1
|
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
|
2
|
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; 16:101261. [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] [MESH Headings] [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
|
3
|
Zhang Y, Ma T, Lu X, Hua H, Wu L, Chen Z. Mechanical mechanics-reclaiming a new battlefield for chronic liver disease. J Adv Res 2025:S2090-1232(25)00346-7. [PMID: 40379238 DOI: 10.1016/j.jare.2025.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/17/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND In the 21st century, significant breakthroughs have been made in the research of chronic liver disease. New biochemical markers of pathogenicity and corresponding drugs continue to emerge. However, current treatment strategies remain unsatisfactory due to complex pathological changes in the liver, including vascular dysfunction, myofibroblast-like transition, and local tissue necrosis in liver sinusoids. These challenges have created an urgent need for innovative, synergistic treatments. Mechanical mechanics is a growing field, with increasing evidence suggesting that mechanical signals play a role similar to that of biochemical markers. These signals influence response speed, conduction intensity, and functional diversity in regulating cell activities. AIM OF REVIEW This review summarizes the three main mechanical characteristics involved in the progression of "liver fibrosis-cirrhosis-hepatocellular carcinoma" and provides an in-depth interpretation of several mechanically-related targets. Finally, current and cutting-edge therapeutic strategies are proposed from a cellular perspective. Despite the many challenges that remain, this review is both relevant and significant.
Collapse
Affiliation(s)
- Yiheng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Tianle Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - XingXing Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Haibing Hua
- Department of Gastroenterology, Jiangyin Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Jiangyin 214400, China.
| | - Li Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhipeng Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
4
|
Park JE, Jo J, Xu K, Lee SA, Han SB, Lee Y, Cho WK, Li B, Kim SH, Kim DH. Attenuated Nuclear Tension Regulates Progerin-Induced Mechanosensitive Nuclear Wrinkling and Chromatin Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2502375. [PMID: 40344643 DOI: 10.1002/advs.202502375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Hutchinson-Gilford progeria syndrome, caused by a mutation in the LMNA gene, leads to increased levels of truncated prelamin A, progerin, in the nuclear membrane. The accumulation of progerin results in defective nuclear morphology and is associated with altered expression of linker of the nucleoskeleton and cytoskeleton complex proteins, which are critical for nuclear signal transduction via molecular coupling between the extranuclear cytoskeleton and lamin-associated nuclear envelope. However, the molecular mechanisms underlying progerin accumulation-induced nuclear deformation and its effects on intranuclear chromosomal organization remain unclear. Here, the spatiotemporal evolution of nuclear wrinkles is analyzed in response to variations in substrate stiffness using a doxycycline-inducible progerin expression system. It is found that cytoskeletal tension regulates the onset of progerin-induced nuclear envelope wrinkling and that the molecular interaction between SUN1 and LMNA controls the actomyosin-dependent attenuation of nuclear tension. Genome-wide analysis of chromatin accessibility and gene expression further suggests that an imbalance in force between the intra- and extranuclear spaces induces nuclear deformation, which specifically regulates progeria-associated gene expression via modification of mechanosensitive signaling pathways. The findings highlight the crucial role of nuclear lamin-cytoskeletal connectivity in bridging nuclear mechanotransduction and the biological aging process.
Collapse
Affiliation(s)
- Ji-Eun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Juhyeon Jo
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Kun Xu
- Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
| | - Sun-Ah Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - YigJi Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Won-Ki Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Bo Li
- Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
- Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, 02841, South Korea
| |
Collapse
|
5
|
Wang Y, Jiang J, Shang K, Xu X, Sun J. Turning "trashed" genomic loci into treasurable sites for integrating chimeric antigen receptors in T and NK cells. Mol Ther 2025; 33:1368-1379. [PMID: 39980196 PMCID: PMC11997492 DOI: 10.1016/j.ymthe.2025.02.028] [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: 07/05/2024] [Revised: 10/11/2024] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Chimeric antigen receptor (CAR)-based immune cell therapy involves genetically engineering immune cells, such as T cells and natural killer (NK) cells, to express CARs that can specifically recognize target antigens. This modification enables T/NK cells to selectively eliminate tumor cells following adoptive transfer. One common approach to stably integrate CARs into the genome of T/NK cells is through retroviral or lentiviral vectors. However, these vectors mediate semi-random gene integration, posing risks such as oncogenic mutations, gene silencing, and variable CAR expression levels. Targeted integration of CAR genes into the specific genomic locus could overcome these limitations, but identifying the optimal integration sites to maximize the safety and efficacy of CAR-T/NK cell products remains a critical question. Improper integration sites may disturb the endogenous genes surrounding the integration sites, raising safety concerns. Additionally, regulatory elements at the integration sites, such as promoters, can influence the expression level of CAR genes, thus affecting the efficacy of CAR-T/NK cells. In this review, we summarized current strategies for selecting integration sites and promoters in the engineering of CAR-T/NK cells to achieve potent anti-tumor efficacy in preclinical studies.
Collapse
Affiliation(s)
- Yajie Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Jie Jiang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Kai Shang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Xiaobao Xu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
6
|
Guan G, Coates DE, Sun Q, Cannon RD, Mei L. Atomic Force Microscopy for Revealing Oncological Nanomechanobiology and Thermodynamics. ACS NANO 2025; 19:10862-10877. [PMID: 40084655 DOI: 10.1021/acsnano.4c14837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Atomic force microscopy (AFM) is powerful nanobiotechnology for characterizing the nanotopographic and nanobiomechanical properties of live cells. Current limitations in AFM analysis of nanomechanobiology include the unjustified selection of nesting indices and filters, leading to the inaccurate reporting of waviness and roughness parameters, and inadequacies in the selection of the mathematical model for the Young's modulus. Critical biomechanical factors such as total deformation energy, elastic energy, and plastic energy are often overlooked. Here we refine and optimize the selection of the nesting index and filters for cellular analysis and develop an artificial intelligence-based classifier that can differentiate between normal and cancer cells. The application of AFM for detecting surface waviness and roughness, further enhanced by artificial intelligence (AI), represents a substantial advancement in cancer diagnostics. Although still in the experimental phase, AFM holds the potential to revolutionize cell biology and oncology by facilitating early cancer detection and advancing precision medicine. Moreover, this study's innovative exploration of the relationship between cellular nanomechanobiology and thermodynamics introduces important perspectives on cancer cell behavior at the nanoscale, unlocking opportunities for therapeutic interventions and cutting-edge oncological research. This paradigm shift may significantly influence the future trajectory of cancer biology and therapy.
Collapse
Affiliation(s)
- Guangzhao Guan
- Department of Oral Diagnostic and Surgical Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Dawn E Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9054, New Zealand
| | - Qing Sun
- Department of Oral Diagnostic and Surgical Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Richard D Cannon
- Department of Oral Sciences, Sir John Walsh Research Institute, University of Otago, Dunedin 9054, New Zealand
| | - Li Mei
- Department of Oral Sciences, Sir John Walsh Research Institute, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
7
|
Chen X, Ji X, Lao Z, Pan B, Qian Y, Yang W. Role of YAP/TAZ in bone diseases: A transductor from mechanics to biology. J Orthop Translat 2025; 51:13-23. [PMID: 39902099 PMCID: PMC11787699 DOI: 10.1016/j.jot.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Wolff's Law and the Mechanostat Theory elucidate how bone tissues detect and convert mechanical stimuli into biological signals, crucial for maintaining bone equilibrium. Abnormal mechanics can lead to diseases such as osteoporosis, osteoarthritis, and nonunion fractures. However, the detailed molecular mechanisms by which mechanical cues are transformed into biological responses in bone remain underexplored. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of bone homeostasis, are instrumental in this process. Emerging research highlights bone cells' ability to sense various mechanical stimuli and relay these signals intracellularly. YAP/TAZ are central in receiving these mechanical cues and converting them into signals that influence bone cell behavior. Abnormal YAP/TAZ activity is linked to several bone pathologies, positioning these proteins as promising targets for new treatments. Thus, this review aims to provide an in-depth examination of YAP/TAZ's critical role in the interpretation of mechanical stimuli to biological signals, with a special emphasis on their involvement in bone cell mechanosensing, mechanotransduction, and mechanoresponse. The translational potential of this article: Clinically, appropriate stress stimulation promotes fracture healing, while bed rest can lead to disuse osteoporosis and excessive stress can cause osteoarthritis or bone spurs. Recent advancements in the understanding of YAP/TAZ-mediated mechanobiological signal transduction in bone diseases have been significant, yet many aspects remain unknown. This systematic review summarizes current research progress, identifies unaddressed areas, and highlights potential future research directions. Advancements in this field facilitate a deeper understanding of the molecular mechanisms underlying bone mechanics regulation and underscore the potential of YAP/TAZ as therapeutic targets for bone diseases such as fractures, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Xing Ji
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
8
|
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
|
9
|
Wu X, Fei W, Shen T, Ye L, Li C, Chu S, Liu M, Cheng X, Qin J. Unveiling the potential of biomechanics in pioneering innovative strategies for cancer therapy. Theranostics 2025; 15:2903-2932. [PMID: 40083943 PMCID: PMC11898300 DOI: 10.7150/thno.108605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 03/16/2025] Open
Abstract
Mechanical force transmission is pivotal in tumor biology, profoundly affecting cancer cell behaviors such as proliferation, metastasis, and resistance to therapy. To explore novel biomechanical-based therapeutic strategies for cancer treatment, this paper deciphers the advances in biomechanical measurement approaches and the impact of biomechanical signals on fundamental oncological processes such as tumor microenvironment remodeling, angiogenesis, metastasis, and drug resistance. Then, the mechanisms of biomechanical signal transduction of tumor cells are demonstrated to identify novel targets for tumor therapy. Additionally, this study proposes a novel tumor treatment strategy, the biomechanical regulation tumor nanotherapeutics, including smart biomaterials designed to disturb mechanical signaling pathways and innovative nanodrugs that interfere transduction of biomechanical signals to improve tumor therapeutic outcomes. These methods mark a departure from conventional pharmacological therapies to novel strategies that utilize mechanical forces to impede tumor progression and enhance tumor responsiveness to treatment. In general, this review highlights the critical role of biomechanical signals in cancer biology from a holistic perspective and underscores the potential of biomechanical interventions as a transformative class of therapeutics. By integrating mechanobiology into the development of cancer treatments, this paper paves the way for more precise and effective strategies that leverage the inherent physical properties of the tumor microenvironment.
