1
|
Jaiswal C, Dey S, Prasad J, Gupta R, Agarwala M, Mandal BB. 3D bioprinted microfluidic based osteosarcoma-on-a chip model as a physiomimetic pre-clinical drug testing platform for anti-cancer drugs. Biomaterials 2025; 320:123267. [PMID: 40138960 DOI: 10.1016/j.biomaterials.2025.123267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Standard chemotherapeutic regimen for osteosarcoma (OS) treatment often leads to poor therapeutic outcome, primarily due to lack of an adequate representative model reflecting native OS structural and cellular complexity, posing a translational gap. Three-dimensional bioprinting (3D-BP) represents an efficient and advanced technique for precise recapitulation of the structural and cellular complexity of OS tumor microenvironment (TME). In the present study, we employed a dual extrusion-based 3D-BP method to develop an improved in vitro OS model consisting of both tumor and stromal components. Additionally, a human physiomimetic microfluidic bioreactor is introduced to mimic the dynamic TME and provide physiologically relevant mechanical stimulation to the cells. The model named TC-OS Dynamic model, demonstrated close resemblance to native OS-TME, validated by in vitro studies. Continuous media flow provided mechanical stimulation in the form of shear stress, positively influencing the growth and aggressiveness of OS. Further, drug screening with the model anticancer drugs (doxorubicin, cis-platin, sorafenib) demonstrated enhanced sensitivity in TC-OS Dynamic model as compared to TC-OS Static model, emphasizing enhanced mass transfer, availability and distribution of anticancer drug due to continuous media flow. Overall, TC-OS Dynamic model holds significant potential as a platform in future for high throughput pre-clinical screening of anticancer drugs.
Collapse
Affiliation(s)
- Chitra Jaiswal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India
| | - Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India
| | - Jayant Prasad
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Raghvendra Gupta
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Manoj Agarwala
- GNRC Institute of Medical Sciences, Guwahati 781039, Assam, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India.
| |
Collapse
|
2
|
Feng Y, Jiang Y, Yang L, Lu D, Li N, Zhang Q, Yang H, Qin H, Zhang J, Gou X, Jiang F. Interactions and communications in lung tumour microenvironment: chemo/radiotherapy resistance mechanisms and therapeutic targets. J Drug Target 2025; 33:817-836. [PMID: 39815747 DOI: 10.1080/1061186x.2025.2453730] [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: 11/17/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
The lung tumour microenvironment (TME) is composed of various cell types, including cancer cells, stromal and immune cells, as well as extracellular matrix (ECM). These cells and surrounding ECM create a stiff, hypoxic, acidic and immunosuppressive microenvironment that can augment the resistance of lung tumours to different forms of cell death and facilitate invasion and metastasis. This environment can induce chemo/radiotherapy resistance by inducing anti-apoptosis mediators such as phosphoinositide 3-kinase (PI3K)/Akt, signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa B (NF-κB), leading to the exhaustion of antitumor immunity and further resistance to chemo/radiotherapy. In addition, lung tumour cells can resist chemo/radiotherapy by boosting multidrug resistance mechanisms and antioxidant defence systems within cancer cells and other TME components. In this review, we discuss the interactions and communications between these different components of the lung TME and also the effects of hypoxia, immune evasion and ECM remodelling on lung cancer resistance. Finally, we review the current strategies in preclinical and clinical studies, including the inhibition of checkpoint molecules, chemoattractants, cytokines, growth factors and immunosuppressive mediators such as programmed death 1 (PD-1), insulin-like growth factor 2 (IGF-2) for targeting the lung TME to overcome resistance to chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Yuan Feng
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ying Jiang
- Department of Neurology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Yang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Danni Lu
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ning Li
- Guangxi University of Chinese Medicine, Nanning, China
| | - Qun Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Haiyan Yang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Huiyuan Qin
- Guangxi University of Chinese Medicine, Nanning, China
| | - Jiaxin Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Xinyun Gou
- Guangxi University of Chinese Medicine, Nanning, China
| | - Feng Jiang
- Science and Technology Department, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
3
|
