had gradually increased both in the static and dynamic control groups, the size of the spheroids were reduced and fragmented in the drug treated groups. Cell viability results showed that panaxatriol exhibited higher cytotoxic effect on cancer cells than healthy cells and the IC50 value was determined as 61.55 µM. Furthermore, panaxatriol has been more effective on single cells around the spheroid structure, whereas less in 3D spheroid tissues with a compact structure in static conditions compared to dynamic systems, where a flow rate of 2 µL/min leading to a shear stress of 0.002 dyne/cm2 was applied. Application of such dynamic systems will contribute to advancing basic research and increasing the predictive accuracy of potential drug molecules, which may accelerate the translation of novel therapeutics to the clinic, possibly decreasing the use of animal models. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-021-00470-7.
Collapse
Affiliation(s)
- Ece Yildiz-Ozturk
- Ege University Translational Pulmonary Research Center (Ege TPRC), 35100 Izmir, Turkey
| | - Pelin Saglam-Metiner
- Faculty of Engineering, Department of Bioengineering, Ege University, 35100 Izmir, Turkey
| | - Ozlem Yesil-Celiktas
- Ege University Translational Pulmonary Research Center (Ege TPRC), 35100 Izmir, Turkey.,Faculty of Engineering, Department of Bioengineering, Ege University, 35100 Izmir, Turkey
| |
Collapse
|
27
|
SenGupta S, Hein LE, Xu Y, Zhang J, Konwerski JR, Li Y, Johnson C, Cai D, Smith JL, Parent CA. Triple-Negative Breast Cancer Cells Recruit Neutrophils by Secreting TGF-β and CXCR2 Ligands. Front Immunol 2021; 12:659996. [PMID: 33912188 PMCID: PMC8071875 DOI: 10.3389/fimmu.2021.659996] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/11/2021] [Indexed: 12/22/2022] Open
Abstract
Tumor associated neutrophils (TANs) are frequently detected in triple-negative breast cancer (TNBC). Recent studies also reveal the importance of neutrophils in promoting tumor progression and metastasis during breast cancer. However, the mechanisms regulating neutrophil trafficking to breast tumors are less clear. We sought to determine whether neutrophil trafficking to breast tumors is determined directly by the malignant potential of cancer cells. We found that tumor conditioned media (TCM) harvested from highly aggressive, metastatic TNBC cells induced a polarized morphology and robust neutrophil migration, while TCM derived from poorly aggressive estrogen receptor positive (ER+) breast cancer cells had no activity. In a three-dimensional (3D) type-I collagen matrix, neutrophils migrated toward TCM from aggressive breast cancer cells with increased velocity and directionality. Moreover, in a neutrophil-tumor spheroid co-culture system, neutrophils migrated with increased directionality towards spheroids generated from TNBC cells compared to ER+ cells. Based on these findings, we next sought to characterize the active factors secreted by TNBC cell lines. We found that TCM-induced neutrophil migration is dependent on tumor-derived chemokines, and screening TCM elution fractions based on their ability to induce polarized neutrophil morphology revealed the molecular weight of the active factors to be around 12 kDa. TCM from TNBC cell lines contained copious amounts of GRO (CXCL1/2/3) chemokines and TGF-β cytokines compared to ER+ cell-derived TCM. TCM activity was inhibited by simultaneously blocking receptors specific to GRO chemokines and TGF-β, while the activity remained intact in the presence of either single receptor inhibitor. Together, our findings establish a direct link between the malignant potential of breast cancer cells and their ability to induce neutrophil migration. Our study also uncovers a novel coordinated function of TGF-β and GRO chemokines responsible for guiding neutrophil trafficking to the breast tumor.
Collapse
Affiliation(s)
- Shuvasree SenGupta
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Lauren E. Hein
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Yang Xu
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jason Zhang
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jamie R. Konwerski
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ye Li
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Dawen Cai
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Janet L. Smith
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Carole A. Parent
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
28
|
Three-Dimensional Spheroids as In Vitro Preclinical Models for Cancer Research. Pharmaceutics 2020; 12:pharmaceutics12121186. [PMID: 33291351 PMCID: PMC7762220 DOI: 10.3390/pharmaceutics12121186] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/29/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Most cancer biologists still rely on conventional two-dimensional (2D) monolayer culture techniques to test in vitro anti-tumor drugs prior to in vivo testing. However, the vast majority of promising preclinical drugs have no or weak efficacy in real patients with tumors, thereby delaying the discovery of successful therapeutics. This is because 2D culture lacks cell–cell contacts and natural tumor microenvironment, important in tumor signaling and drug response, thereby resulting in a reduced malignant phenotype compared to the real tumor. In this sense, three-dimensional (3D) cultures of cancer cells that better recapitulate in vivo cell environments emerged as scientifically accurate and low cost cancer models for preclinical screening and testing of new drug candidates before moving to expensive and time-consuming animal models. Here, we provide a comprehensive overview of 3D tumor systems and highlight the strategies for spheroid construction and evaluation tools of targeted therapies, focusing on their applicability in cancer research. Examples of the applicability of 3D culture for the evaluation of the therapeutic efficacy of nanomedicines are discussed.
Collapse
|
29
|
Wagner M, Koyasu S. A 3D Skin Melanoma Spheroid-Based Model to Assess Tumor-Immune Cell Interactions. Bio Protoc 2020; 10:e3839. [PMID: 33659488 DOI: 10.21769/bioprotoc.3839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/29/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022] Open
Abstract
Three-dimensional (3D) tumor spheroids have the potential to bridge the gap between two-dimensional (2D) monolayer tumor cell cultures and solid tumors with which they share a significant degree of similarity. However, the progression of solid tumors is often influenced by the dynamic and reciprocal interactions between tumor and immune cells. Here we present a 3D tumor spheroid-based model that might shed new light on understanding the mechanisms of tumor and immune cell interactions. The model first utilizes the hanging drop assay, which serves as one of the simplest methods for generating 3D spheroids and requires no specialized equipment. Next, pre-established spheroids can be co-cultured either directly or indirectly with an immune cell population of interest. Using skin melanoma, we provide a detailed description of the model, which might hold a significant importance for the development of successful therapeutic strategies.
