1
|
Deng W, Yang X, Yu J, Omari-Siaw E, Xu X. Recent advances of physiochemical cues on surfaces for directing cell fates. Colloids Surf B Biointerfaces 2025; 250:114550. [PMID: 39929022 DOI: 10.1016/j.colsurfb.2025.114550] [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: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/12/2025]
Abstract
Surface modification plays an essential role in dictating cell behavior and fate, as it creates a microenvironment that profoundly influences cell attachment, migration, proliferation, and differentiation. This review aims to the intricate interplay of culture surface properties, including topography, stiffness, charge, and chemical modifications, demonstrating their profound impact on cell destiny. We explore the nuanced responses of cells to varying surface topographies, from nano- to microscale features, elucidating the influence of geometric patterns and roughness. We also investigate the impact of substrate stiffness, highlighting the way cells perceive and respond to mechanical cues mimicking their native environments. The role of surface charge is examined, revealing how electrostatic interactions influence cell adhesion, signaling, and cell fate decisions. Finally, we delve into the diverse effects of chemical modifications, including the presentation of bioactive molecules, growth factors, and extracellular matrix (ECM) components, demonstrating their ability to guide cell behavior and stimulate specific cellular responses. This review offers comprehensive insights into the important role of surface properties in shaping cell fate, offering promising avenues for developing sophisticated cell culture platforms for applications in drug discovery, regenerative medicine, and fundamental research.
Collapse
Affiliation(s)
- Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Xiufen Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutical Science, Kumasi Technical University, PO Box 854, Kumasi, Ashanti, Ghana
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| |
Collapse
|
2
|
Li Y, Yang Y, Wang X, Li L, Zhou M. Extracellular osmolarity regulates osteoblast migration through the TRPV4-Rho/ROCK signaling. Commun Biol 2025; 8:515. [PMID: 40155775 PMCID: PMC11953337 DOI: 10.1038/s42003-025-07946-8] [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: 09/26/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
For precise bone formation, osteoblasts need to accurately migrate to specific sites guided by various biochemical and mechanical cues. During this migration, fluctuations in extracellular osmolarity may arise from shifts in the surrounding fluid environment. However, as a main regulator of cell morphology and function, whether the extracellular osmolarity change may affect osteoblast migration remains unclear. Here, we provide evidence showing that changes in extracellular osmolarity significantly impact osteoblast migration, with a hypotonic environment enhancing it while a hypertonic environment inhibiting it. Further, our findings reveal that a hypotonic treatment increases intracellular pressure, activating the Transient Receptor Potential Vanilloid 4 (TRPV4) channel. This activation of TRPV4 modulates stress fibers, focal adhesions (FAs), and cell polarity through the Rho/ROCK signaling pathway, ultimately impacting osteoblast migration. Our findings provide valuable insights into the significant influence of extracellular osmolarity on osteoblast migration, which has potential implications for enhancing our understanding of bone remodeling.
Collapse
Affiliation(s)
- Yijie Li
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Yanyan Yang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Xiaohuan Wang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China.
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Mouwang Zhou
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China.
| |
Collapse
|
3
|
Yuan Z, Lin B, Wang C, Yan Z, Yang F, Su H. Collagen remodeling-mediated signaling pathways and their impact on tumor therapy. J Biol Chem 2025; 301:108330. [PMID: 39984051 PMCID: PMC11957794 DOI: 10.1016/j.jbc.2025.108330] [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: 08/27/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
In addition to their traditional roles in maintaining tissue morphology and organ development, emerging evidence suggests that collagen (COL) remodeling-referring to dynamic changes in the quantity, stiffness, arrangements, cleavage states, and homo-/hetero-trimerization of COLs-serves as a key signaling mechanism that governs tumor growth and metastasis. COL receptors act as switches, linking various forms of COL remodeling to different cell types during cancer progression, including cancer cells, immune cells, and cancer-associated fibroblasts. In this review, we summarize recent findings on the signaling pathways mediated by COL arrangement, cleavage, and trimerization states (both homo- and hetero-), as well as the roles of the primary COL receptors-integrin, DDR1/2, LAIR-1/2, MRC2, and GPVI-in cancer progression. We also discuss the latest therapeutic strategies targeting COL fragments, cancer-associated fibroblasts, and COL receptors, including integrins, DDR1/2, and LAIR1/2. Understanding the pathways modulated by COL remodeling and COL receptors in various pathological contexts will pave the way for developing new precision therapies.
Collapse
Affiliation(s)
- Zihang Yuan
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, MOE Innovation Center for Basic Research in Tumor Immunotherapy, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Bo Lin
- Liver Cancer Institute, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chunlan Wang
- Liver Cancer Institute, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhaoyue Yan
- The Department of Stomatology, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong, China
| | - Fei Yang
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, MOE Innovation Center for Basic Research in Tumor Immunotherapy, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Hua Su
- Liver Cancer Institute, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Baster Z, Russell L, Rajfur Z. A Review of Talin- and Integrin-Dependent Molecular Mechanisms in Cancer Invasion and Metastasis. Int J Mol Sci 2025; 26:1798. [PMID: 40076426 PMCID: PMC11899650 DOI: 10.3390/ijms26051798] [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: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer is the second most common cause of death in the world, representing one of the main economic burdens in health care and research. The effort of research has mainly focused on limiting the growth of a localized tumor, but most recently, there has been more attention focused on restricting the spreading of the cancer via invasion and metastasis. The signaling pathways behind these two processes share many molecules with physiological pathways regulating cell adhesion and migration, and, moreover, adhesion and migration processes themselves underlie tumor potential for invasion. In this work, we reviewed the latest literature about cancer development and invasion and their regulation by cell migration- and adhesion-related proteins, with a specific focus on talins and integrins. We also summarized the most recent developments and approaches to anti-cancer therapies, concentrating on cell migration-related therapies.
Collapse
Affiliation(s)
- Zbigniew Baster
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Laboratory for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lindsay Russell
- Undergraduate Program, Barnard College of Columbia University, New York, NY 10027, USA;
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348 Kraków, Poland
| |
Collapse
|
5
|
Chastney MR, Kaivola J, Leppänen VM, Ivaska J. The role and regulation of integrins in cell migration and invasion. Nat Rev Mol Cell Biol 2025; 26:147-167. [PMID: 39349749 DOI: 10.1038/s41580-024-00777-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 01/29/2025]
Abstract
Integrin receptors are the main molecular link between cells and the extracellular matrix (ECM) as well as mediating cell-cell interactions. Integrin-ECM binding triggers the formation of heterogeneous multi-protein assemblies termed integrin adhesion complexes (IACs) that enable integrins to transform extracellular cues into intracellular signals that affect many cellular processes, especially cell motility. Cell migration is essential for diverse physiological and pathological processes and is dysregulated in cancer to favour cell invasion and metastasis. Here, we discuss recent findings on the role of integrins in cell migration with a focus on cancer cell dissemination. We review how integrins regulate the spatial distribution and dynamics of different IACs, covering classical focal adhesions, emerging adhesion types and adhesion regulation. We discuss the diverse roles integrins have during cancer progression from cell migration across varied ECM landscapes to breaching barriers such as the basement membrane, and eventual colonization of distant organs.
Collapse
Affiliation(s)
- Megan R Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jasmin Kaivola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Veli-Matti Leppänen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Life Technologies, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Western Finnish Cancer Center (FICAN West), University of Turku, Turku, Finland.
- Foundation for the Finnish Cancer Institute, Helsinki, Finland.
| |
Collapse
|
6
|
Wang Y, Wang Y, Zhu Y, Yu P, Zhou F, Zhang A, Gu Y, Jin R, Li J, Zheng F, Yu A, Ye D, Xu Y, Liu YJ, Saw TB, Hu G, Lim CT, Yu FX. Angiomotin cleavage promotes leader formation and collective cell migration. Dev Cell 2025; 60:101-118.e7. [PMID: 39389053 DOI: 10.1016/j.devcel.2024.09.013] [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: 07/31/2023] [Revised: 05/22/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
Collective cell migration (CCM) is involved in multiple biological processes, including embryonic morphogenesis, angiogenesis, and cancer invasion. However, the molecular mechanisms underlying CCM, especially leader cell formation, are poorly understood. Here, we show that a signaling pathway regulating angiomotin (AMOT) cleavage plays a role in CCM, using mammalian epithelial cells and mouse models. In a confluent epithelial monolayer, full-length AMOT localizes at cell-cell junctions and limits cell motility. After cleavage, the C-terminal fragment of AMOT (AMOT-CT) translocates to the cell-matrix interface to promote the maturation of focal adhesions (FAs), generate traction force, and induce leader cell formation. Meanwhile, decreased full-length AMOT at cell-cell junctions leads to tissue fluidization and coherent migration of cell collectives. Hence, the cleavage of AMOT serves as a molecular switch to generate polarized contraction, promoting leader cell formation and CCM.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yebin Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yuwen Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Pengcheng Yu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Fanhui Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Anlan Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yuan Gu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Ruxin Jin
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jin Li
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Fengyun Zheng
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Aijuan Yu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Dan Ye
- Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yanhui Xu
- Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yan-Jun Liu
- Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Thuan Beng Saw
- Research Center for Industries of the Future and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China; Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore 117599, Singapore
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Yu H, Jafari M, Mujahid A, Garcia CF, Shah J, Sinha R, Huang Y, Shakiba D, Hong Y, Cheraghali D, Pryce JRS, Sandler JA, Elson EL, Sacks JM, Genin GM, Alisafaei F. Expansion limits of meshed split-thickness skin grafts. Acta Biomater 2025; 191:325-335. [PMID: 39581335 DOI: 10.1016/j.actbio.2024.11.038] [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: 06/27/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Split-thickness skin grafts are widely used to treat chronic wounds. Procedure design requires surgeons to predict how much a patch of the patient's own skin expands when it is meshed with rows of slits and stretched over a larger wound area. Accurate prediction of graft expansion remains a challenge, with current models overestimating the actual expansion, leading to suboptimal outcomes. Inspired by the principles of mechanical metamaterials, we developed a model that distinguishes between the kinematic rearrangement of structural elements and their stretching, providing a more accurate prediction of skin graft expansion. Our model was validated against extensive data from skin graft surgeries, demonstrating vastly superior predictive capability compared to existing methods. This metamaterial-inspired approach enables informed decision-making for potentially improving healing outcomes. STATEMENT OF SIGNIFICANCE: Accurately predicting the expansion of meshed skin grafts is crucial for minimizing patient trauma and optimizing healing outcomes in reconstructive surgery. However, current quantitative models, which treat grafts as tessellated trusses of rigid bars, fail to accurately estimate graft expansion. We have uncovered the mechanisms underlying skin graft expansion and developed a straightforward method based on these findings. This method, designed for practical use by surgeons, provides accurate predictions of graft expansion, as validated against extensive data from skin graft surgeries.
