1
|
Asadi Tokmedash M, Kim C, Chavda AP, Li A, Robins J, Min J. Engineering multifunctional surface topography to regulate multiple biological responses. Biomaterials 2025; 319:123136. [PMID: 39978049 PMCID: PMC11893264 DOI: 10.1016/j.biomaterials.2025.123136] [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: 09/27/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Surface topography or curvature plays a crucial role in regulating cell behavior, influencing processes such as adhesion, proliferation, and gene expression. Recent advancements in nano- and micro-fabrication techniques have enabled the development of biomimetic systems that mimic native extracellular matrix (ECM) structures, providing new insights into cell-adhesion mechanisms, mechanotransduction, and cell-environment interactions. This review examines the diverse applications of engineered topographies across multiple domains, including antibacterial surfaces, immunomodulatory devices, tissue engineering scaffolds, and cancer therapies. It highlights how nanoscale features like nanopillars and nanospikes exhibit bactericidal properties, while many microscale patterns can direct stem cell differentiation and modulate immune cell responses. Furthermore, we discuss the interdisciplinary use of topography for combined applications, such as the simultaneous regulation of immune and tissue cells in 2D and 3D environments. Despite significant advances, key knowledge gaps remain, particularly regarding the effects of topographical cues on multicellular interactions and dynamic 3D contexts. This review summarizes current fabrication methods, explores specific and interdisciplinary applications, and proposes future research directions to enhance the design and utility of topographically patterned biomaterials in clinical and experimental settings.
Collapse
Affiliation(s)
| | - Changheon Kim
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ajay P Chavda
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Adrian Li
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jacob Robins
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jouha Min
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
2
|
Li X, Zhao H, Jiang E, Liu P, Chen Y, Wang Y, Li J, Wu Y, Liu Z, Shang Z. ITGB1/FERMT1 mechanoactivation enhances CD44 characteristic stemness in oral squamous cell carcinoma via ubiquitin-dependent CK1α degradation. Oncogene 2025; 44:1530-1544. [PMID: 40044983 DOI: 10.1038/s41388-025-03317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/30/2025] [Accepted: 02/18/2025] [Indexed: 05/15/2025]
Abstract
Cancer stem cells (CSCs) contribute to chemotherapy resistance and poor prognosis, posing significant challenges in the treatment of oral squamous cell carcinoma. The extracellular matrix (ECM)-constructed microenvironment remodels the niche of CSCs. Yet mechanisms by which biophysical properties of ECM relate to CSCs remain undefined. Here, our findings link ECM mechanical stimuli to CSCs phenotype transition, and propose that ECM stiffening mechanoactivates tumor cells to dedifferentiate and acquire CD44+ stem cell-like characteristics through noncanonical mechanotransduction. ITGB1 senses and transduces biomechanical signals, while FERMT1 acts as an intracellular mechanotransduction downstream, activating CSCs. Mechanistically, FERMT1 promotes the proteasomal degradation of CK1α by E3 ubiquitin ligase MIB1, thereby triggering Wnt signaling pathway. Combining targeted ECM softening with mechanotransduction inhibition strategy significantly attenuates tumor stemness and chemoresistance in vivo. Therefore, our findings highlight the role of ECM in regulating CSCs via biomechanical-dependent manner, suggesting the ECM/ITGB1/FERMT1/Wnt axis as a promising therapeutic target for CSCs therapy.
Collapse
Affiliation(s)
- Xiang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Erhui Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Pan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ji Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yufei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhenan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Wu KP, Yan ZJ, Zhuang XX, Hua JL, Li MX, Huang K, Qi YX. Dynamic structure and function of nuclear pore protein complex: Potential roles of lipid and lamins regulated nuclear membrane curvature. Int J Biol Macromol 2025:144104. [PMID: 40350114 DOI: 10.1016/j.ijbiomac.2025.144104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
The nuclear pore complex (NPC), a massive and highly sophisticated protein assembly, forms a channel embedded in the nuclear envelope (NE) of eukaryotic cells. As a critical gateway, the NPC mediates the bidirectional transport of macromolecules between the cytoplasm and the nucleus. Here, we overview the structure and transport function of this protein complex, and highlight the selective barrier model of NPC transport functional modules. Nuclear membrane curvature (NMC) is a critical parameter for quantifying nuclear deformation. We discuss the mechanism by which NMC regulates dynamic NPC structure, function and distribution. Furthermore we highlight the role of two key factors, i.e. lipid composition and lamins distribution, in NPC dynamics while elucidating their regulatory mechanisms. The investigations on the dynamic structure and function of NPC modulated by NMC provide a new avenue for understanding the role of NPC in different pathological conditions. This knowledge could contribute to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Kun-Peng Wu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, 200240 Shanghai, China
| | - Zhi-Jie Yan
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, 200240 Shanghai, China
| | - Xiao-Xi Zhuang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, 200240 Shanghai, China
| | - Jin-Liang Hua
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, 200240 Shanghai, China
| | - Meng-Xiao Li
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, 200240 Shanghai, China
| | - Kai Huang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, 200240 Shanghai, China.
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, 200240 Shanghai, China.
| |
Collapse
|
4
|
Hilai K, Grubich D, Akrawi M, Zhu H, Zaghloul R, Shi C, Do M, Zhu D, Zhang J. Mechanical Evolution of Metastatic Cancer Cells in 3D Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2403242. [PMID: 40116569 PMCID: PMC12051740 DOI: 10.1002/smll.202403242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 01/01/2025] [Indexed: 03/23/2025]
Abstract
Cellular biomechanics plays a critical role in cancer metastasis and tumor progression. Existing studies on cancer cell biomechanics are mostly conducted in flat 2D conditions, where cells' behavior can differ considerably from those in 3D physiological environments. Despite great advances in developing 3D in vitro models, probing cellular elasticity in 3D conditions remains a major challenge for existing technologies. In this work, optical Brillouin microscopy is utilized to longitudinally acquire mechanical images of growing cancerous spheroids over the period of 8 days. The dense mechanical mapping from Brillouin microscopy enables us to extract spatially resolved and temporally evolving mechanical features that were previously inaccessible. Using an established machine learning algorithm, it is demonstrated that incorporating these extracted mechanical features significantly improves the classification accuracy of cancer cells, from 74% to 95%. Building on this finding, a deep learning pipeline capable of accurately differentiating cancerous spheroids from normal ones solely using Brillouin images have been developed, suggesting the mechanical features of cancer cells can potentially serve as a new biomarker in cancer classification and detection.
Collapse
Affiliation(s)
- Karlin Hilai
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Daniil Grubich
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Marcus Akrawi
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
- Department of Physics and AstronomyWayne State UniversityDetroitMI48201USA
| | - Hui Zhu
- Department of Computer ScienceWayne State UniversityDetroitMI48202USA
| | - Razanne Zaghloul
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Chenjun Shi
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
- Department of Biomedical EngineeringInstitute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48823USA
| | - Man Do
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Dongxiao Zhu
- Department of Computer ScienceWayne State UniversityDetroitMI48202USA
| | - Jitao Zhang
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
- Department of Biomedical EngineeringInstitute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48823USA
| |
Collapse
|
5
|
Tuieng RJ, Disney C, Cartmell SH, Kirwan CC, Eckersley A, Newham E, Gupta HS, Hoyland JA, Lee PD, Sherratt MJ. Impact of therapeutic X-ray exposure on collagen I and associated proteins. Acta Biomater 2025; 197:294-311. [PMID: 40058620 DOI: 10.1016/j.actbio.2025.03.004] [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: 05/18/2024] [Revised: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 03/23/2025]
Abstract
Biological tissues are exposed to X-rays in medical applications (such as diagnosis and radiotherapy) and in research studies (for example microcomputed X-ray tomography: microCT). Radiotherapy may deliver doses up to 50Gy to both tumour and healthy tissues, resulting in undesirable clinical side effects which can compromise quality of life. Whilst cellular responses to X-rays are relatively well-characterised, X-ray-induced structural damage to the extracellular matrix (ECM) is poorly understood. This study tests the hypotheses that ECM proteins and ECM-rich tissues (purified collagen I and rat tail tendons respectively) are structurally compromised by exposure to X-ray doses used in breast radiotherapy. Protein gel electrophoresis demonstrated that breast radiotherapy equivalent doses can induce fragmentation of the constituent α chains in solubilised purified collagen I. However, assembly into fibrils, either in vitro or in vivo, prevented X-ray-induced fragmentation but not structural changes (as characterised by LC-MS/MS and peptide location fingerprinting: PLF). In subsequent experiments exposure to higher (synchrotron) X-ray doses induced substantial fragmentation of solubilised and fibrillar (chicken tendon) collagen I. LC-MS/MS and PLF analysis of synchrotron-irradiated tendon identified structure-associated changes in collagens I, VI, XII, proteoglycans including aggrecan, decorin, and fibromodulin, and the elastic fibre component fibulin-1. Thus, exposure to radiotherapy X-rays can affect the structure of key tissue ECM components, although additional studies will be required to understand dose dependent effects. STATEMENT OF SIGNIFICANCE: Biological systems are routinely exposed to X-rays during medical treatments (radiotherapy) and in imaging studies (microCT). Whilst the impact of ionising radiation on cells is well characterised, the interactions between X-rays and the extracellular matrix are not. Here, we show that relatively low dose breast radiotherapy X-rays are sufficient to affect the structure of collagen I in both its solubilised and fibrillar forms. Although the impact of intermediate X-ray doses on extracellular proteins was not determined, the high dose exposures which are achievable using a synchrotron source had an even greater effect on the structure of collagen I molecules and, in tendon, on the structures of many accessory extracellular matrix proteins, The unwanted side effects of radiotherapy may therefore be due to not only cellular damage but also damage to the surrounding matrix.
Collapse
Affiliation(s)
- Ren Jie Tuieng
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK; Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 118415
| | - Catherine Disney
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK; Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Sarah H Cartmell
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering and The Henry Royce Institute, Royce Hub Building, The University of Manchester, M13 9PL, Manchester, UK
| | - Cliona C Kirwan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oglesby Cancer Research Building, Manchester Cancer Research Centre, Wilmslow Road, Manchester M20 4BX, UK; The Nightingale Breast Cancer Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, M23 9LT, Manchester, UK
| | - Alexander Eckersley
- Manchester Cell-Matrix Centre, Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK
| | - Elis Newham
- School of Engineering and Materials Science & Institute of Bioengineering, Queen Mary University of London, London E1 4NS, UK; Section Palaeontology, Institute of Geosciences, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Himadri S Gupta
- School of Engineering and Materials Science & Institute of Bioengineering, Queen Mary University of London, London E1 4NS, UK
| | - Judith A Hoyland
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK
| | - Peter D Lee
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - Michael J Sherratt
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK.