Collapse
Affiliation(s)
- Marek Wagner
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
The Tumor Suppressor CYLD Inhibits Mammary Epithelial to Mesenchymal Transition by the Coordinated Inhibition of YAP/TAZ and TGF Signaling. Cancers (Basel) 2020; 12:cancers12082047. [PMID: 32722292 PMCID: PMC7466024 DOI: 10.3390/cancers12082047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Downregulation of the cylindromatosis (CYLD) tumor suppressor has been associated with breast cancer development and progression. Here, we report a critical role for CYLD in maintaining the phenotype of mammary epithelial cells in vitro and in vivo. CYLD downregulation or inactivation induced an epithelial to mesenchymal transition of mammary epithelial cells that was dependent on the concomitant activation of the transcription factors Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) and transforming growth factor beta (TGF)signaling. CYLD inactivation enhanced the nuclear localization of YAP/TAZ and the phosphorylation of Small Mothers Against Decapentaplegic (SMAD)2/3 proteins in confluent cell culture conditions. Consistent with these findings were the hyperplastic alterations of CYLD-deficient mouse mammary epithelia, which were associated with enhanced nuclear expression of the YAP/TAZ transcription factors. Furthermore, in human breast cancer samples, downregulation of CYLD expression correlates with enhanced YAP/TAZ-regulated target gene expression. Our results identify CYLD as a critical regulator of a signaling node that prevents the coordinated activation of YAP/TAZ and the TGF pathway in mammary epithelial cells, in order to maintain their phenotypic identity and homeostasis. Consequently, they provide a novel conceptual framework that supports and explains a causal implication of deficient CYLD expression in aggressive human breast cancers.
Collapse
|
31
|
Mendonsa AM, Bandyopadhyay C, Gumbiner BM. p120-catenin phosphorylation status alters E-cadherin mediated cell adhesion and ability of tumor cells to metastasize. PLoS One 2020; 15:e0235337. [PMID: 32589661 PMCID: PMC7319294 DOI: 10.1371/journal.pone.0235337] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/12/2020] [Indexed: 01/06/2023] Open
Abstract
p120-catenin is considered to be a tumor suppressor because it stabilizes E-cadherin levels at the cell surface. p120-catenin phosphorylation is increased in several types of cancer, but the role of phosphorylation in cancer is unknown. The phosphorylation state of p120-catenin is important in controlling E-cadherin homophilic binding strength which maintains epithelial junctions. Because decreased cell-cell adhesion is associated with increased cancer metastasis we hypothesize that p120-catenin phosphorylation at specific Serine and Threonine residues alters the E-cadherin binding strength between tumor cells and thereby affect the ability of tumor cells to leave the primary tumor and metastasize to distant sites. In this study we show that expression of the p120-catenin phosphorylation dead mutant, by converting six Serine and Threonine sites to Alanine, leads to enhanced E-cadherin adhesive binding strength in tumor cells. We observed a decrease in the ability of tumor cells expressing the p120-catenin phosphorylation mutant to migrate and invade using in-vitro models of cancer progression. Further, tumor cells expressing the phosphorylation mutant form of p120-catenin demonstrated a decrease in ability to metastasize to the lungs using an in-vivo orthotopic mammary fat pad injection model of breast cancer development and metastasis. This suggests that regulation of p120-catenin phosphorylation at the cell surface is important in mediating cell-adhesion, thereby impacting cancer progression and metastasis.
Collapse
Affiliation(s)
- Alisha M. Mendonsa
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Chirosree Bandyopadhyay
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Barry M. Gumbiner
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
32
|
Ruiz MC, Kljun J, Turel I, Di Virgilio AL, León IE. Comparative antitumor studies of organoruthenium complexes with 8-hydroxyquinolines on 2D and 3D cell models of bone, lung and breast cancer. Metallomics 2020; 11:666-675. [PMID: 30839008 DOI: 10.1039/c8mt00369f] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The purpose of this work was to screen the antitumor actions of two metal organoruthenium-8-hydroxyquinolinato (Ru-hq) complexes to find a potential novel agent for bone, lung and breast chemotherapies. We showed that ruthenium compounds (1 and 2) impaired the cell viability of human bone (MG-63), lung (A549) and breast (MCF7) cancer cells with greater selectivity and specificity than cisplatin. Besides, complexes 1 and 2 decreased proliferation, migration and invasion on cell monolayers at lower concentrations (2.5-10 μM). In addition, both compounds induced genotoxicity revealed by the micronucleus test, which led to G2/M cell cycle arrest and induced the tumor cells to undergo apoptosis. On the other hand, in multicellular 3D models (multicellular spheroids; MCS), 1 and 2 overcame CDDP presenting lower IC50 values only in MCS of lung origin. Moreover, 1 outperformed 2 in MCS of bone and breast origin. Finally, our findings revealed that both compounds inhibited the cell invasion of multicellular spheroids, showing that complex 1 exhibited the most important antimetastatic action. Taken together, these results indicate that compound 1 is an interesting candidate to be tested on in vivo models as a novel strategy for anticancer therapy.
Collapse
Affiliation(s)
- Maria C Ruiz
- Inorganic Chemistry Center (CEQUINOR, CONICET), Exact School Sciences, National University of La Plata, Bv 120 1465, 1900 La Plata, Argentina.
| | | | | | | | | |
Collapse
|
33
|
Gomes I, de Almeida BP, Dâmaso S, Mansinho A, Correia I, Henriques S, Cruz-Duarte R, Vilhais G, Félix P, Alves P, Corredeira P, Barbosa-Morais NL, Costa L, Casimiro S. Expression of receptor activator of NFkB (RANK) drives stemness and resistance to therapy in ER+HER2- breast cancer. Oncotarget 2020; 11:1714-1728. [PMID: 32477461 PMCID: PMC7233807 DOI: 10.18632/oncotarget.27576] [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: 11/07/2019] [Accepted: 04/10/2020] [Indexed: 12/30/2022] Open
Abstract
The role of RANKL-RANK pathway in progesterone-driven mammary carcinogenesis and triple negative breast cancer tumorigenesis has been well characterized. However, and despite evidences of the existence of RANK-positive hormone receptor (HR)-positive breast tumors, the implication of RANK expression in HR-positive breast cancers has not been addressed before. Here, we report that RANK pathway affects the expression of cell cycle regulators and decreases sensitivity to fulvestrant of estrogen receptor (ER)-positive (ER+)/HER2- breast cancer cells, MCF-7 and T47D. Moreover, RANK overexpressing cells had a staminal and mesenchymal phenotype, with decreased proliferation rate and decreased susceptibility to chemotherapy, but were more invasive in vivo. In silico analysis of the transcriptome of human breast tumors, confirmed the association between RANK expression and stem cell and mesenchymal markers in ER+HER2- tumors. Importantly, exposure of ER+HER2- cells to continuous RANK pathway activation by exogenous RANKL, in vitro and in vivo, induced a negative feedback effect, independent of RANK levels, leading to the downregulation of HR and increased resistance to hormone therapy. These results suggest that ER+HER2- RANK-positive cells may constitute an important reservoir of slow cycling, therapy-resistance cancer cells; and that RANK pathway activation is deleterious in all ER+HER2- breast cancer cells, independently of RANK levels.
