1
|
Wang Y, Shi P, Liu G, Chen W, Wang YJ, Hu Y, Yang A, Wei T, Chen YC, Liang L, Liu Z, Liu YJ, Wu C. Espin enhances confined cell migration by promoting filopodia formation and contributes to cancer metastasis. EMBO Rep 2025:10.1038/s44319-025-00437-1. [PMID: 40185977 DOI: 10.1038/s44319-025-00437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 02/22/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025] Open
Abstract
Genes regulating the finger-like cellular protrusions-filopodia have long been implicated in cancer metastasis. However, depleting the flat lamellipodia but retaining filopodia drastically hampers cell migration on spread surface, obscuring the role of filopodia in cell motility. It has been noticed recently that cells under confinement may employ distinct migratory machineries. However, the regulating factors have mainly been focused on cell blebbing, nuclear deformation and cell rear contractility, without much emphasis on cell protrusions and even less on filopodia. Here, by micropore-based screening, we identified espin as an active regulator for confined migration and that its overexpression was associated with metastasis. In comparison to fascin, espin showed stronger actin bundling in vitro and induced shorter and thicker filopodia in cells. Combining the imaging-compatible microchannels and DNA-based tension probes, we uncovered that espin overexpression induced excessive filopodia at the leading edge and along the sides, exerting force for confined migration. Our results demonstrate an important role for filopodia and the regulating protein-espin in confined cell migration and shed new light on cytoskeletal mechanisms underlying metastasis.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Peng Shi
- Cancer Institute, Suzhou Medical College, Soochow University, 215000, Suzhou, Jiangsu, China.
| | - Geyao Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Wei Chen
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei Province, China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China
| | - Yiping Hu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Ao Yang
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Tonghua Wei
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Yu-Chen Chen
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China
| | - Ling Liang
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Zheng Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei Province, China.
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China.
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China.
| |
Collapse
|
2
|
Guan G, Coates DE, Sun Q, Cannon RD, Mei L. Atomic Force Microscopy for Revealing Oncological Nanomechanobiology and Thermodynamics. ACS NANO 2025; 19:10862-10877. [PMID: 40084655 DOI: 10.1021/acsnano.4c14837] [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: 03/16/2025]
Abstract
Atomic force microscopy (AFM) is powerful nanobiotechnology for characterizing the nanotopographic and nanobiomechanical properties of live cells. Current limitations in AFM analysis of nanomechanobiology include the unjustified selection of nesting indices and filters, leading to the inaccurate reporting of waviness and roughness parameters, and inadequacies in the selection of the mathematical model for the Young's modulus. Critical biomechanical factors such as total deformation energy, elastic energy, and plastic energy are often overlooked. Here we refine and optimize the selection of the nesting index and filters for cellular analysis and develop an artificial intelligence-based classifier that can differentiate between normal and cancer cells. The application of AFM for detecting surface waviness and roughness, further enhanced by artificial intelligence (AI), represents a substantial advancement in cancer diagnostics. Although still in the experimental phase, AFM holds the potential to revolutionize cell biology and oncology by facilitating early cancer detection and advancing precision medicine. Moreover, this study's innovative exploration of the relationship between cellular nanomechanobiology and thermodynamics introduces important perspectives on cancer cell behavior at the nanoscale, unlocking opportunities for therapeutic interventions and cutting-edge oncological research. This paradigm shift may significantly influence the future trajectory of cancer biology and therapy.
Collapse
Affiliation(s)
- Guangzhao Guan
- Department of Oral Diagnostic and Surgical Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Dawn E Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9054, New Zealand
| | - Qing Sun
- Department of Oral Diagnostic and Surgical Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Richard D Cannon
- Department of Oral Sciences, Sir John Walsh Research Institute, University of Otago, Dunedin 9054, New Zealand
| | - Li Mei
- Department of Oral Sciences, Sir John Walsh Research Institute, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
3
|
Zhang K, Constantinou AP, O'Connell C, Georgiou TK, Gelmi A. A thermoresponsive PEG-based methacrylate triblock terpolymer as a bioink for 3D bioprinting. J Mater Chem B 2025; 13:3593-3601. [PMID: 39973333 DOI: 10.1039/d4tb02572e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Thermoresponsive polymers have been extensively reported for their use in tissue engineering and drug delivery applications. They have a wide range of thermoresponsive and rheological properties controlled by their structural characteristics, such as composition and architecture. Here, the considerable potential of a PEG based, non-ionic triblock thermoresponsive copolymer, namely OEGMA30013-b-BuMA22-b-DEGMA12 as a bioink for 3D printing with cell encapsulation is identified. The rheological tests showed that the gel transition temperature is 8 °C with 35% w/w concentration in PBS. The printability and cytotoxicity of the thermoresponsive gel were characterised and compared with those of commercial thermoresponsive polymer Pluronic®F127 in detail. Specifically, the 35% w/w triblock copolymer presented great printability with a printing speed of 450 mm min-1 at 37 °C, and was less cytotoxic than F127 at both 20% and 30% w/w concentrations. A one-layer structure of human mesenchymal stem cell (hMSC) embedded triblock copolymer was successfully printed onto a glass slide at 37 °C. This provides an option to create a scaffold for stem cell culture and programming for further tissue engineering applications via direct printing of a cell-laden thermoresponsive polymer.
Collapse
Affiliation(s)
- Kaiwen Zhang
- School of Science, RMIT University, VIC, 3000, Australia.
| | | | - Cathal O'Connell
- School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC, 3065, Australia
| | | | - Amy Gelmi
- School of Science, RMIT University, VIC, 3000, Australia.
| |
Collapse
|
4
|
Li Y, Sun Y, Yu K, Li Z, Miao H, Xiao W. Keratin: A potential driver of tumor metastasis. Int J Biol Macromol 2025; 307:141752. [PMID: 40049479 DOI: 10.1016/j.ijbiomac.2025.141752] [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: 10/08/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Keratins, as essential components of intermediate filaments in epithelial cells, play a crucial role in maintaining cell structure and function. In various malignant epithelial tumors, abnormal keratin expression is frequently observed and serves not only as a diagnostic marker but also closely correlates with tumor progression. Extensive research has demonstrated that keratins are pivotal in multiple stages of tumor metastasis, including responding to mechanical forces, evading the immune system, reprogramming metabolism, promoting angiogenesis, and resisting apoptosis. Here we emphasize that keratins significantly enhance the migratory and invasive capabilities of tumor cells, making them critical drivers of tumor metastasis. These findings highlight the importance of targeting keratins as a strategic approach to combat tumor metastasis, thereby advancing our understanding of their role in cancer progression and offering new therapeutic opportunities.
Collapse
Affiliation(s)
- Yuening Li
- Army Medical University, Chongqing, China
| | - Yiming Sun
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Kun Yu
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zhixi Li
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Jinfeng Laboratory, Chongqing, China.
| | - Weidong Xiao
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| |
Collapse
|
5
|
Wang X, Bai Y, Zhang X, Li W, Yang J, Hu N. Hydrodynamic efficient cell capture and pairing method on microfluidic cell electrofusion chip. APL Bioeng 2025; 9:016111. [PMID: 40051781 PMCID: PMC11884867 DOI: 10.1063/5.0250472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
Cell fusion is a widely employed process in various biological procedures, demonstrating significant application value in biotechnology. Cell pairing is a crucial manipulation for cell fusion. Standard fusion techniques, however, often provide poor and random cell contact, leading to low yields. In this study, we present a novel microfluidic device that utilizes a three-path symmetrical channel hydrodynamic capture method to achieve high-efficiency cell capture and pairing. The device contains several symmetrical channels and capture units, enabling three-path capture of two kinds of cells. To better understand the conditions necessary for effective cell pairing, we established a theoretical model of the three-path trapping flow field and conducted a qualitative force analysis on cells. Using K562 cells to explore the effect of different volumetric flow ratios of symmetric channels on cell capture and pairing efficiency, we finally got the optimized structure and obtained a single-cell capture efficiency of approximately 95.6 ± 2.0% and a cell pairing efficiency of approximately 83.3 ± 8.8%. Subsequently, electrofusion experiments were carried out on the paired cells, resulting in a fusion efficiency of approximately 77.8 ± 9.6%.
Collapse
Affiliation(s)
- Xuefeng Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Yaqi Bai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Xiaoling Zhang
- School of Smart Health, Chongqing College of Electronic Engineering, Chongqing 401331, China
| | - Wei Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Jun Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Ning Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| |
Collapse
|
6
|
Mehanna LE, Boyd JD, Remus-Williams S, Racca NM, Spraggins DP, Grady ME, Berron BJ. Improvement of cellular pattern organization and clarity through centrifugal force. Biomed Mater 2025; 20:025025. [PMID: 39746325 PMCID: PMC11823422 DOI: 10.1088/1748-605x/ada508] [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] [Received: 07/18/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/04/2025]
Abstract
Rapid and strategic cell placement is necessary for high throughput tissue fabrication. Current adhesive cell patterning systems rely on fluidic shear flow to remove cells outside of the patterned regions, but limitations in washing complexity and uniformity prevent adhesive patterns from being widely applied. Centrifugation is commonly used to study the adhesive strength of cells to various substrates; however, the approach has not been applied to selective cell adhesion systems to create highly organized cell patterns. This study shows centrifugation as a promising method to wash cellular patterns after selective binding of cells to the surface has taken place. After patterning H9C2 cells using biotin-streptavidin as a model adhesive patterning system and washing with centrifugation, there is a significant number of cells removed outside of the patterned areas of the substrate compared to the initial seeding, while there is not a significant number removed from the desired patterned areas. This method is effective in patterning multiple size and linear structures from line widths of 50-200 μm without compromising immediate cell viability below 80%. We also test this procedure on a variety of tube-forming cell lines (MPCs, HUVECs) on various tissue-like surface materials (collagen 1 and Matrigel) with no significant differences in their respective tube formation metrics when the cells were seeded directly on their unconjugated surface versus patterned and washed through centrifugation. This result demonstrates that our patterning and centrifugation system can be adapted to a variety of cell types and substrates to create patterns tailored to many biological applications.