| |
Collapse
|
6
|
Liu T, Liang H, Li Y, Liao W, Deng J, Zhang L, Shen F. The increased matrix stiffness caused by LOXL2 activates Piezo1 channels to promote the migration and invasion of cervical cancer cells. Discov Oncol 2025; 16:644. [PMID: 40304808 PMCID: PMC12044143 DOI: 10.1007/s12672-025-02456-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/21/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Lymph node metastasis in cervical cancer (CC) is a significant contributor to mortality associated with this disease. Notably, CC with lymph node metastasis exhibits greater stiffness upon palpation compared to CC without such metastasis. Lysyl oxidase-like 2 (LOXL2), a member of the lysyl oxidase (LOX) family, is capable of catalyzing the crosslinking of extracellular matrix (ECM) components. Additionally, Piezo1 is a mechanosensitive ion channel protein in mammals that can detect mechanical stimuli and regulate cellular behavior, making it a critical protein in tumor development. This prompted us to explore the relationship between the progression of CC and the roles of Piezo1 from a biomechanical perspective. METHODS Young's modulus of tissue was measured by atomic force microscope (AFM). The collagen coated polyacrylamide hydrogel (PA gel) system was prepared to mimic the soft and stiff substrates in vitro. The efficacy of Piezo1 was evaluated in vitro using transwell assay, immunofluorescence, and western blot analysis. Experiments in vivo have also confirmed the effect of matrix hardness on CC progression and on Piezo1. RESULTS We quantitatively confirmed that CC with lymph node metastases was more rigid and more abundant in connective tissue proliferation than CC without lymph node metastases, and further demonstrated that stromal stiffness significantly modulated CC progression. Remarkably, Piezo1 has been identified as a potent mechanosensitive gene capable of responding to environmental stiffness, thereby mediating stiffness-regulated CC progression through the regulation of the Piezo1 channel protein. In vivo, the LOXL2 inhibitor not only effectively inhibited the growth of tumors in vivo, but also reduced the expression of Piezo1 in tumors by reducing the matrix stiffness. CONCLUSION These data suggest that targeting extracellular matrix (ECM) stiffness may hinder the progress of CC. Notably, targeting Piezo1 may offer promising clinical value for CC therapy.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 239 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Hua Liang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 239 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Yang Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 239 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Wenxin Liao
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 239 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Juexiao Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 239 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Lanyue Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 239 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Fujin Shen
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 239 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| |
Collapse
|
7
|
Finger AM, Hendley AM, Figueroa D, Gonzalez H, Weaver VM. Tissue mechanics in tumor heterogeneity and aggression. Trends Cancer 2025:S2405-8033(25)00096-2. [PMID: 40307158 DOI: 10.1016/j.trecan.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/10/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Tumorigenesis ensues within a heterogeneous tissue microenvironment that promotes malignant transformation, metastasis and treatment resistance. A major feature of the tumor microenvironment is the heterogeneous population of cancer-associated fibroblasts and myeloid cells that stiffen the extracellular matrix. The heterogeneously stiffened extracellular matrix in turn activates cellular mechanotransduction and creates a hypoxic and metabolically hostile microenvironment. The stiffened extracellular matrix and elevated mechanosignaling also drive tumor aggression by fostering tumor cell growth, survival, and invasion, compromising antitumor immunity, expanding cancer stem cell frequency, and increasing mutational burden, which promote intratumor heterogeneity. Delineating the molecular mechanisms whereby tissue mechanics regulate these phenotypes should help to clarify the basis for tumor heterogeneity and cancer aggression and identify novel therapeutic targets that could improve patient outcome. Here, we discuss the role of the extracellular matrix in driving cancer aggression through its impact on tumor heterogeneity.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Malov, Denmark
| | - Audrey Marie Hendley
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143
| | - Diego Figueroa
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Hugo Gonzalez
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | - Valerie Marie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143; Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Tseng PL, Sun W, Salem A, Alaklobie M, Macfarlane SC, Gad AK, Collins MO, Erdmann KS. Mechanical control of the alternative splicing factor PTBP1 regulates extracellular matrix stiffness induced proliferation and cell spreading. iScience 2025; 28:112273. [PMID: 40241749 PMCID: PMC12002664 DOI: 10.1016/j.isci.2025.112273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/23/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Cells sense mechanical cues and convert them into biochemical responses to regulate biological processes such as embryonic development, aging, cellular homeostasis, and disease progression. In this study, we introduce a large-scale, systematic approach to identify proteins with mechanosensitive nuclear localization, highlighting their potential roles in mechanotransduction. Among the proteins identified, we focus here on the splicing factor PTBP1. We demonstrate that its nuclear abundance is regulated by mechanical cues such as cell density, size, and extracellular matrix (ECM) stiffness and that PTBP1 medicates the mechanosensitive alternative splicing of the endocytic adapter protein Numb. Furthermore, we show that PTBP1 and Numb alternative splicing is critical for ECM stiffness-induced epithelial cell spreading and proliferation as well as for mesenchymal stem cell differentiation into osteoblasts on a stiff matrix. Our results underscore the emerging role of alternative splicing in mechanotransduction and provide novel mechanistic insights into how matrix stiffness modulates cellular mechanoresponses.
Collapse
Affiliation(s)
- Pei-Li Tseng
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Weiwei Sun
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Ahmed Salem
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- Department of Biomedical Laboratory Science, Sheba University, Sheba, Libya
| | - Mubarak Alaklobie
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 67714, Saudi Arabia
| | - Sarah C. Macfarlane
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield S10 2TN, UK
| | - Annica K.B. Gad
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield S10 2TN, UK
- Department of Oncology-Pathology, Karolinska Institutet, Anna Steckséns gata 30A, 171 64 Solna, Sweden
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| | - Mark O. Collins
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- biOMICS Mass Spectrometry Facility, University of Sheffield, Sheffield S10 2TN, UK
| | - Kai S. Erdmann
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
9
|
Mierke CT. Softness or Stiffness What Contributes to Cancer and Cancer Metastasis? Cells 2025; 14:584. [PMID: 40277910 PMCID: PMC12026216 DOI: 10.3390/cells14080584] [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: 03/11/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Beyond the genomic and proteomic analysis of bulk and single cancer cells, a new focus of cancer research is emerging that is based on the mechanical analysis of cancer cells. Therefore, several biophysical techniques have been developed and adapted. The characterization of cancer cells, like human cancer cell lines, started with their mechanical characterization at mostly a single timepoint. A universal hypothesis has been proposed that cancer cells need to be softer to migrate and invade tissues and subsequently metastasize in targeted organs. Thus, the softness of cancer cells has been suggested to serve as a universal physical marker for the malignancy of cancer types. However, it has turned out that there exists the opposite phenomenon, namely that stiffer cancer cells are more migratory and invasive and therefore lead to more metastases. These contradictory results question the universality of the role of softness of cancer cells in the malignant progression of cancers. Another problem is that the various biophysical techniques used can affect the mechanical properties of cancer cells, making it even more difficult to compare the results of different studies. Apart from the instrumentation, the culture and measurement conditions of the cancer cells can influence the mechanical measurements. The review highlights the main advances of the mechanical characterization of cancer cells, discusses the strength and weaknesses of the approaches, and questions whether the passive mechanical characterization of cancer cells is still state-of-the art. Besides the cell models, conditions and biophysical setups, the role of the microenvironment on the mechanical characteristics of cancer cells is presented and debated. Finally, combinatorial approaches to determine the malignant potential of tumors, such as the involvement of the ECM, the cells in a homogeneous or heterogeneous association, or biological multi-omics analyses, together with the dynamic-mechanical analysis of cancer cells, are highlighted as new frontiers of research.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Sciences, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
10
|
Zhang M, Zhang B. Extracellular matrix stiffness: mechanisms in tumor progression and therapeutic potential in cancer. Exp Hematol Oncol 2025; 14:54. [PMID: 40211368 PMCID: PMC11984264 DOI: 10.1186/s40164-025-00647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
Tumor microenvironment (TME) is a complex ecosystem composed of both cellular and non-cellular components that surround tumor tissue. The extracellular matrix (ECM) is a key component of the TME, performing multiple essential functions by providing mechanical support, shaping the TME, regulating metabolism and signaling, and modulating immune responses, all of which profoundly influence cell behavior. The quantity and cross-linking status of stromal components are primary determinants of tissue stiffness. During tumor development, ECM stiffness not only serves as a barrier to hinder drug delivery but also promotes cancer progression by inducing mechanical stimulation that activates cell membrane receptors and mechanical sensors. Thus, a comprehensive understanding of how ECM stiffness regulates tumor progression is crucial for identifying potential therapeutic targets for cancer. This review examines the effects of ECM stiffness on tumor progression, encompassing proliferation, migration, metastasis, drug resistance, angiogenesis, epithelial-mesenchymal transition (EMT), immune evasion, stemness, metabolic reprogramming, and genomic stability. Finally, we explore therapeutic strategies that target ECM stiffness and their implications for tumor progression.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China
| | - Bin Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China.
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.
| |
Collapse
|
11
|
Botticelli M, Metzcar J, Phillips T, Cox S, Keshavanarayana P, Spill F. A hybrid computational model of cancer spheroid growth with ribose-induced collagen stiffening. Front Bioeng Biotechnol 2025; 13:1515962. [PMID: 40271351 PMCID: PMC12014586 DOI: 10.3389/fbioe.2025.1515962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025] Open
Abstract
Metastasis, the leading cause of death in cancer patients, arises when cancer cells disseminate from a primary solid tumour to distant organs. Growth and invasion of the solid tumour often involve collective cell migration, which is profoundly influenced by cell-cell interactions and the extracellular matrix (ECM). The ECM's biochemical composition and mechanical properties, such as stiffness, regulate cancer cell behaviour and migration dynamics. Mathematical modelling serves as a pivotal tool for studying and predicting these complex dynamics, with hybrid discrete-continuous models offering a powerful approach by combining agent-based representations of cells with continuum descriptions of the surrounding microenvironment. In this study, we investigate the impact of ECM stiffness, modulated via ribose-induced collagen cross-linking, on cancer spheroid growth and invasion. We employed a hybrid discrete-continuous model implemented in PhysiCell to simulate spheroid dynamics, successfully replicating three-dimensional in vitro experiments. The model incorporates detailed representations of cell-cell and cell-ECM interactions, ECM remodelling, and cell proliferation. Our simulations align with experimental observations of two breast cancer cell lines, non-invasive MCF7 and invasive HCC 1954, under varying ECM stiffness conditions. The results demonstrate that increased ECM stiffness due to ribose-induced cross-linking inhibits spheroid invasion in invasive cells, whereas non-invasive cells remain largely unaffected. Furthermore, our simulations show that higher ECM degradation by the cells not only enables spheroid growth and invasion but also facilitates the formation of multicellular protrusions. Conversely, increasing the maximum speed that cells can reach due to cell-ECM interactions enhances spheroid growth while promoting single-cell invasion. This hybrid modelling approach enhances our understanding of the interplay between cancer cell migration, proliferation, and ECM mechanical properties, paving the way for future studies incorporating additional ECM characteristics and microenvironmental conditions.
Collapse
Affiliation(s)
- Margherita Botticelli
- School of Mathematics, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - John Metzcar
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
| | - Thomas Phillips
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| | - Susan Cox
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| | - Pradeep Keshavanarayana
- School of Mathematics, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Computational Medicine, University College London, London, United Kingdom
| | - Fabian Spill
- School of Mathematics, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
12
|
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N. Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment. Cell Syst 2025:101261. [PMID: 40220761 DOI: 10.1016/j.cels.2025.101261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Daniel León-Periñán
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivano Legnini
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Human Technopole, Milan, Italy
| | - Ilan Theurillat
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anastasiya Boltengagen
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | - Jose Nimo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | | | - Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany; Aignostics GmbH, Berlin, Germany; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | | | | | | - Yan Liang
- NanoString® Technologies, Inc, Seattle, WA, USA
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany; Institute of Pathology, Ludwig Maximilians Universität, Munich, Germany
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany; NeuroCure Cluster of Excellence, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; National Center for Tumor Diseases (NCT), Site Berlin, Berlin, Germany.
| |
Collapse
|
13
|
Heiss J, Treacher N, Lamichhane A, Magar AR, Tavana H. Extracellular Fibronectin and Hyaluronic Acid Effects on Tumorigenic Functions of Triple-Negative Breast Cancer Cells. J Biomed Mater Res A 2025; 113:e37906. [PMID: 40192931 DOI: 10.1002/jbm.a.37906] [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: 10/22/2024] [Revised: 02/21/2025] [Accepted: 03/17/2025] [Indexed: 05/17/2025]
Abstract
The extracellular matrix (ECM) in solid tumors provides structural support and signaling cues to cancer cells. Altered ECM in tumors promotes local invasion of cancer cells, a key step toward metastasis. Engineered tumor models that are used to study cancer invasion often focus on the effects of an individual ECM molecule on specific functions of cancer cells. However, how different components of ECM in a complex tumor model may co-regulate cancer invasion and the underlying signaling pathways is understudied. We developed a 3D tumor model of triple-negative breast cancer (TNBC) to study the effects of fibronectin and hyaluronic acid, alone or in combination, on TNBC cell invasion of collagen-based hydrogels. Our focus on these molecules was due to their significance in breast tumors, disease progression, and association with worse outcomes in patients. Results showed that fibronectin and hyaluronic acid significantly increase collagen invasion of TNBC cells and oncogenic signaling but not in combination, potentially due to differences in the microstructure of the hydrogels. Fibronectin and hyaluronic acid in composite hydrogels also promoted drug resistance and cancer stemness. This study demonstrated the utility of a 3D tumor model for functional and mechanistic studies to define complex effects of ECM in solid cancers.