Collapse
Affiliation(s)
- Inês Gomes
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Bernardo P. de Almeida
- Nuno Morais Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Current affiliation: Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Sara Dâmaso
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, Lisboa, Portugal
| | - André Mansinho
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, Lisboa, Portugal
| | - Inês Correia
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Sara Henriques
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Raquel Cruz-Duarte
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Guilherme Vilhais
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Pedro Félix
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Patrícia Alves
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Patrícia Corredeira
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Nuno L. Barbosa-Morais
- Nuno Morais Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Luis Costa
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Serviço de Oncologia, Hospital de Santa Maria-CHULN, Lisboa, Portugal
| | - Sandra Casimiro
- Luis Costa Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
34
|
Balsa LM, Ruiz MC, Santa Maria de la Parra L, Baran EJ, León IE. Anticancer and antimetastatic activity of copper(II)-tropolone complex against human breast cancer cells, breast multicellular spheroids and mammospheres. J Inorg Biochem 2019; 204:110975. [PMID: 31911364 DOI: 10.1016/j.jinorgbio.2019.110975] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/13/2019] [Accepted: 12/24/2019] [Indexed: 12/11/2022]
Abstract
The goal of this work was to display the anticancer and antimetastatic activity of a copper(II) with tropolone (trp), complex [Cu(trp)2] toward human breast cancer cells in monolayer (2D) and spheroids (3D). Cytotoxicity assays against MCF7 (IC50(complex) = 5.2 ± 1.8 μM, IC50(CDDP) = 19.3 ± 2.1 μM) and MDA-MB-231 (IC50(complex) = 4.0 ± 0.2 μM, IC50(CDDP) = 27.0 ± 1.9 μM) demonstrate that [Cu(trp)2] exert greater antitumor potency than cisplatin (CDDP) on 2D and 3D human breast cancer cell models. Besides, [Cu(trp)2] inhibits cell migration by reducing the metalloproteinases activities and the compound undergoes the breast cancer cells to apoptosis at lower concentrations (2.5-10 μM). Moreover, [Cu(trp)2] overcame CDDP presenting an IC50 value 26-fold more lower against breast multicellular spheroids ((IC50(complex) = 4.9 μM, IC50(CDDP) = 130 μM)). Also, our results showed that [Cu(trp)2] inhibited the cell migration and cell invasion of breast multicellular spheroids, showing that [Cu(trp)2] exhibited antimetastatic properties. On the other hand, [Cu(trp)2] reduced mammosphere forming capacity affecting the size and number of mammospheres. Taken together, [Cu(trp)2] exhibited anticancer and antimetastatic properties on monolayer (2D) and spheroids (3D) derived from human breast cancer cells.
Collapse
Affiliation(s)
- Lucia M Balsa
- Inorganic Chemistry Center (CEQUINOR, CONICET), Exact School Sciences, National University of La Plata, Bv 120 1465, 1900 La Plata, Argentina
| | - Maria C Ruiz
- Inorganic Chemistry Center (CEQUINOR, CONICET), Exact School Sciences, National University of La Plata, Bv 120 1465, 1900 La Plata, Argentina
| | - Lucia Santa Maria de la Parra
- Inorganic Chemistry Center (CEQUINOR, CONICET), Exact School Sciences, National University of La Plata, Bv 120 1465, 1900 La Plata, Argentina
| | - Enrique J Baran
- Inorganic Chemistry Center (CEQUINOR, CONICET), Exact School Sciences, National University of La Plata, Bv 120 1465, 1900 La Plata, Argentina
| | - Ignacio E León
- Inorganic Chemistry Center (CEQUINOR, CONICET), Exact School Sciences, National University of La Plata, Bv 120 1465, 1900 La Plata, Argentina.
| |
Collapse
|
35
|
3D multicellular models to study the regulation and roles of acid-base transporters in breast cancer. Biochem Soc Trans 2019; 47:1689-1700. [PMID: 31803922 DOI: 10.1042/bst20190131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022]
Abstract
As a result of elevated metabolic rates and net acid extrusion in the rapidly proliferating cancer cells, solid tumours are characterized by a highly acidic microenvironment, while cancer cell intracellular pH is normal or even alkaline. Two-dimensional (2D) cell monocultures, which have been used extensively in breast cancer research for decades, cannot precisely recapitulate the rich environment and complex processes occurring in tumours in vivo. The use of such models can consequently be misleading or non-predictive for clinical applications. Models mimicking the tumour microenvironment are particularly pivotal for studying tumour pH homeostasis, which is profoundly affected by the diffusion-limited conditions in the tumour. To advance the understanding of the mechanisms and consequences of dysregulated acid-base homeostasis in breast cancer, clinically relevant models that incorporate the unique microenvironment of these tumours are required. The development of three-dimensional (3D) cell cultures has provided new tools for basic research and pre-clinical approaches, allowing the culture of breast cancer cells under conditions that closely resemble tumour growth in a living organism. Here we provide an overview of the main 3D techniques relevant for breast cancer cell culture. We discuss the advantages and limitations of the classical 3D models as well as recent advances in 3D culture techniques, focusing on how these culture methods have been used to study acid-base transport in breast cancer. Finally, we outline future directions of 3D culture technology and their relevance for studies of acid-base transport.
Collapse
|
36
|
Gong F, Chen MF, Chen J, Li C, Zhou C, Hong P, Sun S, Qian ZJ. Boiled Abalone Byproduct Peptide Exhibits Anti-Tumor Activity in HT1080 Cells and HUVECs by Suppressing the Metastasis and Angiogenesis in Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8855-8867. [PMID: 31343893 DOI: 10.1021/acs.jafc.9b03005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Abalone (Haliotis discus hannai) is a precious seafood in the market. It has been reported that biological active substances derived from abalone have anti-oxidative, anti-inflammatory, anti-bacterial, and anti-thrombosis potential. However, there were few studies to assess whether they have anti-cancer potential. In this study, we evaluated the anti-metastasis and anti-pro-angiogenic factors and mechanism of action of boiled abalone byproduct peptide (BABP, EMDEAQDPSEW) in human fibrosarcoma (HT1080) cells and human umbilical vein endothelial cells (HUVECs). The results demonstrated that BABP treatment significantly lowers migration and the invasion of HT1080 cells and HUVECs. BABP inhibits phorbol 12-myristate 13-acetate (PMA)-induced matrix metalloproteinase (MMP) expression and activity by blocking mitogen-activated protein kinases (MAPKs) and NF-κB signaling and hypoxia-induced vascular endothelial growth factor (VEGF) secretion and hypoxia inducible factor (HIF)-1α accumulation through suppressing the AKT/mTOR signal pathway. BABP treatment inhibits VEGF-induced VEGFR-2 expression and tube formation in HUVECs. The effect of BABP on anti-metastatic and anti-vascular activity in HT1080 cells and HUVECs revealed that BABP may be a potential pharmacophore for tumor therapy in the future.