Collapse
Affiliation(s)
- Xiaodong Wu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Weidong Fei
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Tao Shen
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lei Ye
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chaoqun Li
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Siran Chu
- Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Mingqi Liu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiaodong Cheng
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou, 310006, China
| | - Jiale Qin
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou, 310006, China
| |
Collapse
|
10
|
Clevenger AJ, Collier CA, Gorley JPM, Colijn S, McFarlin MK, Solberg SC, Kopetz ES, Stratman AN, Raghavan SA. Oncogenic KRAS Mutations Confer a Unique Mechanotransduction Response to Peristalsis in Colorectal Cancer Cells. Mol Cancer Res 2025; 23:128-142. [PMID: 39485528 PMCID: PMC11802306 DOI: 10.1158/1541-7786.mcr-24-0624] [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: 09/11/2024] [Revised: 10/04/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Colorectal cancer tumors start as polyps on the inner lining of the colorectum, in which they are exposed to the mechanics of peristalsis. Our previous work leveraged a custom-built peristalsis bioreactor to demonstrate that colonic peristalsis led to cancer stem cell enrichment in colorectal cancer cells. However, this malignant mechanotransductive response was confined to select colorectal cancer lines that harbored an oncogenic mutation in the Kirsten rat sarcoma virus (KRAS) gene. In this study, we explored the involvement of activating KRAS mutations on peristalsis-associated mechanotransduction in colorectal cancer. Peristalsis enriched cancer stem cell marker Leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) in KRAS mutant lines in a Wnt ligand-independent manner. Conversely, LGR5 enrichment in wild-type KRAS lines exposed to peristalsis were minimal. LGR5 enrichment downstream of peristalsis translated to increased tumorigenicity in vivo. Differences in mechanotransduction were apparent via unbiased gene set enrichment analysis, in which many unique pathways were enriched in wild-type versus mutant lines. Peristalsis also triggered β-catenin nuclear localization independent of Wnt ligands, particularly in KRAS mutant lines. The involvement of KRAS was validated via gain and loss of function strategies. Peristalsis-induced β-catenin activation and LGR5 enrichment depended on the activation of the MEK/ERK cascade. Taken together, our results demonstrated that oncogenic KRAS mutations conferred a unique peristalsis-associated mechanotransduction response to colorectal cancer cells, resulting in cancer stem cell enrichment and increased tumorigenicity. These mechanosensory connections can be leveraged in improving the sensitivity of emerging therapies that target oncogenic KRAS. Implications: Oncogenic KRAS empowers colorectal cancer cells to harness the mechanics of colonic peristalsis for malignant gain independent of other cooperating signals.
Collapse
Affiliation(s)
| | - Claudia A. Collier
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - John Paul M. Gorley
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - Sarah Colijn
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Maygan K. McFarlin
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - Spencer C. Solberg
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - E. Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX
| | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Shreya A. Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| |
Collapse
|
11
|
Monaghan-Benson E, Aureille J, Guilluy C. ECM stiffness regulates lung fibroblast survival through RasGRF1-dependent signaling. J Biol Chem 2025; 301:108161. [PMID: 39793891 PMCID: PMC11835592 DOI: 10.1016/j.jbc.2025.108161] [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: 07/03/2024] [Revised: 12/20/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Extracellular matrix stiffness is one of the multiple mechanical signals that alter cellular behavior. During studies exploring the effect of matrix rigidity on lung fibroblast survival, we discovered that enhanced survival on stiff substrates is dependent on elevated Ras activity, owing to the activation of the guanine nucleotide exchange factor, RasGRF1. Mechanistically, we found that the increased Ras activity lead to the activation of both the AKT and ERK pathways. Pharmacological inhibition of AKT or ERK signaling attenuates the elevated survival observed on stiff substrates. AKT signaling regulates the phosphorylation and inactivation of the transcription factor FOXO3a. RNAi experiments demonstrate that FOXO3a activity is critical for the cell death observed on soft substrates. Additionally, downregulation of FOXO3a activity on stiff substrate leads to the degradation of the proapoptotic protein Bim. Depletion of Bim increased the survival of cells on soft substrates. Together, our data show that enhanced matrix stiffness activates a RasGRF1/Ras signaling cascade that regulates the activity of AKT and ERK-dependent FOXO3a and Bim expression to alter cell survival.
Collapse
Affiliation(s)
- Elizabeth Monaghan-Benson
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Julien Aureille
- Institute for Advanced Biosciences Centre de recherche UGA, INSERM U1209, CNRS UMR, Grenoble, France
| | - Christophe Guilluy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
12
|
Dolgitzer D, Plaza-Rodríguez AI, Iglesias MA, Jacob MAC, Todd BA, Robinson DN, Iglesias PA. A continuum model of mechanosensation based on contractility kit assembly. Biophys J 2025; 124:62-76. [PMID: 39521955 PMCID: PMC11739882 DOI: 10.1016/j.bpj.2024.10.020] [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: 07/30/2024] [Revised: 10/07/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
The ability of cells to sense and respond to mechanical forces is crucial for navigating their environment and interacting with neighboring cells. Myosin II and cortexillin I form complexes known as contractility kits (CKs) in the cytosol, which facilitate a cytoskeletal response by accumulating locally at the site of inflicted stress. Here, we present a computational model for mechanoresponsiveness in Dictyostelium, analyzing the role of CKs within the mechanoresponsive mechanism grounded in experimentally measured parameters. Our model further elaborates on the established distributions and channeling of contractile proteins before and after mechanical force application. We rigorously validate our computational findings by comparing the responses of wild-type cells, null mutants, overexpression mutants, and cells deficient in CK formation to mechanical stresses. Parallel in vivo experiments measuring myosin II cortical distributions at equilibrium provide additional validation. Our results highlight the essential functions of CKs in cellular mechanosensitivity and suggest new insights into the regulatory dynamics of mechanoresponsiveness.
Collapse
Affiliation(s)
- David Dolgitzer
- Department of Physics and Astronomy, The Johns Hopkins University, Baltimore, Maryland.
| | - Alma I Plaza-Rodríguez
- Oncology-Quantitative Sciences Department, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Miguel A Iglesias
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey
| | - Mark Allan C Jacob
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Bethany A Todd
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Pablo A Iglesias
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Electrical and Computer Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
13
|
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
|
14
|
Cassani M, Fernandes S, Pagliari S, Cavalieri F, Caruso F, Forte G. Unraveling the Role of the Tumor Extracellular Matrix to Inform Nanoparticle Design for Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409898. [PMID: 39629891 PMCID: PMC11727388 DOI: 10.1002/advs.202409898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Indexed: 01/14/2025]
Abstract
The extracellular matrix (ECM)-and its mechanobiology-regulates key cellular functions that drive tumor growth and development. Accordingly, mechanotherapy is emerging as an effective approach to treat fibrotic diseases such as cancer. Through restoring the ECM to healthy-like conditions, this treatment aims to improve tissue perfusion, facilitating the delivery of chemotherapies. In particular, the manipulation of ECM is gaining interest as a valuable strategy for developing innovative treatments based on nanoparticles (NPs). However, further progress is required; for instance, it is known that the presence of a dense ECM, which hampers the penetration of NPs, primarily impacts the efficacy of nanomedicines. Furthermore, most 2D in vitro studies fail to recapitulate the physiological deposition of matrix components. To address these issues, a comprehensive understanding of the interactions between the ECM and NPs is needed. This review focuses on the main features of the ECM and its complex interplay with NPs. Recent advances in mechanotherapy are discussed and insights are offered into how its combination with nanomedicine can help improve nanomaterials design and advance their clinical translation.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Stefania Pagliari
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
15
|
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
|
16
|
Martin P, Pardo-Pastor C, Jenkins RG, Rosenblatt J. Imperfect wound healing sets the stage for chronic diseases. Science 2024; 386:eadp2974. [PMID: 39636982 PMCID: PMC7617408 DOI: 10.1126/science.adp2974] [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: 09/08/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Although the age of the genome gave us much insight about how our organs fail with disease, it also suggested that diseases do not arise from mutations alone; rather, they develop as we age. In this Review, we examine how wound healing might act to ignite disease. Wound healing works well when we are younger, repairing damage from accidents, environmental assaults, and battles with pathogens. Yet, with age and accumulation of mutations and tissue damage, the repair process can devolve, leading to inflammation, fibrosis, and neoplastic signaling. We discuss healthy wound responses and how our bodies might misappropriate these pathways in disease. Although we focus predominantly on epithelial-based (lung and skin) diseases, similar pathways might operate in cardiac, muscle, and neuronal diseases.
Collapse
Affiliation(s)
- Paul Martin
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Carlos Pardo-Pastor
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - R Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart & Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London, UK
| | - Jody Rosenblatt
- The Randall and Cancer Centres King's College London, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
17
|
Faure LM, Gómez‐González M, Baguer O, Comelles J, Martínez E, Arroyo M, Trepat X, Roca‐Cusachs P. 3D Micropatterned Traction Force Microscopy: A Technique to Control 3D Cell Shape While Measuring Cell-Substrate Force Transmission. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406932. [PMID: 39443837 PMCID: PMC11633470 DOI: 10.1002/advs.202406932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/20/2024] [Indexed: 10/25/2024]
Abstract
Cell shape and function are intimately linked, in a way that is mediated by the forces exerted between cells and their environment. The relationship between cell shape and forces has been extensively studied for cells seeded on flat 2D substrates, but not for cells in more physiological 3D settings. Here, a technique called 3D micropatterned traction force microscopy (3D-µTFM) to confine cells in 3D wells of defined shape, while simultaneously measuring the forces transmitted between cells and their microenvironment is demonstrated. This technique is based on the 3D micropatterning of polyacrylamide wells and on the calculation of 3D traction force from their deformation. With 3D-µTFM, it is shown that MCF10A breast epithelial cells exert defined, reproducible patterns of forces on their microenvironment, which can be both contractile and extensile. Cells switch from a global contractile to extensile behavior as their volume is reduced are further shown. The technique enables the quantitative study of cell mechanobiology with full access to 3D cellular forces while having accurate control over cell morphology and the mechanical conditions of the microenvironment.