Cai S, Wei X, Li Q, Jiang Z, Li L. Smart materials in pharmacological drug development: Neutrophils and its constituents for drug delivery and consequent antitumor effects. Mol Immunol 2025; 183:18-32. [PMID: 40318595 DOI: 10.1016/j.molimm.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/17/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Neutrophil-based drug delivery systems for targeted therapy of cancer have been studied widely in the recent past. Chemotactic cytokines including colony-stimulating factors (CSFs) recruit various immune cells including the neutrophils to the tumor microenvironment (TME) leading to enhanced metastasis. These cytokines can be targeted effectively by immunotherapeutic agents such as checkpoint inhibitors and mAbs that can lead to systemic toxicity. To minimize the systemic adverse effects, camouflaged nanoparticles can be used significantly as alternative therapeutic agents. The neutrophil-interacting NPs and neutrophil membrane coated NPs have been exploited recently for their antitumor properties in vitro and pose limited systemic adverse effects in vivo. Neutrophil-derived exosomes derived from immune cells can efficiently escape immune-surveillance and pass through the blood-brain barrier. They possess several intrinsic properties in drug delivery as they are nano-sized, extremely biocompatible, non-immunogenic, biodegradable, stable and can carry targeting agents with limited toxicity and display antitumor properties. Also, neutrophil-based nanotherapy is dependent on factors such as neutrophil kinetics and the physicochemical properties of NPs such as size, shape, and surface chemistry. Therefore, neutrophil-based drug delivery for cancer therapy via the use of polymer nanoparticles is widely studied as their clinical appliance in nanomedicine is still at its infancy.
Collapse
Affiliation(s)
- Shengjie Cai
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Xuehan Wei
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Qian Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China; Department of Oncology, Ganyu Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu 222000, China
| | - Ziyu Jiang
- Department of Oncology, Lianyungang Integrated Traditional Chinese and Western Medicine Clinical College, Nanjing University of Chinese Medicine, Nanjing 222002, China; Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China.
| | - Lingchang Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China.
| |
Collapse
|
4
|
Fabiano ED, Poole JM, Reinhart-King CA. Mechanometabolism: recent findings on the intersection of cell adhesion, cell migration, and metabolism. Am J Physiol Cell Physiol 2025; 328:C1866-C1879. [PMID: 40271988 DOI: 10.1152/ajpcell.00892.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 11/26/2024] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
Chemical and mechanical cues within the extracellular matrix (ECM) can initiate intracellular signaling that changes an array of fundamental cell functions. In recent work, studies of cell-ECM adhesion have deepened to include the influence of the physical ECM on cell metabolism. Since many biological processes involve metabolic programs, changes to cellular metabolism in response to cues in the ECM can have marked effects on cell health. In this review, we describe molecular mechanisms associated with cell-ECM adhesion that are key players in metabolism-induced changes to cell behaviors, including migration. We first review how changes to metabolite availability in the extracellular environment or manipulation of metabolic machinery in cells impact focal adhesions. We then connect this work to recent findings regarding the reverse relationship, namely, how the manipulation of focal adhesion proteins or integrins feeds back to alter cell metabolism. Finally, we consider the latest findings from studies that describe how the mechanical properties of the ECM, primarily stiffness and confinement, alter cellular metabolism. We identify key areas of future investigation that may elucidate the molecular drivers that permit cells to respond to mechanical and chemical ECM cues by reprogramming their metabolism to better inform future diagnostics and therapeutics for disease states.
Collapse
Affiliation(s)
- Emily D Fabiano
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
- Department of Bioengineering, Rice University, Houston, Texas, United States
| | - Jenna M Poole
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States
| | - Cynthia A Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
- Department of Bioengineering, Rice University, Houston, Texas, United States
| |
Collapse
|
5
|
Bubna-Litic M, Mayor R. Beyond mechanosensing: How cells sense and shape their physical environment during development. Curr Opin Cell Biol 2025; 94:102514. [PMID: 40209565 DOI: 10.1016/j.ceb.2025.102514] [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: 11/27/2024] [Revised: 03/01/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025]
Abstract
The role of mechanics as a regulator of cell behaviour and embryo development has been widely recognised. However, much of the focus in mechanobiology during embryo development has been on how the mechanical properties of a cell affect its behaviour and fate determination. We discuss the role of mechanosignalling in development and propose that an equally important aspect of embryo mechanobiology is understanding how dynamic changes in tissue mechanics are regulated. Comparably to how chemical signals influence the fate of responding tissues during embryonic induction, we suggest that embryonic cell populations can alter the mechanical properties of adjacent tissues in a process we name 'actuation'. Several examples of embryonic actuation and mechanical feedback are discussed.