Collapse
Affiliation(s)
- Jacob Heiss
- Department of Biomedical Engineering, University of Akron, Akron, Ohio, USA
| | - Nina Treacher
- Department of Biomedical Engineering, University of Akron, Akron, Ohio, USA
| | - Astha Lamichhane
- Department of Biomedical Engineering, University of Akron, Akron, Ohio, USA
| | - Anju Rana Magar
- Department of Biomedical Engineering, University of Akron, Akron, Ohio, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, University of Akron, Akron, Ohio, USA
| |
Collapse
|
14
|
Wei X, Ge Y, Zheng Y, Zhao S, Zhou Y, Chang Y, Wang N, Wang X, Zhang J, Zhang X, Hu L, Tan Y, Jia Q. Hybrid EMT Phenotype and Cell Membrane Tension Promote Colorectal Cancer Resistance to Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413882. [PMID: 39985376 PMCID: PMC12005738 DOI: 10.1002/advs.202413882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/26/2025] [Indexed: 02/24/2025]
Abstract
Intratumoral heterogeneity, including epithelial-mesenchymal transition (EMT), is one major cause of therapeutic resistance. The induction of ferroptosis, an iron-dependent death, has the potential in overcoming this resistance to traditional treatment modalities. However, the roles of distinct EMT phenotypes in ferroptosis remain an enigma. This study reports that 3D soft fibrin microenvironment confers colorectal cancer (CRC) cells hybrid EMT phenotype and high level of resistance to ferroptosis. The activation of histone acetylation and WNT/β-catenin signaling drives this EMT phenotypic transition, which promotes the defense of 3D CRCs against ferroptosis via glutathione peroxidases/ferritin signaling axis. Unexpectedly, E-cadherin knockout in 3D but not 2D CRCs mediates an integrin β3 marked-late hybrid EMT state and further enhances the resistance to ferroptosis via integrin-mediated tension and mitochondrial reprogramming. The inhibition of integrin αvβ3-mediated tension and WNT/β-catenin-mediated hybrid EMT sensitizes 3D CRCs with and without E-cadherin deficiency to ferroptosis in vivo, respectively. Further, the EMT phenotype of patient-derived tumoroids is associated with CRC therapeutic resistance. In summary, this study uncovers previously unappreciated roles of hybrid EMT and cell membrane tension in ferroptosis, which not only predict the treatment efficacy but also potentiate the development of new ferroptosis-based targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xiaowei Wei
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yutong Ge
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Yaolin Zheng
- Department of RespiratoryCritical Care and Sleep MedicineXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamen361102China
| | - Sunyan Zhao
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yuhan Zhou
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yuhan Chang
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Nuofan Wang
- School of MedicineSoutheast UniversityNanjing210009China
| | - Xiumei Wang
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Juan Zhang
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Xuanchang Zhang
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Liqiao Hu
- Guangzhou National LaboratoryGuangzhou510005China
| | - Youhua Tan
- The Hong Kong Polytechnic UniversityShenzhen Research InstituteShenzhen518000China
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Qiong Jia
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| |
Collapse
|
15
|
Sun X, Hu X. Unveiling Matrix Metalloproteinase 13's Dynamic Role in Breast Cancer: A Link to Physical Changes and Prognostic Modulation. Int J Mol Sci 2025; 26:3083. [PMID: 40243781 PMCID: PMC11988641 DOI: 10.3390/ijms26073083] [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: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The biomechanical properties of the extracellular matrix (ECM) including its stiffness, viscoelasticity, collagen architecture, and temperature constitute critical biomechanical cues governing breast cancer progression. Matrix metalloproteinase 13 (MMP13) is an important marker of breast cancer and plays important roles in matrix remodelling and cell metastasis. Emerging evidence highlights MMP13 as a dynamic modulator of the ECM's physical characteristics through dual mechanoregulatory mechanisms. While MMP13-mediated collagen degradation facilitates microenvironmental softening, thus promoting tumour cell invasion, paradoxically, its crosstalk with cancer-associated fibroblasts (CAFs) and tumour-associated macrophages (TAMs) drives pathological stromal stiffening via aberrant matrix deposition and crosslinking. This biomechanical duality is amplified through feedforward loops with an epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) populations, mediated by signalling axes such as TGF-β/Runx2. Intriguingly, MMP13 exhibits context-dependent mechanomodulatory effects, demonstrating anti-fibrotic activity and inhibiting the metastasis of breast cancer. At the same time, angiogenesis and increased metabolism are important mechanisms through which MMP13 promotes a temperature increase in breast cancer. Targeting the spatiotemporal regulation of MMP13's mechanobiological functions may offer novel therapeutic strategies for disrupting the tumour-stroma vicious cycle.
Collapse
Affiliation(s)
- Xiaomeng Sun
- Queen Mary School, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China;
| | - Xiaojuan Hu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China;
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China
| |
Collapse
|
16
|
Ishihara S, Enomoto A, Sakai A, Iida T, Tange S, Kioka N, Nukuda A, Nagasato AI, Yasuda M, Tokino T, Haga H. Stiff extracellular matrix activates the transcription factor ATF5 to promote the proliferation of cancer cells. iScience 2025; 28:112057. [PMID: 40124511 PMCID: PMC11928855 DOI: 10.1016/j.isci.2025.112057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/20/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer tissues are stiffer than normal tissues. Carcinogenesis stiffens the extracellular matrix (ECM) of cancerous tissues, to which cancer cells respond by activating transcription factors, such as YAP/TAZ, Twist1, and β-catenin, which further elevate their malignancy. However, these transcription factors are also expressed in normal tissues. Therefore, inhibiting these factors in order to treat cancer may lead to severe side effects. Here, we show that activating transcription factor 5 (ATF5), highly expressed in tumors, is activated by ECM stiffness and promotes the proliferation of cancer cells, including that of pancreatic cancer cells and lung cancer cells. In addition, ATF5 suppressed the expression of early growth response 1 (EGR1), thereby accelerating cancer cell proliferation. Stiff ECMs trigger the JAK-MYC pathway which activates ATF5. JAK activation was actomyosin independent whereas MYC induction was actomyosin dependent. These results demonstrate the critical role played by ATF5 in the mechanotransduction process seen in cancers.
Collapse
Affiliation(s)
- Seiichiro Ishihara
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Akihiro Sakai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550 Japan
| | - Shoichiro Tange
- Department of Medical Genome Sciences, Cancer Research Institute, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Akihiro Nukuda
- Transdisciplinary Life Science Course, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810 Japan
| | - Ayaka Ichikawa Nagasato
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Motoaki Yasuda
- Department of Oral Molecular Microbiology, Division of Oral Pathobiological Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060-8586, Japan
| | - Takashi Tokino
- Department of Medical Genome Sciences, Cancer Research Institute, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Hisashi Haga
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| |
Collapse
|
17
|
Marohl T, Atkins KA, Wang L, Janes KA. PCSK5 M452I is a recessive hypomorph exclusive to MCF10DCIS.com cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.641323. [PMID: 40093128 PMCID: PMC11908202 DOI: 10.1101/2025.03.03.641323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The most widely used cell line for studying ductal carcinoma in situ (DCIS) premalignancy is the transformed breast epithelial cell line, MCF10DCIS.com. During its original clonal isolation and selection, MCF10DCIS.com acquired a heterozygous M452I mutation in the proprotein convertase PCSK5, which has never been reported in any human cancer. The mutation is noteworthy because PCSK5 matures GDF11, a TGFβ-superfamily ligand that suppresses progression of triple-negative breast cancer. We asked here whether PCSK5M452I and its activity toward GDF11 might contribute to the unique properties of MCF10DCIS.com. Using an optimized in-cell GDF11 maturation assay, we found that overexpressed PCSK5M452I was measurably active but at a fraction of the wildtype enzyme. In a PCSK5 -/- clone of MCF10DCIS.com reconstituted with different PCSK5 alleles, PCSK5M452I was mildly defective in anterograde transport. However, the multicellular organization of PCSK5M452I addback cells in 3D matrigel cultures was significantly less compact than wildtype and indistinguishable from a PCSK5T288P null allele. Growth of intraductal MCF10DCIS.com xenografts was similarly impaired along with the frequency of comedo necrosis and stromal activation. In no setting did PCSK5M452I exhibit gain-of-function activity, leading us to conclude that it is hypomorphic and thus compensated by the remaining wildtype allele in MCF10DCIS.com. Implications This work reassures that an exotic PCSK5 mutation is not responsible for the salient characteristics of the MCF10DCIS.com cell line.
Collapse
Affiliation(s)
- Taylor Marohl
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Kristen A. Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
18
|
Baude JA, Li MD, Jackson SM, Sharma A, Walter DI, Stowers RS. Engineered basement membrane mimetic hydrogels to study mammary epithelial morphogenesis and invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640825. [PMID: 40093166 PMCID: PMC11908212 DOI: 10.1101/2025.02.28.640825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Reconstituted basement membrane (rBM) products like Matrigel are widely used in 3D culture models of epithelial tissues and cancer. However, their utility is hindered by key limitations, including batch variability, xenogenic contaminants, and a lack of tunability. To address these challenges, we engineered a 3D basement membrane (eBM) matrix by conjugating defined extracellular matrix (ECM) adhesion peptides (IKVAV, YIGSR, RGD) to an alginate hydrogel network with precisely tunable stiffness and viscoelasticity. We optimized the mechanical and biochemical properties of the engineered basement membranes (eBMs) to support mammary acinar morphogenesis in MCF10A cells, similar to rBM. We found that IKVAV-modified, fast-relaxing (τ1/2 = 30-150 s), and soft (E = 200 Pa) eBMs best promoted polarized acinar structures. Clusters became invasive and lost polarity only when the IKVAV-modified eBM exhibited both similar stiffness to a malignant breast tumor (E = 4000 Pa) and slow stress relaxation (τ1/2 = 600-1100 s). Notably, tumor-like stiffness alone was not sufficient to drive invasion in fast stress relaxing matrices modified with IKVAV. In contrast, RGD-modified matrices promoted a malignant phenotype regardless of mechanical properties. We also utilized this system to interrogate the mechanism driving acinar and tumorigenic phenotypes in response to microenvironmental parameters. A balance in activity between β1- and β4-integrins was observed in the context of IKVAV-modified eBMs, prompting further investigation into the downstream mechanisms. We found differences in hemidesmosome formation and production of endogenous laminin in response to peptide type, stress relaxation, and stiffness. We also saw that inhibiting either focal adhesion kinase or hemidesmosome signaling in IKVAV eBMs prevented acinus formation. This eBM matrix is a powerful, reductionist, xenogenic-free system, offering a robust platform for both fundamental research and translational applications in tissue engineering and disease modeling.