Collapse
Affiliation(s)
- Lauren E Mehanna
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, United States of America
| | - James D Boyd
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Shelley Remus-Williams
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Nicole M Racca
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Dawson P Spraggins
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Martha E Grady
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Brad J Berron
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, United States of America
| |
Collapse
|
7
|
d'Agata L, Rassinoux P, Gounou C, Bouvet F, Bouragba D, Mamchaoui K, Bouter A. A Novel Assay Reveals the Early Setting-Up of Membrane Repair Machinery in Human Skeletal Muscle Cells. J Cell Biochem 2025; 126:e30662. [PMID: 39348239 PMCID: PMC11729639 DOI: 10.1002/jcb.30662] [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] [Received: 04/28/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 10/02/2024]
Abstract
Defect in membrane repair contributes to the development of muscular dystrophies such as limb girdle muscular dystrophy (LGMD) type R2 or R12. Nevertheless, many other muscular dystrophies may also result from a defect in this process. Identifying these pathologies requires the development of specific methods to inflict sarcolemma damage on a large number of cells and rapidly analyze their response. We adapted a protocol hitherto used to study the behavior of cancer cells to mechanical constraint. This method is based on forcing the passage of cells through a thin needle, which induces shear stress. Due to size considerations, this method requires working with mononuclear muscle cells instead of myotubes or muscle fibers. Although functional sarcolemma repair was thought to be restricted to myotubes and muscle fibers, we show here that 24h-differentiated myoblasts express a complete machinery capable of addressing membrane damage. At this stage, muscle cells do not yet form myotubes, revealing that the membrane repair machinery is set up early throughout the differentiation process. When submitted to the shear-stress assay, these cells were observed to repair membrane damage in a Ca2+-dependent manner, as previously reported. We show that this technique is able to identify the absence of membrane resealing in muscle cells from patient suffering from LGMDR2. The proposed technique provides therefore a suitable method for identifying cellular dysregulations in membrane repair of dystrophic human muscle cells.
Collapse
Affiliation(s)
- Léna d'Agata
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248PessacFrance
| | | | - Céline Gounou
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248PessacFrance
| | - Flora Bouvet
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248PessacFrance
| | - Dounia Bouragba
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en MyologieParisFrance
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en MyologieParisFrance
| | - Anthony Bouter
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248PessacFrance
| |
Collapse
|
8
|
Qiu Y, Gao T, Smith BR. Mechanical deformation and death of circulating tumor cells in the bloodstream. Cancer Metastasis Rev 2024; 43:1489-1510. [PMID: 38980581 PMCID: PMC11900898 DOI: 10.1007/s10555-024-10198-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
The circulation of tumor cells through the bloodstream is a significant step in tumor metastasis. To better understand the metastatic process, circulating tumor cell (CTC) survival in the circulation must be explored. While immune interactions with CTCs in recent decades have been examined, research has yet to sufficiently explain some CTC behaviors in blood flow. Studies related to CTC mechanical responses in the bloodstream have recently been conducted to further study conditions under which CTCs might die. While experimental methods can assess the mechanical properties and death of CTCs, increasingly sophisticated computational models are being built to simulate the blood flow and CTC mechanical deformation under fluid shear stresses (FSS) in the bloodstream.Several factors contribute to the mechanical deformation and death of CTCs as they circulate. While FSS can damage CTC structure, diverse interactions between CTCs and blood components may either promote or hinder the next metastatic step-extravasation at a remote site. Overall understanding of how these factors influence the deformation and death of CTCs could serve as a basis for future experiments and simulations, enabling researchers to predict CTC death more accurately. Ultimately, these efforts can lead to improved metastasis-specific therapeutics and diagnostics specific in the future.
Collapse
Affiliation(s)
- Yunxiu Qiu
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, 48824, USA
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Tong Gao
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Department of Computational Mathematics, Science, and Engineering, East Lansing, MI, 48824, USA
| | - Bryan Ronain Smith
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, 48824, USA.
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
9
|
Cardenas-Benitez B, Hurtado R, Luo X, Lee AP. Three-dimensional isotropic imaging of live suspension cells enabled by droplet microvortices. Proc Natl Acad Sci U S A 2024; 121:e2408567121. [PMID: 39436653 PMCID: PMC11536124 DOI: 10.1073/pnas.2408567121] [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] [Received: 04/30/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Fast, nondestructive three-dimensional (3D) imaging of live suspension cells remains challenging without substrate treatment or fixation, precluding scalable single-cell morphometry with minimal alterations. While optical sectioning techniques achieve 3D live cell imaging, lateral versus depth resolution differences further complicate analysis. We present a scalable microfluidic method capable of 3D fluorescent isotropic imaging of live, nonadherent cells suspended inside picoliter droplets with high-speed single-cell volumetric readout (800 to 1,200 slices in 5 to 8 s) and near-diffraction limit resolution (~216 nm). The platform features a droplet trap array that leverages flow-induced droplet interfacial shear to generate intradroplet microvortices, which rotate single cells on their axis to enable optical projection tomography (OPT)-based imaging. This allows gentle (~1 mPa shear stress) observation of cells encapsulated inside nontoxic isotonic buffer droplets, facilitating scalable OPT acquisition by simultaneous spinning of hundreds of cells. We demonstrate 3D imaging of live myeloid and lymphoid cells in suspension, including K562 cells, as well as naive and activated T cells-small cells prone to movement in their suspended phenotype. Our fully suspended, orientation-independent cell morphometry, driven by isotropic imaging and spherical harmonic analysis, enabled the study of primary T cells across various immunological activation states. This approach unveiled six distinct nuclear content distributions, contrasting with conventional 2D images that typically portray spheroid and bean-like nuclear shapes associated with lymphocytes. Our arrayed-droplet OPT technology is capable of isotropic, single live-cell 3D imaging, with the potential to perform large-scale morphometry of immune cell effector function states while providing compatibility with microfluidic droplet operations.
Collapse
Affiliation(s)
- Braulio Cardenas-Benitez
- Department of Biomedical Engineering, University of California, Irvine, CA92697
- Center for Advanced Design & Manufacturing of Integrated Microfluidics, University of California, Irvine, CA92697
| | - Richard Hurtado
- Department of Biomedical Engineering, University of California, Irvine, CA92697
- Center for Advanced Design & Manufacturing of Integrated Microfluidics, University of California, Irvine, CA92697
| | - Xuhao Luo
- Department of Biomedical Engineering, University of California, Irvine, CA92697
- Center for Advanced Design & Manufacturing of Integrated Microfluidics, University of California, Irvine, CA92697
| | - Abraham P. Lee
- Department of Biomedical Engineering, University of California, Irvine, CA92697
- Center for Advanced Design & Manufacturing of Integrated Microfluidics, University of California, Irvine, CA92697
- Department of Mechanical and Aerospace Engineering, University of California, Irvine, CA92697
| |
Collapse
|
10
|
Mand M, Hahn O, Meyer J, Peters K, Seitz H. Investigation of the Effect of High Shear Stress on Mesenchymal Stem Cells Using a Rotational Rheometer in a Small-Angle Cone-Plate Configuration. Bioengineering (Basel) 2024; 11:1011. [PMID: 39451387 PMCID: PMC11504001 DOI: 10.3390/bioengineering11101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Within the healthy human body, cells reside within the physiological environment of a tissue compound. Here, they are subject to constant low levels of mechanical stress that can influence the growth and differentiation of the cells. The liposuction of adipose tissue and the subsequent isolation of mesenchymal stem/stromal cells (MSCs), for example, are procedures that induce a high level of mechanical shear stress. As MSCs play a central role in tissue regeneration by migrating into regenerating areas and driving regeneration through proliferation and tissue-specific differentiation, they are increasingly used in therapeutic applications. Consequently, there is a strong interest in investigating the effects of shear stress on MSCs. In this study, we present a set-up for applying high shear rates to cells based on a rotational rheometer with a small-angle cone-plate configuration. This set-up was used to investigate the effect of various shear stresses on human adipose-derived MSCs in suspension. The results of the study show that the viability of the cells remained unaffected up to 18.38 Pa for an exposure time of 5 min. However, it was observed that intense shear stress damaged the cells, with longer treatment durations increasing the percentage of cell debris.
Collapse
Affiliation(s)
- Mario Mand
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18059 Rostock, Germany
| | - Olga Hahn
- Institute of Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany; (O.H.); (K.P.)
| | | | - Kirsten Peters
- Institute of Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany; (O.H.); (K.P.)
- Department of Life, Light and Matter, University of Rostock, 18059 Rostock, Germany
| | - Hermann Seitz
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18059 Rostock, Germany
- Department of Life, Light and Matter, University of Rostock, 18059 Rostock, Germany
| |
Collapse
|
11
|
Zhou S, Xu H, Duan Y, Tang Q, Huang H, Bi F. Survival mechanisms of circulating tumor cells and their implications for cancer treatment. Cancer Metastasis Rev 2024; 43:941-957. [PMID: 38436892 DOI: 10.1007/s10555-024-10178-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Metastasis remains the principal trigger for relapse and mortality across diverse cancer types. Circulating tumor cells (CTCs), which originate from the primary tumor or its metastatic sites, traverse the vascular system, serving as precursors in cancer recurrence and metastasis. Nevertheless, before CTCs can establish themselves in the distant parenchyma, they must overcome significant challenges present within the circulatory system, including hydrodynamic shear stress (HSS), oxidative damage, anoikis, and immune surveillance. Recently, there has been a growing body of compelling evidence suggesting that a specific subset of CTCs can persist within the bloodstream, but the precise mechanisms of their survival remain largely elusive. This review aims to present an outline of the survival challenges encountered by CTCs and to summarize the recent advancements in understanding the underlying survival mechanisms, suggesting their implications for cancer treatment.
Collapse
Affiliation(s)
- Shuang Zhou
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huanji Xu
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yichun Duan
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qiulin Tang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huixi Huang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Feng Bi
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
12
|
Lopez-Cavestany M, Wright OA, Reckhorn NT, Carter AT, Jayawardana K, Nguyen T, Briggs DP, Koktysh DS, Esteban Linares A, Li D, King MR. Superhydrophobic Array Devices for the Enhanced Formation of 3D Cancer Models. ACS NANO 2024; 18:23637-23654. [PMID: 39150223 PMCID: PMC11363216 DOI: 10.1021/acsnano.4c08132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
During the metastatic cascade, cancer cells travel through the bloodstream as circulating tumor cells (CTCs) to a secondary site. Clustered CTCs have greater shear stress and treatment resistance, yet their biology remains poorly understood. We therefore engineered a tunable superhydrophobic array device (SHArD). The SHArD-C was applied to culture a clinically relevant model of CTC clusters. Using our device, we cultured a model of cancer cell aggregates of various sizes with immortalized cancer cell lines. These exhibited higher E-cadherin expression and are significantly more capable of surviving high fluid shear stress-related forces compared to single cells and model clusters grown using the control method, helping to explain why clustering may provide a metastatic advantage. Additionally, the SHArD-S, when compared with the AggreWell 800 method, provides a more consistent spheroid-forming device culturing reproducible sizes of spheroids for multiple cancer cell lines. Overall, we designed, fabricated, and validated an easily tunable engineered device which grows physiologically relevant three-dimensional (3D) cancer models containing tens to thousands of cells.