Collapse
Affiliation(s)
- Fang Gong
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - Mei-Fang Chen
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - Jiali Chen
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - ChengYong Li
- School of Chemistry and Environment , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
- Shenzhen Institute of Guangdong Ocean University , Shenzhen , Guangdong 518114 , China
| | - ChunXia Zhou
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - PengZhi Hong
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - ShengLi Sun
- School of Chemistry and Environment , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - Zhong-Ji Qian
- School of Chemistry and Environment , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
- Shenzhen Institute of Guangdong Ocean University , Shenzhen , Guangdong 518114 , China
| |
Collapse
|
37
|
Wang W, Friedland SC, Guo B, O’Dell MR, Alexander WB, Whitney-Miller CL, Agostini-Vulaj D, Huber AR, Myers JR, Ashton JM, Dunne RF, Steiner LA, Hezel AF. ARID1A, a SWI/SNF subunit, is critical to acinar cell homeostasis and regeneration and is a barrier to transformation and epithelial-mesenchymal transition in the pancreas. Gut 2019; 68:1245-1258. [PMID: 30228219 PMCID: PMC6551318 DOI: 10.1136/gutjnl-2017-315541] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Here, we evaluate the contribution of AT-rich interaction domain-containing protein 1A (ARID1A), the most frequently mutated member of the SWItch/sucrose non-fermentable (SWI/SNF) complex, in pancreatic homeostasis and pancreatic ductal adenocarcinoma (PDAC) pathogenesis using mouse models. DESIGN Mice with a targeted deletion of Arid1a in the pancreas by itself and in the context of two common genetic alterations in PDAC, Kras and p53, were followed longitudinally. Pancreases were examined and analysed for proliferation, response to injury and tumourigenesis. Cancer cell lines derived from these models were analysed for clonogenic, migratory, invasive and transcriptomic changes. RESULTS Arid1a deletion in the pancreas results in progressive acinar-to-ductal metaplasia (ADM), loss of acinar mass, diminished acinar regeneration in response to injury and ductal cell expansion. Mutant Kras cooperates with homozygous deletion of Arid1a, leading to intraductal papillary mucinous neoplasm (IPMN). Arid1a loss in the context of mutant Kras and p53 leads to shorter tumour latency, with the resulting tumours being poorly differentiated. Cancer cell lines derived from Arid1a-mutant tumours are more mesenchymal, migratory, invasive and capable of anchorage-independent growth; gene expression analysis showed activation of epithelial-mesenchymal transition (EMT) and stem cell identity pathways that are partially dependent on Arid1a loss for dysregulation. CONCLUSIONS ARID1A plays a key role in pancreatic acinar homeostasis and response to injury. Furthermore, ARID1A restrains oncogenic KRAS-driven formation of premalignant proliferative IPMN. Arid1a-deficient PDACs are poorly differentiated and have mesenchymal features conferring migratory/invasive and stem-like properties.
Collapse
Affiliation(s)
- Wenjia Wang
- Department of Medicine, Hematology and Oncology Division, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Scott C Friedland
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, USA
| | - Bing Guo
- Department of Medicine, Hematology and Oncology Division, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael R O’Dell
- Department of Medicine, Hematology and Oncology Division, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - William B Alexander
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, USA
| | - Christa L Whitney-Miller
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Diana Agostini-Vulaj
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Aaron R Huber
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Jason R Myers
- Genomics Research Center, University of Rochester Medical Center, Rochester, New York, USA
| | - John M Ashton
- Genomics Research Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Richard F Dunne
- Department of Medicine, Hematology and Oncology Division, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Laurie A Steiner
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Aram F Hezel
- Department of Medicine, Hematology and Oncology Division, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
38
|
Hu Q, Myers M, Fang W, Yao M, Brummer G, Hawj J, Smart C, Berkland C, Cheng N. Role of ALDH1A1 and HTRA2 expression in CCL2/CCR2-mediated breast cancer cell growth and invasion. Biol Open 2019; 8:bio.040873. [PMID: 31208996 PMCID: PMC6679398 DOI: 10.1242/bio.040873] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chemokines mediate immune cell trafficking during tissue development, wound healing and infection. The chemokine CCL2 is best known to regulate macrophage recruitment during wound healing, infection and inflammatory diseases. While the importance of CCL2/CCR2 signaling in macrophages during cancer progression is well documented, we recently showed that CCL2-mediated breast cancer progression depends on CCR2 expression in carcinoma cells. Using 3D Matrigel: Collagen cultures of SUM225 and DCIS.com breast cancer cells, this study characterized the mechanisms of CCL2/CCR2 signaling in cell growth and invasion. SUM225 cells, which expressed lower levels of CCR2 than DCIS.com cells, formed symmetrical spheroids in Matrigel: Collagen, and were not responsive to CCL2 treatment. DCIS.com cells formed asymmetric cell clusters in Matrigel: Collagen. CCL2 treatment increased growth, decreased expression of E-cadherin and increased TWIST1 expression. CCR2 overexpression in SUM225 cells increased responsiveness to CCL2 treatment, enhancing growth and invasion. These phenotypes corresponded to increased expression of Aldehyde Dehydrogenase 1A1 (ALDH1A1) and decreased expression of the mitochondrial serine protease HTRA2. CCR2 deficiency in DCIS.com cells inhibited CCL2-mediated growth and invasion, corresponding to decreased ALDH1A1 expression and increased HTRA2 expression. ALDH1A1 and HTRA2 expression were modulated in CCR2-deficient and CCR2-overexpressing cell lines. We found that ALDH1A1 and HTRA2 regulates CCR2-mediated breast cancer cell growth and cellular invasion in a CCL2/CCR2 context-dependent manner. These data provide novel insight on the mechanisms of chemokine signaling in breast cancer cell growth and invasion, with important implications on targeted therapeutics for anti-cancer treatment. This article has an associated First Person interview with the first author of the paper. Summary: Chemokines are known to regulate immune cell recruitment during inflammation. This report characterizes novel molecular mechanisms through which CCL2/CCR2 chemokine signaling in breast cancer cells regulates growth and invasion.