Collapse
Affiliation(s)
- Laura M. Faure
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
| | - Manuel Gómez‐González
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
| | - Ona Baguer
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
- Department of Biomedical SciencesUniversity of BarcelonaC. Casanova 143Barcelona08034Spain
| | - Jordi Comelles
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
- Department of Electronics and Biomedical EngineeringUniversity of BarcelonaC. Martí Franquès 1Barcelona08028Spain
| | - Elena Martínez
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
- Department of Electronics and Biomedical EngineeringUniversity of BarcelonaC. Martí Franquès 1Barcelona08028Spain
- Centro de Investigación Biomédica en Red en BioingenieríaBiomateriales y Nanomedicina (CIBER‐BBN)Avenida Monforte de Lemos 3‐5Madrid28029Spain
| | - Marino Arroyo
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
- Laboratori de Càlcul Numèric (LaCàN)Universitat Politècnica de Catalunya (UPC)Jordi Girona 1‐3Barcelona08036Spain
- Institut de Matemàtiques de la UPC–BarcelonaTech (IMTech)Pau Gargallo 14Barcelona08028Spain
- Centre Internacional de Mètodes Numèrics en Enginyeria (CIMNE)Gran Capità S/NBarcelona08034Spain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
- Department of Biomedical SciencesUniversity of BarcelonaC. Casanova 143Barcelona08034Spain
- Centro de Investigación Biomédica en Red en BioingenieríaBiomateriales y Nanomedicina (CIBER‐BBN)Avenida Monforte de Lemos 3‐5Madrid28029Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Lluis Companys 23Barcelona08010Spain
| | - Pere Roca‐Cusachs
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)C. Baldiri Reixac 10‐12Barcelona08028Spain
- Department of Biomedical SciencesUniversity of BarcelonaC. Casanova 143Barcelona08034Spain
| |
Collapse
|
18
|
Yu S, Wang S, Wang X, Xu X. The axis of tumor-associated macrophages, extracellular matrix proteins, and cancer-associated fibroblasts in oncogenesis. Cancer Cell Int 2024; 24:335. [PMID: 39375726 PMCID: PMC11459962 DOI: 10.1186/s12935-024-03518-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024] Open
Abstract
The extracellular matrix (ECM) is a complex, dynamic network of multiple macromolecules that serve as a crucial structural and physical scaffold for neighboring cells. In the tumor microenvironment (TME), ECM proteins play a significant role in mediating cellular communication between cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). Revealing the ECM modification of the TME necessitates the intricate signaling cascades that transpire among diverse cell populations and ECM proteins. The advent of single-cell sequencing has enabled the identification and refinement of specific cellular subpopulations, which has substantially enhanced our comprehension of the intricate milieu and given us a high-resolution perspective on the diversity of ECM proteins. However, it is essential to integrate single-cell data and establish a coherent framework. In this regard, we present a comprehensive review of the relationships among ECM, TAMs, and CAFs. This encompasses insights into the ECM proteins released by TAMs and CAFs, signaling integration in the TAM-ECM-CAF axis, and the potential applications and limitations of targeted therapies for CAFs. This review serves as a reliable resource for focused therapeutic strategies while highlighting the crucial role of ECM proteins as intermediates in the TME.
Collapse
Affiliation(s)
- Shuhong Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Siyu Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xuanyu Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ximing Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
19
|
Tan M, Song B, Zhao X, Du J. The role and mechanism of compressive stress in tumor. Front Oncol 2024; 14:1459313. [PMID: 39351360 PMCID: PMC11439826 DOI: 10.3389/fonc.2024.1459313] [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: 07/04/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Recent research has revealed the important role of mechanical forces in the initiation and progression of tumors. The interplay between mechanical and biochemical cues affects the function and behavior of tumor cells during the development of solid tumors, especially their metastatic potential. The compression force generated by excessive cell proliferation and the tumor microenvironment widely regulates the progression of solid tumor disease. Tumor cells can sense alterations in compressive stress through diverse mechanosensitive components and adapt their mechanical characteristics accordingly to adapt to environmental changes. Here, we summarize the current role of compressive stress in regulating tumor behavior and its biophysical mechanism from the mechanobiological direction.
Collapse
Affiliation(s)
- Min Tan
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Bingqi Song
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xinbin Zhao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
| | - Jing Du
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
20
|
Villares E, Gerecht S. Engineered Biomaterials and Model Systems to Study YAP/TAZ in Cancer. ACS Biomater Sci Eng 2024; 10:5550-5561. [PMID: 39190867 DOI: 10.1021/acsbiomaterials.4c01170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The transcriptional coactivators yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are master regulators involved in a multitude of cancer types and a wide range of tumorigenic events, including cancer stem cell renewal, invasion, metastasis, tumor precursor emergence, and drug resistance. YAP/TAZ are known to be regulated by several external cues and stimuli, such as extracellular matrix stiffness, cell spreading, cell geometry, and shear stress. Therefore, there is a need in the field of cancer research to develop and design relevant in vitro models that can accurately reflect the complex biochemical and biophysical cues of the tumor microenvironment central to the YAP/TAZ signaling nexus. While much progress has been made, this remains a major roadblock to advancing research in this field. In this review, we highlight the current engineered biomaterials and in vitro model systems that can be used to advance our understanding of how YAP/TAZ shapes several aspects of cancer. We begin by discussing current 2D and 3D hydrogel systems that model the YAP/TAZ response to ECM stiffness. We then examine the current trends in organoid culture systems and the use of microfluidics to model the effects of cellular density and shear stress on YAP/TAZ. Finally, we analyze the ongoing pitfalls of the present models used and important future directions in engineering systems that will advance our current knowledge of YAP/TAZ in cancer.
Collapse
Affiliation(s)
- Emma Villares
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| |
Collapse
|
21
|
Karunasagara S, Taghizadeh A, Kim SH, Kim SJ, Kim YJ, Taghizadeh M, Kim MY, Oh KY, Lee JH, Kim HS, Hyun J, Kim HW. Tissue Mechanics and Hedgehog Signaling Crosstalk as a Key Epithelial-Stromal Interplay in Cancer Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400063. [PMID: 38976559 PMCID: PMC11425211 DOI: 10.1002/advs.202400063] [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: 01/03/2024] [Revised: 05/30/2024] [Indexed: 07/10/2024]
Abstract
Epithelial-stromal interplay through chemomechanical cues from cells and matrix propels cancer progression. Elevated tissue stiffness in potentially malignant tissues suggests a link between matrix stiffness and enhanced tumor growth. In this study, employing chronic oral/esophageal injury and cancer models, it is demonstrated that epithelial-stromal interplay through matrix stiffness and Hedgehog (Hh) signaling is key in compounding cancer development. Epithelial cells actively interact with fibroblasts, exchanging mechanoresponsive signals during the precancerous stage. Specifically, epithelial cells release Sonic Hh, activating fibroblasts to produce matrix proteins and remodeling enzymes, resulting in tissue stiffening. Subsequently, basal epithelial cells adjacent to the stiffened tissue become proliferative and undergo epithelial-to-mesenchymal transition, acquiring migratory and invasive properties, thereby promoting invasive tumor growth. Notably, transcriptomic programs of oncogenic GLI2, mechano-activated by actin cytoskeletal tension, govern this process, elucidating the crucial role of non-canonical GLI2 activation in orchestrating the proliferation and mesenchymal transition of epithelial cells. Furthermore, pharmacological intervention targeting tissue stiffening proves highly effective in slowing cancer progression. These findings underscore the impact of epithelial-stromal interplay through chemo-mechanical (Hh-stiffness) signaling in cancer development, and suggest that targeting tissue stiffness holds promise as a strategy to disrupt chemo-mechanical feedback, enabling effective cancer treatment.
Collapse
Affiliation(s)
- Shanika Karunasagara
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Ali Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Sang-Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Chemistry, College of Science & Technology, Dankook University, Cheonan, 31116, Republic of Korea
| | - So Jung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Yong-Jae Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Mohsen Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Moon-Young Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Kyu-Young Oh
- Department of Oral Pathology, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
22
|
Moro-López M, Farré R, Otero J, Sunyer R. Trusting the forces of our cell lines. Cells Dev 2024; 179:203931. [PMID: 38852676 DOI: 10.1016/j.cdev.2024.203931] [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: 02/29/2024] [Revised: 05/03/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Cells isolated from their native tissues and cultured in vitro face different selection pressures than those cultured in vivo. These pressures induce a profound transformation that reshapes the cell, alters its genome, and transforms the way it senses and generates forces. In this perspective, we focus on the evidence that cells cultured on conventional polystyrene substrates display a fundamentally different mechanobiology than their in vivo counterparts. We explore the role of adhesion reinforcement in this transformation and to what extent it is reversible. We argue that this mechanoadaptation is often understood as a mechanical memory. We propose some strategies to mitigate the effects of on-plastic culture on mechanobiology, such as organoid-inspired protocols or mechanical priming. While isolating cells from their native tissues and culturing them on artificial substrates has revolutionized biomedical research, it has also transformed cellular forces. Only by understanding and controlling them, we can improve their truthfulness and validity.
Collapse
Affiliation(s)
- Marina Moro-López
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-RES), Barcelona, Spain; Institut Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Jorge Otero
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-RES), Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sunyer
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería (CIBER-BBN), Barcelona, Spain.
| |
Collapse
|
23
|
Papavassiliou KA, Gargalionis AN, Papavassiliou AG. Direct YAP/TAZ-TEAD inhibitor paves the way toward realizing cancer mechanomedicine. Pharmacol Res 2024; 206:107287. [PMID: 38944219 DOI: 10.1016/j.phrs.2024.107287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024]
Affiliation(s)
- Kostas A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios N Gargalionis
- Laboratory of Clinical Biochemistry, 'Attikon' University General Hospital, Medical School, National and Kapodistrian University of Athens, Chaidari, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
24
|
Graham K, Lienau P, Bader B, Prechtl S, Naujoks J, Lesche R, Weiske J, Kuehnlenz J, Brzezinka K, Potze L, Zanconato F, Nicke B, Montebaur A, Bone W, Golfier S, Kaulfuss S, Kopitz C, Pilari S, Steuber H, Hayat S, Kamburov A, Steffen A, Schlicker A, Buchgraber P, Braeuer N, Font NA, Heinrich T, Kuhnke L, Nowak-Reppel K, Stresemann C, Steigemann P, Walter AO, Blotta S, Ocker M, Lakner A, von Nussbaum F, Mumberg D, Eis K, Piccolo S, Lange M. Discovery of YAP1/TAZ pathway inhibitors through phenotypic screening with potent anti-tumor activity via blockade of Rho-GTPase signaling. Cell Chem Biol 2024; 31:1247-1263.e16. [PMID: 38537632 DOI: 10.1016/j.chembiol.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/08/2024] [Accepted: 02/27/2024] [Indexed: 07/21/2024]
Abstract
This study describes the identification and target deconvolution of small molecule inhibitors of oncogenic Yes-associated protein (YAP1)/TAZ activity with potent anti-tumor activity in vivo. A high-throughput screen (HTS) of 3.8 million compounds was conducted using a cellular YAP1/TAZ reporter assay. Target deconvolution studies identified the geranylgeranyltransferase-I (GGTase-I) complex as the direct target of YAP1/TAZ pathway inhibitors. The small molecule inhibitors block the activation of Rho-GTPases, leading to subsequent inactivation of YAP1/TAZ and inhibition of cancer cell proliferation in vitro. Multi-parameter optimization resulted in BAY-593, an in vivo probe with favorable PK properties, which demonstrated anti-tumor activity and blockade of YAP1/TAZ signaling in vivo.