Collapse
Affiliation(s)
- Haomin Yu
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, USA
| | - Mohammad Jafari
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering, New Jersey Institute of Technology, USA
| | - Aliza Mujahid
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering, New Jersey Institute of Technology, USA
| | - Chelsea F Garcia
- Department of Mechanical Engineering, New Jersey Institute of Technology, USA
| | - Jaisheel Shah
- Department of Mechanical Engineering, New Jersey Institute of Technology, USA
| | - Riya Sinha
- Department of Mechanical Engineering, New Jersey Institute of Technology, USA
| | - Yuxuan Huang
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Biomedical Engineering, Washington University in St. Louis, USA
| | - Delaram Shakiba
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Pathology, Johns Hopkins University, USA
| | - Yuan Hong
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, USA
| | - Danial Cheraghali
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering, New Jersey Institute of Technology, USA
| | - John R S Pryce
- Department of Mechanical Engineering, New Jersey Institute of Technology, USA
| | - Jacob A Sandler
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, USA
| | - Elliot L Elson
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, USA; Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine, USA
| | - Justin M Sacks
- Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, USA
| | - Guy M Genin
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, USA; Department of Biomedical Engineering, Washington University in St. Louis, USA.
| | - Farid Alisafaei
- NSF Science and Technology Center for Engineering Mechanobiology, USA; Department of Mechanical Engineering, New Jersey Institute of Technology, USA.
| |
Collapse
|
8
|
Mousso T, Rice K, Tumenbayar BI, Pham K, Heo Y, Heo SC, Lee K, Lombardo AT, Bae Y. Survivin modulates stiffness-induced vascular smooth muscle cell motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.628062. [PMID: 39713437 PMCID: PMC11661181 DOI: 10.1101/2024.12.11.628062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Arterial stiffness is a key contributor to cardiovascular diseases, including atherosclerosis, restenosis, and coronary artery disease, it has been characterized to be associated with the aberrant migration of vascular smooth muscle cells (VSMCs). However, the underlying molecular mechanisms driving VSMC migration in stiff environments remain incompletely understood. We recently demonstrated that survivin, a member of the inhibitor of apoptosis protein family, is highly expressed in both mouse and human VSMCs cultured on stiff polyacrylamide hydrogels, where it modulates stiffness-mediated cell cycle progression and proliferation. However, its role in stiffness-dependent VSMC migration remains unknown. To assess its impact on migration, we performed time-lapse video microscopy on VSMCs seeded on fibronectin-coated soft and stiff polyacrylamide hydrogels, mimicking the physiological stiffness of normal and diseased arteries, with either survivin inhibition or overexpression. We observed that VSMC motility increased under stiff conditions, while pharmacologic or siRNA-mediated inhibition of survivin reduced stiffness-stimulated migration to rates similar to those observed under soft conditions. Further investigation revealed that cells on stiff hydrogels exhibited greater directional movement and robust lamellipodial protrusion compared to those on soft hydrogels. Interestingly, survivin-inhibited cells on stiff hydrogels showed reduced directional persistence and lamellipodial protrusion compared to control cells. We also examined whether survivin overexpression alone is sufficient to induce cell migration on soft hydrogels, and found that survivin overexpression modestly increased cell motility and partially rescued the lack of directional persistence compared to GFP-expressing control VSMCs on soft hydrogels. In conclusion, our findings demonstrate that survivin plays a key role in regulating stiffness-induced VSMC migration, suggesting that targeting survivin and its signaling pathways could offer therapeutic strategies for addressing arterial stiffness in cardiovascular diseases.
Collapse
Affiliation(s)
- Thomas Mousso
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY 14203, USA
| | - Kalina Rice
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY 14203, USA
| | - Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Khanh Pham
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY 14203, USA
| | - Yuna Heo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY 14203, USA
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Chin Heo
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kwonmoo Lee
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Andrew T Lombardo
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY 14203, USA
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
9
|
Zhang Y, Dong X, Zhang Y, Chen Z, Zhou G, Chen N, Shen W, Yang K, Pei P. Biomaterials to regulate tumor extracellular matrix in immunotherapy. J Control Release 2024; 376:149-166. [PMID: 39389365 DOI: 10.1016/j.jconrel.2024.10.010] [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: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The tumor extracellular matrix (ECM) provides physical support and influences tumor development, metastasis, and the tumor microenvironment, creating barriers to immune drug delivery and cell infiltration. Therefore, modulating or degrading the ECM is of significant importance to enhance the efficacy of tumor immunotherapy. This manuscript initially summarizes the main strategies and mechanisms of biomaterials in modulating various components of the ECM, including collagen, fibronectin, hyaluronic acid, and in remodeling the ECM. Subsequently, it discusses the benefits of biomaterials for immunotherapy following ECM modulation, such as promoting the infiltration of drugs and immune cells, regulating immune cell function, and alleviating the immunosuppressive microenvironment. The manuscript also briefly introduces the application of biomaterials that utilize and mimic the ECM for tumor immunotherapy. Finally, it addresses the current challenges and future directions in this field, providing a comprehensive overview of the potential and innovation in leveraging biomaterials to enhance cancer treatment outcomes. Our work will offer a comprehensive overview of ECM modulation strategies and their application in biomaterials to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Yujie Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xuexue Dong
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zetong Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Ni Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Jiangsu, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province 230022, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| |
Collapse
|
10
|
Cho Y, Doh J. The extracellular matrix in solid tumor immunotherapy. Trends Immunol 2024; 45:705-714. [PMID: 39164157 DOI: 10.1016/j.it.2024.07.009] [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: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024]
Abstract
The extracellular matrix (ECM) of solid tumors impacts the antitumor activities of CD8+ T and natural killer (NK) cells in a variety of ways. Cell motility is restricted by the tumor ECM which creates physical barriers. The tumor ECM directly alter the phenotypes and functions of cytotoxic lymphocytes, and indirectly influences immunological synapse-mediated interactions between cytotoxic lymphocytes and cancer cells. Therefore, strategies to improve solid tumor immunotherapy should be established by considering complex ternary interactions between cytotoxic lymphocytes, cancer cells, and the tumor ECM. Novel bioengineering tools approximating key characteristics of the tumor ECM, such as in vitro reconstituted 3D ECMs and microfluidics are valuable from a fundamental study viewpoint and from a translational perspective, aiming to enable systematic screening approaches.
Collapse
Affiliation(s)
- Yongbum Cho
- Research Institute of Advanced Materials (RIAM), Seoul National University, Seoul, South Korea
| | - Junsang Doh
- Research Institute of Advanced Materials (RIAM), Seoul National University, Seoul, South Korea; Department of Materials Science and Engineering, Institute of Engineering Research, BioMAX, Soft Foundry Institute, Seoul National University, Seoul, South Korea.
| |
Collapse
|
11
|
Yadav S, Joshi R, Majumder A. Concave/Convex Curvature of Anisotropic Grooves Differentially Alters Cellular Morphology, Adhesion, and Proliferation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:17590-17600. [PMID: 39132850 DOI: 10.1021/acs.langmuir.4c01896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Curvature is an integral part of the complex in vivo tissue architecture across various length scales. Therefore, several in vitro models with a patterned curvature in different length scales have been developed to understand the role of this in cellular behavior. At the subcellular scale, wavy patterns have been reported wherein concave and convex grooves are adjacently present. However, the independent effect of continuous subcellular concave and convex shapes has not been reported, mainly owing to the limitations in fabricating such patterns. In this study, we developed continuous concave and convex grooves on polydimethylsiloxane (PDMS) using a Dracaena sanderiana (bamboo) leaf as a template. The first (negative) replica from the abaxial side of the bamboo leaf, which imparted concave grooves on PDMS, was subsequently used as a template to fabricate a positive replica of the leaf, resulting in convex grooves of the same size and arrangement as the concave grooves. We examined the influence of the groove curvature on the morphology of bone marrow-derived human mesenchymal stem cells (BM-hMSCs) and skeletal muscle cells (C2C12). BM-hMSCs and C2C12 cells aligned on both concave and convex grooves as compared to the random orientation on a flat substrate. The significant difference was observed in the morphology of both cells, in terms of area, aspect ratio, number, and length of protrusions on concave and convex patterns. We found that the number of protrusions was also dependent on the ratio of cell to pattern length scale for convex-shaped grooves but independent of length scale for concave-shaped grooves. The proliferation of BM-hMSCs was also found to be different on concave and convex shapes. Therefore, this study shows the importance of (1) convex and concave curvatures of the subcellular length scale in cellular response, (2) dependence on the ratio of cell and curvature length scale, and (3) use of natural templates for overcoming fabrication challenges.
Collapse
Affiliation(s)
- Shital Yadav
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai400076,India
| | - Rohit Joshi
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai400076,India
| | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai400076,India
| |
Collapse
|
12
|
Ding DY, Jiang SY, Zu YX, Yang Y, Gan XJ, Yuan SX, Zhou WP. Collagen in hepatocellular carcinoma: A novel biomarker and therapeutic target. Hepatol Commun 2024; 8:e0489. [PMID: 38967581 PMCID: PMC11227359 DOI: 10.1097/hc9.0000000000000489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024] Open
Abstract
HCC is globally recognized as a major health threat. Despite significant progress in the development of treatment strategies for liver cancer, recurrence, metastasis, and drug resistance remain key factors leading to a poor prognosis for the majority of liver cancer patients. Thus, there is an urgent need to develop effective biomarkers and therapeutic targets for HCC. Collagen, the most abundant and diverse protein in the tumor microenvironment, is highly expressed in various solid tumors and plays a crucial role in the initiation and progression of tumors. Recent studies have shown that abnormal expression of collagen in the tumor microenvironment is closely related to the occurrence, development, invasion, metastasis, drug resistance, and treatment of liver cancer, making it a potential therapeutic target and a possible diagnostic and prognostic biomarker for HCC. This article provides a comprehensive review of the structure, classification, and origin of collagen, as well as its role in the progression and treatment of HCC and its potential clinical value, offering new insights into the diagnosis, treatment, and prognosis assessment of liver cancer.