Collapse
Affiliation(s)
- Qingting Hu
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Megan Myers
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Fang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Min Yao
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Gage Brummer
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Justin Hawj
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Curtis Smart
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Nikki Cheng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA .,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
39
|
Vimentin activation in early apoptotic cancer cells errands survival pathways during DNA damage inducer CPT treatment in colon carcinoma model. Cell Death Dis 2019; 10:467. [PMID: 31197132 PMCID: PMC6565729 DOI: 10.1038/s41419-019-1690-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/24/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Epithelial to mesenchymal transitions (EMT) is a preparatory process for cancer cells to attain motility and further metastasis to distant sites. Majority of DNA damaging drugs have shown to develop EMT as one of the major mechanisms to attain drug resistance. Here we sought to understand the resistance/survival instincts of cancer cells during initial phase of drug treatment. We provide a tangible evidence of stimulation of EMT factors in Apc knockout colorectal carcinoma model. Our results implied that CPT-treated Apc knockout cohorts depicted increased pro-invasive and pro-survival factors (Vimentin/pser38Vimentin & NFκB). Moreover, by cell sorting experiment, we have observed the expression of Vimentin in early apoptotic cells (AnnexinV positive) from 36 to 48 h of CPT treatment. We also observed the expression of chimeric Sec-AnnexinV-mvenus protein in migrated cells on transwell membrane recapitulating signatures of early apoptosis. Notably, induction of Vimentin-mediated signaling (by CPT) delayed apoptosis progression in cells conferring survival responses by modulating the promoter activity of NFκB. Furthermore, our results unveiled a novel link between Vimentin and ATM signaling, orchestrated via binding interaction between Vimentin and ATM kinase. Finally, we observed a significant alteration of crypt-villus morphology upon combination of DIM (EMT inhibitor) with CPT nullified the background EMT signals thus improving the efficacy of the DNA damaging agent. Thus, our findings revealed a resistance strategy of cancer cells within a very initial period of drug treatment by activating EMT program, which hinders the cancer cells to achieve later phases of apoptosis thus increasing the chances of early migration.
Collapse
|
40
|
A Transcriptomic Insight into the Impact of Colon Cancer Cells on Mast Cells. Int J Mol Sci 2019; 20:ijms20071689. [PMID: 30987352 PMCID: PMC6480031 DOI: 10.3390/ijms20071689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/27/2019] [Accepted: 04/01/2019] [Indexed: 12/13/2022] Open
Abstract
Mast cells (MCs) are one of the first immune cells recruited to a tumor. It is well recognized that MCs accumulate in colon cancer lesion and their density is associated with the clinical outcomes. However, the molecular mechanism of how colon cancer cells may modify MC function is still unclear. In this study, primary human MCs were generated from CD34+ progenitor cells and a 3D coculture model was developed to study the interplay between colon cancer cells and MCs. By comparing the transcriptomic profile of colon cancer-cocultured MCs versus control MCs, we identified a number of deregulated genes, such as MMP-2, VEGF-A, PDGF-A, COX2, NOTCH1 and ISG15, which contribute to the enrichment of cancer-related pathways. Intriguingly, pre-stimulation with a TLR2 agonist prior to colon cancer coculture induced upregulation of multiple interferon-inducible genes as well as MHC molecules in MCs. Our study provides an alternative approach to study the influence of colon cancer on MCs. The transcriptome signature of colon cancer-cocultured MCs may potentially reflect the mechanism of how colon cancer cells educate MCs to become pro-tumorigenic in the initial phase and how a subsequent inflammatory signal—e.g., TLR2 ligands—may modify their responses in the cancer milieu.
Collapse
|
41
|
Melissaridou S, Wiechec E, Magan M, Jain MV, Chung MK, Farnebo L, Roberg K. The effect of 2D and 3D cell cultures on treatment response, EMT profile and stem cell features in head and neck cancer. Cancer Cell Int 2019; 19:16. [PMID: 30651721 PMCID: PMC6332598 DOI: 10.1186/s12935-019-0733-1] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/08/2019] [Indexed: 01/20/2023] Open
Abstract
Background Head and Neck Squamous Cell Carcinoma (HNSCC) tumors are often resistant to therapies. Therefore searching for predictive markers and new targets for treatment in clinically relevant in vitro tumor models is essential. Five HNSCC-derived cell lines were used to assess the effect of 3D culturing compared to 2D monolayers in terms of cell proliferation, response to anti-cancer therapy as well as expression of EMT and CSC genes. Methods The viability and proliferation capacity of HNSCC cells as well as induction of apoptosis in tumor spheroids cells after treatment was assessed by MTT assay, crystal violet- and TUNEL assay respectively. Expression of EMT and CSC markers was analyzed on mRNA (RT-qPCR) and protein (Western blot) level. Results We showed that HNSCC cells from different tumors formed spheroids that differed in size and density in regard to EMT-associated protein expression and culturing time. In all spheroids, an up regulation of CDH1, NANOG and SOX2 was observed in comparison to 2D but changes in the expression of EGFR and EMT markers varied among the cell lines. Moreover, most HNSCC cells grown in 3D showed decreased sensitivity to cisplatin and cetuximab (anti-EGFR) treatment. Conclusions Taken together, our study points at notable differences between these two cellular systems in terms of EMT-associated gene expression profile and drug response. As the 3D cell cultures imitate the in vivo behaviour of neoplastic cells within the tumor, our study suggest that 3D culture model is superior to 2D monolayers in the search for new therapeutic targets. Electronic supplementary material The online version of this article (10.1186/s12935-019-0733-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Styliani Melissaridou
- 1Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Emilia Wiechec
- 1Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mustafa Magan
- 1Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Otorhinolaryngology in Linköping, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Linköping, Sweden
| | - Mayur Vilas Jain
- 1Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,3Department of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Man Ki Chung
- Department of Otorhinolaryngology-Head & Neck Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Lovisa Farnebo
- 1Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Otorhinolaryngology in Linköping, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Linköping, Sweden
| | - Karin Roberg
- 1Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Otorhinolaryngology in Linköping, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Linköping, Sweden
| |
Collapse
|
42
|
Iwai S, Kishimoto S, Amano Y, Nishiguchi A, Matsusaki M, Takeshita A, Akashi M. Three-dimensional cultured tissue constructs that imitate human living tissue organization for analysis of tumor cell invasion. J Biomed Mater Res A 2018; 107:292-300. [PMID: 29280265 PMCID: PMC6587574 DOI: 10.1002/jbm.a.36319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/09/2017] [Accepted: 12/21/2017] [Indexed: 01/17/2023]
Abstract
Preventing cancer metastasis requires a thorough understanding of cancer cell invasion. These phenomena occur in human 3-D living tissues. To this end, we developed a human cell-based three-dimensional (3-D) cultured tissue constructs that imitate in vivo human tissue organization. We investigated whether our 3-D cell culture system can be used to analyze the invasion of human oral squamous cell carcinoma (OSCC) cells. The 3-D tissue structure consisted of five layers of normal human dermal fibroblasts along with human dermal lymphatic endothelial cell tubes and was generated by the cell accumulation technique and layer-by-layer assembly using fibronectin and gelatin. OSCC cells with different lymph metastatic capacity were inoculated on the 3-D tissues and their invasion through the 3-D tissue structure was observed. Conventional methods of analyzing cell migration and invasion, that is, 2-D culture-based transwell and Matrigel assays were also used for comparison. The results using the 3-D cultured tissue constructs were comparable to those obtained using conventional assays; moreover, use of the 3-D system enabled visualization of differential invasion capacities of cancer cells. These results indicate that our 3-D cultured tissue constructs can be a useful tool for analysis of cancer cell invasion in a setting that reflects the in vivo tissue organization. © 2018 The Authors. Journal of Biomedical Materials Research Part A published by Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 292-300, 2019.