Collapse
Affiliation(s)
- Maria Lopez-Cavestany
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Olivia A. Wright
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Noah T. Reckhorn
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Alexandria T. Carter
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - Kalana Jayawardana
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Tin Nguyen
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Dayrl P. Briggs
- Center
for Nanophase Materials Science, Oak Ridge
National Laboratories, Knoxville, Tennessee 37830, United States
| | - Dmitry S. Koktysh
- Vanderbilt
Institute for Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Alberto Esteban Linares
- Department
of Mechanical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Deyu Li
- Department
of Mechanical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Michael R. King
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| |
Collapse
|
13
|
Raskov H, Orhan A, Agerbæk MØ, Gögenur I. The impact of platelets on the metastatic potential of tumour cells. Heliyon 2024; 10:e34361. [PMID: 39114075 PMCID: PMC11305202 DOI: 10.1016/j.heliyon.2024.e34361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
In cancer, activation of platelets by tumor cells is critical to disease progression. Development of precise antiplatelet targeting may improve outcomes from anticancer therapy. Alongside a distinct shift in functionality such as pro-metastatic and pro-coagulant properties, platelet production is often accelerated significantly early in carcinogenesis and the cancer-associated thrombocytosis increases the risk of metastasis formation and thromboembolic events. Tumor-activated platelets facilitate the proliferation of migrating tumor cells and shield them from immune surveillance and physical stress during circulation. Additionally, platelet-tumor cell interactions promote tumor cell intravasation, intravascular arrest, and extravasation through a repertoire of adhesion molecules, growth factors and angiogenic factors. Particularly, the presence of circulating tumor cell (CTC) clusters in association with platelets is a negative prognostic indicator. The contribution of platelets to the metastatic process is an area of intense investigation and this review provides an overview of the advances in understanding platelet-tumor cell interactions and their contribution to disease progression. Also, we review the potential of targeting platelets to interfere with the metastatic process.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Mette Ørskov Agerbæk
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Li W, Guo Z, Zhou Z, Zhou Z, He H, Sun J, Zhou X, Chin YR, Zhang L, Yang M. Distinguishing high-metastasis-potential circulating tumor cells through fluidic shear stress in a bloodstream-like microfluidic circulatory system. Oncogene 2024; 43:2295-2306. [PMID: 38858591 DOI: 10.1038/s41388-024-03075-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
Circulating tumor cells (CTCs) play a critical role as initiators in tumor metastasis, which unlocks an irreversible process of cancer progression. Regarding the fluid environment of intravascular CTCs, a comprehensive understanding of the impact of hemodynamic shear stress on CTCs is of profound significance but remains vague. Here, we report a microfluidic circulatory system that can emulate the CTC microenvironment to research the responses of typical liver cancer cells to varying levels of fluid shear stress (FSS). We observe that HepG2 cells surviving FSS exhibit a marked overexpression of TLR4 and TPPP3, which are shown to be associated with the colony formation, migration, and anti-apoptosis abilities of HepG2. Furthermore, overexpression of these two genes in another liver cancer cell line with normally low TLR4 and TPPP3 expression, SK-Hep-1 cells, by lentivirus-mediated transfection also confirms the critical role of TLR4 and TPPP3 in improving colony formation, migration, and survival capability under a fluid environment. Interestingly, in vivo experiments show SK-Hep-1 cells, overexpressed with these genes, have enhanced metastatic potential to the liver and lungs in mouse models via tail vein injection. Mechanistically, TLR4 and TPPP3 upregulated by FSS may increase FSS-mediated cell survival and metastasis through the p53-Bax signaling pathway. Moreover, elevated levels of these genes correlate with poorer overall survival in liver cancer patients, suggesting that our findings could offer new therapeutic strategies for early cancer diagnosis and targeted treatment development.
Collapse
Affiliation(s)
- Wenxiu Li
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Zhengjun Guo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhengdong Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Huimin He
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Y Rebecca Chin
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Liang Zhang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China.
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China.
| |
Collapse
|
15
|
Filipe EC, Velayuthar S, Philp A, Nobis M, Latham SL, Parker AL, Murphy KJ, Wyllie K, Major GS, Contreras O, Mok ETY, Enriquez RF, McGowan S, Feher K, Quek L, Hancock SE, Yam M, Tran E, Setargew YFI, Skhinas JN, Chitty JL, Phimmachanh M, Han JZR, Cadell AL, Papanicolaou M, Mahmodi H, Kiedik B, Junankar S, Ross SE, Lam N, Coulson R, Yang J, Zaratzian A, Da Silva AM, Tayao M, Chin IL, Cazet A, Kansara M, Segara D, Parker A, Hoy AJ, Harvey RP, Bogdanovic O, Timpson P, Croucher DR, Lim E, Swarbrick A, Holst J, Turner N, Choi YS, Kabakova IV, Philp A, Cox TR. Tumor Biomechanics Alters Metastatic Dissemination of Triple Negative Breast Cancer via Rewiring Fatty Acid Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307963. [PMID: 38602451 PMCID: PMC11186052 DOI: 10.1002/advs.202307963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/11/2024] [Indexed: 04/12/2024]
Abstract
In recent decades, the role of tumor biomechanics on cancer cell behavior at the primary site has been increasingly appreciated. However, the effect of primary tumor biomechanics on the latter stages of the metastatic cascade, such as metastatic seeding of secondary sites and outgrowth remains underappreciated. This work sought to address this in the context of triple negative breast cancer (TNBC), a cancer type known to aggressively disseminate at all stages of disease progression. Using mechanically tuneable model systems, mimicking the range of stiffness's typically found within breast tumors, it is found that, contrary to expectations, cancer cells exposed to softer microenvironments are more able to colonize secondary tissues. It is shown that heightened cell survival is driven by enhanced metabolism of fatty acids within TNBC cells exposed to softer microenvironments. It is demonstrated that uncoupling cellular mechanosensing through integrin β1 blocking antibody effectively causes stiff primed TNBC cells to behave like their soft counterparts, both in vitro and in vivo. This work is the first to show that softer tumor microenvironments may be contributing to changes in disease outcome by imprinting on TNBC cells a greater metabolic flexibility and conferring discrete cell survival advantages.
Collapse
|
16
|
Fabiano AR, Robbins SC, Knoblauch SV, Rowland SJ, Dombroski JA, King MR. Multiplex, high-throughput method to study cancer and immune cell mechanotransduction. Commun Biol 2024; 7:674. [PMID: 38824207 PMCID: PMC11144229 DOI: 10.1038/s42003-024-06327-x] [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] [Received: 12/07/2023] [Accepted: 05/14/2024] [Indexed: 06/03/2024] Open
Abstract
Studying cellular mechanoresponses during cancer metastasis is limited by sample variation or complex protocols that current techniques require. Metastasis is governed by mechanotransduction, whereby cells translate external stimuli, such as circulatory fluid shear stress (FSS), into biochemical cues. We present high-throughput, semi-automated methods to expose cells to FSS using the VIAFLO96 multichannel pipetting device custom-fitted with 22 G needles, increasing the maximum FSS 94-fold from the unmodified tips. Specifically, we develop protocols to semi-automatically stain live samples and to fix, permeabilize, and intracellularly process cells for flow cytometry analysis. Our first model system confirmed that the pro-apoptotic effects of TRAIL therapeutics in prostate cancer cells can be enhanced via FSS-induced Piezo1 activation. Our second system implements this multiplex methodology to show that FSS exposure (290 dyn cm-2) increases activation of murine bone marrow-derived dendritic cells. These methodologies greatly improve the mechanobiology workflow, offering a high-throughput, multiplex approach.
Collapse
Affiliation(s)
- Abigail R Fabiano
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Spencer C Robbins
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Samantha V Knoblauch
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Schyler J Rowland
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Jenna A Dombroski
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN, 37212, USA.
| |
Collapse
|
17
|
Xin Y, Hu B, Li K, Hu G, Zhang C, Chen X, Tang K, Du P, Tan Y. Circulating tumor cells with metastasis-initiating competence survive fluid shear stress during hematogenous dissemination through CXCR4-PI3K/AKT signaling. Cancer Lett 2024; 590:216870. [PMID: 38614386 DOI: 10.1016/j.canlet.2024.216870] [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: 01/11/2024] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 04/15/2024]
Abstract
To seed lethal secondary lesions, circulating tumor cells (CTCs) must survive all rate-limiting factors during hematogenous dissemination, including fluid shear stress (FSS) that poses a grand challenge to their survival. We thus hypothesized that CTCs with the ability to survive FSS in vasculature might hold metastasis-initiating competence. This study reported that FSS of physiologic magnitude selected a small subpopulation of suspended tumor cells in vitro with the traits of metastasis-initiating cells, including stemness, migration/invasion potential, cellular plasticity, and biophysical properties. These shear-selected cells generated local and metastatic tumors at the primary and distal sites efficiently, implicating their metastasis competence. Mechanistically, FSS activated the mechanosensitive protein CXCR4 and the downstream PI3K/AKT signaling, which were essential in shear-mediated selection of metastasis-competent CTCs. In summary, these findings conclude that CTCs with metastasis-initiating competence survive FSS during hematogenous dissemination through CXCR4-PI3K/AKT signaling, which may provide new therapeutic targets for the early prevention of tumor metastasis.
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Bing Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Guanshuo Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Cunyu Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Kai Tang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Pengyu Du
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China.
| |
Collapse
|
18
|
Bai Y, Zhang X, Wang X, Xu M, Yang J, Hu N. Controllable and Stable Fusion Strategy on Microfluidics. Anal Chem 2024; 96:4437-4445. [PMID: 38501259 DOI: 10.1021/acs.analchem.3c04592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
This paper presents a microfluidic device with 200 cell "cage" structures. Based on this microfluidics device, a new simple and stable electrofusion method was developed. Under hydrodynamic force, the cells moved to the desired "cage" cell capture structure and efficiently formed cell pairs (∼80.0 ± 4.6%). Intimate intercellular connectivity was induced by the precise modulation of hypotonic solution substitution and the microstructure, which ensured no cell movement or displacement during the cell electroporation/electrofusion process. It also guaranteed repeated electroporation occurring in the same contact region and provided a stable cell membrane recombination and a cell fusion microenvironment. When the pulse signal was applied, a high fusion efficiency of ∼88.3 ± 0.6% was demonstrated on the microfluidic device.