Collapse
Affiliation(s)
- Dong-yang Ding
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Shu-ya Jiang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Yun-xi Zu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Yuan Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Xiao-jie Gan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Sheng-xian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| | - Wei-ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, P. R. China
| |
Collapse
|
13
|
Wang X, Yang Y, Wang Y, Lu C, Hu X, Kawazoe N, Yang Y, Chen G. Focal adhesion and actin orientation regulated by cellular geometry determine stem cell differentiation via mechanotransduction. Acta Biomater 2024; 182:81-92. [PMID: 38734287 DOI: 10.1016/j.actbio.2024.05.017] [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: 12/31/2023] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Tuning cell adhesion geometry can affect cytoskeleton organization and the distribution of cytoskeleton forces, which play critical roles in controlling cell functions. To elucidate the geometrical relationship with cytoskeleton force distribution, it is necessary to control cell morphology. In this study, a series of dextral vortex micropatterns were prepared to precisely control cell morphology for investigating the influence of the curvature degree of adhesion curves on intracellular force distribution and stem cell differentiation at a sub-cellular level. Peripherial actin filaments of micropatterned cells were assembled along the adhesion curves and showed different orientations, filament thicknesses and densities. Focal adhesion and cytoskeleton force distribution were dependent on the curvature degree. Intracellular force distribution was also regulated by adhesion curves. The cytoskeleton and force distribution affected the osteogenic differentiation of mesenchymal stem cells through a YAP/TAZ-mediated mechanotransduction process. Thus, regulation of cell adhesion curvature, especially at cytoskeletal filament level, is critical for cell function manipulation. STATEMENT OF SIGNIFICANCE: In this study, a series of dextral micro-vortexes were prepared and used for the culture of human mesenchymal stem cells (hMSCs) to precisely control adhesive curvatures (0°, 30°, 60°, and 90°). The single MSCs on the micropatterns had the same size and shape but showed distinct focal adhesion (FA) and cytoskeleton orientations. Cellular nanomechanics were observed to be correlated with the curvature degrees, subsequently influencing nuclear morphological features. As a consequence, the localization of the mechanotransduction sensor and activator-YAP/TAZ was affected, influencing osteogenic differentiation. The results revealed the pivotal role of adhesive curvatures in the manipulation of stem cell differentiation via the machanotransduction process, which has rarely been investigated.
Collapse
Affiliation(s)
- Xinlong Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Yingjun Yang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Yongtao Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Chengyu Lu
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Xiaohong Hu
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan.
| |
Collapse
|
14
|
Ashworth JC, Cox TR. The importance of 3D fibre architecture in cancer and implications for biomaterial model design. Nat Rev Cancer 2024; 24:461-479. [PMID: 38886573 DOI: 10.1038/s41568-024-00704-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2024] [Indexed: 06/20/2024]
Abstract
The need for improved prediction of clinical response is driving the development of cancer models with enhanced physiological relevance. A new concept of 'precision biomaterials' is emerging, encompassing patient-mimetic biomaterial models that seek to accurately detect, treat and model cancer by faithfully recapitulating key microenvironmental characteristics. Despite recent advances allowing tissue-mimetic stiffness and molecular composition to be replicated in vitro, approaches for reproducing the 3D fibre architectures found in tumour extracellular matrix (ECM) remain relatively unexplored. Although the precise influences of patient-specific fibre architecture are unclear, we summarize the known roles of tumour fibre architecture, underlining their implications in cell-matrix interactions and ultimately clinical outcome. We then explore the challenges in reproducing tissue-specific 3D fibre architecture(s) in vitro, highlighting relevant biomaterial fabrication techniques and their benefits and limitations. Finally, we discuss imaging and image analysis techniques (focussing on collagen I-optimized approaches) that could hold the key to mapping tumour-specific ECM into high-fidelity biomaterial models. We anticipate that an interdisciplinary approach, combining materials science, cancer research and image analysis, will elucidate the role of 3D fibre architecture in tumour development, leading to the next generation of patient-mimetic models for mechanistic studies and drug discovery.
Collapse
Affiliation(s)
- Jennifer C Ashworth
- School of Veterinary Medicine & Science, Sutton Bonington Campus, University of Nottingham, Leicestershire, UK.
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK.
- Cancer Ecosystems Program, The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
| | - Thomas R Cox
- Cancer Ecosystems Program, The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
- The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia.
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
15
|
Golo M, Newman PLH, Kempe D, Biro M. Mechanoimmunology in the solid tumor microenvironment. Biochem Soc Trans 2024; 52:1489-1502. [PMID: 38856041 DOI: 10.1042/bst20231427] [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: 03/28/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem that adjoins the cancer cells within solid tumors and comprises distinct components such as extracellular matrix, stromal and immune cells, blood vessels, and an abundance of signaling molecules. In recent years, the mechanical properties of the TME have emerged as critical determinants of tumor progression and therapeutic response. Aberrant mechanical cues, including altered tissue architecture and stiffness, contribute to tumor progression, metastasis, and resistance to treatment. Moreover, burgeoning immunotherapies hold great promise for harnessing the immune system to target and eliminate solid malignancies; however, their success is hindered by the hostile mechanical landscape of the TME, which can impede immune cell infiltration, function, and persistence. Consequently, understanding TME mechanoimmunology - the interplay between mechanical forces and immune cell behavior - is essential for developing effective solid cancer therapies. Here, we review the role of TME mechanics in tumor immunology, focusing on recent therapeutic interventions aimed at modulating the mechanical properties of the TME to potentiate T cell immunotherapies, and innovative assays tailored to evaluate their clinical efficacy.
Collapse
Affiliation(s)
- Matteo Golo
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter L H Newman
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maté Biro
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
16
|
Zhang T, Chen R, Su X, Wang M, Lu Q. Integrated analysis of purine metabolism assists in predicting prognosis and treatment decisions for patients with lung adenocarcinoma. Heliyon 2024; 10:e29290. [PMID: 38601636 PMCID: PMC11004420 DOI: 10.1016/j.heliyon.2024.e29290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
The incidence of lung cancer, especially lung adenocarcinoma (LUAD), has recently increased. Targeted therapy and immunotherapy combined with conventional treatment have shown surprising benefits in enhancing the LUAD patient's prognosis. For the purpose of guiding treatment planning and the prognosis of LUAD, more research is required. The particular aim of this work was to establish a purine metabolism scoring (PMS) model for the purpose of individually forecasting treatment outcomes and overall survival for patients who have LUAD. Clinical and whole genome data were obtained from the TCGA-LUAD cohort via "UCSC". The 25 driver purine metabolism-related prognostic genes were determined founded on univariate Cox regression. Then PMS was developed through stepwise LASSO Cox regression. Survival analysis indicated that patients who have PMS experienced worse outcomes. We validated the PGM2 effect on lung adenocarcinoma malignancy in in vitro experiments. Univariate as well as multivariate Cox regression suggested that PMS was an independent prognostic indicator for LUAD patients, which was confirmed in subgroup analysis. Functional assay demonstrated that immune response as well as cytotoxicity pathways have a connection with lower PMS, and patients who have low PMS possess an active immune microenvironment. Moreover, the LUAD patients who have low PMS showed greater sensitivity to immunotherapy, targeted therapy, as well as chemotherapy. Knockdown of PGM2 was discovered to decrease the proliferation, invasion, as well as migration of lung adenocarcinoma cells in an in vitro assay. Pertaining to this particular research, we created a PMS model and conducted a thorough analysis of purine metabolism in LUAD in order to determine prognosis and offer recommendations for treatment. This finding offered a fresh concept for the clinical management of LUAD and novel therapy protocols.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Respiratory and Critical Care Medicine, Yixing Hospital Affiliated to Jiangsu University, Wuxi, 214221, China
| | - Ruhua Chen
- Department of Respiratory and Critical Care Medicine, Yixing Hospital Affiliated to Jiangsu University, Wuxi, 214221, China
| | - Xiangyu Su
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 21009, China
| | - Meng Wang
- Department of Respiratory and Critical Care Medicine, Yixing Hospital Affiliated to Jiangsu University, Wuxi, 214221, China
| | - Qin Lu
- Department of Respiratory and Critical Care Medicine, Yixing Hospital Affiliated to Jiangsu University, Wuxi, 214221, China
| |
Collapse
|
17
|
Burgess JK, Weiss DJ, Westergren-Thorsson G, Wigen J, Dean CH, Mumby S, Bush A, Adcock IM. Extracellular Matrix as a Driver of Chronic Lung Diseases. Am J Respir Cell Mol Biol 2024; 70:239-246. [PMID: 38190723 DOI: 10.1165/rcmb.2023-0176ps] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
The extracellular matrix (ECM) is not just a three-dimensional scaffold that provides stable support for all cells in the lungs, but also an important component of chronic fibrotic airway, vascular, and interstitial diseases. It is a bioactive entity that is dynamically modulated during tissue homeostasis and disease, that controls structural and immune cell functions and drug responses, and that can release fragments that have biological activity and that can be used to monitor disease activity. There is a growing recognition of the importance of considering ECM changes in chronic airway, vascular, and interstitial diseases, including 1) compositional changes, 2) structural and organizational changes, and 3) mechanical changes and how these affect disease pathogenesis. As altered ECM biology is an important component of many lung diseases, disease models must incorporate this factor to fully recapitulate disease-driver pathways and to study potential novel therapeutic interventions. Although novel models are evolving that capture some or all of the elements of the altered ECM microenvironment in lung diseases, opportunities exist to more fully understand cell-ECM interactions that will help devise future therapeutic targets to restore function in chronic lung diseases. In this perspective article, we review evolving knowledge about the ECM's role in homeostasis and disease in the lung.
Collapse
Affiliation(s)
- Janette K Burgess
- Department of Pathology and Medical Biology
- Groningen Research Institute for Asthma and COPD, and
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont
| | | | - Jenny Wigen
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Andrew Bush
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
- Centre for Pediatrics and Child Health, Imperial College and Royal Brompton Hospital, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| |
Collapse
|
18
|
Massey A, Stewart J, Smith C, Parvini C, McCormick M, Do K, Cartagena-Rivera AX. Mechanical properties of human tumour tissues and their implications for cancer development. NATURE REVIEWS. PHYSICS 2024; 6:269-282. [PMID: 38706694 PMCID: PMC11066734 DOI: 10.1038/s42254-024-00707-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 05/07/2024]
Abstract
The mechanical properties of cells and tissues help determine their architecture, composition and function. Alterations to these properties are associated with many diseases, including cancer. Tensional, compressive, adhesive, elastic and viscous properties of individual cells and multicellular tissues are mostly regulated by reorganization of the actomyosin and microtubule cytoskeletons and extracellular glycocalyx, which in turn drive many pathophysiological processes, including cancer progression. This Review provides an in-depth collection of quantitative data on diverse mechanical properties of living human cancer cells and tissues. Additionally, the implications of mechanical property changes for cancer development are discussed. An increased knowledge of the mechanical properties of the tumour microenvironment, as collected using biomechanical approaches capable of multi-timescale and multiparametric analyses, will provide a better understanding of the complex mechanical determinants of cancer organization and progression. This information can lead to a further understanding of resistance mechanisms to chemotherapies and immunotherapies and the metastatic cascade.