Collapse
Affiliation(s)
- Soichi Iwai
- Department of Oral and Maxillofacial Surgery II, Graduate School of Dentistry, Osaka University, 1-8, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Satoko Kishimoto
- Department of Oral and Maxillofacial Surgery II, Graduate School of Dentistry, Osaka University, 1-8, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yuto Amano
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Akihiro Nishiguchi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamada-oka, Suita, Osaka 565-0871, Japan.,PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama, Japan
| | - Akinori Takeshita
- Department of Oral and Maxillofacial Surgery II, Graduate School of Dentistry, Osaka University, 1-8, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Mitsuru Akashi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamada-oka, Suita, Osaka 565-0871, Japan.,Graduate School of Frontier Biosciences, Osaka University, 1-3, Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
43
|
Hao J, Zhang Y, Yan X, Yan F, Sun Y, Zeng J, Waigel S, Yin Y, Fraig MM, Egilmez NK, Suttles J, Kong M, Liu S, Cleary MP, Sauter E, Li B. Circulating Adipose Fatty Acid Binding Protein Is a New Link Underlying Obesity-Associated Breast/Mammary Tumor Development. Cell Metab 2018; 28:689-705.e5. [PMID: 30100196 PMCID: PMC6221972 DOI: 10.1016/j.cmet.2018.07.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/24/2018] [Accepted: 07/11/2018] [Indexed: 12/26/2022]
Abstract
It is clear that obesity increases the risk of many types of cancer, including breast cancer. However, the underlying molecular mechanisms by which obesity is linked to cancer risk remain to be defined. Herein, we report that circulating adipose fatty acid binding protein (A-FABP) promotes obesity-associated breast cancer development. Using clinical samples, we demonstrated that circulating A-FABP levels were significantly increased in obese patients with breast cancer in comparison with those without breast cancer. Circulating A-FABP released by adipose tissue directly targeted mammary tumor cells, enhancing tumor stemness and aggressiveness through activation of the IL-6/STAT3/ALDH1 pathway. Importantly, genetic deletion of A-FABP successfully reduced tumor ALHD1 activation and obesity-associated mammary tumor growth and development in different mouse models. Collectively, these data suggest circulating A-FABP as a new link between obesity and breast cancer risk, thereby revealing A-FABP as a potential new therapeutic target for treatment of obesity-associated cancers.
Collapse
Affiliation(s)
- Jiaqing Hao
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock Street, Louisville, KY 40202, USA
| | - Yuwen Zhang
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock Street, Louisville, KY 40202, USA
| | - Xiaofang Yan
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, USA
| | - Fei Yan
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Yanwen Sun
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock Street, Louisville, KY 40202, USA
| | - Jun Zeng
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock Street, Louisville, KY 40202, USA; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Sabine Waigel
- Genomics Facility and Life Tech Supply Center, University of Louisville, Louisville, KY, USA
| | - Yanhui Yin
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Mostafa M Fraig
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY, USA
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock Street, Louisville, KY 40202, USA
| | - Jill Suttles
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock Street, Louisville, KY 40202, USA
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, USA
| | - Shujun Liu
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Margot P Cleary
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Edward Sauter
- Hartford Healthcare Cancer Institute, Hartford, CT, USA
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock Street, Louisville, KY 40202, USA.
| |
Collapse
|
44
|
Hersi HM, Raulf N, Gaken J, Folarin N, Tavassoli M. MicroRNA-9 inhibits growth and invasion of head and neck cancer cells and is a predictive biomarker of response to plerixafor, an inhibitor of its target CXCR4. Mol Oncol 2018; 12:2023-2041. [PMID: 29959873 PMCID: PMC6275261 DOI: 10.1002/1878-0261.12352] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/15/2018] [Accepted: 06/28/2018] [Indexed: 12/16/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are associated with poor morbidity and mortality. Current treatment strategies are highly toxic and do not benefit over 50% of patients. There is therefore a crucial need for predictive and/or prognostic biomarkers to allow treatment stratification for individual patients. One class of biomarkers that has recently gained importance are microRNA (miRNA). MiRNA are small, noncoding molecules which regulate gene expression post‐transcriptionally. We performed miRNA expression profiling of a cohort of head and neck tumours with known clinical outcomes. The results showed miR‐9 to be significantly downregulated in patients with poor treatment outcome, indicating its role as a potential biomarker in HNSCC. Overexpression of miR‐9 in HNSCC cell lines significantly decreased cellular proliferation and inhibited colony formation in soft agar. Conversely, miR‐9 knockdown significantly increased both these features. Importantly, endogenous CXCR4 expression levels, a known target of miR‐9, inversely correlated with miR‐9 expression in a panel of HNSCC cell lines tested. Induced overexpression of CXCR4 in low expressing cells increased proliferation, colony formation and cell cycle progression. Moreover, CXCR4‐specific ligand, CXCL12, enhanced cellular proliferation, migration, colony formation and invasion in CXCR4‐overexpressing and similarly in miR‐9 knockdown cells. CXCR4‐specific inhibitor plerixafor abrogated the oncogenic phenotype of CXCR4 overexpression as well as miR‐9 knockdown. Our data demonstrate a clear role for miR‐9 as a tumour suppressor microRNA in HNSCC, and its role seems to be mediated through CXCR4 suppression. MiR‐9 knockdown, similar to CXCR4 overexpression, significantly promoted aggressive HNSCC tumour cell characteristics. Our results suggest CXCR4‐specific inhibitor plerixafor as a potential therapeutic agent, and miR‐9 as a possible predictive biomarker of treatment response in HNSCC.
Collapse
Affiliation(s)
| | - Nina Raulf
- Department of Molecular Oncology, King's College London, UK
| | - Joop Gaken
- Department of Haematological Medicine, The Rayne Institute, King's College London, UK
| | | | | |
Collapse
|
45
|
Muenzner JK, Kunze P, Lindner P, Polaschek S, Menke K, Eckstein M, Geppert CI, Chanvorachote P, Baeuerle T, Hartmann A, Schneider-Stock R. Generation and characterization of hepatocellular carcinoma cell lines with enhanced cancer stem cell potential. J Cell Mol Med 2018; 22:6238-6248. [PMID: 30280520 PMCID: PMC6237557 DOI: 10.1111/jcmm.13911] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/20/2018] [Indexed: 12/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes for cancer-related death worldwide with rapidly increasing incidence and mortality rates. As for other types of cancers, also in HCC cancer stem cells (CSCs) are thought to be responsible for tumour initiation, progression and therapy failure. However, as rare subpopulations of tumour tissue, CSCs are difficult to isolate, thus making the development of suitable and reliable model systems necessary. In our study, we generated HepG2 subclones with enriched CSC potential by application of the spheroid formation method and subsequent single-cell cloning. Analyses in several 2D and 3D cell culture systems as well as a panel of functional assays both in vitro and in vivo revealed that the generated subclones displayed characteristic and sustained features of tumour initiating cells as well as highly aggressive properties related to tumour progression and metastasis. These characteristics could clearly be correlated with the expression of CSC markers that might have prognostic value in the clinical HCC setting. Therefore, we conclude that our CSC enriched HepG2 clones certainly represent suitable model systems to study the role of CSCs during HCC initiation, progression and drug resistance.