Collapse
Affiliation(s)
- Yaqi Bai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Xiaoling Zhang
- School of Smart Health, Chongqing College of Electronic Engineering, Chongqing 401331, China
| | - Xuefeng Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Mengli Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Jun Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Ning Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing 400044, China
| |
Collapse
|
19
|
Kerry J, Specker EJ, Mizzoni M, Brumwell A, Fell L, Goodbrand J, Rosen MN, Uniacke J. Autophagy-dependent alternative splicing of ribosomal protein S24 produces a more stable isoform that aids in hypoxic cell survival. FEBS Lett 2024; 598:503-520. [PMID: 38281767 DOI: 10.1002/1873-3468.14804] [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] [Received: 11/05/2023] [Revised: 12/08/2023] [Accepted: 12/24/2023] [Indexed: 01/30/2024]
Abstract
Cells remodel splicing and translation machineries to mount specialized gene expression responses to stress. Here, we show that hypoxic human cells in 2D and 3D culture models increase the relative abundance of a longer mRNA variant of ribosomal protein S24 (RPS24L) compared to a shorter mRNA variant (RPS24S) by favoring the inclusion of a 22 bp cassette exon. Mechanistically, RPS24L and RPS24S are induced and repressed, respectively, by distinct pathways in hypoxia: RPS24L is induced in an autophagy-dependent manner, while RPS24S is reduced by mTORC1 repression in a hypoxia-inducible factor-dependent manner. RPS24L produces a more stable protein isoform that aids in hypoxic cell survival and growth, which could be exploited by cancer cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Jenna Kerry
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Erin J Specker
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Morgan Mizzoni
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Andrea Brumwell
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Leslie Fell
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Jenna Goodbrand
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Michael N Rosen
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - James Uniacke
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| |
Collapse
|
20
|
Mendez J, Toker A. Minimizing Shear Stress in Cell Signaling Studies. Curr Protoc 2024; 4:e1019. [PMID: 38506433 DOI: 10.1002/cpz1.1019] [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: 03/21/2024]
Abstract
Cellular signal transduction comprises a complex series of biochemical reactions by which extracellular signals such as growth factors, hormones, cytokines, and neurotransmitters are translated into specific intracellular responses. Signal transduction is mediated by protein kinase phosphorylation cascades that culminate in the regulation of numerous cellular responses, including division, differentiation, migration, and survival. Importantly, signal relay pathways are dysregulated in human diseases, making the study of signal transduction important for both uncovering basic biology and understanding pathophysiology. Established laboratory cell culture models are useful for studying signal transduction mechanisms, but differences in sample handling procedures can introduce unwanted variability in experimental outcomes and conclusions. One such potential source of experimental variability is the introduction of fluid shear stress upon handling of tissue culture cells. Fluid shear stress triggers a wide range of cellular responses in adherent cell culture, including stimulating the production of cyclic AMP, potentiating the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), and ultimately inducing changes in the gene expression of growth and remodeling factors. Further, mechanical stress on cells is physiologically relevant to the development of many pathologies. Here, we describe a detailed protocol for cell lysis and protein extraction that minimizes shear stress induced by classical cell harvest protocols. We also highlight the impact of fluid shear stress by using immunoblotting to assess ERK pathway activation as a readout for this protocol. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Gentle cell lysis and protein extraction Basic Protocol 2: Immunoblotting for cell signaling readouts by SDS-PAGE.
Collapse
Affiliation(s)
- Josefina Mendez
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Zhang Y, O'Mahony A, He Y, Barber T. Hydrodynamic shear stress' impact on mammalian cell properties and its applications in 3D bioprinting. Biofabrication 2024; 16:022003. [PMID: 38277669 DOI: 10.1088/1758-5090/ad22ee] [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: 03/03/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
As an effective cell assembly method, three-dimensional bioprinting has been widely used in building organ models and tissue repair over the past decade. However, different shear stresses induced throughout the entire printing process can cause complex impacts on cell integrity, including reducing cell viability, provoking morphological changes and altering cellular functionalities. The potential effects that may occur and the conditions under which these effects manifest are not clearly understood. Here, we review systematically how different mammalian cells respond under shear stress. We enumerate available experimental apparatus, and we categorise properties that can be affected under disparate stress patterns. We also summarise cell damaging mathematical models as a predicting reference for the design of bioprinting systems. We concluded that it is essential to quantify specific cell resistance to shear stress for the optimisation of bioprinting systems. Besides, as substantial positive impacts, including inducing cell alignment and promoting cell motility, can be generated by shear stress, we suggest that we find the proper range of shear stress and actively utilise its positive influences in the development of future systems.
Collapse
Affiliation(s)
- Yani Zhang
- School of Mechanical Engineering, UNSW, Sydney, NSW 2052, Australia
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Aidan O'Mahony
- Inventia Life Science Pty Ltd, Alexandria, Sydney, NSW 2015, Australia
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Tracie Barber
- School of Mechanical Engineering, UNSW, Sydney, NSW 2052, Australia
| |
Collapse
|
22
|
Emmons MF, Bennett RL, Riva A, Gupta K, Carvalho LADC, Zhang C, Macaulay R, Dupéré-Richér D, Fang B, Seto E, Koomen JM, Li J, Chen YA, Forsyth PA, Licht JD, Smalley KSM. HDAC8-mediated inhibition of EP300 drives a transcriptional state that increases melanoma brain metastasis. Nat Commun 2023; 14:7759. [PMID: 38030596 PMCID: PMC10686983 DOI: 10.1038/s41467-023-43519-1] [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] [Received: 08/24/2022] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Melanomas can adopt multiple transcriptional states. Little is known about the epigenetic drivers of these cell states, limiting our ability to regulate melanoma heterogeneity. Here, we identify stress-induced HDAC8 activity as driving melanoma brain metastasis development. Exposure of melanocytes and melanoma cells to multiple stresses increases HDAC8 activation leading to a neural crest-stem cell transcriptional state and an amoeboid, invasive phenotype that increases seeding to the brain. Using ATAC-Seq and ChIP-Seq we show that increased HDAC8 activity alters chromatin structure by increasing H3K27ac and enhancing accessibility at c-Jun binding sites. Functionally, HDAC8 deacetylates the histone acetyltransferase EP300, causing its enzymatic inactivation. This, in turn, increases binding of EP300 to Jun-transcriptional sites and decreases binding to MITF-transcriptional sites. Inhibition of EP300 increases melanoma cell invasion, resistance to stress and increases melanoma brain metastasis development. HDAC8 is identified as a mediator of transcriptional co-factor inactivation and chromatin accessibility that drives brain metastasis.
Collapse
Affiliation(s)
- Michael F Emmons
- Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Richard L Bennett
- UF Health Cancer Center, 2033 Mowry Road, University of Florida, Gainesville, FL, 32610, USA
| | - Alberto Riva
- Bioinformatics Core, Interdisciplinary Center for Biotechnology Research, University of Florida, 2033 Mowry Road, Gainesville, FL, 32610, USA
| | - Kanchan Gupta
- UF Health Cancer Center, 2033 Mowry Road, University of Florida, Gainesville, FL, 32610, USA
| | | | - Chao Zhang
- Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Robert Macaulay
- Department of Neuro-Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Daphne Dupéré-Richér
- UF Health Cancer Center, 2033 Mowry Road, University of Florida, Gainesville, FL, 32610, USA
| | - Bin Fang
- Proteomics & Metabolomics Core, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Edward Seto
- Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, George Washington Cancer Center, George Washington University, 2300 Eye Street, Washington, DC, 20037, USA
| | - John M Koomen
- Department of Molecular Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Jiannong Li
- Department of Bioinformatics and Biostatistics, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Y Ann Chen
- Department of Bioinformatics and Biostatistics, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Peter A Forsyth
- Department of Neuro-Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Jonathan D Licht
- UF Health Cancer Center, 2033 Mowry Road, University of Florida, Gainesville, FL, 32610, USA
| | - Keiran S M Smalley
- Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
23
|
Ildiz ES, Gvozdenovic A, Kovacs WJ, Aceto N. Travelling under pressure - hypoxia and shear stress in the metastatic journey. Clin Exp Metastasis 2023; 40:375-394. [PMID: 37490147 PMCID: PMC10495280 DOI: 10.1007/s10585-023-10224-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023]
Abstract
Cancer cell invasion, intravasation and survival in the bloodstream are early steps of the metastatic process, pivotal to enabling the spread of cancer to distant tissues. Circulating tumor cells (CTCs) represent a highly selected subpopulation of cancer cells that tamed these critical steps, and a better understanding of their biology and driving molecular principles may facilitate the development of novel tools to prevent metastasis. Here, we describe key research advances in this field, aiming at describing early metastasis-related processes such as collective invasion, shedding, and survival of CTCs in the bloodstream, paying particular attention to microenvironmental factors like hypoxia and mechanical stress, considered as important influencers of the metastatic journey.
Collapse
Affiliation(s)
- Ece Su Ildiz
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Werner J Kovacs
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland.
| |
Collapse
|
24
|
Zhou M, Li K, Luo KQ. Shear Stress Drives the Cleavage Activation of Protease-Activated Receptor 2 by PRSS3/Mesotrypsin to Promote Invasion and Metastasis of Circulating Lung Cancer Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301059. [PMID: 37395651 PMCID: PMC10477893 DOI: 10.1002/advs.202301059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/04/2023] [Indexed: 07/04/2023]
Abstract
When circulating tumor cells (CTCs) travel in circulation, they can be killed by detachment-induced anoikis and fluidic shear stress (SS)-mediated apoptosis. Circulatory treatment, which can make CTCs detached but also generate SS, can increase metastasis of cancer cells. To identify SS-specific mechanosensors without detachment impacts, a microfluidic circulatory system is used to generate arteriosus SS and compare transcriptome profiles of circulating lung cancer cells with suspended cells. Half of the cancer cells can survive SS damage and show higher invasion ability. Mesotrypsin (PRSS3), protease-activated receptor 2 (PAR2), and the subunit of activating protein 1, Fos-related antigen 1 (FOSL1), are upregulated by SS, and their high expression is responsible for promoting invasion and metastasis. SS triggers PRSS3 to cleave the N-terminal inhibitory domain of PAR2 within 2 h. As a G protein-coupled receptor, PAR2 further activates the Gαi protein to turn on the Src-ERK/p38/JNK-FRA1/cJUN axis to promote the expression of epithelial-mesenchymal transition markers, and also PRSS3, which facilitates metastasis. Enriched PRSS3, PAR2, and FOSL1 in human tumor samples and their correlations with worse outcomes reveal their clinical significance. PAR2 may serve as an SS-specific mechanosensor cleavable by PRSS3 in circulation, which provides new insights for targeting metastasis-initiating CTCs.