Collapse
Affiliation(s)
- Andrew Massey
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Stewart
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Cameron Parvini
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Moira McCormick
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kun Do
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Chowdhury D, Mistry A, Maity D, Bhatia R, Priyadarshi S, Wadan S, Chakraborty S, Haldar S. Pan-cancer analyses suggest kindlin-associated global mechanochemical alterations. Commun Biol 2024; 7:372. [PMID: 38548811 PMCID: PMC10978987 DOI: 10.1038/s42003-024-06044-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/11/2024] [Indexed: 04/01/2024] Open
Abstract
Kindlins serve as mechanosensitive adapters, transducing extracellular mechanical cues to intracellular biochemical signals and thus, their perturbations potentially lead to cancer progressions. Despite the kindlin involvement in tumor development, understanding their genetic and mechanochemical characteristics across different cancers remains elusive. Here, we thoroughly examined genetic alterations in kindlins across more than 10,000 patients with 33 cancer types. Our findings reveal cancer-specific alterations, particularly prevalent in advanced tumor stage and during metastatic onset. We observed a significant co-alteration between kindlins and mechanochemical proteome in various tumors through the activation of cancer-related pathways and adverse survival outcomes. Leveraging normal mode analysis, we predicted structural consequences of cancer-specific kindlin mutations, highlighting potential impacts on stability and downstream signaling pathways. Our study unraveled alterations in epithelial-mesenchymal transition markers associated with kindlin activity. This comprehensive analysis provides a resource for guiding future mechanistic investigations and therapeutic strategies targeting the roles of kindlins in cancer treatment.
Collapse
Affiliation(s)
- Debojyoti Chowdhury
- Department of Chemical and Biological Sciences, S.N. Bose National Centre for Basic Sciences, Kolkata, West Bengal, 700106, India.
| | - Ayush Mistry
- Department of Biology, Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana, 131029, India
| | - Debashruti Maity
- Department of Chemical and Biological Sciences, S.N. Bose National Centre for Basic Sciences, Kolkata, West Bengal, 700106, India
| | - Riti Bhatia
- Department of Biology, Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana, 131029, India
| | - Shreyansh Priyadarshi
- Department of Biology, Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana, 131029, India
| | - Simran Wadan
- Department of Biology, Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana, 131029, India
| | - Soham Chakraborty
- Department of Biology, Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana, 131029, India
| | - Shubhasis Haldar
- Department of Chemical and Biological Sciences, S.N. Bose National Centre for Basic Sciences, Kolkata, West Bengal, 700106, India.
- Department of Biology, Trivedi School of Biosciences, Ashoka University, Sonepat, Haryana, 131029, India.
- Technical Research Centre, S.N. Bose National Centre for Basic Sciences, Kolkata, West Bengal, 700106, India.
| |
Collapse
|
20
|
Sadhu RK, Luciano M, Xi W, Martinez-Torres C, Schröder M, Blum C, Tarantola M, Villa S, Penič S, Iglič A, Beta C, Steinbock O, Bodenschatz E, Ladoux B, Gabriele S, Gov NS. A minimal physical model for curvotaxis driven by curved protein complexes at the cell's leading edge. Proc Natl Acad Sci U S A 2024; 121:e2306818121. [PMID: 38489386 PMCID: PMC10963004 DOI: 10.1073/pnas.2306818121] [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: 04/27/2023] [Accepted: 01/29/2024] [Indexed: 03/17/2024] Open
Abstract
Cells often migrate on curved surfaces inside the body, such as curved tissues, blood vessels, or highly curved protrusions of other cells. Recent in vitro experiments provide clear evidence that motile cells are affected by the curvature of the substrate on which they migrate, preferring certain curvatures to others, termed "curvotaxis." The origin and underlying mechanism that gives rise to this curvature sensitivity are not well understood. Here, we employ a "minimal cell" model which is composed of a vesicle that contains curved membrane protein complexes, that exert protrusive forces on the membrane (representing the pressure due to actin polymerization). This minimal-cell model gives rise to spontaneous emergence of a motile phenotype, driven by a lamellipodia-like leading edge. By systematically screening the behavior of this model on different types of curved substrates (sinusoidal, cylinder, and tube), we show that minimal ingredients and energy terms capture the experimental data. The model recovers the observed migration on the sinusoidal substrate, where cells move along the grooves (minima), while avoiding motion along the ridges. In addition, the model predicts the tendency of cells to migrate circumferentially on convex substrates and axially on concave ones. Both of these predictions are verified experimentally, on several cell types. Altogether, our results identify the minimization of membrane-substrate adhesion energy and binding energy between the membrane protein complexes as key players of curvotaxis in cell migration.
Collapse
Affiliation(s)
- Raj Kumar Sadhu
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Marine Luciano
- Department of Biochemistry, University of Geneva, Geneva4 CH-1211, Switzerland
- Mechanobiology & Biomaterials Group, Research Institute for Biosciences, Center of Innovation and Research in Materials and Polymers, University of Mons, MonsB-7000, Belgium
| | - Wang Xi
- Universite Paris Cite, CNRS, Institut Jacques Monod, ParisF-75013, France
| | | | - Marcel Schröder
- Department of Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen37077, Germany
| | - Christoph Blum
- Department of Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen37077, Germany
| | - Marco Tarantola
- Department of Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen37077, Germany
| | - Stefano Villa
- Department of Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen37077, Germany
| | - Samo Penič
- Laboratory of Physics, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana1000, Slovenia
| | - Aleš Iglič
- Laboratory of Physics, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana1000, Slovenia
| | - Carsten Beta
- Institute of Physics and Astronomy, University of Potsdam, Potsdam14476, Germany
- Nano Life Science Institute, Kanazawa University, Kanazawa920-1192, Japan
| | - Oliver Steinbock
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL32306-4390
| | - Eberhard Bodenschatz
- Department of Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen37077, Germany
| | - Benoît Ladoux
- Universite Paris Cite, CNRS, Institut Jacques Monod, ParisF-75013, France
| | - Sylvain Gabriele
- Mechanobiology & Biomaterials Group, Research Institute for Biosciences, Center of Innovation and Research in Materials and Polymers, University of Mons, MonsB-7000, Belgium
| | - Nir S. Gov
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot7610001, Israel
| |
Collapse
|
21
|
Nizamoglu M, Alleblas F, Koster T, Borghuis T, Vonk JM, Thomas MJ, White ES, Watson CK, Timens W, El Kasmi KC, Melgert BN, Heijink IH, Burgess JK. Three dimensional fibrotic extracellular matrix directs microenvironment fiber remodeling by fibroblasts. Acta Biomater 2024; 177:118-131. [PMID: 38350556 DOI: 10.1016/j.actbio.2024.02.008] [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: 10/13/2023] [Revised: 01/12/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF), for which effective treatments are limited, results in excessive and disorganized deposition of aberrant extracellular matrix (ECM). An altered ECM microenvironment is postulated to contribute to disease progression through inducing profibrotic behavior of lung fibroblasts, the main producers and regulators of ECM. Here, we examined this hypothesis in a 3D in vitro model system by growing primary human lung fibroblasts in ECM-derived hydrogels from non-fibrotic (control) or IPF lung tissue. Using this model, we compared how control and IPF lung-derived fibroblasts responded in control and fibrotic microenvironments in a combinatorial manner. Culture of fibroblasts in fibrotic hydrogels did not alter in the overall amount of collagen or glycosaminoglycans but did cause a drastic change in fiber organization compared to culture in control hydrogels. High-density collagen percentage was increased by control fibroblasts in IPF hydrogels at day 7, but decreased at day 14. In contrast, IPF fibroblasts only decreased the high-density collagen percentage at day 14, which was accompanied by enhanced fiber alignment in IPF hydrogels. Similarly, stiffness of fibrotic hydrogels was increased only by control fibroblasts by day 14 while those of control hydrogels were not altered by fibroblasts. These data highlight how the ECM-remodeling responses of fibroblasts are influenced by the origin of both the cells and the ECM. Moreover, by showing how the 3D microenvironment plays a crucial role in directing cells, our study paves the way in guiding future investigations examining fibrotic processes with respect to ECM remodeling responses of fibroblasts. STATEMENT OF SIGNIFICANCE: In this study, we investigated the influence of the altered extracellular matrix (ECM) in Idiopathic Pulmonary Fibrosis (IPF), using a 3D in vitro model system composed of ECM-derived hydrogels from both IPF and control lungs, seeded with human IPF and control lung fibroblasts. While our results indicated that fibrotic microenvironment did not change the overall collagen or glycosaminoglycan content, it resulted in a dramatically alteration of fiber organization and mechanical properties. Control fibroblasts responded differently from IPF fibroblasts, highlighting the unique instructive role of the fibrotic ECM and the interplay with fibroblast origin. These results underscore the importance of 3D microenvironments in guiding pro-fibrotic responses, offering potential insights for future IPF therapies as well as other fibrotic diseases and cancer.
Collapse
Affiliation(s)
- Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Frederique Alleblas
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Taco Koster
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Theo Borghuis
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Judith M Vonk
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - Matthew J Thomas
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Eric S White
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, United States
| | - Carolin K Watson
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Karim C El Kasmi
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Barbro N Melgert
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, Groningen, the Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, the Netherlands.
| |
Collapse
|
22
|
Liu Y, Zhao T, Xu Z, Dai N, Zhao Q, Liang Y, Geng S, Lei M, Xu F, Wang L, Cheng B. Influence of Curvature on Cell Motility and Morphology during Cancer Migration in Confined Microchannels. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9956-9967. [PMID: 38349958 DOI: 10.1021/acsami.4c00196] [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: 02/15/2024]
Abstract
Microchannels often serve as highways for cancer migration, and their topology largely determines the migration efficiency. Curvature, a topological parameter in biological systems, has recently been reported to be efficient in guiding cell polarization and migration. Curvature varies widely along curved microchannels, while its influence on cell migration remains elusive. Here, we recapitulated the curved microchannels, as observed in clinical tumor tissues with hydrogels, and studied how cancer cells respond to curvature. We found that cells bend more significantly in a larger curvature and exhibit less spreading as well as lower motility. The underlying mechanism is probably based on the hindrance of the movement of cytoskeletal molecules at the curved microchannel walls. Collectively, our results demonstrated that the accelerated actin retrograde flow rate under local curvature has an effective negative regulation on cell motility and morphology, leading to shortened and bent cell morphologies as well as hampered cell migration efficiency.