Collapse
Affiliation(s)
- Jane A Baude
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
| | - Megan D Li
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
| | - Sabrina M Jackson
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
| | - Abhishek Sharma
- University of California, Santa Barbara, Department of Mechanical Engineering
| | - Daniella I Walter
- University of California, Santa Barbara, Department of Mechanical Engineering
| | - Ryan S Stowers
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
- University of California, Santa Barbara, Department of Mechanical Engineering
- University of California, Santa Barbara, Department of Bioengineering
| |
Collapse
|
19
|
Shang C, Zhang J, Huang Y. Prediction of axillary lymph node metastasis in T1 breast cancer using diffuse optical tomography, strain elastography and molecular markers. Quant Imaging Med Surg 2025; 15:2162-2174. [PMID: 40160647 PMCID: PMC11948403 DOI: 10.21037/qims-24-1664] [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: 08/11/2024] [Accepted: 02/04/2025] [Indexed: 04/02/2025]
Abstract
Background Early-stage breast cancer (BC) presents a certain risk of axillary lymph node (ALN) metastasis (ALNM), leading to different individualized treatment. Preoperative non-invasive prediction to determine ALN status is of great significance for avoiding ineffective axillary surgery. Tumor total hemoglobin concentration (TTHC), strain ratio (SR), and Ki-67 expression are associated with ALNM in BC, but few studies have focused on T1 BC. This study aimed to explore the usefulness of these factors individually and in combination for the preoperative prediction of ALNM in T1 BC. Methods This was a cross-sectional study. A total of 122 patients with T1 BC were enrolled. TTHC and SR were assessed preoperatively using diffuse optical tomography and strain elastography, respectively. All patients were pathologically evaluated to determine ALN status. Univariate analysis and logistic regression trend test were performed to identify independent predictors. A combined model of imaging-pathological parameters for ALNM was developed. Results Histopathological analysis indicated that 56 patients (45.9%) exhibited ALNM. The fully adjusted model demonstrated a significant trend correlating increased TTHC (P<0.01 for trend), SR (P<0.001 for trend), and Ki-67 expression (P=0.004 for trend) with ALNM. The area under the receiver operating characteristic curves (AUCs) of TTHC, SR, and Ki-67 expression for predicting ALNM were 0.707 [95% confidence interval (CI): 0.61-0.80], 0.718 (95% CI: 0.63-0.81) and 0.642 (95% CI: 0.56-0.73), respectively. The integration of imaging parameters (TTHC and SR) and Ki-67 expression yielded superior predictive performance for ALN status compared to each parameter individually, as evidenced by an AUC of 0.837 (95% CI: 0.76-0.91). The Hosmer-Lemeshow test P value was 0.880, demonstrating good calibration. In the subgroup analysis, the model exhibited positive predictive capabilities (AUC >0.75) across various subgroups, including molecular subtype, pathological type, and grade. Notably, the predictive performance was particularly enhanced in patients with invasive lobular carcinomas and triple-negative BC, with AUC values exceeding 0.90 for both subgroups. Conclusions The study found that TTHC ≥185.75 µmol/L, SR ≥3.93 and Ki-67 expression ≥20% were strongly associated with ALNM in patients with T1 BC. The combined imaging-pathological model might provide a convenient preoperative method for predicting ALN status, which can assist clinicians in individualizing management for patients with early-stage BC.
Collapse
Affiliation(s)
- Cong Shang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing Zhang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Huang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Yao Y, Lv R, Dong J, Chen Q. CAFs-derived TIAM1 Promotes OSCC Cell Growth and Metastasis by Regulating ZEB2. Cell Biochem Biophys 2025; 83:729-740. [PMID: 39256253 DOI: 10.1007/s12013-024-01505-4] [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] [Accepted: 08/25/2024] [Indexed: 09/12/2024]
Abstract
Previous studies have suggested that cancer-associated fibroblasts (CAFs) within the tumor microenvironment are a critical factor in tumorigenesis and tumor development. However, the regulatory mechanisms of CAFs on oral squamous cell carcinoma (OSCC) are poorly defined. A CAF-conditioned medium (CAF-CM) was collected and applied to culture OSCC cells. Then, cell viability, proliferation, migration, and invasion were evaluated using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), Transwell, and scratch healing assays. T-Lymphoma Invasion and Metastasis 1 (TIAM1), zinc finger E-box-binding homeobox 2 (ZEB2), E-cadherin, and increased N-cadherin protein levels were determined using western blot. TIAM1 and ZEB2 mRNA levels were measured using real-time quantitative polymerase chain reaction (RT-qPCR). Their interaction was analyzed using Co-immunoprecipitation (Co-IP) assay. SCC25 cells with or without (TIAM1-silencing) CAFs were subcutaneously inoculated in nude mice to assess the effect of TIAM1 in CAFs on OSCC tumor growth in vivo. CAFs expedited OSCC cell proliferation, migration, invasion, and EMT. TIAM1 and ZEB2 expression were upregulated in OSCC patients and OSCC cells, and the TIAM1 level was much higher in CAFs than in OSCC cells. Furthermore, TIAM1 knockdown in CAFs might partly abolish the promotion of CAFs on OSCC cell development, implying that TIAM1 might be secreted by CAFs into the culture medium to exert its effects inside OSCCs. TIAM1 might increase ZEB2 expression, and ZEB2 upregulation might partly reverse the repression of TIAM1 silencing in CAFs on OSCC cell malignant behaviors. In vivo studies confirmed that CAFs accelerated OSCC tumor growth, these effects were partially counteracted by TIAM1 downregulation. Overall, TIAM1 secreted by CAFs could expedite OSCC cell growth and metastasis by regulating ZEB2, providing a promising therapeutic target for OSCC treatment.
Collapse
Affiliation(s)
- Yao Yao
- Department of stomatology, Jingzhou Central Hospital, Jingzhou City, Hubei Province, China.
- Department of stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China.
| | - Ruya Lv
- Department of stomatology, Jingzhou Central Hospital, Jingzhou City, Hubei Province, China
- Department of stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Jingjing Dong
- Department of stomatology, Jingzhou Central Hospital, Jingzhou City, Hubei Province, China
- Department of stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Qi'an Chen
- Department of stomatology, Jingzhou Central Hospital, Jingzhou City, Hubei Province, China
- Department of stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| |
Collapse
|
21
|
Andrieu C, Danesin C, Montigny A, Rey M, Baqué K, Bibonne A, Alfandari D, Theveneau E. Delamination of chick cephalic neural crest cells requires an MMP14-dependent downregulation of Cadherin-6B. Differentiation 2025; 142:100836. [PMID: 39828493 DOI: 10.1016/j.diff.2025.100836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Matrix Metalloproteinases (MMPs) are known for their role in matrix remodeling via their catalytic activities in the extracellular space. Interestingly, these enzymes can also play less expected roles in cell survival, polarity and motility via other substrates (e.g. receptors, chemokines), through an intracellular localization (e.g. the nucleus) or via non-catalytic functions. Most of these unconventional functions are yet to be functionally validated in a physiological context. Here, we used the delamination of the cephalic Neural Crest (NC) cells of the chicken embryo, a well described experimental model of epithelial-mesenchymal transition (EMT), to study the in vivo function of MMP14 (a.k.a MT1-MMP). MMP14 is a transmembrane MMP known for its importance in cell invasion and often associated with poor prognosis in cancer. We found that MMP14 is expressed and required for cephalic NC delamination. More specifically, MMP14 is necessary for the downregulation of Cadherin-6B and a co-inhibition of Cadherin-6B and MMP14 expressions is sufficient to restore NC delamination. Cadherin-6B is normally repressed by Snail2. Surprisingly, in MMP14 knockdown this lack of Cadherin-6B repression occurs in the context of a normal expression and nuclear import of Snail2. We further show that MMP14 is not detected in the nucleus and that Snail2 and MMP14 do not physically interact. These data reveals that a yet to be identified MMP14-dependent signaling event is required for the Snail2-dependent repression of Cadherin-6B. In conclusion, this work provides an in vivo example of atypical regulation of Cadherins by an MMP which emphasizes the importance and diversity of non-canonical functions of MMPs.
Collapse
Affiliation(s)
- Cyril Andrieu
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Cathy Danesin
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Audrey Montigny
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Marie Rey
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Klara Baqué
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Anne Bibonne
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Dominique Alfandari
- University of Massachusetts Amherst, Dept. of Veterinary and Animal Sciences, Amherst, MA, 01003, USA
| | - Eric Theveneau
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France.
| |
Collapse
|
22
|
Li CX, Zhao ZX, Su DB, Yin DC, Ye YJ. In vitro regulation of collective cell migration: Understanding the role of physical and chemical microenvironments. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:23-40. [PMID: 39612952 DOI: 10.1016/j.pbiomolbio.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
Collective cell migration is the primary mode of cellular movement during embryonic morphogenesis, tissue repair and regeneration, and cancer invasion. Distinct from single-cell migration, collective cell migration involves complex intercellular signaling cascades and force transmission. Consequently, cell collectives exhibit intricate and diverse migration patterns under the influence of the microenvironment in vivo. Investigating the patterns and mechanisms of collective cell migration within complex environmental factors in vitro is essential for elucidating collective cell migration in vivo. This review elucidates the influence of physical and chemical factors in vitro microenvironment on the migration patterns and efficiency of cell collectives, thereby enhancing our comprehension of the phenomenon. Furthermore, we concisely present the effects of characteristic properties of common biomaterials on collective cell migration during tissue repair and regeneration, as well as the features and applications of tumor models of different dimensions (2D substrate or 3D substrate) in vitro. Finally, we highlight the challenges facing the research of collective cell migration behaviors in vitro microenvironment and propose that modulating collective cell migration may represent a potential strategy to promote tissue repair and regeneration and to control tumor invasion and metastasis.
Collapse
Affiliation(s)
- Chang-Xing Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
23
|
Tang K, Zheng Y, Hu G, Xin Y, Li K, Zhang C, Chen X, Zhang B, Li X, Hu B, Jia Q, Zheng YP, Yang M, Tan Y. Local soft niches in mechanically heterogeneous primary tumors promote brain metastasis via mechanotransduction-mediated HDAC3 activity. SCIENCE ADVANCES 2025; 11:eadq2881. [PMID: 40009679 PMCID: PMC11864190 DOI: 10.1126/sciadv.adq2881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
Tumor cells with organ-specific metastasis traits arise in primary lesions with substantial variations of local niche mechanics owing to intratumoral heterogeneity. However, the roles of mechanically heterogeneous primary tumor microenvironment in metastatic organotropism remain an enigma. This study reports that persistent priming in soft but not stiff niches that mimic primary tumor mechanical heterogeneity induces transcriptional reprogramming reminiscent of neuron and promotes the acquisition of brain metastatic potential. Soft-primed cells generate brain metastases in vivo through enhanced transendothelial migration across blood-brain barrier and brain colonization, which is further supported by the findings that tumor cells residing in local soft niches of primary xenografts exhibit brain metastatic tropism. Mechanistically, soft niches suppress cytoskeleton-nucleus-mediated mechanotransduction, which promotes histone deacetylase 3 activity. Inhibiting histone deacetylase 3 abolishes niche softness-induced brain metastatic ability. Collectively, this study uncovers a previously unappreciated role of local niche softness within primary tumors in brain metastasis, highlighting the significance of primary tumor mechanical heterogeneity in metastatic organotropism.