Collapse
Affiliation(s)
- Keith Graham
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Philip Lienau
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Benjamin Bader
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Stefan Prechtl
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Jan Naujoks
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Ralf Lesche
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Joerg Weiske
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Julia Kuehnlenz
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Krzysztof Brzezinka
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Lisette Potze
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Francesca Zanconato
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Barbara Nicke
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Anna Montebaur
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Wilhelm Bone
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Sven Golfier
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Stefan Kaulfuss
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Charlotte Kopitz
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Sabine Pilari
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Holger Steuber
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Sikander Hayat
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Atanas Kamburov
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Andreas Steffen
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Andreas Schlicker
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Philipp Buchgraber
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Nico Braeuer
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Nuria Aiguabella Font
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Tobias Heinrich
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Lara Kuhnke
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Katrin Nowak-Reppel
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Carlo Stresemann
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Patrick Steigemann
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Annette O Walter
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Simona Blotta
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Matthias Ocker
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Ashley Lakner
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Franz von Nussbaum
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Dominik Mumberg
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Knut Eis
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy; IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Martin Lange
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany.
| |
Collapse
|
25
|
Chang Z, Li LY, Shi ZJ, Liu W, Xu GK. Beyond stiffness: Multiscale viscoelastic features as biomechanical markers for assessing cell types and states. Biophys J 2024; 123:1869-1881. [PMID: 38835167 PMCID: PMC11267428 DOI: 10.1016/j.bpj.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
Cell mechanics are pivotal in regulating cellular activities, diseases progression, and cancer development. However, the understanding of how cellular viscoelastic properties vary in physiological and pathological stimuli remains scarce. Here, we develop a hybrid self-similar hierarchical theory-microrheology approach to accurately and efficiently characterize cellular viscoelasticity. Focusing on two key cell types associated with livers fibrosis-the capillarized liver sinusoidal endothelial cells and activated hepatic stellate cells-we uncover a universal two-stage power-law rheology characterized by two distinct exponents, αshort and αlong. The mechanical profiles derived from both exponents exhibit significant potential for discriminating among diverse cells. This finding suggests a potential common dynamic creep characteristic across biological systems, extending our earlier observations in soft tissues. Using a tailored hierarchical model for cellular mechanical structures, we discern significant variations in the viscoelastic properties and their distribution profiles across different cell types and states from the cytoplasm (elastic stiffness E1 and viscosity η), to a single cytoskeleton fiber (elastic stiffness E2), and then to the cell level (transverse expansion stiffness E3). Importantly, we construct a logistic-regression-based machine-learning model using the dynamic parameters that outperforms conventional cell-stiffness-based classifiers in assessing cell states, achieving an area under the curve of 97% vs. 78%. Our findings not only advance a robust framework for monitoring intricate cell dynamics but also highlight the crucial role of cellular viscoelasticity in discerning cell states across a spectrum of liver diseases and prognosis, offering new avenues for developing diagnostic and therapeutic strategies based on cellular viscoelasticity.
Collapse
Affiliation(s)
- Zhuo Chang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Li-Ya Li
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhi-Jun Shi
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Wenjia Liu
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Guang-Kui Xu
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
26
|
Mancini A, Gentile MT, Pentimalli F, Cortellino S, Grieco M, Giordano A. Multiple aspects of matrix stiffness in cancer progression. Front Oncol 2024; 14:1406644. [PMID: 39015505 PMCID: PMC11249764 DOI: 10.3389/fonc.2024.1406644] [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: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
The biophysical and biomechanical properties of the extracellular matrix (ECM) are crucial in the processes of cell differentiation and proliferation. However, it is unclear to what extent tumor cells are influenced by biomechanical and biophysical changes of the surrounding microenvironment and how this response varies between different tumor forms, and over the course of tumor progression. The entire ensemble of genes encoding the ECM associated proteins is called matrisome. In cancer, the ECM evolves to become highly dysregulated, rigid, and fibrotic, serving both pro-tumorigenic and anti-tumorigenic roles. Tumor desmoplasia is characterized by a dramatic increase of α-smooth muscle actin expressing fibroblast and the deposition of hard ECM containing collagen, fibronectin, proteoglycans, and hyaluronic acid and is common in many solid tumors. In this review, we described the role of inflammation and inflammatory cytokines, in desmoplastic matrix remodeling, tumor state transition driven by microenvironment forces and the signaling pathways in mechanotransduction as potential targeted therapies, focusing on the impact of qualitative and quantitative variations of the ECM on the regulation of tumor development, hypothesizing the presence of matrisome drivers, acting alongside the cell-intrinsic oncogenic drivers, in some stages of neoplastic progression and in some tumor contexts, such as pancreatic carcinoma, breast cancer, lung cancer and mesothelioma.
Collapse
Affiliation(s)
- Alessandro Mancini
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- BioUp Sagl, Lugano, Switzerland
| | - Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University “Giuseppe De Gennaro,” Casamassima, Bari, Italy
| | - Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, NA, Italy
- Sbarro Health Research Organization (S.H.R.O.) Italia Foundation ETS, Candiolo, TO, Italy
| | - Michele Grieco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
27
|
Liu Y, Okesola BO, Osuna de la Peña D, Li W, Lin M, Trabulo S, Tatari M, Lawlor RT, Scarpa A, Wang W, Knight M, Loessner D, Heeschen C, Mata A, Pearce OMT. A Self-Assembled 3D Model Demonstrates How Stiffness Educates Tumor Cell Phenotypes and Therapy Resistance in Pancreatic Cancer. Adv Healthc Mater 2024; 13:e2301941. [PMID: 38471128 PMCID: PMC11468796 DOI: 10.1002/adhm.202301941] [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: 08/17/2023] [Revised: 01/16/2024] [Indexed: 03/14/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense and stiff extracellular matrix (ECM) associated with tumor progression and therapy resistance. To further the understanding of how stiffening of the tumor microenvironment (TME) contributes to aggressiveness, a three-dimensional (3D) self-assembling hydrogel disease model is developed based on peptide amphiphiles (PAs, PA-E3Y) designed to tailor stiffness. The model displays nanofibrous architectures reminiscent of native TME and enables the study of the invasive behavior of PDAC cells. Enhanced tuneability of stiffness is demonstrated by interacting thermally annealed aqueous solutions of PA-E3Y (PA-E3Yh) with divalent cations to create hydrogels with mechanical properties and ultrastructure similar to native tumor ECM. It is shown that stiffening of PA-E3Yh hydrogels to levels found in PDAC induces ECM deposition, promotes epithelial-to-mesenchymal transition (EMT), enriches CD133+/CXCR4+ cancer stem cells (CSCs), and subsequently enhances drug resistance. The findings reveal how a stiff 3D environment renders PDAC cells more aggressive and therefore more faithfully recapitulates in vivo tumors.
Collapse
Affiliation(s)
- Ying Liu
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Babatunde O. Okesola
- School of Life SciencesFaculty of Medicine and Health SciencesUniversity of NottinghamNottinghamNG7 2RDUK
| | - David Osuna de la Peña
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Weiqi Li
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Meng‐Lay Lin
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Sara Trabulo
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Marianthi Tatari
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Rita T. Lawlor
- Department of Diagnostics and Public HealthSection of PathologyUniversity of VeronaVerona37134Italy
- ARC‐NetApplied Research on Cancer CentreUniversity of VeronaVerona37134Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public HealthSection of PathologyUniversity of VeronaVerona37134Italy
- ARC‐NetApplied Research on Cancer CentreUniversity of VeronaVerona37134Italy
| | - Wen Wang
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Martin Knight
- Centre for BioengineeringSchool of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
- Centre for Predictive in vitro ModelsQueen Mary University of LondonLondonE1 4NSUK
| | - Daniela Loessner
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
- Department of Chemical and Biological EngineeringFaculty of EngineeringMonash UniversityMelbourneVIC3800Australia
- Department of Materials Science and EngineeringFaculty of EngineeringMonash UniversityMelbourneVIC3800Australia
- Department of Anatomy and Developmental BiologyFaculty of MedicineNursing and Health SciencesMonash UniversityMelbourneVIC3800Australia
| | - Christopher Heeschen
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute – FPO – IRCCSCandiolo (TO)10060Italy
| | - Alvaro Mata
- School of PharmacyUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
- Biodiscovery InstituteUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
- Department of Chemical and Environmental EngineeringUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
| | | |
Collapse
|
28
|
Gupta P, Kayal S, Tanimura N, Pothapragada SP, Senapati HK, Devendran P, Fujita Y, Bi D, Das T. Mechanical imbalance between normal and transformed cells drives epithelial homeostasis through cell competition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.27.559723. [PMID: 37961252 PMCID: PMC10635021 DOI: 10.1101/2023.09.27.559723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Cell competition in epithelial tissue eliminates transformed cells expressing activated oncoproteins to maintain epithelial homeostasis. Although the process is now understood to be of mechanochemical origin, direct mechanical characterization and associated biochemical underpinnings are lacking. Here, we employ tissue-scale stress and compressibility measurements and theoretical modeling to unveil a mechanical imbalance between normal and transformed cells, which drives cell competition. In the mouse intestinal epithelium and epithelial monolayer, transformed cells get compacted during competition. Stress microscopy reveals an emergent compressive stress at the transformed loci leading to this compaction. A cell-based self-propelled Voronoi model predicts that this compressive stress originates from a difference in the collective compressibility of the competing populations. A new collective compressibility measurement technique named gel compression microscopy then elucidates a two-fold higher compressibility of the transformed population than the normal population. Mechanistically, weakened cell-cell adhesions due to reduced junctional abundance of E-cadherin in the transformed cells render them collectively more compressible than normal cells. Taken together, our findings unveil a mechanical basis for epithelial homeostasis against oncogenic transformations with implications in epithelial defense against cancer.