Collapse
Affiliation(s)
- Yan Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi 710049, PR China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Shaanxi 710049, PR China
| | - Tianyu Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhao Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi 710049, PR China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Shaanxi 710049, PR China
| | - Ningman Dai
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi 710049, PR China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Shaanxi 710049, PR China
| | - Qiang Zhao
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Shaanxi 710049, PR China
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi 710049, PR China
| | - Yutong Liang
- College of Medicine, Xi'an International University, Xi'an, Shaanxi 710077, PR China
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi 710049, PR China
| | - Ming Lei
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi 710049, PR China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Shaanxi 710049, PR China
| | - Lin Wang
- College of Medicine, Xi'an International University, Xi'an, Shaanxi 710077, PR China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation Universities of Shaanxi Province, Xi'an 710077, China
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi 710049, PR China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Shaanxi 710049, PR China
| |
Collapse
|
23
|
Zhang Y, Zheng H, Xu M, Maeda N, Tsunedomi R, Kishi H, Nagano H, Kobayashi S. Fyn-Mediated Paxillin Tyrosine 31 Phosphorylation Regulates Migration and Invasion of Breast Cancer Cells. Int J Mol Sci 2023; 24:15980. [PMID: 37958964 PMCID: PMC10647795 DOI: 10.3390/ijms242115980] [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: 10/09/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Metastasis is the leading cause of death in breast cancer patients due to the lack of effective therapies. Elevated levels of paxillin expression have been observed in various cancer types, with tyrosine phosphorylation shown to play a critical role in driving cancer cell migration. However, the specific impact of the distinct tyrosine phosphorylation events of paxillin in the progression of breast cancer remains to be fully elucidated. Here, we found that paxillin overexpression in breast cancer tissue is associated with a patient's poor prognosis. Paxillin knockdown inhibited the migration and invasion of breast cancer cells. Furthermore, the phosphorylation of paxillin tyrosine residue 31 (Tyr31) was significantly increased upon the TGF-β1-induced migration and invasion of breast cancer cells. Inhibiting Fyn activity or silencing Fyn decreases paxillin Tyr31 phosphorylation. The wild-type and constitutively active Fyn directly phosphorylate paxillin Tyr31 in an in vitro system, indicating that Fyn directly phosphorylates paxillin Tyr31. Additionally, the non-phosphorylatable mutant of paxillin at Tyr31 reduces actin stress fiber formation, migration, and invasion of breast cancer cells. Taken together, our results provide direct evidence that Fyn-mediated paxillin Tyr31 phosphorylation is required for breast cancer migration and invasion, suggesting that targeting paxillin Tyr31 phosphorylation could be a potential therapeutic strategy for mitigating breast cancer metastasis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan;
| | - Huanyu Zheng
- Department of Gastroenterological, Breast and Endocrine Surgery, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan (H.N.)
| | - Ming Xu
- Department of Gastroenterological, Breast and Endocrine Surgery, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan (H.N.)
| | - Noriko Maeda
- Department of Gastroenterological, Breast and Endocrine Surgery, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan (H.N.)
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan (H.N.)
| | - Hiroko Kishi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan;
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan (H.N.)
| | - Sei Kobayashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan;
| |
Collapse
|
24
|
Cho Y, Kim J, Park J, Doh J. Surface nanotopography and cell shape modulate tumor cell susceptibility to NK cell cytotoxicity. MATERIALS HORIZONS 2023; 10:4532-4540. [PMID: 37559559 DOI: 10.1039/d3mh00367a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Natural killer (NK) cells are innate cytotoxic lymphocytes exerting cytotoxicity against virally infected cells and tumor cells. NK cell cytotoxicity is primarily determined by biochemical signals received from ligands expressed on target cell surfaces, but it is also possible that biophysical environments of tumor cells, such as nanoscale surface topography typically existing on extracellular matrixes (ECMs) or cell morphology determined by ECM spaces or cell density, regulate NK cell cytotoxicity. In this study, micro/nanofabrication technology was applied to examine this possibility. Tumor cells were plated on flat or nanogrooved surfaces, or micropatterned into circular or elliptical geometries, and the effects of surface topography and tumor cell morphology on NK cell cytotoxicity were investigated. NK cells exhibited significantly higher cytotoxicity against tumor cells on nanogrooved surfaces or tumor cells in elliptical patterns than tumor cells on flat surfaces or tumor cells in circular patterns, respectively. The amounts of stress fiber formation in tumor cells positively correlated with NK cell cytotoxicity, indicating that increased cellular tension of tumor cells, either mediated by nanogrooved surfaces or elongated morphologies, was a key factor regulating NK cell cytotoxicity. These results may provide insight into the design of NK cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Yongbum Cho
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology, 77, Cheongam-ro, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - JangHyuk Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul, South Korea.
| | - Jeehun Park
- SOFT Foundry Institute, Seoul National University, Seoul, South Korea.
| | - Junsang Doh
- Department of Materials Science and Engineering, Seoul National University, Seoul, South Korea.
- SOFT Foundry Institute, Seoul National University, Seoul, South Korea.
- Institute of Engineering Research, BioMAX, Seoul National University, Seoul, South Korea
| |
Collapse
|
25
|
Su H, Karin M. Collagen architecture and signaling orchestrate cancer development. Trends Cancer 2023; 9:764-773. [PMID: 37400314 DOI: 10.1016/j.trecan.2023.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 07/05/2023]
Abstract
The tumor microenvironment (TME) controls tumor progression and maintenance. Accordingly, tumor-centric cancer treatment must adjust to being more holistic and TME-centric. Collagens are the most abundant TME proteins, and their dynamic remodeling profoundly affects both TME architecture and tumor development. Recent evidence shows that in addition to being structural elements, collagens are an important source of nutrients and decisive growth controlling and immunoregulatory signals. This review focuses on macropinocytosis-dependent collagen support of cancer cell metabolism and the role of collagen fiber remodeling and trimer heterogeneity in control of tumor bioenergetics, growth, progression, and response to therapy. If properly translated, these basic advances may alter the future of cancer treatment.
Collapse
Affiliation(s)
- Hua Su
- Institutes of Biomedical Sciences, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
26
|
Baschieri F, Illand A, Barbazan J, Zajac O, Henon C, Loew D, Dingli F, Vignjevic DM, Lévêque-Fort S, Montagnac G. Fibroblasts generate topographical cues that steer cancer cell migration. SCIENCE ADVANCES 2023; 9:eade2120. [PMID: 37585527 PMCID: PMC10431708 DOI: 10.1126/sciadv.ade2120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 07/14/2023] [Indexed: 08/18/2023]
Abstract
Fibroblasts play a fundamental role in tumor development. Among other functions, they regulate cancer cells' migration through rearranging the extracellular matrix, secreting soluble factors, and establishing direct physical contacts with cancer cells. Here, we report that migrating fibroblasts deposit on the substrate a network of tubular structures that serves as a guidance cue for cancer cell migration. Such membranous tubular network, hereafter called tracks, is stably anchored to the substrate in a β5-integrin-dependent manner. We found that cancer cells specifically adhere to tracks by using clathrin-coated structures that pinch and engulf tracks. Tracks thus represent a spatial memory of fibroblast migration paths that is read and erased by cancer cells directionally migrating along them. We propose that fibroblast tracks represent a topography-based intercellular communication system capable of steering cancer cell migration.
Collapse
Affiliation(s)
- Francesco Baschieri
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Abigail Illand
- Université Paris Saclay, CNRS, Institut des sciences moléculaires d’Orsay, UMR8214, Orsay, France
| | - Jorge Barbazan
- Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Olivier Zajac
- Institut Curie, UMR144, PSL Research University, Centre Universitaire, Paris, France
| | - Clémence Henon
- Inserm U981, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | | | - Sandrine Lévêque-Fort
- Université Paris Saclay, CNRS, Institut des sciences moléculaires d’Orsay, UMR8214, Orsay, France
| | - Guillaume Montagnac
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
27
|
Lichtenberg JY, Tran S, Hwang PY. Mechanical factors driving cancer progression. Adv Cancer Res 2023; 160:61-81. [PMID: 37704291 DOI: 10.1016/bs.acr.2023.06.002] [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] [Indexed: 09/15/2023]
Abstract
A fundamental step of tumor metastasis is tumor cell migration away from the primary tumor site. One mode of migration that is essential but still understudied is collective invasion, the process by which clusters of cells move in a coordinated fashion. In recent years, there has been growing interest to understand factors regulating collective invasion, with increasing number of studies investigating the biomechanical regulation of collective invasion. In this review we discuss the dynamic relationship between tumor microenvironment cues and cell response by first covering mechanical factors in the microenvironment and second, discussing the mechanosensing pathways utilized by cells in collective clusters to dynamically respond to mechanical matrix cues. Finally, we discuss model systems that have been developed which have increased our understanding of the mechanical factors contributing to tumor progression.
Collapse
Affiliation(s)
- Jessanne Y Lichtenberg
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Sydnie Tran
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Priscilla Y Hwang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
28
|
Zhang H, Fan J, Maclin JM, Wan LQ. The Actin Crosslinker Fascin Regulates Cell Chirality. Adv Biol (Weinh) 2023; 7:e2200240. [PMID: 36658789 PMCID: PMC10293081 DOI: 10.1002/adbi.202200240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Indexed: 01/21/2023]
Abstract
The left-right (L-R) asymmetry of the cells, or cell chirality, is a well-known intrinsic property derived from the dynamic organization of the actin cytoskeleton. Cell chirality can be regulated by actin-binding proteins such as α-actinin-1 and can also be mediated by certain signaling pathways, such as protein kinase C (PKC) signaling. Fascin, an actin crosslinker known to mediate parallel bundling of actin filaments, appears as a prominent candidate in cell chirality regulation, given its role in facilitating cell migration as an important PKC substrate. Here, it is shown that the chirality of NIH/3T3 cells can be altered by PKC activation and fascin manipulation. With either small-molecule drug inhibition or genetic knockdown of fascin, the chirality of 3T3 cells is reversed from a clockwise (CW) bias to a counterclockwise (CCW) bias on ring-shaped micropatterns, accompanied by the reversal in cell directional migration. The Ser-39 fascin-actin binding sites are further explored in cell chirality regulation. The findings of this study reveal the critical role of fascin as an important intermediator in cell chirality, shedding novel insights into the mechanisms of L-R asymmetric cell migration and multicellular morphogenesis.