Collapse
Affiliation(s)
- Kai Tang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Yufan Zheng
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Guanshuo Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Cunyu Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Bai Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xueyi Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Bing Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Qiong Jia
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, 210006
| | - Yong-ping Zheng
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| |
Collapse
|
24
|
Zhao Y, Yu H, Li J, Qian J, Li M, Zhang X, Wang M, Wang Y, Dong Y, You Y, Zhou Q, Gao D, Zhao Y, Liu B, Chen R, Ren Z, Wang Z, Zhang K, Cui J. A glucose-enriched lung pre-metastatic niche triggered by matrix stiffness-tuned exosomal miRNAs in hepatocellular carcinoma. Nat Commun 2025; 16:1736. [PMID: 39966385 PMCID: PMC11836368 DOI: 10.1038/s41467-025-56878-8] [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: 03/05/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Apart from the classic features, it is almost unknown whether there exist other new pathological features during pre-metastatic niche formation in hepatocellular carcinoma (HCC). Our previous works have highlighted the contribution of increased matrix stiffness to lung pre-metastatic niche formation and metastasis in HCC. However, whether increased matrix stiffness influences glucose metabolism and supply of lung pre-metastatic niche remains largely unclear. Here we uncover the underlying mechanism by which matrix stiffness-tuned exosomal miRNAs as the major contributor modulate glucose enrichment during lung pre-metastatic niche formation through decreasing the glucose uptake and consumption of lung fibroblasts and increasing angiogenesis and vascular permeability. Our findings suggest that glucose enrichment, a new characteristic of the lung pre-metastatic niche triggered by matrix stiffness-tuned exosomal miRNAs, is essential for the colonization and survival of metastatic tumor cells, as well as subsequent metastatic foci growth.
Collapse
Affiliation(s)
- Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Hongmei Yu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Jiajun Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Jiali Qian
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Mimi Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Yaohui Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, PR China
| | - Yinying Dong
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Yang You
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Qiwen Zhou
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Binbin Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Rongxin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Kezhi Zhang
- Department of Hepatobiliary Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, PR China.
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China.
| |
Collapse
|
25
|
Shah S, Osuala KO, Brock EJ, Ji K, Sloane BF, Mattingly RR. Three-Dimensional Models: Biomimetic Tools That Recapitulate Breast Tissue Architecture and Microenvironment to Study Ductal Carcinoma In Situ Transition to Invasive Ductal Breast Cancer. Cells 2025; 14:220. [PMID: 39937011 PMCID: PMC11817749 DOI: 10.3390/cells14030220] [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: 12/24/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Diagnosis of ductal carcinoma in situ (DCIS) presents a challenge as we cannot yet distinguish between those lesions that remain dormant from cases that may progress to invasive ductal breast cancer (IDC) and require therapeutic intervention. Our overall interest is to develop biomimetic three-dimensional (3D) models that more accurately recapitulate the structure and characteristics of pre-invasive breast cancer in order to study the underlying mechanisms driving malignant progression. These models allow us to mimic the microenvironment to investigate many aspects of mammary cell biology, including the role of the extracellular matrix (ECM), the interaction between carcinoma-associated fibroblasts (CAFs) and epithelial cells, and the dynamics of cytoskeletal reorganization. In this review article, we outline the significance of 3D culture models as reliable pre-clinical tools that mimic the in vivo tumor microenvironment and facilitate the study of DCIS lesions as they progress to invasive breast cancer. We also discuss the role of CAFs and other stromal cells in DCIS transition as well as the clinical significance of emerging technologies like tumor-on-chip and co-culture models.
Collapse
Affiliation(s)
- Seema Shah
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
| | | | - Ethan J. Brock
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
| | - Kyungmin Ji
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Bonnie F. Sloane
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Raymond R. Mattingly
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
26
|
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
|
27
|
Wu S, Pan JW, Citartan M, Tang TH, Teo SH, Ch'ng ES. Profiling Precursor microRNAs of Breast Cancer From Total RNA Sequencing Data to Gain Insights Into Their Roles and Prognostic Values. Genes Chromosomes Cancer 2025; 64:e70027. [PMID: 39936877 DOI: 10.1002/gcc.70027] [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: 09/07/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Breast cancer, a molecularly heterogeneous disease, is classified into hormone receptor-positive luminal breast cancer (LBC), human epidermal growth factor receptor 2-positive breast cancer, and triple-negative breast cancer (TNBC). Precursor microRNAs (pre-miRNAs), typically form hairpin structures with a length from 65 to 80 bases, are shown to play crucial roles in breast cancer carcinogenesis. We hypothesized that these pre-miRNAs could have been sequenced in total RNA sequencing (RNA-seq) and developed a novel algorithm to profile pre-miRNAs from raw total RNA-seq data. A total of 907 breast cancer samples curated by Malaysian Breast Cancer Genetic Study (MyBrCa) were profiled using this algorithm and a comparison was made between pre-miRNA profiles and mature miRNA profiles obtained from The Cancer Genome Atlas (TCGA) dataset. We explored differentially expressed pre-miRNAs in TNBC in comparison to LBC and conducted downstream functional analyses of the target genes. A prognostic signature was built by LASSO-Cox regression on selected pre-miRNAs and validated internally and externally by MyBrCa and TCGA datasets, respectively. As a result, 10 common differentially expressed pre-miRNAs were identified. Functional analyses of these pre-miRNAs captured certain aggressive TNBC behaviors. Importantly, a pre-miRNA signature composed of 4 out of these 10 pre-miRNAs significantly prognosticated the breast cancer patients in the MyBrCa cohort and TCGA cohort, independent of conventional prognostic factors. In conclusion, this novel algorithm allows profiling pre-miRNAs from raw total RNA-seq data, which could be cross-validated with mature miRNA profiles for cross-platform comparison. The findings of this study underscore the importance of pre-miRNAs in breast cancer carcinogenesis and as prognostic factors.
Collapse
Affiliation(s)
- Sen Wu
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | | | - Marimuthu Citartan
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Thean-Hock Tang
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Soo-Hwang Teo
- Cancer Research Malaysia, Subang Jaya, Malaysia
- Faculty of Medicine, University Malaya Cancer Research Institute, University of Malaya, Kuala Lumpur, Malaysia
| | - Ewe Seng Ch'ng
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
28
|
Hou H, Liu X, Liu J, Wang Y. Carbohydrate polymer-based nanoparticles with cell membrane camouflage for cancer therapy: A review. Int J Biol Macromol 2025; 289:138620. [PMID: 39674458 DOI: 10.1016/j.ijbiomac.2024.138620] [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: 10/08/2024] [Revised: 11/21/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Recent developments in biomimetic nanoparticles, specifically carbohydrate polymer-coated cell membrane nanoparticles, have demonstrated considerable promise in treating cancer. These systems improve drug delivery by imitating natural cell actions, enhancing biocompatibility, and decreasing immune clearance. Conventional drug delivery methods frequently face challenges with non-specific dispersal and immune detection, which can hinder their efficiency and safety. These biomimetic nanoparticles improve target specificity, retention times, and therapeutic efficiency by using biological components like chitosan, hyaluronic acid, and alginate. Chitosan-based nanoparticles, which come from polysaccharides found in nature, have self-assembly abilities that make them better drug carriers. Hyaluronic acid helps target tissues more effectively, especially in cancer environments where there are high levels of hyaluronic acid receptors. Alginate-based systems also enhance drug delivery by being biocompatible and degradable, making them ideal choices for advanced therapeutic uses. Moreover, these particles hold potential for overcoming resistance to multiple drugs and boosting the body's immune reaction to tumors through precise delivery and decreased side effects of chemotherapy drugs. This review delves into the possibilities of using carbohydrate polymer-functionalized nanoparticles and their impact on enhancing the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Haijia Hou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuejian Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Liu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yudong Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
29
|
Wani TU, Kim HY, Lee GH, Lim YJ, Chae HJ, Kim JY, Yoon H. Mechanistic insights into epithelial-mesenchymal transition mediated cisplatin resistance in ovarian cancer. Sci Rep 2025; 15:3053. [PMID: 39856185 PMCID: PMC11760351 DOI: 10.1038/s41598-025-87388-8] [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: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is designated as one of the prime causes of chemoresistance in many cancers. In our previous study we established that cisplatin resistance in ovarian cancer (OC) is associated with EMT using sensitive OV90 cells and its resistant counterparts OV90CisR1 and OV90CisR2. In this study, we revealed through RNAseq analysis that ITGA1 can play essential part in EMT mediated cisplatin resistance in OC. We found large number of EMT related terms predominant in the top gene ontologies (GO). We also found Extracellular matrix (ECM) and actin cytoskeleton genes highly altered in the resistant cells. This was further confirmed by the protein-protein interaction (PPI) analysis where we identified that the core ECM components e.g., collagen, fibronectin, metalloproteases and integrins possessed most interactions. The pathway analysis revealed the Wnt signaling as the leading pathway. Since integrins have significant interaction with Wnt signaling, we focused our study on integrins among which, ITGA1, ITGA6, ITGA11 and ITGAV were primarily altered. We validated our results by western blotting and found that ITGA1 was highly expressed in resistant cells. Additionally, the high ABCA5 (efflux transporter) expression in resistant cells also supports the EMT proposition. The western blotting also revealed high β-catenin expression in resistant cells confirming the high Wnt signaling activity. Further, we induced xenograft tumors in nude mice. The histopathological analysis confirmed the aggressive nature of resistant tumors and showed the presence of necrotic core which could be implicated to EMT. Finally, the immunohistochemical staining confirmed the high protein expression in resistant tumor.
Collapse
Affiliation(s)
- Taha Umair Wani
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju-si, 52828, Gyeongnam, Republic of Korea.
| | - Hyun-Yi Kim
- NGeneS Inc, Ansan-si, 15495, Gyeonggi-do, Republic of Korea
| | - Geum-Hwa Lee
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju-si, 54907, Jeollabuk-do, Republic of Korea
| | - Young Je Lim
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju-si, 54907, Jeollabuk-do, Republic of Korea
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju-si, 54896, Jeollabuk-do, Republic of Korea
| | - Han-Jung Chae
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju-si, 54896, Jeollabuk-do, Republic of Korea
| | - Ji-Ye Kim
- Department of Pathology, Ilsan Paik Hospital, Inje University, College of Medicine, Goyang-si, 10380, Gyeonggi-do, Republic of Korea.
| | - Hyonok Yoon
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju-si, 52828, Gyeongnam, Republic of Korea.
- Department of Pharmacology and Physiology, Drexel University, College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
30
|
Lin WJ, Yu H, Pathak A. Gradients in cell density and shape transitions drive collective cell migration into confining environments. SOFT MATTER 2025; 21:719-728. [PMID: 39784299 PMCID: PMC11715644 DOI: 10.1039/d3sm01240a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/27/2024] [Indexed: 01/12/2025]
Abstract
Epithelial cell collectives migrate through tissue interfaces and crevices to orchestrate development processes, tumor invasion, and wound healing. Naturally, the traversal of cell collective through confining environments involves crowding due to narrowing spaces, which seems tenuous given the conventional inverse relationship between cell density and migration. However, the physical transitions required to overcome such epithelial densification for migration across confinements remain unclear. Here, in a system of contiguous microchannels of varying confinements, we show that epithelial (MCF10A) monolayers accumulate higher cell density and undergo fluid-like shape transitions before entering narrower channels. However, overexpression of breast cancer oncogene ErbB2 did not require such accumulation of cell density to migrate across matrix confinement. While wild-type MCF10A cells migrated faster in narrow channels, this confinement sensitivity was reduced after +ErbB2 mutation or with constitutively active RhoA. This physical interpretation of collective cell migration as density and shape transitions in granular matter could advance our understanding of complex living systems.