Collapse
Affiliation(s)
- Julienne K Muenzner
- Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Philipp Kunze
- Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Pablo Lindner
- Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Sandra Polaschek
- Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Kira Menke
- Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Markus Eckstein
- Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Carol I Geppert
- Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tobias Baeuerle
- Preclinical Imaging Platform Erlangen (PIPE), Institute of Radiology, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Regine Schneider-Stock
- Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
46
|
Yu Y, Blokhuis B, Derks Y, Kumari S, Garssen J, Redegeld F. Human mast cells promote colon cancer growth via bidirectional crosstalk: studies in 2D and 3D coculture models. Oncoimmunology 2018; 7:e1504729. [PMID: 30377568 PMCID: PMC6205014 DOI: 10.1080/2162402x.2018.1504729] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 07/19/2018] [Accepted: 07/21/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation drives the development of colorectal cancer (CRC), where tumor-infiltrating immune cells interact with cancer cells in a dynamic crosstalk. Mast cells (MC), one of earliest recruited immune cells, accumulate in CRC tissues and their density is correlated with cancer progression. However, the exact contribution of MC in CRC and their interaction with colon cancer cells is poorly understood. Here, we investigated the impact of primary human MC and their mediators on colon cancer growth using 2D and 3D coculture models. Primary human MC were generated from peripheral CD34+ stem cells. Transwell chambers were used to analyze MC chemotaxis to colon cancer. Colon cancer cells HT29 and Caco2 differentially recruited MC by releasing CCL15 or SCF, respectively. Using BrdU proliferation assays, we demonstrated that MC can directly support colon cancer proliferation and this effect was mediated by their cellular crosstalk. 3D coculture models with cancer spheroids further confirmed the pro-tumor effect of MC on colon cancer growth, where direct cell-cell contact is dispensable and increased production of multiple soluble mediators was detected. Moreover, TLR2 stimulation of MC promoted stronger growth of colon cancer spheroids. By examining the transcriptome profile of colon cancer-cocultured MC versus control MC, we identified several MC marker genes, which were deregulated in expression. Our study provides an advanced in vitro model to investigate the role of human MC in cancer. Our data support the detrimental role of MC in CRC development and provide a molecular insight into the cellular crosstalk between MC and colon cancer cells.
Collapse
Affiliation(s)
- Yingxin Yu
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Bart Blokhuis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Yvonne Derks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Sangeeta Kumari
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,Department of Immunology, Nutricia Research, Utrecht, The Netherlands
| | - Frank Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
47
|
Cattin S, Ramont L, Rüegg C. Characterization and In Vivo Validation of a Three-Dimensional Multi-Cellular Culture Model to Study Heterotypic Interactions in Colorectal Cancer Cell Growth, Invasion and Metastasis. Front Bioeng Biotechnol 2018; 6:97. [PMID: 30065926 PMCID: PMC6056662 DOI: 10.3389/fbioe.2018.00097] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third cause of cancer-related mortality in industrialized countries. Local invasion and metastasis formation are events associated with poor prognosis for which today there are no effective therapeutic options. Invasion and metastasis are strongly modulated by cells of the tumor microenvironment (TME), in particular fibroblasts and endothelial cells. Unraveling interactions between tumor cells and cells of the TME may identify novel mechanisms and therapeutic targets to prevent or treat metastasis. We report here the development and in vivo validation of a 3D tumor spheroid model to study the interactions between CRC cells, fibroblasts and endothelial cells in vitro. Co-cultured fibroblasts promoted SW620 and HCT116 CRC spheroid invasion, and this was prevented by the SRC and FGFR kinase inhibitors Dasatinib and Erdafitinib, respectively. To validate these findings in vivo, we injected SW620 cells alone or together with fibroblasts orthotopically in the caecum of mice. Co-injection with fibroblasts promoted lung metastasis growth, which was fully reversed by treatment with Dasatinib or Erdafitinib. Co-culture of SW620 or HCT116 CRC spheroids with endothelial cells suppressed spheroid growth while it had no effect on cancer cell migration or invasion. Consistent with this in vitro effect, co-injected endothelial cells significantly inhibited primary tumor growth in vivo. From these experiments we conclude that effects on cancer cell invasion and growth induced by co-cultured TME cells and drug treatment in the 3D spheroid model in vitro, are predictive of in vivo effects. The 3D spheroid model may be considered as an attractive model to study the effect of heterotypic cellular interactions and drug activities on cancer cells, as animal testing alternative. This model may be adapted and further developed to include different types of cancer and host cells and to investigate additional functions and drugs.
Collapse
Affiliation(s)
- Sarah Cattin
- Department of Oncology, Faculty of Science and Medicine, Immunology and Microbiology, University of Fribourg, Fribourg, Switzerland
| | - Laurent Ramont
- Laboratory of Medical and Molecular Biology, Centre National de la Recherche Scientifique, Reims, France
| | - Curzio Rüegg
- Department of Oncology, Faculty of Science and Medicine, Immunology and Microbiology, University of Fribourg, Fribourg, Switzerland.,Swiss Integrative Center for Human Health, Fribourg, Switzerland
| |
Collapse
|
48
|
Fernandez HR, Gadre SM, Tan M, Graham GT, Mosaoa R, Ongkeko MS, Kim KA, Riggins RB, Parasido E, Petrini I, Pacini S, Cheema A, Varghese R, Ressom HW, Zhang Y, Albanese C, Üren A, Paige M, Giaccone G, Avantaggiati ML. The mitochondrial citrate carrier, SLC25A1, drives stemness and therapy resistance in non-small cell lung cancer. Cell Death Differ 2018; 25:1239-1258. [PMID: 29651165 PMCID: PMC6030199 DOI: 10.1038/s41418-018-0101-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/08/2018] [Accepted: 03/02/2018] [Indexed: 12/21/2022] Open
Abstract
Therapy resistance represents a clinical challenge for advanced non-small cell lung cancer (NSCLC), which still remains an incurable disease. There is growing evidence that cancer-initiating or cancer stem cells (CSCs) provide a reservoir of slow-growing dormant populations of cells with tumor-initiating and unlimited self-renewal ability that are left behind by conventional therapies reigniting post-therapy relapse and metastatic dissemination. The metabolic pathways required for the expansion of CSCs are incompletely defined, but their understanding will likely open new therapeutic opportunities. We show here that lung CSCs rely upon oxidative phosphorylation for energy production and survival through the activity of the mitochondrial citrate transporter, SLC25A1. We demonstrate that SLC25A1 plays a key role in maintaining the mitochondrial pool of citrate and redox balance in CSCs, whereas its inhibition leads to reactive oxygen species build-up thereby inhibiting the self-renewal capability of CSCs. Moreover, in different patient-derived tumors, resistance to cisplatin or to epidermal growth factor receptor (EGFR) inhibitor treatment is acquired through SLC25A1-mediated implementation of mitochondrial activity and induction of a stemness phenotype. Hence, a newly identified specific SLC25A1 inhibitor is synthetic lethal with cisplatin or with EGFR inhibitor co-treatment and restores antitumor responses to these agents in vitro and in animal models. These data have potential clinical implications in that they unravel a metabolic vulnerability of drug-resistant lung CSCs, identify a novel SLC25A1 inhibitor and, lastly, provide the first line of evidence that drugs, which block SLC25A1 activity, when employed in combination with selected conventional antitumor agents, lead to a therapeutic benefit.