Collapse
Affiliation(s)
- Haokang Zhang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Jie Fan
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI 48128, USA
| | - Joshua M.A. Maclin
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Biological Science, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Biological Science, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
29
|
Alneyadi A, Nizami ZN, Aburawi HE, Hisaindee S, Nawaz M, Attoub S, Ramadan G, Benhalilou N, Al Azzani M, Elmahi Y, Almeqbali A, Muhammad K, Eid AH, Vijayan R, Iratni R. Synthesis of New Chromene Derivatives Targeting Triple-Negative Breast Cancer Cells. Cancers (Basel) 2023; 15:2682. [PMID: 37345018 DOI: 10.3390/cancers15102682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 05/06/2023] [Indexed: 06/23/2023] Open
Abstract
Breast cancer continues to be the leading cause of cancer-related deaths among women worldwide. The most aggressive type of breast cancer is triple-negative breast cancer (TNBC). Indeed, not only does TNBC not respond well to several chemotherapeutic agents, but it also frequently develops resistance to various anti-cancer drugs, including taxane mitotic inhibitors. This necessitates the search for newer, more efficacious drugs. In this study, we synthesized two novel chromene derivatives (C1 and C2) and tested their efficacy against a battery of luminal type A and TNBC cell lines. Our results show that C1 and C2 significantly and specifically inhibited TNBC cell viability but had no effect on the luminal A cell type. In addition, these novel compounds induced mitotic arrest, cell multinucleation leading to senescence, and apoptotic cell death through the activation of the extrinsic pathway. We also showed that the underlying mechanisms for these actions of C1 and C2 involved inhibition of microtubule polymerization and disruption of the F-actin cytoskeleton. Furthermore, both compounds significantly attenuated migration of TNBC cells and inhibited angiogenesis in vitro. Finally, we performed an in silico analysis, which revealed that these novel variants bind to the colchicine binding site in β-tubulin. Taken together, our data highlight the potential chemotherapeutic properties of two novel chromene compounds against TNBC.
Collapse
Affiliation(s)
- Aysha Alneyadi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Hanan E Aburawi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Soleiman Hisaindee
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Samir Attoub
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
| | - Gaber Ramadan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Nehla Benhalilou
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Yassine Elmahi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Aysha Almeqbali
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Ranjit Vijayan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
30
|
Pant B, Park M, Kim AA. Electrospun Nanofibers for Dura Mater Regeneration: A Mini Review on Current Progress. Pharmaceutics 2023; 15:pharmaceutics15051347. [PMID: 37242589 DOI: 10.3390/pharmaceutics15051347] [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: 03/24/2023] [Revised: 04/14/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
Dural defects are a common problem in neurosurgical procedures and should be repaired to avoid complications such as cerebrospinal fluid leakage, brain swelling, epilepsy, intracranial infection, and so on. Various types of dural substitutes have been prepared and used for the treatment of dural defects. In recent years, electrospun nanofibers have been applied for various biomedical applications, including dural regeneration, due to their interesting properties such as a large surface area to volume ratio, porosity, superior mechanical properties, ease of surface modification, and, most importantly, similarity with the extracellular matrix (ECM). Despite continuous efforts, the development of suitable dura mater substrates has had limited success. This review summarizes the investigation and development of electrospun nanofibers with particular emphasis on dura mater regeneration. The objective of this mini-review article is to give readers a quick overview of the recent advances in electrospinning for dura mater repair.
Collapse
Affiliation(s)
- Bishweshwar Pant
- Carbon Composite Energy Nanomaterials Research Center, Woosuk University, Wanju 55338, Republic of Korea
- Woosuk Institute of Smart Convergence Life Care (WSCLC), Woosuk University, Wanju 55338, Republic of Korea
- Department of Automotive Engineering, Woosuk University, Wanju 55338, Republic of Korea
| | - Mira Park
- Carbon Composite Energy Nanomaterials Research Center, Woosuk University, Wanju 55338, Republic of Korea
- Woosuk Institute of Smart Convergence Life Care (WSCLC), Woosuk University, Wanju 55338, Republic of Korea
- Department of Automotive Engineering, Woosuk University, Wanju 55338, Republic of Korea
| | - Allison A Kim
- Department of Healthcare Management, Woosong University, Daejon 34606, Republic of Korea
| |
Collapse
|
31
|
Phung TKN, Mitchel JA, O'Sullivan MJ, Park JA. Quantification of basal stem cell elongation and stress fiber accumulation in the pseudostratified airway epithelium during the unjamming transition. Biol Open 2023; 12:bio059727. [PMID: 37014330 PMCID: PMC10151827 DOI: 10.1242/bio.059727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/16/2023] [Indexed: 04/05/2023] Open
Abstract
Under homeostatic conditions, epithelial cells remain non-migratory. However, during embryonic development and pathological conditions, they become migratory. The mechanism underlying the transition of the epithelial layer between non-migratory and migratory phases is a fundamental question in biology. Using well-differentiated primary human bronchial epithelial cells that form a pseudostratified epithelium, we have previously identified that a confluent epithelial layer can transition from a non-migratory to migratory phase through an unjamming transition (UJT). We previously defined collective cellular migration and apical cell elongation as hallmarks of UJT. However, other cell-type-specific changes have not been previously studied in the pseudostratified airway epithelium, which consists of multiple cell types. Here, we focused on the quantifying morphological changes in basal stem cells during the UJT. Our data demonstrate that during the UJT, airway basal stem cells elongated and enlarged, and their stress fibers elongated and aligned. These morphological changes observed in basal stem cells correlated to the previously defined hallmarks of the UJT. Moreover, basal cell and stress fiber elongation were observed prior to apical cell elongation. Together, these morphological changes indicate that basal stem cells in pseudostratified airway epithelium are actively remodeling, presumably through accumulation of stress fibers during the UJT.
Collapse
Affiliation(s)
- Thien-Khoi N. Phung
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jennifer A. Mitchel
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Biology, Wesleyan University, Middletown, CT 06459, USA
| | - Michael J. O'Sullivan
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
32
|
Lei X, Miao S, Wang X, Gao Y, Wu H, Cheng P, Song Y, Bi L, Pei G. Microgroove Cues Guiding Fibrogenesis of Stem Cells via Intracellular Force. ACS APPLIED MATERIALS & INTERFACES 2023; 15:16380-16393. [PMID: 36961871 DOI: 10.1021/acsami.2c20903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Groove patterns are widely used in material surface modifications. However, the independent role of ditches/ridges in regulating fibrosis of soft tissues is not well-understood, especially the lack of linkage evidence in vitro and in vivo. Herein, two kinds of combinational microgroove chips with the gradient ditch/ridge width were fabricated by photolithography technology, termed R and G groups, respectively. In group R, the ridge width was 1, 5, 10, and 30 μm, with a ditch width of 30 μm; in group G, the groove width was 5, 10, 20, and 30 μm, and the ridge width was 5 μm. The effect of microgrooves on the morphology, proliferation, and expression of fibrous markers of stem cells was systematically investigated in vitro. Moreover, thicknesses of fibrous capsules were evaluated after chips were implanted into the muscular pouches of rats for 5 months. The results show that microgrooves have almost no effect on cell proliferation but significantly modulate the morphology of cells and focal adhesions (FAs) in vitro, as well as fibrosis differentiation. In particular, the differentiation of stem cells is attenuated after the intracellular force caused by stress fibers and FAs is interfered by drugs, such as rotenone and blebbistatin. Histological analysis shows that patterns of high intracellular force can apparently stimulate soft tissue fibrosis in vivo. This study not only reveals the specific rules and mechanisms of ditch/ridge regulating stem cell behaviors but also offers insight into tailoring implant surface patterns to induce controlled soft tissue fibrosis.
Collapse
Affiliation(s)
- Xing Lei
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
- Department of Orthopedic Surgery, Linyi People's Hospital, Linyi 276000, China
| | - Sheng Miao
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Xiuli Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Yi Gao
- Southern University of Science and Technology Hospital, No. 6019 Liuxian Street, Xili Avenue, Nanshan District, Shenzhen 518055, China
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hao Wu
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Pengzhen Cheng
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Yue Song
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Long Bi
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Guoxian Pei
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
- Southern University of Science and Technology Hospital, No. 6019 Liuxian Street, Xili Avenue, Nanshan District, Shenzhen 518055, China
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
33
|
McGowan SE. Discoidin domain receptor-2 enhances secondary alveolar septation in mice by activating integrins and modifying focal adhesions. Am J Physiol Lung Cell Mol Physiol 2023; 324:L307-L324. [PMID: 36719983 PMCID: PMC9988528 DOI: 10.1152/ajplung.00169.2022] [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: 05/30/2022] [Revised: 12/20/2022] [Accepted: 01/09/2023] [Indexed: 02/02/2023] Open
Abstract
The extracellular matrix (ECM) of the pulmonary parenchyma must maintain the structural relationships among resident cells during the constant distortion imposed by respiration. This dictates that both the ECM and cells adapt to changes in shape, while retaining their attachment. Membrane-associated integrins and discoidin domain receptors (DDR) bind collagen and transmit signals to the cellular cytoskeleton. Although the contributions of DDR2 to collagen deposition and remodeling during osseous development are evident, it is unclear how DDR2 contributes to lung development. Using mice (smallie, Slie/Slie, DDR2Δ) bearing a spontaneous inactivating deletion within the DDR2 coding region, we observed a decrease in gas-exchange surface area and enlargement of alveolar ducts. Compared with fibroblasts isolated from littermate controls, DDR2Δ fibroblasts, spread more slowly, developed fewer lamellipodia, and were less responsive to the rigidity of neighboring collagen fibers. Activated β1-integrin (CD29) was reduced in focal adhesions (FA) of DDR2Δ fibroblasts, less phospho-zyxin localized to and fewer FA developed over ventral actin stress fibers, and the adhesions had a lower aspect ratio compared with controls. However, DDR2 deletion did not reduce cellular displacement of the ECM. Our findings indicate that DDR2, in concert with collagen-binding β1-integrins, regulates the timing and location of focal adhesion formation and how lung fibroblasts respond to ECM rigidity. Reduced rigidity sensing and mechano-responsiveness may contribute to the distortion of alveolar ducts, where the fiber cable-network is enriched and tensile forces are concentrated. Strategies targeting DDR2 could help guide fibroblasts to locations where tensile forces organize parenchymal repair.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| |
Collapse
|
34
|
Cho IS, Gupta P, Mostafazadeh N, Wong SW, Saichellappa S, Lenzini S, Peng Z, Shin J. Deterministic Single Cell Encapsulation in Asymmetric Microenvironments to Direct Cell Polarity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206014. [PMID: 36453581 PMCID: PMC9875620 DOI: 10.1002/advs.202206014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Indexed: 06/17/2023]
Abstract
Various signals in tissue microenvironments are often unevenly distributed around cells. Cellular responses to asymmetric cell-matrix adhesion in a 3D space remain generally unclear and are to be studied at the single-cell resolution. Here, the authors developed a droplet-based microfluidic approach to manufacture a pure population of single cells in a microscale layer of compartmentalized 3D hydrogel matrices with a tunable spatial presentation of ligands at the subcellular level. Cells elongate with an asymmetric presentation of the integrin adhesion ligand Arg-Gly-Asp (RGD), while cells expand isotropically with a symmetric presentation of RGD. Membrane tension is higher on the side of single cells interacting with RGD than on the side without RGD. Finite element analysis shows that a non-uniform isotropic cell volume expansion model is sufficient to recapitulate the experimental results. At a longer timescale, asymmetric ligand presentation commits mesenchymal stem cells to the osteogenic lineage. Cdc42 is an essential mediator of cell polarization and lineage specification in response to asymmetric cell-matrix adhesion. This study highlights the utility of precisely controlling 3D ligand presentation around single cells to direct cell polarity for regenerative engineering and medicine.