Collapse
Affiliation(s)
- Muya Zhou
- Department of Biomedical Sciences, Faculty of Health SciencesUniversity of MacauTaipaMacao SAR999078China
| | - Koukou Li
- Department of Biomedical Sciences, Faculty of Health SciencesUniversity of MacauTaipaMacao SAR999078China
| | - Kathy Qian Luo
- Department of Biomedical Sciences, Faculty of Health SciencesUniversity of MacauTaipaMacao SAR999078China
- Ministry of Education Frontiers Science Center for Precision OncologyUniversity of MacauTaipaMacao SAR999078China
| |
Collapse
|
25
|
Kaya U, Gopireddy S, Urbanetz N, Kreitmayer D, Gutheil E, Nopens I, Verwaeren J. Quantifying the hydrodynamic stress for bioprocesses. Biotechnol Prog 2023; 39:e3367. [PMID: 37293967 DOI: 10.1002/btpr.3367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023]
Abstract
Hydrodynamic stress is an influential physical parameter for various bioprocesses, affecting the performance and viability of the living organisms. However, different approaches are in use in various computational and experimental studies to calculate this parameter (including its normal and shear subcomponents) from velocity fields without a consensus on which one is the most representative of its effect on living cells. In this letter, we investigate these different methods with clear definitions and provide our suggested approach which relies on the principal stress values providing a maximal distinction between the shear and normal components. Furthermore, a numerical comparison is presented using the computational fluid dynamics simulation of a stirred and sparged bioreactor. It is demonstrated that for this specific bioreactor, some of these methods exhibit quite similar patterns throughout the bioreactor-therefore can be considered equivalent-whereas some of them differ significantly.
Collapse
Affiliation(s)
- Umut Kaya
- Supply Chain Operations, Pharmaceutical Development, Daiichi Sankyo Europe GmbH, Pfaffenhofen, Germany
- Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| | - Srikanth Gopireddy
- Supply Chain Operations, Pharmaceutical Development, Daiichi Sankyo Europe GmbH, Pfaffenhofen, Germany
- Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| | - Nora Urbanetz
- Supply Chain Operations, Pharmaceutical Development, Daiichi Sankyo Europe GmbH, Pfaffenhofen, Germany
| | - Diana Kreitmayer
- Supply Chain Operations, Pharmaceutical Development, Daiichi Sankyo Europe GmbH, Pfaffenhofen, Germany
| | - Eva Gutheil
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | - Ingmar Nopens
- Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| | - Jan Verwaeren
- Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| |
Collapse
|
26
|
Wang W, Hayes PR, Ren X, Taylor RE. Synthetic Cell Armor Made of DNA Origami. NANO LETTERS 2023; 23:7076-7085. [PMID: 37463308 PMCID: PMC10416349 DOI: 10.1021/acs.nanolett.3c01878] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/11/2023] [Indexed: 07/20/2023]
Abstract
The bioengineering applications of cells, such as cell printing and multicellular assembly, are directly limited by cell damage and death due to a harsh environment. Improved cellular robustness thus motivates investigations into cell encapsulation, which provides essential protection. Here we target the cell-surface glycocalyx and cross-link two layers of DNA nanorods on the cellular plasma membrane to form a modular and programmable nanoshell. We show that the DNA origami nanoshell modulates the biophysical properties of cell membranes by enhancing the membrane stiffness and lowering the lipid fluidity. The nanoshell also serves as armor to protect cells and improve their viability against mechanical stress from osmotic imbalance, centrifugal forces, and fluid shear stress. Moreover, it enables mediated cell-cell interactions for effective and robust multicellular assembly. Our results demonstrate the potential of the nanoshell, not only as a cellular protection strategy but also as a platform for cell and cell membrane manipulation.
Collapse
Affiliation(s)
- Weitao Wang
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Peter R. Hayes
- Department
of Chemical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Xi Ren
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Rebecca E. Taylor
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
27
|
Sugier HR, Bellebon L, Aider JL, Larghero J, Peltzer J, Martinaud C. Feasibility of an acoustophoresis-based system for a high-throughput cell washing: application to bioproduction. Cytotherapy 2023; 25:891-899. [PMID: 37269272 DOI: 10.1016/j.jcyt.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/12/2023] [Accepted: 05/08/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND AIMS These last decades have seen the emergence and development of cell-based therapies, notably those based on mesenchymal stromal cells (MSCs). The advancement of these promising treatments requires increasing the throughput of processed cell for industrialization in order to reduce production costs. Among the various bioproduction challenges, downstream processing, including medium exchange, cell washing, cell harvesting and volume reduction, remains a critical step for which improvements are needed. Typically, these processes are performed by centrifugation. However, this approach limits the automation, especially in small batch productions where it is performed manually in open system. METHODS An acoustophoresis-based system was developed for cell washing. The cells were transferred from one stream to another via the acoustic forces and were collected in a different medium. The optimal flow rates of the different streams were assessed using red blood cells suspended in an albumin solution. Finally, the impact of acoustic washing on adipose tissue-derived MSCs (AD-MSCs) transcriptome was investigated by RNA-sequencing. RESULTS With a single passage through the acoustic device at input flow rate of 45 mL/h, the albumin removal was up to 90% while recovering 99% of RBCs. To further increase the protein removal, a loop washing in two steps was performed and has allowed an albumin removal ≥99% and a red blood cell/AD-MSCs recovery of 99%. After loop washing of AD-MSCs, only two genes, HES4 and MIR-3648-1, were differently expressed compared with the input. CONCLUSIONS In this study, we developed a continuous cell-washing system based on acoustophoresis. The process allows a theoretically high cell throughput while inducing little gene expression changes. These results indicate that cell washing based on acoustophoresis is a relevant and promising solution for numerous applications in cell manufacturing.
Collapse
Affiliation(s)
- Hugo R Sugier
- Aenitis Technologies, Paris, France; Institut André Lwoff, INSERM UMR-MD 1197, Villejuif, France.
| | - Ludovic Bellebon
- Laboratoire PMMH, UMR7636 CNRS, ESPCI Paris - PSL, Paris Sciences Lettres, Sorbonne Université, Paris, France
| | - Jean-Luc Aider
- Laboratoire PMMH, UMR7636 CNRS, ESPCI Paris - PSL, Paris Sciences Lettres, Sorbonne Université, Paris, France
| | - Jérôme Larghero
- Université de Paris, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France; Unité de Thérapie Cellulaire, INSERM U976, Centre d'investigation clinique de Biothérapies CBT501, Paris, France
| | | | | |
Collapse
|
28
|
Kurma K, Alix-Panabières C. Mechanobiology and survival strategies of circulating tumor cells: a process towards the invasive and metastatic phenotype. Front Cell Dev Biol 2023; 11:1188499. [PMID: 37215087 PMCID: PMC10196185 DOI: 10.3389/fcell.2023.1188499] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Metastatic progression is the deadliest feature of cancer. Cancer cell growth, invasion, intravasation, circulation, arrest/adhesion and extravasation require specific mechanical properties to allow cell survival and the completion of the metastatic cascade. Circulating tumor cells (CTCs) come into contact with the capillary bed during extravasation/intravasation at the beginning of the metastatic cascade. However, CTC mechanobiology and survival strategies in the bloodstream, and specifically in the microcirculation, are not well known. A fraction of CTCs can extravasate and colonize distant areas despite the biomechanical constriction forces that are exerted by the microcirculation and that strongly decrease tumor cell survival. Furthermore, accumulating evidence shows that several CTC adaptations, via molecular factors and interactions with blood components (e.g., immune cells and platelets inside capillaries), may promote metastasis formation. To better understand CTC journey in the microcirculation as part of the metastatic cascade, we reviewed how CTC mechanobiology and interaction with other cell types in the bloodstream help them to survive the harsh conditions in the circulatory system and to metastasize in distant organs.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (E LBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (E LBS), Hamburg, Germany
| |
Collapse
|
29
|
Lee JE, Lee P, Yoon YC, Han BS, Ko S, Park MS, Lee YJ, Kim SE, Cho YJ, Lim JH, Ryu JK, Shim S, Kim DK, Jung KH, Hong SS. Vactosertib, TGF-β receptor I inhibitor, augments the sensitization of the anti-cancer activity of gemcitabine in pancreatic cancer. Biomed Pharmacother 2023; 162:114716. [PMID: 37086509 DOI: 10.1016/j.biopha.2023.114716] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 04/24/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits a pronounced extracellular matrix (ECM)-rich response, which is produced by an excessive amount of transforming growth factor β (TGF-β), resulting in tumor progression and metastasis. In addition, TGF-β signaling contributes to rapidly acquired resistance and incomplete response to gemcitabine. Recently, selective inhibitors of the TGF-β signaling pathway have shown promise in PDAC treatment, particularly as an option for augmenting responses to chemotherapy. Here, we investigated the synergistic anticancer effects of a small-molecule TGF-β receptor I kinase inhibitor (vactosertib/EW-7197) in the presence of gemcitabine, and its mechanism of action in pancreatic cancer. Vactosertib sensitized pancreatic cancer cells to gemcitabine by synergistically inhibiting their viability. Importantly, the combination of vactosertib and gemcitabine significantly attenuated the expression of major ECM components, including collagens, fibronectin, and α-SMA, in pancreatic cancer compared with gemcitabine alone. This resulted in potent induction of mitochondrial-mediated apoptosis, gemcitabine-mediated cytotoxicity, and inhibition of tumor ECM by vactosertib. Additionally, the combination decreased metastasis through inhibition of migration and invasion, and exhibited synergistic anti-cancer activity by inhibiting the TGF-β/Smad2 pathway in pancreatic cancer cells. Furthermore, co-treatment significantly suppressed tumor growth in orthotopic models. Therefore, our findings demonstrate that vactosertib synergistically increased the antitumor activity of gemcitabine via inhibition of ECM component production by inhibiting the TGF-β/Smad2 signaling pathway. This suggests that the combination of vactosertib and gemcitabine may be a potential treatment option for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Ji Eun Lee
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Pureunchowon Lee
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Young-Chan Yoon
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Beom Seok Han
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Soyeon Ko
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Min Seok Park
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Yun Ji Lee
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Sang Eun Kim
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Ye Jin Cho
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Joo Han Lim
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Ji-Kan Ryu
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea
| | - Soyeon Shim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Dae-Kee Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Kyung Hee Jung
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea.
| | - Soon-Sun Hong
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, Republic of Korea.
| |
Collapse
|
30
|
Mechanotransduction in tumor dynamics modeling. Phys Life Rev 2023; 44:279-301. [PMID: 36841159 DOI: 10.1016/j.plrev.2023.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Mechanotherapy is a groundbreaking approach to impact carcinogenesis. Cells sense and respond to mechanical stimuli, translating them into biochemical signals in a process known as mechanotransduction. The impact of stress on tumor growth has been studied in the last three decades, and many papers highlight the role of mechanics as a critical self-inducer of tumor fate at the in vitro and in vivo biological levels. Meanwhile, mathematical models attempt to determine laws to reproduce tumor dynamics. This review discusses biological mechanotransduction mechanisms and mathematical-biomechanical models together. The aim is to provide a common framework for the different approaches that have emerged in the literature from the perspective of tumor avascularity and to provide insight into emerging mechanotherapies that have attracted interest in recent years.