Collapse
Affiliation(s)
- Harvey R Fernandez
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Shreyas M Gadre
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Mingjun Tan
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Garrett T Graham
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Rami Mosaoa
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Martin S Ongkeko
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Kyu Ah Kim
- Chemistry and Biochemistry Department, George Mason University, Fairfax, VA, USA
| | - Rebecca B Riggins
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Erika Parasido
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Iacopo Petrini
- Department of Clinical and Experimental Medicine, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine University of Pisa, Pisa, Italy
| | - Simone Pacini
- Department of Clinical and Experimental Medicine, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine University of Pisa, Pisa, Italy
| | - Amrita Cheema
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Rency Varghese
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Habtom W Ressom
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Yuwen Zhang
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Christopher Albanese
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Aykut Üren
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Mikell Paige
- Chemistry and Biochemistry Department, George Mason University, Fairfax, VA, USA
| | - Giuseppe Giaccone
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA
| | - Maria Laura Avantaggiati
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington D.C, 20057, USA.
| |
Collapse
|
49
|
Hall MK, Weidner DA, Whitman AA, Schwalbe RA. Lack of complex type N-glycans lessens aberrant neuronal properties. PLoS One 2018; 13:e0199202. [PMID: 29902282 PMCID: PMC6002081 DOI: 10.1371/journal.pone.0199202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/25/2018] [Indexed: 01/26/2023] Open
Abstract
Modifications in surface glycans attached to proteins via N-acetylglucosamine-β1-N-asparagine linkage have been linked to tumor development and progression. These modifications include complex N-glycans with high levels of branching, fucose and sialic acid residues. Previously, we silenced Mgat2 in neuroblastoma (NB) cells, which halted the conversion of hybrid type N-glycans to complex type, to generate a novel cell line, NB_1(-Mgat2). By comparing the aberrant cell properties of the NB_1(-Mgat2) cell line to the parental cell line (NB_1), we investigated the impact of eliminating complex type N-glycans on NB cell behavior. Further, the N-glycosylation pathway in the NB_1(-Mgat2) cell line was rescued by transiently transfecting cells with Mgat2, thus creating the NB_1(-/+Mgat2) cell line. Changes in the N-glycosylation pathway were verified by enhanced binding of E-PHA and L-PHA to proteins in the rescued cell line relative to those of the NB_1(-Mgat2) cell line. Also, western blotting of total membranes from the rescued cell line ectopically expressing a voltage-gated K+ channel (Kv3.1b) revealed that N-glycans of Kv3.1b were processed to complex type. By employment of various cell lines, we demonstrated that reduction of the complex type N-glycans diminished anchorage-independent cell growth, and enhanced cell-cell interactions. Two independent cell invasion assays showed that cell invasiveness was markedly lessened by lowering the levels of complex type N-glycans while cell mobility was only slightly modified. Neurites of NB cells were shortened by the absence of complex type N-glycans. Cell proliferation was reduced in NB cells with lowered levels of complex type N-glycans which resulted from hindered progression through G1+Go phases of the cell cycle. Overall, our results illustrate that reducing the ratio of complex to hybrid types of N-glycans diminishes aberrant NB cell behavior and thereby has a suppressive effect in cell proliferation, and cell dissociation and invasion phases of NB.
Collapse
Affiliation(s)
- M. Kristen Hall
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Douglas A. Weidner
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Austin A. Whitman
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Ruth A. Schwalbe
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| |
Collapse
|
50
|
Wang F, Li L, Piontek K, Sakaguchi M, Selaru FM. Exosome miR-335 as a novel therapeutic strategy in hepatocellular carcinoma. Hepatology 2018; 67:940-954. [PMID: 29023935 PMCID: PMC5826829 DOI: 10.1002/hep.29586] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 09/02/2017] [Accepted: 10/03/2017] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is a common and deadly cancer. Most cases of HCC arise in a cirrhotic/fibrotic liver, indicating that environment may play a paramount role in cancer genesis. Previous studies from our group and others have shown that, in desmoplastic cancers, there is a rich intercellular communication between activated, cancer-associated fibroblasts and cancer cells. Moreover, extracellular vesicles (EVs), or exosomes, have been identified as an important arm of this intercellular communication platform. Finally, these studies have shown that EVs can carry microRNA (miR) species in vivo and deliver them to desmoplastic cancers. The precise role played by activated liver fibroblasts/stellate cells in HCC development is insufficiently known. Based on previous studies, it appears plausible that activated fibroblasts produce signals carried by EVs that promote HCC genesis. In the current study, we first hypothesized and then demonstrated that stellate cell-derived EVs 1) can be loaded with an miR species of choice (miR-335-5p); 2) are taken up by HCC cells in vitro and more importantly in vivo; 3) can supply the miR-335-5p cargo to recipient HCC cells in vitro as well as in vivo; and 4) inhibit HCC cell proliferation and invasion in vitro as well as induce HCC tumor shrinkage in vivo. Finally, we identified messenger RNA targets for miR-335 that are down-regulated after treatment with EV-miR-335-5p. This study informs potential therapeutic strategies in HCC, whereby stellate cell-derived EVs are loaded with therapeutic nucleic acids and delivered in vivo. (Hepatology 2018;67:940-954).
Collapse
Affiliation(s)
- Fang Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Ling Li
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Klaus Piontek
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Masazumi Sakaguchi
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Florin M. Selaru
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
- Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, Maryland, USA
- The Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|