Collapse
Affiliation(s)
- Ik Sung Cho
- Department of Pharmacology and Regenerative MedicineUniversity of Illinois at Chicago College of MedicineChicagoIL60612USA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Prerak Gupta
- Department of Pharmacology and Regenerative MedicineUniversity of Illinois at Chicago College of MedicineChicagoIL60612USA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Nima Mostafazadeh
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Sing Wan Wong
- Department of Pharmacology and Regenerative MedicineUniversity of Illinois at Chicago College of MedicineChicagoIL60612USA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Saiumamaheswari Saichellappa
- Department of Pharmacology and Regenerative MedicineUniversity of Illinois at Chicago College of MedicineChicagoIL60612USA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Stephen Lenzini
- Department of Pharmacology and Regenerative MedicineUniversity of Illinois at Chicago College of MedicineChicagoIL60612USA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Zhangli Peng
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| | - Jae‐Won Shin
- Department of Pharmacology and Regenerative MedicineUniversity of Illinois at Chicago College of MedicineChicagoIL60612USA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIL60607USA
| |
Collapse
|
35
|
Xia N, Jin D, Pan C, Zhang J, Yang Z, Su L, Zhao J, Wang L, Zhang L. Dynamic morphological transformations in soft architected materials via buckling instability encoded heterogeneous magnetization. Nat Commun 2022; 13:7514. [PMID: 36473857 PMCID: PMC9727123 DOI: 10.1038/s41467-022-35212-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
The geometric reconfigurations in three-dimensional morphable structures have a wide range of applications in flexible electronic devices and smart systems with unusual mechanical, acoustic, and thermal properties. However, achieving the highly controllable anisotropic transformation and dynamic regulation of architected materials crossing different scales remains challenging. Herein, we develop a magnetic regulation approach that provides an enabling technology to achieve the controllable transformation of morphable structures and unveil their dynamic modulation mechanism as well as potential applications. With buckling instability encoded heterogeneous magnetization profiles inside soft architected materials, spatially and temporally programmed magnetic inputs drive the formation of a variety of anisotropic morphological transformations and dynamic geometric reconfiguration. The introduction of magnetic stimulation could help to predetermine the buckling states of soft architected materials, and enable the formation of definite and controllable buckling states without prolonged magnetic stimulation input. The dynamic modulations can be exploited to build systems with switchable fluidic properties and are demonstrated to achieve capabilities of fluidic manipulation, selective particle trapping, sensitivity-enhanced biomedical analysis, and soft robotics. The work provides new insights to harness the programmable and dynamic morphological transformation of soft architected materials and promises benefits in microfluidics, programmable metamaterials, and biomedical applications.
Collapse
Affiliation(s)
- Neng Xia
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Dongdong Jin
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China.
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, China.
| | - Chengfeng Pan
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiachen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Zhengxin Yang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Lin Su
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinsheng Zhao
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Liu Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Li Zhang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China.
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK T Stone Robotics Institute, The Chinese University of Hong Kong, Hong Kong, China.
- Department of Surgery, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China.
| |
Collapse
|
36
|
The Forces behind Directed Cell Migration. BIOPHYSICA 2022. [DOI: 10.3390/biophysica2040046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Directed cell migration is an essential building block of life, present when an embryo develops, a dendritic cell migrates toward a lymphatic vessel, or a fibrotic organ fails to restore its normal parenchyma. Directed cell migration is often guided by spatial gradients in a physicochemical property of the cell microenvironment, such as a gradient in chemical factors dissolved in the medium or a gradient in the mechanical properties of the substrate. Single cells and tissues sense these gradients, establish a back-to-front polarity, and coordinate the migration machinery accordingly. Central to these steps we find physical forces. In some cases, these forces are integrated into the gradient sensing mechanism. Other times, they transmit information through cells and tissues to coordinate a collective response. At any time, they participate in the cellular migratory system. In this review, we explore the role of physical forces in gradient sensing, polarization, and coordinating movement from single cells to multicellular collectives. We use the framework proposed by the molecular clutch model and explore to what extent asymmetries in the different elements of the clutch can lead to directional migration.
Collapse
|
37
|
Holmes C, Varas J, San Martín S, Egaña JT. Towards an In Vitro 3D Model for Photosynthetic Cancer Treatment: A Study of Microalgae and Tumor Cell Interactions. Int J Mol Sci 2022; 23:13550. [PMID: 36362338 PMCID: PMC9657947 DOI: 10.3390/ijms232113550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/05/2022] [Accepted: 10/20/2022] [Indexed: 09/08/2024] Open
Abstract
As hypoxic tumors show resistance to several clinical treatments, photosynthetic microorganisms have been recently suggested as a promising safe alternative for oxygenating the tumor microenvironment. The relationship between organisms and the effect microalgae have on tumors is still largely unknown, evidencing the need for a simple yet representative model for studying photosynthetic tumor oxygenation in a reproducible manner. Here, we present a 3D photosynthetic tumor model composed of human melanoma cells and the microalgae Chlamydomonas reinhardtii, both seeded into a collagen scaffold, which allows for the simultaneous study of both cell types. This work focuses on the biocompatibility and cellular interactions of the two cell types, as well as the study of photosynthetic oxygenation of the tumor cells. It is shown that both cell types are biocompatible with one another at cell culture conditions and that a 10:1 ratio of microalgae to cells meets the metabolic requirement of the tumor cells, producing over twice the required amount of oxygen. This 3D tumor model provides an easy-to-use in vitro resource for analyzing the effects of photosynthetically produced oxygen on a tumor microenvironment, thus opening various potential research avenues.
Collapse
Affiliation(s)
- Christopher Holmes
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7821093, Chile
| | - Juan Varas
- Biomedical Research Center, School of Medicine, Universidad de Valparaiso, Viña del Mar 2520000, Chile
| | - Sebastián San Martín
- Biomedical Research Center, School of Medicine, Universidad de Valparaiso, Viña del Mar 2520000, Chile
| | - José Tomás Egaña
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7821093, Chile
| |
Collapse
|
38
|
Vasudevan J, Jiang K, Fernandez J, Lim CT. Extracellular matrix mechanobiology in cancer cell migration. Acta Biomater 2022; 163:351-364. [PMID: 36243367 DOI: 10.1016/j.actbio.2022.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/11/2022] [Accepted: 10/06/2022] [Indexed: 11/01/2022]
Abstract
The extracellular matrix (ECM) is pivotal in modulating tumor progression. Besides chemically stimulating tumor cells, it also offers physical support that orchestrates the sequence of events in the metastatic cascade upon dynamically modulating cell mechanosensation. Understanding this translation between matrix biophysical cues and intracellular signaling has led to rapid growth in the interdisciplinary field of cancer mechanobiology in the last decade. Substantial efforts have been made to develop novel in vitro tumor mimicking platforms to visualize and quantify the mechanical forces within the tissue that dictate tumor cell invasion and metastatic growth. This review highlights recent findings on tumor matrix biophysical cues such as fibrillar arrangement, crosslinking density, confinement, rigidity, topography, and non-linear mechanics and their implications on tumor cell behavior. We also emphasize how perturbations in these cues alter cellular mechanisms of mechanotransduction, consequently enhancing malignancy. Finally, we elucidate engineering techniques to individually emulate the mechanical properties of tumors that could help serve as toolkits for developing and testing ECM-targeted therapeutics on novel bioengineered tumor platforms. STATEMENT OF SIGNIFICANCE: Disrupted ECM mechanics is a driving force for transitioning incipient cells to life-threatening malignant variants. Understanding these ECM changes can be crucial as they may aid in developing several efficacious drugs that not only focus on inducing cytotoxic effects but also target specific matrix mechanical cues that support and enhance tumor invasiveness. Designing and implementing an optimal tumor mimic can allow us to predictively map biophysical cue-modulated cell behaviors and facilitate the design of improved lab-grown tumor models with accurately controlled structural features. This review focuses on the abnormal changes within the ECM during tumorigenesis and its implications on tumor cell-matrix mechanoreciprocity. Additionally, it accentuates engineering approaches to produce ECM features of varying levels of complexity which is critical for improving the efficiency of current engineered tumor tissue models.
Collapse
|
39
|
Nakanishi J, Yamamoto S. Static and photoresponsive dynamic materials to dissect physical regulation of cellular functions. Biomater Sci 2022; 10:6116-6134. [PMID: 36111810 DOI: 10.1039/d2bm00789d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent progress in mechanobiology has highlighted the importance of physical cues, such as mechanics, geometry (size), topography, and porosity, in the determination of cellular activities and fates, in addition to biochemical factors derived from their surroundings. In this review, we will first provide an overview of how such fundamental insights are identified by synchronizing the hierarchical nature of biological systems and static materials with tunable physical cues. Thereafter, we will explain the photoresponsive dynamic biomaterials to dissect the spatiotemporal aspects of the dependence of biological functions on physical cues.