Collapse
|
31
|
Kieda J, Appak-Baskoy S, Jeyhani M, Navi M, Chan KWY, Tsai SSH. Microfluidically-generated Encapsulated Spheroids (μ-GELS): An All-Aqueous Droplet Microfluidics Platform for Multicellular Spheroids Generation. ACS Biomater Sci Eng 2023; 9:1043-1052. [PMID: 36626575 DOI: 10.1021/acsbiomaterials.2c00963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Spheroids are three-dimensional clusters of cells that serve as in vitro tumor models to recapitulate in vivo morphology. A limitation of many existing on-chip platforms for spheroid formation is the use of cytotoxic organic solvents as the continuous phase in droplet generation processes. All-aqueous methods do not contain cytotoxic organic solvents but have so far been unable to achieve complete hydrogel gelation on chip. Here, we describe an enhanced droplet microfluidic platform that achieves on-chip gelation of all-aqueous hydrogel multicellular spheroids (MCSs). Specifically, we generate dextran-alginate droplets containing MCF-7 breast cancer cells, surrounded by polyethylene glycol, at a flow-focusing junction. Droplets then travel to a second flow-focusing junction where they interact with calcium chloride and gel on chip to form hydrogel MCSs. On-chip gelation of the MCSs is possible here because of an embedded capillary at the second junction that delays the droplet gelation, which prevents channel clogging problems that would otherwise exist. In drug-free experiments, we demonstrate that MCSs remain viable for 6 days. We also confirm the applicability of this system for cancer drug testing by observing that dose-dependent cell death is achievable using doxorubicin.
Collapse
Affiliation(s)
- Jennifer Kieda
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Sila Appak-Baskoy
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Chemistry and Biology, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| | - Morteza Jeyhani
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| | - Maryam Navi
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Katherine W Y Chan
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Scott S H Tsai
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| |
Collapse
|
32
|
Layachi M, Treizebré A, Hay L, Gilbert D, Pesez J, D’Acremont Q, Braeckmans K, Thommen Q, Courtade E. Novel opto-fluidic drug delivery system for efficient cellular transfection. J Nanobiotechnology 2023; 21:43. [PMID: 36747263 PMCID: PMC9901003 DOI: 10.1186/s12951-023-01797-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Intracellular drug delivery is at the heart of many diagnosis procedures and a key step in gene therapy. Research has been conducted to bypass cell barriers for controlled intracellular drug release and made consistent progress. However, state-of-the-art techniques based on non-viral carriers or physical methods suffer several drawbacks, including limited delivery yield, low throughput or low viability, which are key parameters in therapeutics, diagnostics and drug delivery. Nevertheless, gold nanoparticle (AuNP) mediated photoporation has stood out as a promising approach to permeabilize cell membranes through laser induced Vapour NanoBubble (VNB) generation, allowing the influx of external cargo molecules into cells. However, its use as a transfection technology for the genetic manipulation of therapeutic cells is hindered by the presence of non-degradable gold nanoparticles. Here, we report a new optofluidic method bringing gold nanoparticles in close proximity to cells for photoporation, while avoiding direct contact with cells by taking advantage of hydrodynamic focusing in a multi-flow device. Cells were successfully photoporated with [Formula: see text] efficiency with no significant reduction in cell viability at a throughput ranging from [Formula: see text] to [Formula: see text]. This optofluidic approach provides prospects of translating photoporation from an R &D setting to clinical use for producing genetically engineered therapeutic cells.
Collapse
Affiliation(s)
- Majid Layachi
- grid.464109.e0000 0004 0638 7509Laboratoire Physique des Lasers, Atomes et Molécules - UMR 8523, Université de Lille, 59655 Villeneuve d’Ascq, France ,grid.464109.e0000 0004 0638 7509Institut d’Électronique, de
Microélectronique et de Nanotechnologie - UMR CNRS 8520, Université de Lille, 59655 Villeneuve d’Ascq, France ,grid.121334.60000 0001 2097 0141Present Address: Laboratoire Charles Coulomb - UMR 5221, Université de Montpellier, Montpellier, France
| | - Anthony Treizebré
- grid.464109.e0000 0004 0638 7509Laboratoire Physique des Lasers, Atomes et Molécules - UMR 8523, Université de Lille, 59655 Villeneuve d’Ascq, France ,grid.464109.e0000 0004 0638 7509Institut d’Électronique, de
Microélectronique et de Nanotechnologie - UMR CNRS 8520, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Laurent Hay
- grid.464109.e0000 0004 0638 7509Laboratoire Physique des Lasers, Atomes et Molécules - UMR 8523, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - David Gilbert
- grid.464109.e0000 0004 0638 7509Laboratoire Physique des Lasers, Atomes et Molécules - UMR 8523, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Jean Pesez
- grid.464109.e0000 0004 0638 7509Laboratoire Physique des Lasers, Atomes et Molécules - UMR 8523, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Quentin D’Acremont
- grid.464109.e0000 0004 0638 7509Laboratoire Physique des Lasers, Atomes et Molécules - UMR 8523, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Kevin Braeckmans
- grid.5342.00000 0001 2069 7798Laboratory for General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium
| | - Quentin Thommen
- grid.503422.20000 0001 2242 6780CANTHER - Cancer
Heterogeneity Plasticity and Resistance to Therapies - UMR9020-UMR1277, Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, 59000 Lille, France
| | - Emmanuel Courtade
- Laboratoire Physique des Lasers, Atomes et Molécules - UMR 8523, Université de Lille, 59655, Villeneuve d'Ascq, France.
| |
Collapse
|
33
|
Mazzaglia C, Sheng Y, Rodrigues LN, Lei IM, Shields JD, Huang YYS. Deployable extrusion bioprinting of compartmental tumoroids with cancer associated fibroblasts for immune cell interactions. Biofabrication 2023; 15:025005. [PMID: 36626838 DOI: 10.1088/1758-5090/acb1db] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/10/2023] [Indexed: 01/11/2023]
Abstract
Realizing the translational impacts of three-dimensional (3D) bioprinting for cancer research necessitates innovation in bioprinting workflows which integrate affordability, user-friendliness, and biological relevance. Herein, we demonstrate 'BioArm', a simple, yet highly effective extrusion bioprinting platform, which can be folded into a carry-on pack, and rapidly deployed between bio-facilities. BioArm enabled the reconstruction of compartmental tumoroids with cancer-associated fibroblasts (CAFs), forming the shell of each tumoroid. The 3D printed core-shell tumoroids showedde novosynthesized extracellular matrices, and enhanced cellular proliferation compared to the tumour alone 3D printed spheroid culture. Further, thein vivophenotypes of CAFs normally lost after conventional 2D co-culture re-emerged in the bioprinted model. Embedding the 3D printed tumoroids in an immune cell-laden collagen matrix permitted tracking of the interaction between immune cells and tumoroids, and subsequent simulated immunotherapy treatments. Our deployable extrusion bioprinting workflow could significantly widen the accessibility of 3D bioprinting for replicating multi-compartmental architectures of tumour microenvironment, and for developing strategies in cancer drug testing in the future.
Collapse
Affiliation(s)
| | - Yaqi Sheng
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | | | - Iek Man Lei
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Jacqueline D Shields
- MRC Cancer Unit, University of Cambridge, Cambridge, United Kingdom
- Comprehensive Cancer Centre, King's College London, Great Maze Pond, London, United Kingdom
| | - Yan Yan Shery Huang
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
34
|
Li X, Hu L, Tan C, Wang X, Ran Q, Chen L, Li Z. Platelet-promoting drug delivery efficiency for inhibition of tumor growth, metastasis, and recurrence. Front Oncol 2022; 12:983874. [PMID: 36276066 PMCID: PMC9582853 DOI: 10.3389/fonc.2022.983874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Nanomedicines are considered one of the promising strategies for anticancer therapy; however, the low targeting efficiency of nanomedicines in vivo is a great obstacle to their clinical applications. Camouflaging nanomedicines with either platelet membrane (PM) or platelet would significantly prolong the retention time of nanomedicines in the bloodstream, enhance the targeting ability of nanomedicines to tumor cells, and reduce the off-target effect of nanomedicines in major organs during the anticancer treatment. In the current review, the advantages of using PM or platelet as smart carriers for delivering nanomedicines to inhibit tumor growth, metastasis, and recurrence were summarized. The opportunities and challenges of this camouflaging strategy for anticancer treatment were also discussed.
Collapse
Affiliation(s)
- Xiaoliang Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Lanyue Hu
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Chengning Tan
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xiaojie Wang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Qian Ran
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Li Chen
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- *Correspondence: Li Chen, ; Zhongjun Li,
| | - Zhongjun Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma, Burn and Combined Injuries, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- *Correspondence: Li Chen, ; Zhongjun Li,
| |
Collapse
|
35
|
Bera K, Kiepas A, Zhang Y, Sun SX, Konstantopoulos K. The interplay between physical cues and mechanosensitive ion channels in cancer metastasis. Front Cell Dev Biol 2022; 10:954099. [PMID: 36158191 PMCID: PMC9490090 DOI: 10.3389/fcell.2022.954099] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Physical cues have emerged as critical influencers of cell function during physiological processes, like development and organogenesis, and throughout pathological abnormalities, including cancer progression and fibrosis. While ion channels have been implicated in maintaining cellular homeostasis, their cell surface localization often places them among the first few molecules to sense external cues. Mechanosensitive ion channels (MICs) are especially important transducers of physical stimuli into biochemical signals. In this review, we describe how physical cues in the tumor microenvironment are sensed by MICs and contribute to cancer metastasis. First, we highlight mechanical perturbations, by both solid and fluid surroundings typically found in the tumor microenvironment and during critical stages of cancer cell dissemination from the primary tumor. Next, we describe how Piezo1/2 and transient receptor potential (TRP) channels respond to these physical cues to regulate cancer cell behavior during different stages of metastasis. We conclude by proposing alternative mechanisms of MIC activation that work in tandem with cytoskeletal components and other ion channels to bestow cells with the capacity to sense, respond and navigate through the surrounding microenvironment. Collectively, this review provides a perspective for devising treatment strategies against cancer by targeting MICs that sense aberrant physical characteristics during metastasis, the most lethal aspect of cancer.