Collapse
Affiliation(s)
- Praver Gupta
- Tata Institute of Fundamental Research Hyderabad (TIFRH), Hyderabad 500046, India
| | - Sayantani Kayal
- Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Nobuyuki Tanimura
- Department of Molecular Oncology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto-city, Kyoto 606-8501, Japan
| | - Shilpa P. Pothapragada
- Tata Institute of Fundamental Research Hyderabad (TIFRH), Hyderabad 500046, India
- Present address: Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115. USA
| | - Harish K. Senapati
- Tata Institute of Fundamental Research Hyderabad (TIFRH), Hyderabad 500046, India
- Present address: Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Padmashree Devendran
- Tata Institute of Fundamental Research Hyderabad (TIFRH), Hyderabad 500046, India
| | - Yasuyuki Fujita
- Department of Molecular Oncology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto-city, Kyoto 606-8501, Japan
| | - Dapeng Bi
- Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Tamal Das
- Tata Institute of Fundamental Research Hyderabad (TIFRH), Hyderabad 500046, India
| |
Collapse
|
29
|
Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, Lin D, Wu C. Tumor initiation and early tumorigenesis: molecular mechanisms and interventional targets. Signal Transduct Target Ther 2024; 9:149. [PMID: 38890350 PMCID: PMC11189549 DOI: 10.1038/s41392-024-01848-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 06/20/2024] Open
Abstract
Tumorigenesis is a multistep process, with oncogenic mutations in a normal cell conferring clonal advantage as the initial event. However, despite pervasive somatic mutations and clonal expansion in normal tissues, their transformation into cancer remains a rare event, indicating the presence of additional driver events for progression to an irreversible, highly heterogeneous, and invasive lesion. Recently, researchers are emphasizing the mechanisms of environmental tumor risk factors and epigenetic alterations that are profoundly influencing early clonal expansion and malignant evolution, independently of inducing mutations. Additionally, clonal evolution in tumorigenesis reflects a multifaceted interplay between cell-intrinsic identities and various cell-extrinsic factors that exert selective pressures to either restrain uncontrolled proliferation or allow specific clones to progress into tumors. However, the mechanisms by which driver events induce both intrinsic cellular competency and remodel environmental stress to facilitate malignant transformation are not fully understood. In this review, we summarize the genetic, epigenetic, and external driver events, and their effects on the co-evolution of the transformed cells and their ecosystem during tumor initiation and early malignant evolution. A deeper understanding of the earliest molecular events holds promise for translational applications, predicting individuals at high-risk of tumor and developing strategies to intercept malignant transformation.
Collapse
Affiliation(s)
- Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyi Xiao
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yonglin Yi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyu Wang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Lingxuan Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Changping Laboratory, 100021, Beijing, China
| | - Yanrong Shen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- CAMS Oxford Institute, Chinese Academy of Medical Sciences, 100006, Beijing, China.
| |
Collapse
|
30
|
Mottareale R, Frascogna C, La Verde G, Arrichiello C, Muto P, Netti PA, Fusco S, Panzetta V, Pugliese M. Impact of ionizing radiation on cell-ECM mechanical crosstalk in breast cancer. Front Bioeng Biotechnol 2024; 12:1408789. [PMID: 38903185 PMCID: PMC11187264 DOI: 10.3389/fbioe.2024.1408789] [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: 03/28/2024] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
The stiffness of the extracellular matrix plays a crucial role in cell motility and spreading, influencing cell morphology through cytoskeleton organization and transmembrane proteins' expression. In this context, mechanical characterization of both cells and the extracellular matrix gains prominence for enhanced diagnostics and clinical decision-making. Here, we investigate the combined effect of mechanotransduction and ionizing radiations on altering cells' mechanical properties, analysing mammary cell lines (MCF10A and MDA-MB-231) after X-ray radiotherapy (2 and 10 Gy). We found that ionizing radiations sensitively affect adenocarcinoma cells cultured on substrates mimicking cancerous tissue stiffness (15 kPa), inducing an increased structuration of paxillin-rich focal adhesions and cytoskeleton: this process translates in the augmentation of tension at the actin filaments level, causing cellular stiffness and consequently affecting cytoplasmatic/nuclear morphologies. Deeper exploration of the intricate interplay between mechanical factors and radiation should provide novel strategies to orient clinical outcomes.
Collapse
Affiliation(s)
- Rocco Mottareale
- Department of Physics “E. Pancini”, University of Naples Federico II, Naples, Italy
- Institute of Applied Sciences and Intelligent Systems E. Caianiello (CNR-ISASI), Pozzuoli, Italy
| | - Crescenzo Frascogna
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
| | - Giuseppe La Verde
- Department of Physics “E. Pancini”, University of Naples Federico II, Naples, Italy
| | - Cecilia Arrichiello
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, Naples, Italy
| | - Paolo Muto
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, Naples, Italy
| | - Paolo A. Netti
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Sabato Fusco
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Naples, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Valeria Panzetta
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
31
|
Pratt SJP, Plunkett CM, Kuzu G, Trinh T, Barbara J, Choconta P, Quackenbush D, Huynh T, Smith A, Barnes SW, New J, Pierce J, Walker JR, Mainquist J, King FJ, Elliott J, Hammack S, Decker RS. A high throughput cell stretch device for investigating mechanobiology in vitro. APL Bioeng 2024; 8:026129. [PMID: 38938688 PMCID: PMC11210978 DOI: 10.1063/5.0206852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Mechanobiology is a rapidly advancing field, with growing evidence that mechanical signaling plays key roles in health and disease. To accelerate mechanobiology-based drug discovery, novel in vitro systems are needed that enable mechanical perturbation of cells in a format amenable to high throughput screening. Here, both a mechanical stretch device and 192-well silicone flexible linear stretch plate were designed and fabricated to meet high throughput technology needs for cell stretch-based applications. To demonstrate the utility of the stretch plate in automation and screening, cell dispensing, liquid handling, high content imaging, and high throughput sequencing platforms were employed. Using this system, an assay was developed as a biological validation and proof-of-concept readout for screening. A mechano-transcriptional stretch response was characterized using focused gene expression profiling measured by RNA-mediated oligonucleotide Annealing, Selection, and Ligation with Next-Gen sequencing. Using articular chondrocytes, a gene expression signature containing stretch responsive genes relevant to cartilage homeostasis and disease was identified. The possibility for integration of other stretch sensitive cell types (e.g., cardiovascular, airway, bladder, gut, and musculoskeletal), in combination with alternative phenotypic readouts (e.g., protein expression, proliferation, or spatial alignment), broadens the scope of high throughput stretch and allows for wider adoption by the research community. This high throughput mechanical stress device fills an unmet need in phenotypic screening technology to support drug discovery in mechanobiology-based disease areas.
Collapse
Affiliation(s)
- Stephen J. P. Pratt
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | | | - Guray Kuzu
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Ton Trinh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joshua Barbara
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Paula Choconta
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Doug Quackenbush
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Truc Huynh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Anders Smith
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - S. Whitney Barnes
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joel New
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Pierce
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - John R. Walker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Mainquist
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Frederick J. King
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Jimmy Elliott
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Scott Hammack
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Rebekah S. Decker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| |
Collapse
|
32
|
Yu KX, Yuan WJ, Wang HZ, Li YX. Extracellular matrix stiffness and tumor-associated macrophage polarization: new fields affecting immune exclusion. Cancer Immunol Immunother 2024; 73:115. [PMID: 38693304 PMCID: PMC11063025 DOI: 10.1007/s00262-024-03675-9] [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: 11/12/2023] [Accepted: 03/12/2024] [Indexed: 05/03/2024]
Abstract
In the malignant progression of tumors, there is deposition and cross-linking of collagen, as well as an increase in hyaluronic acid content, which can lead to an increase in extracellular matrix stiffness. Recent research evidence have shown that the extracellular matrix plays an important role in angiogenesis, cell proliferation, migration, immunosuppression, apoptosis, metabolism, and resistance to chemotherapeutic by the alterations toward both secretion and degradation. The clinical importance of tumor-associated macrophage is increasingly recognized, and macrophage polarization plays a central role in a series of tumor immune processes through internal signal cascade, thus regulating tumor progression. Immunotherapy has gradually become a reliable potential treatment strategy for conventional chemotherapy resistance and advanced cancer patients, but the presence of immune exclusion has become a major obstacle to treatment effectiveness, and the reasons for their resistance to these approaches remain uncertain. Currently, there is a lack of exact mechanism on the regulation of extracellular matrix stiffness and tumor-associated macrophage polarization on immune exclusion. An in-depth understanding of the relationship between extracellular matrix stiffness, tumor-associated macrophage polarization, and immune exclusion will help reveal new therapeutic targets and guide the development of clinical treatment methods for advanced cancer patients. This review summarized the different pathways and potential molecular mechanisms of extracellular matrix stiffness and tumor-associated macrophage polarization involved in immune exclusion and provided available strategies to address immune exclusion.
Collapse
Affiliation(s)
- Ke-Xun Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei-Jie Yuan
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Hui-Zhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yong-Xiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
33
|
Lin S, He X, Wang Y, Chen Y, Lin A. Emerging role of lncRNAs as mechanical signaling molecules in mechanotransduction and their association with Hippo-YAP signaling: a review. J Zhejiang Univ Sci B 2024; 25:280-292. [PMID: 38584091 PMCID: PMC11009445 DOI: 10.1631/jzus.b2300497] [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: 07/10/2023] [Accepted: 12/11/2023] [Indexed: 04/09/2024]
Abstract
Cells within tissues are subject to various mechanical forces, including hydrostatic pressure, shear stress, compression, and tension. These mechanical stimuli can be converted into biochemical signals through mechanoreceptors or cytoskeleton-dependent response processes, shaping the microenvironment and maintaining cellular physiological balance. Several studies have demonstrated the roles of Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ) as mechanotransducers, exerting dynamic influence on cellular phenotypes including differentiation and disease pathogenesis. This regulatory function entails the involvement of the cytoskeleton, nucleoskeleton, integrin, focal adhesions (FAs), and the integration of multiple signaling pathways, including extracellular signal-regulated kinase (ERK), wingless/integrated (WNT), and Hippo signaling. Furthermore, emerging evidence substantiates the implication of long non-coding RNAs (lncRNAs) as mechanosensitive molecules in cellular mechanotransduction. In this review, we discuss the mechanisms through which YAP/TAZ and lncRNAs serve as effectors in responding to mechanical stimuli. Additionally, we summarize and elaborate on the crucial signal molecules involved in mechanotransduction.