Collapse
Affiliation(s)
- Wan-Jung Lin
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, USA.
| | - Hongsheng Yu
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, USA.
| | - Amit Pathak
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, USA.
| |
Collapse
|
31
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [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: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
32
|
Peng H, Chao Z, Wang Z, Hao X, Xi Z, Ma S, Guo X, Zhang J, Zhou Q, Qu G, Gao Y, Luo J, Wang Z, Wang J, Li L. Biomechanics in the tumor microenvironment: from biological functions to potential clinical applications. Exp Hematol Oncol 2025; 14:4. [PMID: 39799341 PMCID: PMC11724500 DOI: 10.1186/s40164-024-00591-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
Immune checkpoint therapies have spearheaded drug innovation over the last decade, propelling cancer treatments toward a new era of precision therapies. Nonetheless, the challenges of low response rates and prevalent drug resistance underscore the imperative for a deeper understanding of the tumor microenvironment (TME) and the pursuit of novel targets. Recent findings have revealed the profound impacts of biomechanical forces within the tumor microenvironment on immune surveillance and tumor progression in both murine models and clinical settings. Furthermore, the pharmacological or genetic manipulation of mechanical checkpoints, such as PIEZO1, DDR1, YAP/TAZ, and TRPV4, has shown remarkable potential in immune activation and eradication of tumors. In this review, we delved into the underlying biomechanical mechanisms and the resulting intricate biological meaning in the TME, focusing mainly on the extracellular matrix, the stiffness of cancer cells, and immune synapses. We also summarized the methodologies employed for biomechanical research and the potential clinical translation derived from current evidence. This comprehensive review of biomechanics will enhance the understanding of the functional role of biomechanical forces and provide basic knowledge for the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Hao Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zheng Chao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zefeng Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaodong Hao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zirui Xi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Sheng Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Qiang Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, 810001, Qinghai, China
| | - Guanyu Qu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Yuan Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
- Taikang Tongji (Wuhan) Hospital, 420060, Wuhan, China.
| | - Jing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| | - Le Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| |
Collapse
|
33
|
Narain R, Muncie-Vasic JM, Weaver VM. Forcing the code: tension modulates signaling to drive morphogenesis and malignancy. Genes Dev 2025; 39:163-181. [PMID: 39638568 PMCID: PMC11789492 DOI: 10.1101/gad.352110.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Development and disease are regulated by the interplay between genetics and the signaling pathways stimulated by morphogens, growth factors, and cytokines. Experimental data highlight the importance of mechanical force in regulating embryonic development, tissue morphogenesis, and malignancy. Force not only sculpts tissue movements to drive embryogenesis and morphogenesis but also modifies the context of biochemical signaling and gene expression to regulate cell and tissue fate. Not surprisingly, experiments have demonstrated that perturbations in cell tension drive malignancy and metastasis by altering biochemical signaling and gene expression through modifications in cytoskeletal tension, transmembrane receptor structure and function, and organelle phenotype that enhance cell growth and survival, alter metabolism, and foster cell migration and invasion. At the tissue level, tumor-associated forces disrupt cell-cell adhesions to perturb tissue organization, compromise vascular integrity to induce hypoxia, and interfere with antitumor immunity to foster metastasis and treatment resistance. Exciting new approaches now exist with which to clarify the relationship between mechanotransduction, biochemical signaling, and gene expression in development and disease. Indeed, gaining insight into these interactions is essential to unravel molecular mechanisms that regulate development and clarify the molecular basis of cancer.
Collapse
Affiliation(s)
- Radhika Narain
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California 94720, USA
| | | | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA;
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California 94143
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
34
|
Guan X, Wang F, Zhang D, Qiao P, Qin Y, Wang B. Mid-urethral sling with proper sling tension is an effective treatment for stress urinary incontinence in women after pelvic radiotherapy: a pilot study of case series. Front Surg 2025; 11:1475030. [PMID: 39830372 PMCID: PMC11739098 DOI: 10.3389/fsurg.2024.1475030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Background At present, consensus on the management of female stress urinary incontinence (SUI) after pelvic radiotherapy is lacking. We aim to assess the clinical effects of mid-urethral sling (MUS) for the treatment of SUI after pelvic radiotherapy in women. Methods We conducted a retrospective review of the clinical database of female with SUI after pelvic radiotherapy from June 2015 to February 2022. The clinical efficacy was evaluated by International Consultation on Incontinence Questionnaire-Short Form (ICI-Q-SF) questionnaire, maximum flow rate (Qmax) and postvoid residual (PVR) urine. All patients were reviewed postoperatively in an outpatient clinic. Results We identified 26 patients with mean age of 59.35 ± 7.32 years. All the patients who suffered from SUI had a history of gynaecological malignancies and received pelvic radiotherapy. 21 patients (80.77%, 95% CI: 0.621-0.915) were considered to have successfully improved after surgery, the ICI-Q-SF scores were lower than the pre-operative at 2 weeks, 6 months and 1 year postoperatively (P < 0.01). After 1-year follow-up, none of the patients had mesh erosion. Conclusion SUI following radiotherapy for the treatment of pelvic malignancy can be challenging to manage. MUS is a highly effective and safe option for the treatment of SUI after radiotherapy, additionally, that proper sling tension is the key to the success of the procedure.
Collapse
Affiliation(s)
- Xing Guan
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Fei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Di Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Peng Qiao
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yan Qin
- Department of Urology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi, China
| | - Biao Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
Zhang Y, Hill CM, Leach KA, Grillini L, Deliard S, Offley SR, Gatto M, Picone F, Zucco A, Gardini A. The enhancer module of Integrator controls cell identity and early neural fate commitment. Nat Cell Biol 2025; 27:103-117. [PMID: 39592860 PMCID: PMC11752693 DOI: 10.1038/s41556-024-01556-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/09/2024] [Indexed: 11/28/2024]
Abstract
Lineage-specific transcription factors operate as master orchestrators of developmental processes by activating select cis-regulatory enhancers and proximal promoters. Direct DNA binding of transcription factors ultimately drives context-specific recruitment of the basal transcriptional machinery that comprises RNA polymerase II (RNAPII) and a host of polymerase-associated multiprotein complexes, including the metazoan-specific Integrator complex. Integrator is primarily known to modulate RNAPII processivity and to surveil RNA integrity across coding genes. Here we describe an enhancer module of Integrator that directs cell fate specification by promoting epigenetic changes and transcription factor binding at neural enhancers. Depletion of Integrator's INTS10 subunit upends neural traits and derails cells towards mesenchymal identity. Commissioning of neural enhancers relies on Integrator's enhancer module, which stabilizes SOX2 binding at chromatin upon exit from pluripotency. We propose that Integrator is a functional bridge between enhancers and promoters and a main driver of early development, providing new insight into a growing family of neurodevelopmental syndromes.
Collapse
Affiliation(s)
| | - Connor M Hill
- The Wistar Institute, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelsey A Leach
- The Wistar Institute, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Luca Grillini
- The Wistar Institute, Philadelphia, PA, USA
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Sarah R Offley
- The Wistar Institute, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martina Gatto
- The Wistar Institute, Philadelphia, PA, USA
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | | | | |
Collapse
|
36
|
Zhang X, Al‐Danakh A, Zhu X, Feng D, Yang L, Wu H, Li Y, Wang S, Chen Q, Yang D. Insights into the mechanisms, regulation, and therapeutic implications of extracellular matrix stiffness in cancer. Bioeng Transl Med 2025; 10:e10698. [PMID: 39801760 PMCID: PMC11711218 DOI: 10.1002/btm2.10698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/19/2024] [Accepted: 06/29/2024] [Indexed: 01/03/2025] Open
Abstract
The tumor microenvironment (TME) is critical for cancer initiation, growth, metastasis, and therapeutic resistance. The extracellular matrix (ECM) is a significant tumor component that serves various functions, including mechanical support, TME regulation, and signal molecule generation. The quantity and cross-linking status of ECM components are crucial factors in tumor development, as they determine tissue stiffness and the interaction between stiff TME and cancer cells, resulting in aberrant mechanotransduction, proliferation, migration, invasion, angiogenesis, immune evasion, and treatment resistance. Therefore, broad knowledge of ECM dysregulation in the TME might aid in developing innovative cancer therapies. This review summarized the available information on major ECM components, their functions, factors that increase and decrease matrix stiffness, and related signaling pathways that interplay between cancer cells and the ECM in TME. Moreover, mechanotransduction alters during tumorogenesis, and current drug therapy based on ECM as targets, as well as future efforts in ECM and cancer, are also discussed.
Collapse
Affiliation(s)
- Ximo Zhang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Abdullah Al‐Danakh
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xinqing Zhu
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Dan Feng
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Linlin Yang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Haotian Wu
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yingying Li
- Department of Discipline ConstructionDalian Medical UniversityDalianChina
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of GlycobiologyDalian Medical UniversityDalianChina
| | - Qiwei Chen
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Zhongda Hospital, Medical School Advanced Institute Life HealthSoutheast UniversityNanjingChina
| | - Deyong Yang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of SurgeryHealinghands ClinicDalianChina
| |
Collapse
|
37
|
Lipari S, Sacco P, Cok M, Scognamiglio F, Romano M, Brun F, Giulianini PG, Marsich E, Aachmann FL, Donati I. Hydrogel Elastic Energy: A Stressor Triggering an Adaptive Stress-Mediated Cell Response. Adv Healthc Mater 2025; 14:e2402400. [PMID: 39535422 PMCID: PMC11730662 DOI: 10.1002/adhm.202402400] [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: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The crosstalk between the cells and the extracellular matrix (ECM) is bidirectional and consists of a pushing/pulling stretch exerted by the cells and a mechanical resistance counteracted by the surrounding microenvironment. It is widely recognized that the stiffness of the ECM, its viscoelasticity, and its overall deformation are the most important traits influencing the response of the cells. Here these three parameters are combined into a concept of elastic energy, which in biological terms represents the mechanical feedback that cells perceive when the ECM is deformed. It is shown that elastic energy is a stress factor that influences the response of cells in three-dimensional (3D) cultures. Strikingly, the higher the elastic energy of the matrix and thus the mechanical feedback, the higher the stress state of the cells, which correlates with the formation of G3BP-mediated stress granules. This condition is associated with an increase in alkaline phosphatase (ALP) activity but a decrease in gene expression and is mediated by the nuclear translocation of Yes-associated protein (YAP). This work supports the importance of considering the elastic energy as mechano-controller in regulating cellular stress state in 3D cultures.
Collapse
Affiliation(s)
- Sara Lipari
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Pasquale Sacco
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Michela Cok
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | | | - Maurizio Romano
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Francesco Brun
- Department of Engineering and ArchitectureUniversity of TriesteVia A. Valerio 6/1Trieste34127Italy
| | | | - Eleonora Marsich
- Department of MedicineSurgery and Health SciencesUniversity of TriestePiazza dell'Ospitale 1Trieste34129Italy
| | - Finn L. Aachmann
- Department of Biotechnology and Food ScienceNorwegian Biopolymer Laboratory (NOBIPOL)NTNU Norwegian University of Science and TechnologySem Sælands vei 6/8Trondheim7491Norway
| | - Ivan Donati
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| |
Collapse
|
38
|
Hulahan TS, Angel PM. From ductal carcinoma in situ to invasive breast cancer: the prognostic value of the extracellular microenvironment. J Exp Clin Cancer Res 2024; 43:329. [PMID: 39716322 DOI: 10.1186/s13046-024-03236-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/19/2024] [Indexed: 12/25/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) is a noninvasive breast disease that variably progresses to invasive breast cancer (IBC). Given the unpredictability of this progression, most DCIS patients are aggressively managed similar to IBC patients. Undoubtedly, this treatment paradigm places many DCIS patients at risk of overtreatment and its significant consequences. Historically, prognostic modeling has included the assessment of clinicopathological features and genomic markers. Although these provide valuable insights into tumor biology, they remain insufficient to predict which DCIS patients will progress to IBC. Contemporary work has begun to focus on the microenvironment surrounding the ductal cells for molecular patterns that might predict progression. In this review, extracellular microenvironment alterations occurring with the malignant transformation from DCIS to IBC are detailed. Not only do changes in collagen abundance, organization, and localization mediate the transition to IBC, but also the discrete post-translational regulation of collagen fibers is understood to promote invasion. Other extracellular matrix proteins, such as matrix metalloproteases, decorin, and tenascin C, have been characterized for their role in invasive transformation and further demonstrate the prognostic value of the extracellular matrix. Importantly, these extracellular matrix proteins influence immune cells and fibroblasts toward pro-tumorigenic phenotypes. Thus, the progressive changes in the extracellular microenvironment play a key role in invasion and provide promise for prognostic development.