Collapse
Affiliation(s)
- Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| | - Yuqi Zhang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Sean X. Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| |
Collapse
|
36
|
Lv Y, Wei C, Zhao B. Study on the mechanism of low shear stress restoring the viability of damaged breast tumor cells. Tissue Cell 2022; 79:101947. [DOI: 10.1016/j.tice.2022.101947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/09/2022] [Accepted: 09/23/2022] [Indexed: 11/28/2022]
|
37
|
Focal adhesion kinase priming in pancreatic cancer, altering biomechanics to improve chemotherapy. Biochem Soc Trans 2022; 50:1129-1141. [PMID: 35929603 PMCID: PMC9444069 DOI: 10.1042/bst20220162] [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: 05/12/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
The dense desmoplastic and fibrotic stroma is a characteristic feature of pancreatic ductal adenocarcinoma (PDAC), regulating disease progression, metastasis and response to treatment. Reciprocal interactions between the tumour and stroma are mediated by bidirectional integrin-mediated signalling, in particular by Focal Adhesion Kinase (FAK). FAK is often hyperactivated and overexpressed in aggressive cancers, promoting stromal remodelling and inducing tissue stiffness which can accelerate cancer cell proliferation, survival and chemoresistance. Therapeutic targeting of the PDAC stroma is an evolving area of interest for pre-clinical and clinical research, where a subtle reshaping of the stromal architecture prior to chemotherapy may prove promising in the clinical management of disease and overall patient survival. Here, we describe how transient stromal manipulation (or ‘priming’) via short-term FAK inhibition, rather than chronic treatment, can render PDAC cells exquisitely vulnerable to subsequent standard-of-care chemotherapy. We assess how our priming publication fits with the recent literature and describe in this perspective how this could impact future cancer treatment. This highlights the significance of treatment timing and warrants further consideration of anti-fibrotic therapies in the clinical management of PDAC and other fibrotic diseases.
Collapse
|
38
|
Pereira-Veiga T, Schneegans S, Pantel K, Wikman H. Circulating tumor cell-blood cell crosstalk: Biology and clinical relevance. Cell Rep 2022; 40:111298. [PMID: 36044866 DOI: 10.1016/j.celrep.2022.111298] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 08/09/2022] [Indexed: 01/17/2023] Open
Abstract
Circulating tumor cells (CTCs) are the seeds of distant metastasis, and the number of CTCs detected in the blood of cancer patients is associated with a worse prognosis. CTCs face critical challenges for their survival in circulation, such as anoikis, shearing forces, and immune surveillance. Thus, understanding the mechanisms and interactions of CTCs within the blood microenvironment is crucial for better understanding of metastatic progression and the development of novel treatment strategies. CTCs interact with different hematopoietic cells, such as platelets, red blood cells, neutrophils, macrophages, natural killer (NK) cells, lymphocytes, endothelial cells, and cancer-associated fibroblasts, which can affect CTC survival in blood. This interaction may take place either via direct cell-cell contact or through secreted molecules. Here, we review interactions of CTCs with blood cells and discuss the potential clinical relevance of these interactions as biomarkers or as targets for anti-metastatic therapies.
Collapse
Affiliation(s)
- Thais Pereira-Veiga
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Svenja Schneegans
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
39
|
Greenlee JD, Liu K, Lopez-Cavestany M, King MR. Piezo1 Mechano-Activation Is Augmented by Resveratrol and Differs between Colorectal Cancer Cells of Primary and Metastatic Origin. Molecules 2022; 27:5430. [PMID: 36080197 PMCID: PMC9458129 DOI: 10.3390/molecules27175430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer cells must survive aberrant fluid shear stress (FSS) in the circulation to metastasize. Herein, we investigate the role that FSS has on colorectal cancer cell apoptosis, proliferation, membrane damage, calcium influx, and therapeutic sensitization. We tested this using SW480 (primary tumor) and SW620 cells (lymph node metastasis) derived from the same patient. The cells were exposed to either shear pulses, modeling millisecond intervals of high FSS seen in regions of turbulent flow, or sustained shear to model average magnitudes experienced by circulating tumor cells. SW480 cells were significantly more sensitive to FSS-induced death than their metastatic counterparts. Shear pulses caused significant cell membrane damage, while constant shear decreased cell proliferation and increased the expression of CD133. To investigate the role of mechanosensitive ion channels, we treated cells with the Piezo1 agonist Yoda1, which increased intracellular calcium. Pretreatment with resveratrol further increased the calcium influx via the lipid-raft colocalization of Piezo1. However, minimal changes in apoptosis were observed due to calcium saturation, as predicted via a computational model of apoptosis. Furthermore, SW480 cells had increased levels of Piezo1, calcium influx, and TRAIL-mediated apoptosis compared to SW620 cells, highlighting differences in the mechano-activation of metastatic cells, which may be a necessary element for successful dissemination in vivo.
Collapse
Affiliation(s)
| | | | | | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, 2301 Vanderbilt Place, Nashville, TN 37235-1631, USA
| |
Collapse
|
40
|
KrishnaPriya S, Omer S, Banerjee S, Karunagaran D, Suraishkumar GK. An integrated approach to understand fluid shear stress-driven and reactive oxygen species-mediated metastasis of colon adenocarcinoma through mRNA-miRNA-lncRNA-circRNA networks. Mol Genet Genomics 2022; 297:1353-1370. [PMID: 35831469 DOI: 10.1007/s00438-022-01924-z] [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: 02/17/2021] [Accepted: 07/01/2022] [Indexed: 11/28/2022]
Abstract
Development of colon adenocarcinoma (COAD) metastasis involves several mediators including fluid shear stress (FSS), intracellular ROS levels, and non-coding RNAs. In our present study, we identified and investigated the role of regulatory non-coding RNA molecules specifically involved in COAD metastasis and their association with FSS and ROS. Interactions between the mRNAs associated with FSS and ROS, the corresponding microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) in COAD metastasis were used to generate the mRNA-miRNA-lncRNA-circRNA network. Experimental validation of the identified RNA hubs using quantitative real-time PCR demonstrated a direct effect of the FSS on their expression levels in cancer cells. FSS resulted in the downregulation of HMGA1 and RAN, as well as the upregulation of HSP90AA1, PMAIP1 and BIRC5. Application of shear stress also led to downregulation of hsa-miR-26b-5p and hsa-miR-34a-5p levels in HCT116 cells. Further, functional enrichment and survival analysis of the significant miRNAs, as well as the OncoPrint and the survival analyses of the selected mRNAs were performed. Subsequently, their functional role was also corroborated with existing literature. Ten significant miRNA hubs were identified, out of which hsa-miR-17-5p and hsa-miR-20a-5p were found to interact with lncRNA (CCAT2) while hsa-miR-335 was found to interact with four circRNAs. Fifteen significant miRNAs were identified in 10 different modules suggesting their importance in FSS and ROS-mediated COAD metastasis. Finally, 10 miRNAs and 3 mRNAs associated with FSS and/or ROS were identified as significant overall survival markers; 33 mRNAs were also identified as metastasis-free survival markers whereas 15 mRNAs showed > 10% gene alterations in TCGA-COAD data and may serve as promising therapeutic biomarkers in the COAD metastasis.
Collapse
Affiliation(s)
- Siluveru KrishnaPriya
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, 600036, India
| | - Sonal Omer
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, 600036, India
| | - Satarupa Banerjee
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, 600036, India. .,School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India.
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, 600036, India
| | - G K Suraishkumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, 600036, India
| |
Collapse
|
41
|
Das J, Maiti TK. Fluid shear stress influences invasiveness of HeLa cells through the induction of autophagy. Clin Exp Metastasis 2022; 39:495-504. [PMID: 35211829 DOI: 10.1007/s10585-022-10156-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/09/2022] [Indexed: 11/25/2022]
Abstract
Extravasation of metastatic cells from the blood or lymphatic circulation and formation of secondary tumor at a distant site is a key step of cancer metastasis. In this study, we report the role of hemodynamic shear stresses in fostering the release of pro-extravasation factors through the mediation of autophagy in cervical cancer HeLa cells. HeLa cells were exposed to physiological shear stress through the microfluidic approach adapted in our previous study on the role of hemodynamic shear stresses in survival of HeLa cells. Herein, an optimum number of passes through a cylindrical microchannel was chosen such that the viability of cells was unaffected by shear. Shear-exposed cells were then probed for their invasive and migratory potential through in vitro migration and invasion assays. The dependence of cancer cells on mechanically-induced autophagy for extravasation was further assessed through protein expression studies. Our results suggest that shear stress upregulates autophagy, which fosters paxillin turnover thereby leading to enhanced focal adhesion disassembly and in turn enhanced cell migration. Concurrently, shear stress-induced secretion of pro-invasive factors like MMP-2 and IL-6 were found to be autophagy-dependent thereby hinting at autophagy as a potential therapeutic target in metastatic cancer. Proposed model for mechano-autophagic modulation of extravasation.
Collapse
Affiliation(s)
- Joyjyoti Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, India.
| | - Tapas K Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India
| |
Collapse
|
42
|
Xu Z, Li K, Xin Y, Tan K, Yang M, Wang G, Tan Y. Fluid shear stress regulates the survival of circulating tumor cells via nuclear expansion. J Cell Sci 2022; 135:275517. [PMID: 35510498 DOI: 10.1242/jcs.259586] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/27/2022] [Indexed: 11/20/2022] Open
Abstract
Distant metastasis mainly occurs through hematogenous dissemination, where suspended circulating tumor cells (CTCs) experience a considerable level of fluid shear stress. We recently reported that shear flow induced substantial apoptosis of CTCs, although a small subpopulation could still persist. However, how suspended tumor cells survive in shear flow remains poorly understood. This study finds that fluid shear stress eliminates the majority of suspended CTCs and increases nuclear size, whereas it has no effect on the viability of adherent tumor cells and decreases their nuclear size. Shear flow promotes histone acetylation in suspended tumor cells, the inhibition of which using one drug suppresses shear-induced nuclear expansion, suggesting that shear stress might increase nuclear size through histone acetylation. Suppressing histone acetylation-mediated nuclear expansion enhances shear-induced apoptosis of CTCs. These findings suggest that suspended tumor cells respond to shear stress through histone acetylation-mediated nuclear expansion, which protects CTCs from shear-induced destruction. Our study elucidates a unique mechanism underlying the mechanotransduction of suspended CTCs to shear flow, which might hold therapeutic promise for CTC eradication.
Collapse
Affiliation(s)
- Zichen Xu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.,Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong 999077, China.,Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China.,Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.,Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong 999077, China.,Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.,Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong 999077, China.,Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Kai Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.,Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong 999077, China.,Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Mo Yang
- Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.,Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong 999077, China.,Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
43
|
Abstract
During cancer progression, metastatic dissemination accounts for ∼90% of death in patients. Metastasis occurs upon dissemination of circulating tumor cells (CTC) through body fluids, in particular the bloodstream, and several key steps remain elusive. Although the majority of CTCs travel as single cells, they can form clusters either with themselves (homoclusters) or with other circulating cells (heteroclusters) and thereby increase their metastatic potential. In addition, cancer cell mechanics and mechanical cues from the microenvironment are important factors during metastatic progression. Recent progress in intravital imaging technologies, biophysical methods, and microfluidic-based isolation of CTCs allow now to probe mechanics at single cell resolution while shedding light on key steps of the hematogenous metastatic cascade. In this review, we discuss the importance of CTC mechanics and their correlation with metastatic success and how such development could lead to the identification of therapeutically relevant targets.