Collapse
Affiliation(s)
- Siyi Lin
- College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyu He
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Ying Wang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Yu Chen
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
- Cancer Center, Zhejiang University, Hangzhou 310058, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China.
- International School of Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou 310058, China.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
34
|
Coker ZN, Troyanova-Wood M, Steelman ZA, Ibey BL, Bixler JN, Scully MO, Yakovlev VV. Brillouin microscopy monitors rapid responses in subcellular compartments. PHOTONIX 2024; 5:9. [PMID: 38618142 PMCID: PMC11006764 DOI: 10.1186/s43074-024-00123-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/12/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024]
Abstract
Measurements and imaging of the mechanical response of biological cells are critical for understanding the mechanisms of many diseases, and for fundamental studies of energy, signal and force transduction. The recent emergence of Brillouin microscopy as a powerful non-contact, label-free way to non-invasively and non-destructively assess local viscoelastic properties provides an opportunity to expand the scope of biomechanical research to the sub-cellular level. Brillouin spectroscopy has recently been validated through static measurements of cell viscoelastic properties, however, fast (sub-second) measurements of sub-cellular cytomechanical changes have yet to be reported. In this report, we utilize a custom multimodal spectroscopy system to monitor for the very first time the rapid viscoelastic response of cells and subcellular structures to a short-duration electrical impulse. The cytomechanical response of three subcellular structures - cytoplasm, nucleoplasm, and nucleoli - were monitored, showing distinct mechanical changes despite an identical stimulus. Through this pioneering transformative study, we demonstrate the capability of Brillouin spectroscopy to measure rapid, real-time biomechanical changes within distinct subcellular compartments. Our results support the promising future of Brillouin spectroscopy within the broad scope of cellular biomechanics.
Collapse
Affiliation(s)
- Zachary N. Coker
- Department of Physics & Astronomy, Texas A&M University, 4242 TAMU, College Station, TX 77843 USA
- SAIC, Fort Sam Houston, TX 78234 USA
| | | | - Zachary A. Steelman
- Air Force Research Laboratory, JBSA Fort Sam Houston, Fort Sam Houston, TX 78234 USA
| | - Bennett L. Ibey
- Air Force Research Laboratory, JBSA Fort Sam Houston, Fort Sam Houston, TX 78234 USA
| | - Joel N. Bixler
- Air Force Research Laboratory, JBSA Fort Sam Houston, Fort Sam Houston, TX 78234 USA
| | - Marlan O. Scully
- Department of Physics & Astronomy, Texas A&M University, 4242 TAMU, College Station, TX 77843 USA
- Institute for Quantum Science and Engineering, Texas A&M University, College Station, TX 77843 USA
| | - Vladislav V. Yakovlev
- Department of Physics & Astronomy, Texas A&M University, 4242 TAMU, College Station, TX 77843 USA
- Institute for Quantum Science and Engineering, Texas A&M University, College Station, TX 77843 USA
- Department of Biomedical Engineering, Texas A&M University, 3120 TAMU, 101 Bizzell Street, College Station, TX 77843 USA
| |
Collapse
|
35
|
Guerrero-Barberà G, Burday N, Costell M. Shaping Oncogenic Microenvironments: Contribution of Fibronectin. Front Cell Dev Biol 2024; 12:1363004. [PMID: 38660622 PMCID: PMC11039881 DOI: 10.3389/fcell.2024.1363004] [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: 12/29/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins and glycans, dynamically remodeled and specifically tailored to the structure/function of each organ. The malignant transformation of cancer cells is determined by both cell intrinsic properties, such as mutations, and extrinsic variables, such as the mixture of surrounding cells in the tumor microenvironment and the biophysics of the ECM. During cancer progression, the ECM undergoes extensive remodeling, characterized by disruption of the basal lamina, vascular endothelial cell invasion, and development of fibrosis in and around the tumor cells resulting in increased tissue stiffness. This enhanced rigidity leads to aberrant mechanotransduction and further malignant transformation potentiating the de-differentiation, proliferation and invasion of tumor cells. Interestingly, this fibrotic microenvironment is primarily secreted and assembled by non-cancerous cells. Among them, the cancer-associated fibroblasts (CAFs) play a central role. CAFs massively produce fibronectin together with type I collagen. This review delves into the primary interactions and signaling pathways through which fibronectin can support tumorigenesis and metastasis, aiming to provide critical molecular insights for better therapy response prediction.
Collapse
Affiliation(s)
| | | | - Mercedes Costell
- Departament of Biochemistry and Molecular Biology, Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Valencia, Spain
| |
Collapse
|
36
|
Xing H, Li X. Engineered Nanomaterials for Tumor Immune Microenvironment Modulation in Cancer Immunotherapy. Chemistry 2024:e202400425. [PMID: 38576219 DOI: 10.1002/chem.202400425] [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: 01/30/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/06/2024]
Abstract
Tumor immunotherapy, represented by immune checkpoint blocking and chimeric antigen receptor (CAR) T cell therapy, has achieved promising results in clinical applications. However, it faces challenges that hinder its further development, such as limited response rates and poor tumor permeability. The efficiency of tumor immunotherapy is also closely linked to the structure and function of the immune microenvironment where the tumor resides. Recently, nanoparticle-based tumor immune microenvironment (TIME) modulation strategies have attracted a great deal of attention in cancer immunotherapy. This is primarily due to the distinctive physical characteristics of nanoparticles, which enable them to effectively infiltrate the TIME and selectively modulate its key constituents. This paper reviews recent advances in nanoparticle engineering to improve anti-cancer immunotherapy. Emerging nanoparticle-based approaches for modulating immune cells, tumor stroma, cytokines and immune checkpoints are discussed, aiming to overcome current challenges in the clinic. In addition, integrating immunotherapy with various treatment modalities such as chemotherapy and photodynamic therapy can be facilitated through the utilization of nanoparticles, thereby enhancing the efficacy of cancer treatment. The future challenges and opportunities of using nanomaterials to reeducate the suppressive immune microenvironment of tumors are also discussed, with the aim of anticipating further advancements in this growing field.
Collapse
Affiliation(s)
- Hao Xing
- Department of General Surgery, Naval Medical Center, Naval Medical University, 200052, Shanghai, China
- The First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China
| | - Xiaomin Li
- Department of Chemistry, Laboratory of Advanced Materials, College of Chemistry and Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, 200438, Shanghai, China
| |
Collapse
|
37
|
Liu G, Li B, Qin S, Nice EC, Yang J, Yang L, Huang C. Redox signaling-mediated tumor extracellular matrix remodeling: pleiotropic regulatory mechanisms. Cell Oncol (Dordr) 2024; 47:429-445. [PMID: 37792154 DOI: 10.1007/s13402-023-00884-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND The extracellular matrix (ECM), a fundamental constituent of all tissues and organs, is crucial for shaping the tumor microenvironment. Dysregulation of ECM remodeling has been closely linked to tumor initiation and progression, where specific signaling pathways, including redox signaling, play essential roles. Reactive oxygen species (ROS) are risk factors for carcinogenesis whose excess can facilitate the oxidative damage of biomacromolecules, such as DNA and proteins. Emerging evidence suggests that redox effects can aid the modification, stimulation, and degradation of ECM, thus affecting ECM remodeling. These alterations in both the density and components of the ECM subsequently act as critical drivers for tumorigenesis. In this review, we provide an overview of the functions and primary traits of the ECM, and it delves into our current understanding of how redox reactions participate in ECM remodeling during cancer progression. We also discuss the opportunities and challenges presented by clinical strategies targeting redox-controlled ECM remodeling to overcome cancer. CONCLUSIONS The redox-mediated ECM remodeling contributes importantly to tumor survival, progression, metastasis, and poor prognosis. A comprehensive investigation of the concrete mechanism of redox-mediated tumor ECM remodeling and the combination usage of redox-targeted drugs with existing treatment means may reveal new therapeutic strategy for future antitumor therapies.
Collapse
Affiliation(s)
- Guowen Liu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Jinlin Yang
- Department of Gastroenterology & Hepatology, West China Hospital of Sichuan University, Sichuan Province, No.37 Guoxue Alley, Chengdu, 610041, China.
- Department of Gastroenterology & Hepatology, Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Li Yang
- Department of Gastroenterology & Hepatology, West China Hospital of Sichuan University, Sichuan Province, No.37 Guoxue Alley, Chengdu, 610041, China.
- Department of Gastroenterology & Hepatology, Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China.
| |
Collapse
|
38
|
Ogasawara N, Kano Y, Yoneyama Y, Kobayashi S, Watanabe S, Kirino S, Velez-Bravo FD, Hong Y, Ostapiuk A, Lutsik P, Onishi I, Yamauchi S, Hiraguri Y, Ito G, Kinugasa Y, Ohashi K, Watanabe M, Okamoto R, Tejpar S, Yui S. Discovery of non-genomic drivers of YAP signaling modulating the cell plasticity in CRC tumor lines. iScience 2024; 27:109247. [PMID: 38439969 PMCID: PMC10910304 DOI: 10.1016/j.isci.2024.109247] [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: 09/29/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
In normal intestines, a fetal/regenerative/revival cell state can be induced upon inflammation. This plasticity in cell fate is also one of the current topics in human colorectal cancer (CRC). To dissect the underlying mechanisms, we generated human CRC organoids with naturally selected genetic mutation profiles and exposed them to two different conditions by modulating the extracellular matrix (ECM). Among tested mutation profiles, a fetal/regenerative/revival state was induced following YAP activation via a collagen type I-enriched microenvironment. Mechanistically, YAP transcription was promoted by activating AP-1 and TEAD-dependent transcription and suppressing intestinal lineage-determining transcription via mechanotransduction. The phenotypic conversion was also involved in chemoresistance, which could be potentially resolved by targeting the underlying YAP regulatory elements, a potential target of CRC treatment.