Collapse
Affiliation(s)
- Taylor S Hulahan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Peggi M Angel
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
39
|
Raaijmakers KTPM, Adema GJ, Bussink J, Ansems M. Cancer-associated fibroblasts, tumor and radiotherapy: interactions in the tumor micro-environment. J Exp Clin Cancer Res 2024; 43:323. [PMID: 39696386 DOI: 10.1186/s13046-024-03251-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) represent a group of genotypically non-malignant stromal cells in the tumor micro-environment (TME) of solid tumors that encompasses up to 80% of the tumor volume. Even though the phenotypic diversity and plasticity of CAFs complicates research, it is well-established that CAFs can affect many aspects of tumor progression, including growth, invasion and therapy resistance. Although anti-tumorigenic properties of CAFs have been reported, the majority of research demonstrates a pro-tumorigenic role for CAFs via (in)direct signaling to cancer cells, immunomodulation and extracellular matrix (ECM) remodeling. Following harsh therapeutic approaches such as radio- and/or chemotherapy, CAFs do not die but rather become senescent. Upon conversion towards senescence, many pro-tumorigenic characteristics of CAFs are preserved or even amplified. Senescent CAFs continue to promote tumor cell therapy resistance, modulate the ECM, stimulate epithelial-to-mesenchymal transition (EMT) and induce immunosuppression. Consequently, CAFs play a significant role in tumor cell survival, relapse and potentially malignant transformation of surviving cancer cells following therapy. Modulating CAF functioning in the TME therefore is a critical area of research. Proposed strategies to enhance therapeutic efficacy include reverting senescent CAFs towards a quiescent phenotype or selectively targeting (non-)senescent CAFs. In this review, we discuss CAF functioning in the TME before and during therapy, with a strong focus on radiotherapy. In the future, CAF functioning in the therapeutic TME should be taken into account when designing treatment plans and new therapeutic approaches.
Collapse
Affiliation(s)
- Kris T P M Raaijmakers
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
40
|
Borges A, Chara O. Peeking into the future: inferring mechanics in dynamical tissues. Biochem Soc Trans 2024; 52:2579-2592. [PMID: 39656056 DOI: 10.1042/bst20230225] [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: 05/29/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
Cells exert forces on each other and their environment, shaping the tissue. The resulting mechanical stresses can be determined experimentally or estimated computationally using stress inference methods. Over the years, mechanical stress inference has become a non-invasive, low-cost computational method for estimating the relative intercellular stresses and intracellular pressures of tissues. This mini-review introduces and compares the static and dynamic modalities of stress inference, considering their advantages and limitations. To date, most software has focused on static inference, which requires only a single microscopy image as input. Although applicable in quasi-equilibrium states, this approach neglects the influence that cell rearrangements might have on the inference. In contrast, dynamic stress inference relies on a time series of microscopy images to estimate stresses and pressures. Here, we discuss both static and dynamic mechanical stress inference in terms of their physical, mathematical, and computational foundations and then outline what we believe are promising avenues for in silico inference of the mechanical states of tissues.
Collapse
Affiliation(s)
- Augusto Borges
- Unit Sensory Biology and Organogenesis, Helmholtz Zentrum München, Munich, Germany
- Graduate School of Quantitative Biosciences, Ludwig Maximilian University, Munich, Germany
| | - Osvaldo Chara
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Nottingham LE12, U.K
- Instituto de Tecnología, Universidad Argentina de la Empresa, Buenos Aires, Argentina
| |
Collapse
|
41
|
Lv K, He T. Cancer-associated fibroblasts: heterogeneity, tumorigenicity and therapeutic targets. MOLECULAR BIOMEDICINE 2024; 5:70. [PMID: 39680287 PMCID: PMC11649616 DOI: 10.1186/s43556-024-00233-8] [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: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Cancer, characterized by its immune evasion, active metabolism, and heightened proliferation, comprises both stroma and cells. Although the research has always focused on parenchymal cells, the non-parenchymal components must not be overlooked. Targeting cancer parenchymal cells has proven to be a formidable challenge, yielding limited success on a broad scale. The tumor microenvironment(TME), a critical niche for cancer cell survival, presents a novel way for cancer treatment. Cancer-associated fibroblast (CAF), as a main component of TME, is a dynamically evolving, dual-functioning stromal cell. Furthermore, their biological activities span the entire spectrum of tumor development, metastasis, drug resistance, and prognosis. A thorough understanding of CAFs functions and therapeutic advances holds significant clinical implications. In this review, we underscore the heterogeneity of CAFs by elaborating on their origins, types and function. Most importantly, by elucidating the direct or indirect crosstalk between CAFs and immune cells, the extracellular matrix, and cancer cells, we emphasize the tumorigenicity of CAFs in cancer. Finally, we highlight the challenges encountered in the exploration of CAFs and list targeted therapies for CAF, which have implications for clinical treatment.
Collapse
Affiliation(s)
- Keke Lv
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China
| | - Tianlin He
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
42
|
Chen Z, Wang J, Kankala RK, Jiang M, Long L, Li W, Zou L, Chen A, Liu Y. Decellularized extracellular matrix-based disease models for drug screening. Mater Today Bio 2024; 29:101280. [PMID: 39399243 PMCID: PMC11470555 DOI: 10.1016/j.mtbio.2024.101280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
In vitro drug screening endeavors to replicate cellular states closely resembling those encountered in vivo, thereby maximizing the fidelity of drug effects and responses within the body. Decellularized extracellular matrix (dECM)-based materials offer a more authentic milieu for crafting disease models, faithfully emulating the extracellular components and structural complexities encountered by cells in vivo. This review discusses recent advancements in leveraging dECM-based materials as biomaterials for crafting cell models tailored for drug screening. Initially, we delineate the biological functionalities of diverse ECM components, shedding light on their potential influences on disease model construction. Further, we elucidate the decellularization techniques and methodologies for fabricating cell models utilizing dECM substrates. Then, the article delves into the research strides made in employing dECM-based models for drug screening across a spectrum of ailments, including tumors, as well as heart, liver, lung, and bone diseases. Finally, the review summarizes the bottlenecks, hurdles, and promising research trajectories associated with the dECM materials for drug screening, alongside their prospective applications in personalized medicine. Together, by encapsulating the contemporary research landscape surrounding dECM materials in cell model construction and drug screening, this review underscores the vast potential of dECM materials in drug assessment and personalized therapy.
Collapse
Affiliation(s)
- Zhoujiang Chen
- Institute for Advanced Study, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Ji Wang
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, PR China
| | - Mingli Jiang
- School of Pharmacy, Zunyi Medical University, Zunyi, 563099, Guizhou, PR China
| | - Lianlin Long
- School of Pharmacy, Zunyi Medical University, Zunyi, 563099, Guizhou, PR China
| | - Wei Li
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Liang Zou
- Institute for Advanced Study, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, PR China
| | - Ya Liu
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| |
Collapse
|
43
|
Yuan J, Yang L, Zhang H, Beeraka NM, Zhang D, Wang Q, Wang M, Pr HV, Sethi G, Wang G. Decoding tumor microenvironment: EMT modulation in breast cancer metastasis and therapeutic resistance, and implications of novel immune checkpoint blockers. Biomed Pharmacother 2024; 181:117714. [PMID: 39615165 DOI: 10.1016/j.biopha.2024.117714] [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: 08/14/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Tumor microenvironment (TME) and epithelial-mesenchymal transition (EMT) play crucial roles in the initiation and progression of tumors. TME is composed of various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as non-cellular components like extracellular matrix (ECM) proteins and soluble factors. These elements interact with tumor cells through a complex network of signaling pathways involving cytokines, growth factors, metabolites, and non-coding RNA-carrying exosomes. Hypoxic conditions within the TME further modulate these interactions, collectively influencing tumor growth, metastatic potential, and response to therapy. EMT represents a dynamic and reversible process where epithelial cells undergo phenotypic changes to adopt mesenchymal characteristics in several cancers, including breast cancers. This transformation enhances cell motility and imparts stem cell-like properties, which are closely associated with increased metastatic capability and resistance to conventional cancer treatments. Thus, understanding the crosstalk between the TME and EMT is essential for unraveling the underlying mechanisms of breast cancer metastasis and therapeutic resistance. This review uniquely examines the intricate interplay between the tumor TME and epithelial-mesenchymal transition EMT in driving breast cancer metastasis and treatment resistance. It explores the therapeutic potential of targeting the TME-EMT axis, specifically through CD73-TGF-β dual-blockade, to improve outcomes in triple-negative breast cancer. Additionally, it underscores new strategies to enhance immune checkpoint blockade (ICB) responses by modulating EMT, thereby offering innovative insights for more effective cancer treatment.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Li Yang
- Department of Clinical Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hua Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Narasimha M Beeraka
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu, Andhra Pradesh 515721, India; Department of Studies in Molecular Biology, Faculty of Science and Technology, University of Mysore, Mysore, Karnataka, 570006, India.
| | - Danfeng Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Qun Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Minghua Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hemanth Vikram Pr
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Geng Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
44
|
Fan Y, Chiu A, Zhao F, George JT. Understanding the interplay between extracellular matrix topology and tumor-immune interactions: Challenges and opportunities. Oncotarget 2024; 15:768-781. [PMID: 39513932 PMCID: PMC11546212 DOI: 10.18632/oncotarget.28666] [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: 07/12/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Modern cancer management comprises a variety of treatment strategies. Immunotherapy, while successful at treating many cancer subtypes, is often hindered by tumor immune evasion and T cell exhaustion as a result of an immunosuppressive tumor microenvironment (TME). In solid malignancies, the extracellular matrix (ECM) embedded within the TME plays a central role in T cell recognition and cancer growth by providing structural support and regulating cell behavior. Relative to healthy tissues, tumor associated ECM signatures include increased fiber density and alignment. These and other differentiating features contributed to variation in clinically observed tumor-specific ECM configurations, collectively referred to as Tumor-Associated Collagen Signatures (TACS) 1-3. TACS is associated with disease progression and immune evasion. This review explores our current understanding of how ECM geometry influences the behaviors of both immune cells and tumor cells, which in turn impacts treatment efficacy and cancer evolutionary progression. We discuss the effects of ECM remodeling on cancer cells and T cell behavior and review recent in silico models of cancer-immune interactions.
Collapse
Affiliation(s)
- Yijia Fan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Translational Medical Sciences, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - Alvis Chiu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Jason T. George
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Translational Medical Sciences, Texas A&M University Health Science Center, Houston, TX 77030, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
- Department of Hematopoietic Biology and Malignancy, MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
45
|
Friedman-DeLuca M, Karagiannis GS, Condeelis JS, Oktay MH, Entenberg D. Macrophages in tumor cell migration and metastasis. Front Immunol 2024; 15:1494462. [PMID: 39555068 PMCID: PMC11563815 DOI: 10.3389/fimmu.2024.1494462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a phenotypically diverse, highly plastic population of cells in the tumor microenvironment (TME) that have long been known to promote cancer progression. In this review, we summarize TAM ontogeny and polarization, and then explore how TAMs enhance tumor cell migration through the TME, thus facilitating metastasis. We also discuss how chemotherapy and host factors including diet, obesity, and race, impact TAM phenotype and cancer progression. In brief, TAMs induce epithelial-mesenchymal transition (EMT) in tumor cells, giving them a migratory phenotype. They promote extracellular matrix (ECM) remodeling, allowing tumor cells to migrate more easily. TAMs also provide chemotactic signals that promote tumor cell directional migration towards blood vessels, and then participate in the signaling cascade at the blood vessel that allows tumor cells to intravasate and disseminate throughout the body. Furthermore, while chemotherapy can repolarize TAMs to induce an anti-tumor response, these cytotoxic drugs can also lead to macrophage-mediated tumor relapse and metastasis. Patient response to chemotherapy may be dependent on patient-specific factors such as diet, obesity, and race, as these factors have been shown to alter macrophage phenotype and affect cancer-related outcomes. More research on how chemotherapy and patient-specific factors impact TAMs and cancer progression is needed to refine treatment strategies for cancer patients.