Collapse
Affiliation(s)
- Marina Peralta
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg 67000, France.,Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France.,Equipe Labellisée Ligue Contre le Cancer
| | - Naël Osmani
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg 67000, France.,Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France.,Equipe Labellisée Ligue Contre le Cancer
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg 67000, France.,Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France.,Equipe Labellisée Ligue Contre le Cancer
| |
Collapse
|
44
|
Circulating tumour cells in the -omics era: how far are we from achieving the 'singularity'? Br J Cancer 2022; 127:173-184. [PMID: 35273384 PMCID: PMC9296521 DOI: 10.1038/s41416-022-01768-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/27/2022] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
Over the past decade, cancer diagnosis has expanded to include liquid biopsies in addition to tissue biopsies. Liquid biopsies can result in earlier and more accurate diagnosis and more effective monitoring of disease progression than tissue biopsies as samples can be collected frequently. Because of these advantages, liquid biopsies are now used extensively in clinical care. Liquid biopsy samples are analysed for circulating tumour cells (CTCs), cell-free DNA, RNA, proteins and exosomes. CTCs originate from the tumour, play crucial roles in metastasis and carry information on tumour heterogeneity. Multiple single-cell omics approaches allow the characterisation of the molecular makeup of CTCs. It has become evident that CTCs are robust biomarkers for predicting therapy response, clinical development of metastasis and disease progression. This review describes CTC biology, molecular heterogeneity within CTCs and the involvement of EMT in CTC dynamics. In addition, we describe the single-cell multi-omics technologies that have provided insights into the molecular features within therapy-resistant and metastasis-prone CTC populations. Functional studies coupled with integrated multi-omics analyses have the potential to identify therapies that can intervene the functions of CTCs.
Collapse
|
45
|
Houthaeve G, De Smedt SC, Braeckmans K, De Vos WH. The cellular response to plasma membrane disruption for nanomaterial delivery. NANO CONVERGENCE 2022; 9:6. [PMID: 35103909 PMCID: PMC8807741 DOI: 10.1186/s40580-022-00298-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
Delivery of nanomaterials into cells is of interest for fundamental cell biological research as well as for therapeutic and diagnostic purposes. One way of doing so is by physically disrupting the plasma membrane (PM). Several methods that exploit electrical, mechanical or optical cues have been conceived to temporarily disrupt the PM for intracellular delivery, with variable effects on cell viability. However, apart from acute cytotoxicity, subtler effects on cell physiology may occur as well. Their nature and timing vary with the severity of the insult and the efficiency of repair, but some may provoke permanent phenotypic alterations. With the growing palette of nanoscale delivery methods and applications, comes a need for an in-depth understanding of this cellular response. In this review, we summarize current knowledge about the chronology of cellular events that take place upon PM injury inflicted by different delivery methods. We also elaborate on their significance for cell homeostasis and cell fate. Based on the crucial nodes that govern cell fitness and functionality, we give directions for fine-tuning nano-delivery conditions.
Collapse
Affiliation(s)
- Gaëlle Houthaeve
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
46
|
Liquid Biopsies: Flowing Biomarkers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:341-368. [DOI: 10.1007/978-3-031-04039-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Measuring the density and viscosity of culture media for optimized computational fluid dynamics analysis of in vitro devices. J Mech Behav Biomed Mater 2021; 126:105024. [PMID: 34911025 DOI: 10.1016/j.jmbbm.2021.105024] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/25/2020] [Accepted: 12/02/2021] [Indexed: 12/22/2022]
Abstract
Culture medium is frequently modelled as water in computational fluid dynamics (CFD) analysis of in vitro culture systems involving flow, such as bioreactors and organ-on-chips. However, culture medium can be expected to have different properties to water due to its higher solute content. Furthermore, cellular activities such as metabolism and secretion of ECM proteins alter the composition of culture medium and therefore its properties during culture. As these properties directly determine the hydromechanical stimuli exerted on cells in vitro, these, along with any changes during culture must be known for CFD modelling accuracy and meaningful interpretation of cellular responses. In this study, the density and dynamic viscosity of DMEM and RPMI-1640 media supplemented with typical concentrations of foetal bovine serum (0, 5, 10 and 20% v/v) were measured to serve as a reference for computational design analysis. Any changes in the properties of medium during culture were also investigated with NCI-H460 and HN6 cell lines. The density and dynamic viscosity of the media increased proportional to the % volume of added foetal bovine serum (FBS). Importantly, the viscosity of 5% FBS-supplemented RPMI-1640 was found to increase significantly after 3 days of culture of NCI-H460 and HN6 cell lines, with distinct differences between magnitude of change for each cell line. Finally, these experimentally-derived values were applied in CFD analysis of a simple microfluidic device, which demonstrated clear differences in maximum wall shear stress and pressure between fluid models. Overall, these results highlight the importance of characterizing model-specific properties for CFD design analysis of cell culture systems.
Collapse
|
48
|
Yan HH, Jung KH, Lee JE, Son MK, Fang Z, Park JH, Kim SJ, Kim JY, Lim JH, Hong SS. ANGPTL4 accelerates KRAS G12D-Induced acinar to ductal metaplasia and pancreatic carcinogenesis. Cancer Lett 2021; 519:185-198. [PMID: 34311032 DOI: 10.1016/j.canlet.2021.07.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022]
Abstract
Oncogenic KRASG12D induces neoplastic transformation of pancreatic acinar cells through acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN), and drives pancreatic ductal adenocarcinoma (PDAC). Angiopoietin-like 4 (ANGPTL4) is known to be involved in the regulation of cancer growth and metastasis. However, whether ANGPTL4 affects KRASG12D-mediated ADM and early PDAC intervention remains unknown. In the current study, we investigated the role of ANGPTL4 in KRASG12D-induced ADM, PanIN formation, and PDAC maintenance. We found that ANGPTL4 was highly expressed in human and mouse ADM lesions and contributed to the promotion of KRASG12D-driven ADM in mice. Consistently, ANGPTL4 rapidly induced ADM in three-dimensional culture of acinar cells with KRAS mutation and formed ductal cysts that silenced acinar genes and activated ductal genes, which are characteristic of in vivo ADM/PanIN lesions. We also found that periostin works as a downstream regulator of ANGPTL4-mediated ADM/PDAC. Genetic ablation of periostin diminished the ADM/PanIN phenotype induced by ANGPTL4. A high correlation between ANGPTL4 and periostin was confirmed in human samples. These results demonstrate that ANGPTL4 is critical for ADM/PanIN initiation and PDAC progression through the regulation of periostin. Thus, the ANGPTL4/periostin axis is considered a potential target for ADM-derived PDAC.
Collapse
Affiliation(s)
- Hong Hua Yan
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Kyung Hee Jung
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Ji Eun Lee
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Mi Kwon Son
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Zhenghuan Fang
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Jung Hee Park
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Soo Jung Kim
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Ju Young Kim
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Ju Han Lim
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Medicine, College of Medicine, and Program in Biomedical Sciences & Engineering, Inha University, 366, Seohae-daero, Jung-gu, Incheon, 22332, Republic of Korea.
| |
Collapse
|
49
|
Dombroski JA, Hope JM, Sarna NS, King MR. Channeling the Force: Piezo1 Mechanotransduction in Cancer Metastasis. Cells 2021; 10:2815. [PMID: 34831037 PMCID: PMC8616475 DOI: 10.3390/cells10112815] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/25/2022] Open
Abstract
Cancer metastasis is one of the leading causes of death worldwide, motivating research into identifying new methods of preventing cancer metastasis. Recently there has been increasing interest in understanding how cancer cells transduce mechanical forces into biochemical signals, as metastasis is a process that consists of a wide range of physical forces. For instance, the circulatory system through which disseminating cancer cells must transit is an environment characterized by variable fluid shear stress due to blood flow. Cancer cells and other cells can transduce physical stimuli into biochemical responses using the mechanosensitive ion channel Piezo1, which is activated by membrane deformations that occur when cells are exposed to physical forces. When active, Piezo1 opens, allowing for calcium flux into the cell. Calcium, as a ubiquitous second-messenger cation, is associated with many signaling pathways involved in cancer metastasis, such as angiogenesis, cell migration, intravasation, and proliferation. In this review, we discuss the roles of Piezo1 in each stage of cancer metastasis in addition to its roles in immune cell activation and cancer cell death.
Collapse
Affiliation(s)
| | | | | | - Michael R. King
- King Lab, Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA; (J.A.D.); (J.M.H.); (N.S.S.)
| |
Collapse
|
50
|
Al-Ani A, Toms D, Sunba S, Giles K, Touahri Y, Schuurmans C, Ungrin M. Scaffold-Free Retinal Pigment Epithelium Microtissues Exhibit Increased Release of PEDF. Int J Mol Sci 2021; 22:11317. [PMID: 34768747 PMCID: PMC8583603 DOI: 10.3390/ijms222111317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 12/26/2022] Open
Abstract
The retinal pigmented epithelium (RPE) plays a critical role in photoreceptor survival and function. RPE deficits are implicated in a wide range of diseases that result in vision loss, including age-related macular degeneration (AMD) and Stargardt disease, affecting millions worldwide. Subretinal delivery of RPE cells is considered a promising avenue for treatment, and encouraging results from animal trials have supported recent progression into the clinic. However, the limited survival and engraftment of transplanted RPE cells delivered as a suspension continues to be a major challenge. While RPE delivery as epithelial sheets exhibits improved outcomes, this comes at the price of increased complexity at both the production and transplant stages. In order to combine the benefits of both approaches, we have developed size-controlled, scaffold-free RPE microtissues (RPE-µTs) that are suitable for scalable production and delivery via injection. RPE-µTs retain key RPE molecular markers, and interestingly, in comparison to conventional monolayer cultures, they show significant increases in the transcription and secretion of pigment-epithelium-derived factor (PEDF), which is a key trophic factor known to enhance the survival and function of photoreceptors. Furthermore, these microtissues readily spread in vitro on a substrate analogous to Bruch's membrane, suggesting that RPE-µTs may collapse into a sheet upon transplantation. We anticipate that this approach may provide an alternative cell delivery system to improve the survival and integration of RPE transplants, while also retaining the benefits of low complexity in production and delivery.
Collapse
Affiliation(s)
- Abdullah Al-Ani
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (A.A.-A.); (S.S.); (K.G.)
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB T2N 1N4, Canada
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Derek Toms
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (A.A.-A.); (S.S.); (K.G.)
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Saud Sunba
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (A.A.-A.); (S.S.); (K.G.)
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Kayla Giles
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (A.A.-A.); (S.S.); (K.G.)
| | - Yacine Touahri
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (Y.T.); (C.S.)
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Carol Schuurmans
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (Y.T.); (C.S.)
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mark Ungrin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (A.A.-A.); (S.S.); (K.G.)
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|