Collapse
Affiliation(s)
- Nobuhiko Ogasawara
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yoshihito Kano
- Department of Clinical Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sakurako Kobayashi
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Satoshi Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sakura Kirino
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | - Yourae Hong
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Pavlo Lutsik
- Computational Cancer Biology and Epigenomics, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Iichiroh Onishi
- Department of Diagnostic Pathology, Tokyo Medical and Dental University Hospital, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shinichi Yamauchi
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yui Hiraguri
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Go Ito
- Advanced Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kenichi Ohashi
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mamoru Watanabe
- Advanced Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sabine Tejpar
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Shiro Yui
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
39
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [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: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
40
|
Nakamura F. The Role of Mechanotransduction in Contact Inhibition of Locomotion and Proliferation. Int J Mol Sci 2024; 25:2135. [PMID: 38396812 PMCID: PMC10889191 DOI: 10.3390/ijms25042135] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Contact inhibition (CI) represents a crucial tumor-suppressive mechanism responsible for controlling the unbridled growth of cells, thus preventing the formation of cancerous tissues. CI can be further categorized into two distinct yet interrelated components: CI of locomotion (CIL) and CI of proliferation (CIP). These two components of CI have historically been viewed as separate processes, but emerging research suggests that they may be regulated by both distinct and shared pathways. Specifically, recent studies have indicated that both CIP and CIL utilize mechanotransduction pathways, a process that involves cells sensing and responding to mechanical forces. This review article describes the role of mechanotransduction in CI, shedding light on how mechanical forces regulate CIL and CIP. Emphasis is placed on filamin A (FLNA)-mediated mechanotransduction, elucidating how FLNA senses mechanical forces and translates them into crucial biochemical signals that regulate cell locomotion and proliferation. In addition to FLNA, trans-acting factors (TAFs), which are proteins or regulatory RNAs capable of directly or indirectly binding to specific DNA sequences in distant genes to regulate gene expression, emerge as sensitive players in both the mechanotransduction and signaling pathways of CI. This article presents methods for identifying these TAF proteins and profiling the associated changes in chromatin structure, offering valuable insights into CI and other biological functions mediated by mechanotransduction. Finally, it addresses unanswered research questions in these fields and delineates their possible future directions.
Collapse
Affiliation(s)
- Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
41
|
Jones MJ, Jones MC. Cell cycle control by cell-matrix interactions. Curr Opin Cell Biol 2024; 86:102288. [PMID: 38056140 DOI: 10.1016/j.ceb.2023.102288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
Cell adhesion to the extracellular matrix (ECM) is required for normal cell cycle progression and accurate cell division. However, how cell adhesion to the wide range of ECM proteins found in human tissues influences the cell cycle is not fully understood. The composition and physical properties of the ECM can have profound effects on cell proliferation but can also promote cell cycle exit and quiescence. Furthermore, during tumor development and progression, changes in the ECM can drive both cancer cell proliferation and dormancy. Cell-matrix adhesion is primarily sensed via integrin-associated adhesion complexes, which in turn are regulated by the cell cycle machinery. In particular, cyclin-dependent kinase 1 (CDK1) has been shown to play a crucial role in regulating adhesion complexes during interphase and entry into mitosis. These reciprocal links between cell cycle progression and cell-matrix interactions are now being identified.
Collapse
Affiliation(s)
- Michael J Jones
- Peninsula Medical School, Faculty of Health, Medicine, Dentistry and Human Sciences, University of Plymouth, PL6 8BU, United Kingdom
| | - Matthew C Jones
- Peninsula Medical School, Faculty of Health, Medicine, Dentistry and Human Sciences, University of Plymouth, PL6 8BU, United Kingdom.
| |
Collapse
|
42
|
Strash N, DeLuca S, Janer Carattini GL, Chen Y, Wu T, Helfer A, Scherba J, Wang I, Jain M, Naseri R, Bursac N. Time-dependent effects of BRAF-V600E on cell cycling, metabolism, and function in engineered myocardium. SCIENCE ADVANCES 2024; 10:eadh2598. [PMID: 38266090 PMCID: PMC10807800 DOI: 10.1126/sciadv.adh2598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Candidate cardiomyocyte (CM) mitogens such as those affecting the extracellular signal-regulated kinase (ERK) signaling pathway represent potential targets for functional heart regeneration. We explored whether activating ERK via a constitutively active mutant of B-raf proto-oncogene (BRAF), BRAF-V600E (caBRAF), can induce proproliferative effects in neonatal rat engineered cardiac tissues (ECTs). Sustained CM-specific caBRAF expression induced chronic ERK activation, substantial tissue growth, deficit in sarcomeres and contractile function, and tissue stiffening, all of which persisted for at least 4 weeks of culture. caBRAF-expressing CMs in ECTs exhibited broad transcriptomic changes, shift to glycolytic metabolism, loss of connexin-43, and a promigratory phenotype. Transient, doxycycline-controlled caBRAF expression revealed that the induction of CM cycling is rapid and precedes functional decline, and the effects are reversible only with short-lived ERK activation. Together, direct activation of the BRAF kinase is sufficient to modulate CM cycling and functional phenotype, offering mechanistic insights into roles of ERK signaling in the context of cardiac development and regeneration.
Collapse
Affiliation(s)
| | - Sophia DeLuca
- Department of Cell Biology, Duke University, Durham NC, USA
| | | | - Yifan Chen
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| | - Tianyu Wu
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| | - Abbigail Helfer
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| | - Jacob Scherba
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| | - Isabella Wang
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| | - Mehul Jain
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| | - Ramona Naseri
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| | - Nenad Bursac
- Department of Cell Biology, Duke University, Durham NC, USA
- Department of Biomedical Engineering, Duke University, Durham NC, USA
| |
Collapse
|
43
|
Qiu S, Qiu Y, Deng L, Nie L, Ge L, Zheng X, Jin D, Jin K, Zhou X, Su X, Cai B, Li J, Tu X, Gong L, Liu L, Liu Z, Bao Y, Ai J, Lin T, Yang L, Wei Q. Cell softness reveals tumorigenic potential via ITGB8/AKT/glycolysis signaling in a mice model of orthotopic bladder cancer. Chin Med J (Engl) 2024; 137:209-221. [PMID: 37390491 PMCID: PMC10798691 DOI: 10.1097/cm9.0000000000002710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Bladder cancer, characterized by a high potential of tumor recurrence, has high lifelong monitoring and treatment costs. To date, tumor cells with intrinsic softness have been identified to function as cancer stem cells in several cancer types. Nonetheless, the existence of soft tumor cells in bladder tumors remains elusive. Thus, our study aimed to develop a micro-barrier microfluidic chip to efficiently isolate deformable tumor cells from distinct types of bladder cancer cells. METHODS The stiffness of bladder cancer cells was determined by atomic force microscopy (AFM). The modified microfluidic chip was utilized to separate soft cells, and the 3D Matrigel culture system was to maintain the softness of tumor cells. Expression patterns of integrin β8 (ITGB8), protein kinase B (AKT), and mammalian target of rapamycin (mTOR) were determined by Western blotting. Double immunostaining was conducted to examine the interaction between F-actin and tripartite motif containing 59 (TRIM59). The stem-cell-like characteristics of soft cells were explored by colony formation assay and in vivo studies upon xenografted tumor models. RESULTS Using our newly designed microfluidic approach, we identified a small fraction of soft tumor cells in bladder cancer cells. More importantly, the existence of soft tumor cells was confirmed in clinical human bladder cancer specimens, in which the number of soft tumor cells was associated with tumor relapse. Furthermore, we demonstrated that the biomechanical stimuli arising from 3D Matrigel activated the F-actin/ITGB8/TRIM59/AKT/mTOR/glycolysis pathways to enhance the softness and tumorigenic capacity of tumor cells. Simultaneously, we detected a remarkable up-regulation in ITGB8, TRIM59, and phospho-AKT in clinical bladder recurrent tumors compared with their non-recurrent counterparts. CONCLUSIONS The ITGB8/TRIM59/AKT/mTOR/glycolysis axis plays a crucial role in modulating tumor softness and stemness. Meanwhile, the soft tumor cells become more sensitive to chemotherapy after stiffening, that offers new insights for hampering tumor progression and recurrence.
Collapse
Affiliation(s)
- Shi Qiu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Department of Molecular Oncology, Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Bellinzona 6500, Switzerland
| | - Yaqi Qiu
- Department of Science and Drug Technology, University of Turin, Turin, Italy
| | - Linghui Deng
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liming Ge
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610000, China
| | - Xiaonan Zheng
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Di Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianghong Zhou
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyang Su
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Boyu Cai
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiakun Li
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiang Tu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Lina Gong
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yige Bao
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianhai Lin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
44
|
Frascogna C, Mottareale R, La Verde G, Arrichiello C, Muto P, Netti PA, Pugliese M, Panzetta V. Role of the mechanical microenvironment on CD-44 expression of breast adenocarcinoma in response to radiotherapy. Sci Rep 2024; 14:391. [PMID: 38172135 PMCID: PMC10764959 DOI: 10.1038/s41598-023-50473-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
The biological effects of ionizing radiation are exploited in the clinical practice of radiotherapy to destroy tumour cells while sparing the surrounding normal tissue. While most of the radiotherapy research focused on DNA damage and repair, recently a great attention is going to cells' interactions with the mechanical microenvironment of both malignant and healthy tissues after exposure. In fact, the stiffness of the extracellular matrix can modify cells' motility and spreading through the modulation of transmembrane proteins and surface receptors' expression, such as CD-44. CD-44 receptor has held much interest also in targeted-therapy due to its affinity with hyaluronic acid, which can be used to functionalize biodegradable nanoparticles loaded with chemotherapy drugs for targeted therapy. We evaluated changes in CD-44 expression in two mammary carcinoma cell lines (MCF10A and MDA-MB-231) after exposure to X-ray (2 or 10 Gy). To explore the role of the mechanical microenvironment, we mimicked tissues' stiffness with polyacrylamide's substrates producing two different elastic modulus values (0.5 and 15 kPa). We measured a dose dependent increase in CD-44 relative expression in tumour cells cultured in a stiffer microenvironment. These findings highlight a crucial connection between the mechanical properties of the cell's surroundings and the post-radiotherapy expression of surface receptors.