Collapse
Affiliation(s)
- Jun Nakanishi
- Research Center for Functional Materials, National Institute for Materials Science, Japan. .,Graduate School of Advanced Science and Engineering, Waseda University, Japan.,Graduate School of Advanced Engineering, Tokyo University of Science, Japan
| | - Shota Yamamoto
- Research Center for Functional Materials, National Institute for Materials Science, Japan. .,Graduate School of Arts and Sciences, The University of Tokyo, Japan
| |
Collapse
|
40
|
Vidović T, Ewald CY. Longevity-Promoting Pathways and Transcription Factors Respond to and Control Extracellular Matrix Dynamics During Aging and Disease. FRONTIERS IN AGING 2022; 3:935220. [PMID: 35874275 PMCID: PMC9301135 DOI: 10.3389/fragi.2022.935220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/27/2022] [Indexed: 05/28/2023]
Abstract
Aging is one of the largest risk factors for cancer, type 2 diabetes, osteoarthritis, cardiovascular diseases, and other age-related pathologies. Here, we give a detailed description of the interplay of chronic age-related pathologies with the remodeling of the extracellular matrix during disease development and progression. Longevity-promoting signaling pathways slow or prevent age-related diseases. In particular, we focus on the mTOR signaling pathway, sirtuins, and canonical longevity-promoting transcription factors, such as FOXO, NF-κB, and Nrf2. We extend our analysis using chromatin immunoprecipitation (ChIP) sequencing and transcriptomic data and report that many established and emerging longevity-promoting transcription factors, such as CREB1, FOXO1,3, GATA1,2,3,4, HIF1A, JUN, KLF4, MYC, NFE2L2/Nrf2, RELA/NF-κB, REST, STAT3,5A, and TP53/p53, directly regulate many extracellular matrix genes and remodelers. We propose that modulation of these pathways increases lifespan and protects from age-related diseases in part due to their effects on extracellular matrix remodeling. Therefore, to successfully treat age-related diseases, it is necessary to better understand the connection between extracellular matrix components and longevity pathways.
Collapse
Affiliation(s)
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
41
|
Pasapera AM, Heissler SM, Eto M, Nishimura Y, Fischer RS, Thiam HR, Waterman CM. MARK2 regulates directed cell migration through modulation of myosin II contractility and focal adhesion organization. Curr Biol 2022; 32:2704-2718.e6. [PMID: 35594862 DOI: 10.1016/j.cub.2022.04.088] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/23/2022] [Accepted: 04/28/2022] [Indexed: 12/11/2022]
Abstract
Cancer cell migration during metastasis is mediated by a highly polarized cytoskeleton. MARK2 and its invertebrate homolog Par1B are kinases that regulate the microtubule cytoskeleton to mediate polarization of neurons in mammals and embryos in invertebrates. However, the role of MARK2 in cancer cell migration is unclear. Using osteosarcoma cells, we found that in addition to its known localizations on microtubules and the plasma membrane, MARK2 also associates with the actomyosin cytoskeleton and focal adhesions. Cells depleted of MARK proteins demonstrated that MARK2 promotes phosphorylation of both myosin II and the myosin phosphatase targeting subunit MYPT1 to synergistically drive myosin II contractility and stress fiber formation in cells. Studies with isolated proteins showed that MARK2 directly phosphorylates myosin II regulatory light chain, while its effects on MYPT1 phosphorylation are indirect. Using a mutant lacking the membrane-binding domain, we found that membrane association is required for focal adhesion targeting of MARK2, where it specifically enhances cell protrusion by promoting FAK phosphorylation and formation of focal adhesions oriented in the direction of migration to mediate directionally persistent cell motility. Together, our results define MARK2 as a master regulator of the actomyosin and microtubule cytoskeletal systems and focal adhesions to mediate directional cancer cell migration.
Collapse
Affiliation(s)
- Ana M Pasapera
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, South Drive, Room 4537, MSC 8019, Bethesda, MD 20892, USA
| | - Sarah M Heissler
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, South Drive, Room 4537, MSC 8019, Bethesda, MD 20892, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, 370 W. 9th Avenue, Columbus, OH 43210, USA
| | - Masumi Eto
- Department of Veterinary Medicine, Okayama University of Science, 1-3 Ikoino-oka, Imabari, Ehime 794-8555, Japan
| | - Yukako Nishimura
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, South Drive, Room 4537, MSC 8019, Bethesda, MD 20892, USA; Division of Developmental Physiology, Institute for Genetic Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan
| | - Robert S Fischer
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, South Drive, Room 4537, MSC 8019, Bethesda, MD 20892, USA
| | - Hawa R Thiam
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, South Drive, Room 4537, MSC 8019, Bethesda, MD 20892, USA
| | - Clare M Waterman
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, South Drive, Room 4537, MSC 8019, Bethesda, MD 20892, USA.
| |
Collapse
|
42
|
Kolesnikoff N, Chen CH, Samuel M. Interrelationships between the extracellular matrix and the immune microenvironment that govern epithelial tumour progression. Clin Sci (Lond) 2022; 136:361-377. [PMID: 35260891 PMCID: PMC8907655 DOI: 10.1042/cs20210679] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 12/19/2022]
Abstract
Solid tumours are composed of cancer cells characterised by genetic mutations that underpin the disease, but also contain a suite of genetically normal cells and the extracellular matrix (ECM). These two latter components are constituents of the tumour microenvironment (TME), and are key determinants of tumour biology and thereby the outcomes for patients. The tumour ECM has been the subject of intense research over the past two decades, revealing key biochemical and mechanobiological principles that underpin its role in tumour cell proliferation and survival. However, the ECM also strongly influences the genetically normal immune cells within the microenvironment, regulating not only their proliferation and survival, but also their differentiation and access to tumour cells. Here we review recent advances in our knowledge of how the ECM regulates the tumour immune microenvironment and vice versa, comparing normal skin wound healing to the pathological condition of tumour progression.
Collapse
Affiliation(s)
- Natasha Kolesnikoff
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Chun-Hsien Chen
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
- Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
43
|
Qian K, Zheng XX, Wang C, Huang WG, Liu XB, Xu SD, Liu DK, Liu MY, Lin CS. β-Sitosterol Inhibits Rheumatoid Synovial Angiogenesis Through Suppressing VEGF Signaling Pathway. Front Pharmacol 2022; 12:816477. [PMID: 35295740 PMCID: PMC8918576 DOI: 10.3389/fphar.2021.816477] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Rheumatoid arthritis (RA) is a chronic disabling inflammatory disease that causes synovial angiogenesis in an invasive manner and leads to joint destruction. Currently available pharmacotherapy for RA has unwanted side effects and limitations. Although anti-angiogenic therapy is regarded as a new potential treatment for RA, only a few anti-angiogenic drugs are available. An increasing number of studies have shown that β-sitosterol (BSS) may exert inhibitory effects against angiogenesis. However, the mechanisms involved are still unclear.Methods: Based on the results of the gene set enrichment analysis (GSEA) of the transcriptome data of endothelial cells from RA patients, we evaluated the pharmacological effects of BSS on the tube formation, cell proliferation, and migration of human umbilical vein endothelial cells (HUVECs). Furthermore, the effects of BSS treatment on vascular endothelial growth factor receptor 2 (VEGFR2) were determined using molecular docking and Western blotting. Additionally, in the presence or absence of BSS, synovial angiogenesis and joint destruction of the ankle were investigated in collagen-induced arthritis (CIA) mice. The effect of BSS treatment on VEGFR2/p-VEGFR2 expression was verified through immunohistochemical staining.Results: The immunohistochemistry results revealed that BSS treatment inhibited angiogenesis both in vitro and in vivo. In addition, the results of 5-ethynyl-2′-deoxyuridine and cell cycle analysis showed that BSS treatment suppressed the proliferation of HUVECs, while the Transwell migration and stress fiber assays demonstrated that BSS treatment inhibited the migration of HUVECs. Notably, the inhibitory effect of BSS treatment on VEGFR2/p-VEGFR2 was similar to that of axitinib. In CIA mice, BSS also exerted therapeutic effects on the ankles by reducing the degree of swelling, ameliorating bone and cartilage damage, preventing synovial angiogenesis, and inhibiting VEGFR2 and p-VEGFR2 expression.Conclusion: Therefore, our findings demonstrate that BSS exerts an inhibitory effect on synovial angiogenesis by suppressing the proliferation and migration of endothelial cells, thereby alleviating joint swelling and bone destruction in CIA mice. Furthermore, the underlying therapeutic mechanisms may involve the inhibition of VEGF signaling pathway activation.
Collapse
Affiliation(s)
- Kai Qian
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Postdoctoral Research Station, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Xue-Xia Zheng
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chen Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Xiao-Bao Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shu-Di Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan-Kai Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min-Ying Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Chang-Song Lin, ; Min-Ying Liu,
| | - Chang-Song Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Chang-Song Lin, ; Min-Ying Liu,
| |
Collapse
|
44
|
Physically-based structural modeling of a typical regenerative tissue analog bridges material macroscale continuum and cellular microscale discreteness and elucidates the hierarchical characteristics of cell-matrix interaction. J Mech Behav Biomed Mater 2021; 126:104956. [PMID: 34930707 DOI: 10.1016/j.jmbbm.2021.104956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/22/2021] [Accepted: 11/01/2021] [Indexed: 11/20/2022]
Abstract
This paper presents a comprehensive physically-based structural modelling for the passive and active biomechanical processes in a typical engineered tissue - namely, cell-compacted collagen gel. First, it introduces a sinusoidal curve analog for quantifying the mechanical response of the collagen fibrils and a probability distribution function of the characteristic crimp ratio for taking into account the fibrillar geometric entropic effect. The constitutive framework based on these structural characteristics precisely reproduces the nonlinearity, the viscoelasticity, and fairly captures the Poisson effect exhibiting in the macroscale tensile tests; which, therefore, substantially validates the structural modelling for the analysis of the cell-gel interaction during collagen gel compaction. Second, a deterministic molecular clutch model specific to the interaction between the cell pseudopodium and the collagen network is developed, which emphasizes the dependence of traction force on clutch number altering with the retrograde flow velocity, actin polymeric velocity, and the deformation of the stretched fibril. The modelling reveals the hierarchical features of cellular substrate sensing, i.e. a biphasic traction force response to substrate elasticity begins at the level of individual fibrils and develops into the second biphasic sensing by means of the fibrillar number integration at the whole-cell level. Singular in crossing the realms of continuum and discrete mechanics, the methodologies developed in this study for modelling the filamentous materials and cell-fibril interaction deliver deep insight into the temporospatially dynamic 3D cell-matrix interaction, and are able to bridge the cellular microscale and material macroscale in the exploration of related topics in mechanobiology.
Collapse
|
45
|
QnAs with Clare M. Waterman. Proc Natl Acad Sci U S A 2021; 118:2107569118. [PMID: 34083446 DOI: 10.1073/pnas.2107569118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|