Collapse
Affiliation(s)
- Madeline Friedman-DeLuca
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - George S. Karagiannis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Marilyn and Stanley M. Katz Institute for Immunotherapy of Cancer and Inflammatory Disorders, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - John S. Condeelis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Maja H. Oktay
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - David Entenberg
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
46
|
Barros da Silva P, Zhao X, Bidarra SJ, Nascimento DS, LaLone V, Lourenço BN, Paredes J, Stevens MM, Barrias CC. Tunable Hybrid Hydrogels of Alginate and Cell-Derived dECM to Study the Impact of Matrix Alterations on Epithelial-to-Mesenchymal Transition. Adv Healthc Mater 2024; 13:e2401032. [PMID: 39246099 PMCID: PMC11582509 DOI: 10.1002/adhm.202401032] [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: 03/19/2024] [Revised: 07/27/2024] [Indexed: 09/10/2024]
Abstract
Epithelial-to-mesenchymal transition (EMT) is crucial for tumor progression, being linked to alterations in the extracellular matrix (ECM). Understanding the ECM's role in EMT can uncover new therapeutic targets, yet replicating these interactions in vitro remains challenging. It is shown that hybrid hydrogels of alginate (ALG) and cell-derived decellularized ECM (dECM), with independently tunable composition and stiffness, are useful 3D-models to explore the impact of the breast tumor matrix on EMT. Soft RGD-ALG hydrogels (200 Pa), used as neutral bulk material, supported mammary epithelial cells morphogenesis without spontaneous EMT, allowing to define the gene, protein, and biochemical profiles of cells at different TGFβ1-induced EMT states. To mimic the breast tumor composition, dECM from TGFβ1-activated fibroblasts (adECM) are generated, which shows upregulation of tumor-associated proteins compared to ndECM from normal fibroblasts. Using hybrid adECM-ALG hydrogels, it is shown that the presence of adECM induces partial EMT in normal epithelial cells, and amplifes TGF-β1 effects compared to ALG and ndECM-ALG. Increasing the hydrogel stiffness to tumor-like levels (2.5 kPa) have a synergistic effect, promoting a more evident EMT. These findings shed light on the complex interplay between matrix composition and stiffness in EMT, underscoring the utility of dECM-ALG hydrogels as a valuable in vitro platform for cancer research.
Collapse
Affiliation(s)
- P Barros da Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, 4200-135, Portugal
| | - Xiaoyu Zhao
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Sílvia J Bidarra
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
| | - Diana S Nascimento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4200-135, Portugal
| | - Vernon LaLone
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Department of Materials, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Bianca N Lourenço
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, 4200-135, Portugal
| | - Joana Paredes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- FMUP - Faculdade de Medicina da Universidade do Porto, Porto, 4200-319, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, 4200-135, Portugal
| | - Molly M Stevens
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Department of Materials, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - C C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4200-135, Portugal
| |
Collapse
|
47
|
Wang C, Huangfu Y, Wang J, Lu X, Liu D, Zhang ZL. Microchip construction for migration assays: investigating the impact of physical confinement on cell morphology and motility during vaccinia virus infection. Anal Bioanal Chem 2024; 416:5605-5618. [PMID: 39158632 DOI: 10.1007/s00216-024-05485-5] [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: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024]
Abstract
Vaccinia virus (VACV)-induced cell migration is thought to be closely related to the rapid transmission of viral infection in the body. The limited studies are mainly based on scratch assay using traditional cell culture techniques, which inevitably ignores the influences of extracellular microenvironment. Physical confinement, inherently presenting in vivo, has proven to be a critical extern cue in modulating migration behaviors of multiple cells, while its impacts on VACV-induced cell motility remain unclear. Herein, we developed a migration assay microchip featuring confined microchannel array to investigate the effect of physical confinement on infected cell morphology and motility during VACV infection. Results showed that different from the random cell migration observed in traditional scratch assay on planar substrate, VACV-infected cells exhibited accelerated directionally persistent migration under confinement microenvironment. Moreover, single-directed elongated dominant lamella appeared to contrast distinctly with multiple protrusions stretched in random directions under unconfined condition. Additionally, the Golgi complex tended to relocate behind the nucleus confined within the microchannel axis compared to the classical reorientation pattern. These differences in characteristic subcellular architecture and organelle reorientation of migrating cells revealed cell biological mechanisms underlying altered migration behavior. Collectively, our study demonstrates that physical confinement acting as a guidance cue has profound impacts on VACV-induced migration behaviors, which provides new insight into cell migration behavior and viral rapid spread during VACV infection.
Collapse
Affiliation(s)
- Cheng Wang
- School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China.
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China.
| | - Yueyue Huangfu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ji Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, Hong Kong, 999077, China
| | - Xiaofeng Lu
- School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Dong Liu
- School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
48
|
Lan BQ, Wang YJ, Yu SX, Liu W, Liu YJ. Physical effects of 3-D microenvironments on confined cell behaviors. Am J Physiol Cell Physiol 2024; 327:C1192-C1201. [PMID: 39246142 DOI: 10.1152/ajpcell.00288.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Cell migration is a fundamental and functional cellular process, influenced by a complex microenvironment consisting of different cells and extracellular matrix. Recent research has highlighted that, besides biochemical cues from the microenvironment, physical cues can also greatly alter cellular behavior. However, due to the complexity of the microenvironment, little is known about how the physical interactions between migrating cells and surrounding microenvironment instructs cell movement. Here, we explore various examples of three-dimensional microenvironment reconstruction models in vitro and describe how the physical interplay between migrating cells and the neighboring microenvironment controls cell behavior. Understanding this mechanical cooperation will provide key insights into organ development, regeneration, and tumor metastasis.
Collapse
Affiliation(s)
- Bao-Qiong Lan
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| |
Collapse
|
49
|
Upadhyay A, Bakkalci D, Micalet A, Butler M, Bergin M, Moeendarbary E, Loizidou M, Cheema U. Dense Collagen I as a Biomimetic Material to Track Matrix Remodelling in Renal Carcinomas. ACS OMEGA 2024; 9:41419-41432. [PMID: 39398183 PMCID: PMC11465592 DOI: 10.1021/acsomega.4c04442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
Aims: Renal tissue is a dynamic biophysical microenvironment, regulating healthy function and influencing tumor development. Matrix remodelling is an iterative process and aberrant tissue repair is prominent in kidney fibrosis and cancer. Biomimetic 3D models recapitulating the collagen composition and mechanical fidelity of native renal tissue were developed to investigate cell-matrix interactions in renal carcinomas. Methods: Collagen I and laminin hydrogels were engineered with renal cancer cells (ACHN and 786-O), which underwent plastic compression to generate dense matrices. Mechanical properties were determined using shear rheology and qPCR determined the gene expression of matrix markers. Results: The shear modulus and phase angle of acellular dense collagen I gels (474 Pa and 10.7) are similar to human kidney samples (1410 Pa and 10.5). After 21 days, 786-O cells softened the dense matrix (∼155 Pa), with collagen IV downregulation and upregulation of matrix metalloproteinases (MMP7 and MMP8). ACHN cells were found to be less invasive and stiffened the matrix to ∼1.25 kPa, with gene upregulation of collagen IV and the cross-linking enzyme LOX. Conclusions: Renal cancer cells remodel their biophysical environment, altering the material properties of tissue stroma in 3D models. These models can generate physiologically relevant stiffness to investigate the different matrix remodelling mechanisms utilized by cancer cells.
Collapse
Affiliation(s)
- Anuja Upadhyay
- UCL
Centre for 3D Models of Health and Disease, Division of Surgery and
Interventional Science, University College
London, Charles Bell House, 43-45 Foley Street, W1W 7TS London, United Kingdom
| | - Deniz Bakkalci
- UCL
Centre for 3D Models of Health and Disease, Division of Surgery and
Interventional Science, University College
London, Charles Bell House, 43-45 Foley Street, W1W 7TS London, United Kingdom
| | - Auxtine Micalet
- UCL
Centre for 3D Models of Health and Disease, Division of Surgery and
Interventional Science, University College
London, Charles Bell House, 43-45 Foley Street, W1W 7TS London, United Kingdom
- Department
of Mechanical Engineering, Roberts Building, University College London, WC1E 6BT London, United Kingdom
| | - Matt Butler
- UCB
Pharma, 216 Bath Road, SL1 3WE Slough, United Kingdom
| | | | - Emad Moeendarbary
- Department
of Mechanical Engineering, Roberts Building, University College London, WC1E 6BT London, United Kingdom
| | - Marilena Loizidou
- Division
of Surgery and Interventional Science, University
College London, Royal
Free Campus, Rowland Hill Street, NW3
2PF London, United
Kingdom
| | - Umber Cheema
- UCL
Centre for 3D Models of Health and Disease, Division of Surgery and
Interventional Science, University College
London, Charles Bell House, 43-45 Foley Street, W1W 7TS London, United Kingdom
| |
Collapse
|
50
|
Klabukov I, Smirnova A, Yakimova A, Kabakov AE, Atiakshin D, Petrenko D, Shestakova VA, Sulina Y, Yatsenko E, Stepanenko VN, Ignatyuk M, Evstratova E, Krasheninnikov M, Sosin D, Baranovskii D, Ivanov S, Shegay P, Kaprin AD. Oncomatrix: Molecular Composition and Biomechanical Properties of the Extracellular Matrix in Human Tumors. JOURNAL OF MOLECULAR PATHOLOGY 2024; 5:437-453. [DOI: 10.3390/jmp5040029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
The extracellular matrix is an organized three-dimensional network of protein-based molecules and other macromolecules that provide structural and biochemical support to tissues. Depending on its biochemical and structural properties, the extracellular matrix influences cell adhesion and signal transduction and, in general, can influence cell differentiation and proliferation through specific mechanisms of chemical and mechanical sensing. The development of body tissues during ontogenesis is accompanied by changes not only in cells but also in the composition and properties of the extracellular matrix. Similarly, tumor development in carcinogenesis is accompanied by a continuous change in the properties of the extracellular matrix of tumor cells, called ‘oncomatrix’, as the tumor matures, from the development of the primary focus to the stage of metastasis. In this paper, the characteristics of the composition and properties of the extracellular matrix of tumor tissues are considered, as well as changes to the composition and properties of the matrix during the evolution of the tumor and metastasis. The extracellular matrix patterns of tumor tissues can be used as biomarkers of oncological diseases as well as potential targets for promising anti-tumor therapies.
Collapse
Affiliation(s)
- Ilya Klabukov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anna Smirnova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Anna Yakimova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Alexander E. Kabakov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Dmitri Atiakshin
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Daria Petrenko
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Victoria A. Shestakova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Yana Sulina
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Elena Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Vasiliy N. Stepanenko
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Michael Ignatyuk
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Evstratova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Michael Krasheninnikov
- Scientific and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Dmitry Sosin
- Center for Strategic Planning and Management of Medical and Biological Health Risks of the FMBA of Russia, 119121 Moscow, Russia
| | - Denis Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Sergey Ivanov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Peter Shegay
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|