Collapse
Affiliation(s)
- Crescenzo Frascogna
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
| | - Rocco Mottareale
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
| | - Giuseppe La Verde
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
- Istituto Nazionale di Fisica Nucleare, INFN Sezione di Napoli, Via Cinthia Ed. 6, 80126, Naples, Italy
| | - Cecilia Arrichiello
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 53, 80131, Naples, Italy
| | - Paolo Muto
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 53, 80131, Naples, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
- Interdisciplinary Research Centre On Biomaterials CRIB, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
| | - Mariagabriella Pugliese
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cinthia, 80126, Naples, Italy.
- Istituto Nazionale di Fisica Nucleare, INFN Sezione di Napoli, Via Cinthia Ed. 6, 80126, Naples, Italy.
| | - Valeria Panzetta
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
- Interdisciplinary Research Centre On Biomaterials CRIB, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
| |
Collapse
|
45
|
Selcuk K, Leitner A, Braun L, Le Blanc F, Pacak P, Pot S, Vogel V. Transglutaminase 2 has higher affinity for relaxed than for stretched fibronectin fibers. Matrix Biol 2024; 125:113-132. [PMID: 38135164 DOI: 10.1016/j.matbio.2023.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
Transglutaminase 2 (TG2) plays a vital role in stabilizing extracellular matrix (ECM) proteins through enzymatic crosslinking during tissue growth, repair, and inflammation. TG2 also binds non-covalently to fibronectin (FN), an essential component of the ECM, facilitating cell adhesion, migration, proliferation, and survival. However, the interaction between TG2 and fibrillar FN remains poorly understood, as most studies have focused on soluble or surface-adsorbed FN or FN fragments, which differ in their conformations from insoluble FN fibers. Using a well-established in vitro FN fiber stretch assay, we discovered that the binding of a crosslinking enzyme to ECM fibers is mechano-regulated. TG2 binding to FN is tuned by the mechanical tension of FN fibers, whereby TG2 predominantly co-localizes to low-tension FN fibers, while fiber stretching reduces their affinity for TG2. This mechano-regulated binding relies on the proximity between the N-terminal β-sandwich and C-terminal β-barrels of TG2. Crosslinking mass spectrometry (XL-MS) revealed a novel TG2-FN synergy site within TG2's C-terminal β-barrels that interacts with FN regions located outside of the canonical gelatin binding domain, specifically FNI2 and FNIII14-15. Combining XL-MS distance restraints with molecular docking revealed the mechano-regulated binding mechanism between TG2 and modules FNI7-9 by which mechanical forces regulate TG2-FN interactions. This highlights a previously unrecognized role of TG2 as a tension sensor for FN fibers. This novel interaction mechanism has significant implications in physiology and mechanobiology, including how forces regulate cell adhesion, spreading, migration, phenotype modulation, depending on the tensional state of ECM fibers. Data are available via ProteomeXchange with identifier PXD043976.
Collapse
Affiliation(s)
- Kateryna Selcuk
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Alexander Leitner
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, CH-8093 Zurich, Switzerland
| | - Lukas Braun
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Fanny Le Blanc
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, CH-8093 Zurich, Switzerland
| | - Paulina Pacak
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Simon Pot
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Viola Vogel
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland.
| |
Collapse
|
46
|
Deng H, Shu X, Wang Y, Zhang J, Yin Y, Wu F, He J. Matrix Stiffness Regulated Endoplasmic Reticulum Stress-mediated Apoptosis of Osteosarcoma Cell through Ras Signal Cascades. Cell Biochem Biophys 2023; 81:839-850. [PMID: 37789235 DOI: 10.1007/s12013-023-01184-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/05/2023]
Abstract
The modulating effects of matrix stiffness on spreading and apoptosis of tumor cells have been well recognized. Nevertheless, the detail road map leading to the apoptosis and the underlying mechanisms governing the cell apoptosis have remained to be elucidated. To this aim, we provided a tunable elastic hydrogel matrix that promoted cell adhesion by modifying the surface of polyacrylamide with polydopamine, with stiffness value of 1, 10, 30, and 250 kPa, respectively. While the cell spreading increased and the apoptosis decreased with the matrix stiffness, such modulating effect of matrix on cell spreading exhibited different time evolvement behaviors as a function of stiffness, which likely led to surprisingly similar apoptosis rates for the 30 kPa and 250 kPa samples. Matrix stiffness mediated the spreading and apoptosis of MG-63 cells by regulating cell adhesion to matrix and in particular cytoskeletal organization, which was dependent on Ras, Rap1 and PI3K-Akt signaling pathways and finally led to the apoptosis of cancer cells dominated by endoplasmic reticulum stress pathway. Our results provided an insight into the regulation of tumor cell fate by the mechanical clues of ECM, which would have implication for future cancer research and the design of novel anticancer materials.
Collapse
Affiliation(s)
- Huan Deng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Xuedong Shu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Yao Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Junwei Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Yue Yin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Fang Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China.
| |
Collapse
|
47
|
Yan B, Liao P, Shi L, Lei P. Pan-cancer analyses of senescence-related genes in extracellular matrix characterization in cancer. Discov Oncol 2023; 14:208. [PMID: 37985530 PMCID: PMC10660488 DOI: 10.1007/s12672-023-00828-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
PURPOSE The aged microenvironment plays a crucial role in tumor onset and progression. However, it remains unclear whether and how the aging of the extracellular matrix (ECM) influences cancer onset and progression. Furthermore, the mechanisms and implications of extracellular matrix senescence-related genes (ECM-SRGs) in pan-cancer have not been investigated. METHODS We collected profiling data from over 10,000 individuals, covering 33 cancer types, 750 small molecule drugs, and 24 immune cell types, for a thorough and systematic analysis of ECM-SRGs in cancer. RESULTS We observed a significant correlation between immune cell infiltrates and Gene Set Variation Analysis enrichment scores of ECM-SRGs in 33 cancer types. Moreover, our results revealed significant differences in immune cell infiltration among patients with copy number variations (CNV) and single nucleotide variations (SNV) in ECM-SRGs across various malignancies. Aberrant hypomethylation led to increased ECM-SRGs expression, and in specific malignancies, a connection between ECM-SRGs hypomethylation and adverse patient survival was established. The frequency of CNV and SNV in ECM-SRGs was elevated. We observed a positive correlation between CNV, SNV, and ECM-SRGs expression. Furthermore, a correlation was found between the high frequency of CNV and SNV in ECM-SRGs and poor patient survival in several cancer types. Additionally, the results demonstrated that ECM-SRGs expression could serve as a predictor of patient survival in diverse cancers. Pathway analysis unveiled the role of ECM-SRGs in activating EMT, apoptosis, and the RAS/MAPK signaling pathway while suppressing the cell cycle, hormone AR, and the response to DNA damage signaling pathway. Finally, we conducted searches in the "Genomics of Drug Sensitivity in Cancer" and "Genomics of Therapeutics Response Portal" databases, identifying several drugs that target ECM-SRGs. CONCLUSIONS We conducted a comprehensive evaluation of the genomes and immunogenomics of ECM-SRGs, along with their clinical features in 33 solid tumors. This may provide insights into the relationship between ECM-SRGs and tumorigenesis. Consequently, targeting these ECM-SRGs holds promise as a clinical approach for cancer treatment.
Collapse
Affiliation(s)
- Bo Yan
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Pan Liao
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- The School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Liqiu Shi
- Inner Mongolia Forestry General Hospital, 81 Lincheng North Road, Yakeshi, 022150, Inner Mongolia, China
| | - Ping Lei
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- The School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
48
|
Yan W, Maimaitimin M, Wu Y, Fan Y, Ren S, Zhao F, Cao C, Hu X, Cheng J, Ao Y. Meniscal fibrocartilage regeneration inspired by meniscal maturational and regenerative process. SCIENCE ADVANCES 2023; 9:eadg8138. [PMID: 37939174 PMCID: PMC10631723 DOI: 10.1126/sciadv.adg8138] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Meniscus is a complex and crucial fibrocartilaginous tissue within the knee joint. Meniscal regeneration remains to be a scientific and translational challenge. We clarified that mesenchymal stem cells (MSCs) participated in meniscal maturation and regeneration using MSC-tracing transgenic mice model. Here, inspired by meniscal natural maturational and regenerative process, we developed an effective and translational strategy to facilitate meniscal regeneration by three-dimensionally printing biomimetic meniscal scaffold combining autologous synovium transplant, which contained abundant intrinsic MSCs. We verified that this facilitated anisotropic meniscus-like tissue regeneration and protected cartilage from degeneration in large animal model. Mechanistically, the biomechanics and matrix stiffness up-regulated Piezo1 expression, facilitating concerted activation of calcineurin and NFATc1, further activated YAP-pSmad2/3-SOX9 axis, and consequently facilitated fibrochondrogenesis of MSCs during meniscal regeneration. In addition, Piezo1 induced by biomechanics and matrix stiffness up-regulated collagen cross-link enzyme expression, which catalyzed collagen cross-link and thereby enhanced mechanical properties of regenerated tissue.
Collapse
Affiliation(s)
- Wenqiang Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Maihemuti Maimaitimin
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yue Wu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yifei Fan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Shuang Ren
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Fengyuan Zhao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Chenxi Cao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| |
Collapse
|
49
|
Yu TY, Zhang G, Chai XX, Ren L, Yin DC, Zhang CY. Recent progress on the effect of extracellular matrix on occurrence and progression of breast cancer. Life Sci 2023; 332:122084. [PMID: 37716504 DOI: 10.1016/j.lfs.2023.122084] [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: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Breast cancer (BC) metastasis is an enormous challenge targeting BC therapy. The extracellular matrix (ECM), the principal component of the BC metastasis niche, is the pivotal driver of breast tumor development, whose biochemical and biophysical characteristics have attracted widespread attention. Here, we review the biological effects of ECM constituents and the influence of ECM stiffness on BC metastasis and drug resistance. We provide an overview of the relative signal transduction mechanisms, existing metastasis models, and targeted drug strategies centered around ECM stiffness. It will shed light on exploring more underlying targets and developing specific drugs aimed at ECM utilizing biomimetic platforms, which are promising for breast cancer treatment.
Collapse
Affiliation(s)
- Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo 315103, Zhejiang, PR China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| |
Collapse
|
50
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|