Collapse
Affiliation(s)
- Matyas Bubna-Litic
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
6
|
Ashi L, Taurin S. Computational modeling of breast tissue mechanics and machine learning in cancer diagnostics: enhancing precision in risk prediction and therapeutic strategies. Expert Rev Anticancer Ther 2025:1-14. [PMID: 40380913 DOI: 10.1080/14737140.2025.2508850] [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: 01/14/2025] [Revised: 03/19/2025] [Accepted: 05/16/2025] [Indexed: 05/19/2025]
Abstract
INTRODUCTION Breast cancer remains a significant global health issue. Despite advances in detection and treatment, its complexity is driven by genetic, environmental, and structural factors. Computational methods like Finite Element Modeling (FEM) have transformed our understanding of breast cancer risk and progression. AREAS COVERED Advanced computational approaches in breast cancer research are the focus, with an emphasis on FEM's role in simulating breast tissue mechanics and enhancing precision in therapies such as radiofrequency ablation (RFA). Machine learning (ML), particularly Convolutional Neural Networks (CNNs), has revolutionized imaging modalities like mammograms and MRIs, improving diagnostic accuracy and early detection. AI applications in analyzing histopathological images have advanced tumor classification and grading, offering consistency and reducing inter-observer variability. Explainability tools like Grad-CAM, SHAP, and LIME enhance the transparency of AI-driven models, facilitating their integration into clinical workflows. EXPERT OPINION Integrating FEM and ML represents a paradigm shift in breast cancer management. FEM offers precise modeling of tissue mechanics, while ML excels in predictive analytics and image analysis. Despite challenges such as data variability and limited standardization, synergizing these approaches promises adaptive, personalized care. These computational methods have the potential to redefine diagnostics, optimize treatment, and improve patient outcomes.
Collapse
Affiliation(s)
- Layal Ashi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Sebastien Taurin
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Kingdom of Bahrain
| |
Collapse
|
7
|
Lee JJ, Ng KY, Bakhtiar A. Extracellular matrix: unlocking new avenues in cancer treatment. Biomark Res 2025; 13:78. [PMID: 40426238 PMCID: PMC12117852 DOI: 10.1186/s40364-025-00757-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/05/2025] [Indexed: 05/29/2025] Open
Abstract
The extracellular matrix (ECM) plays a critical role in cancer progression by influencing tumor growth, invasion, and metastasis. This review explores the emerging therapeutic strategies that target the ECM as a novel approach in cancer treatment. By disrupting the structural and biochemical interactions within the tumor microenvironment, ECM-targeted therapies aim to inhibit cancer progression and overcome therapeutic resistance. We examine the current state of ECM research, focusing on key components such as collagen, laminin, fibronectin, periostin, and hyaluronic acid, and their roles in tumor biology. Additionally, we discuss the challenges associated with ECM-targeted therapies, including drug delivery, specificity, and potential side effects, while highlighting recent advancements and future directions. This review underscores the potential of ECM-focused strategies to enhance the efficacy of existing treatments and contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Jia Jing Lee
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia.
| |
Collapse
|
8
|
Peura A, Turpin R, Liu R, Heilala M, Salmela M, Aung J, Mikkonen P, Mutka M, Kovanen PE, Niinikoski L, Meretoja T, Mattson J, Heikkilä P, Palanne P, Kantanen T, Kilpeläinen M, Ukkonen O, Hollmén M, Tervonen TA, Klefström J, Munne PM. Soft matrix promotes immunosuppression in tumor-resident immune cells via COX-FGF2 signaling. Nat Commun 2025; 16:4908. [PMID: 40425576 PMCID: PMC12116891 DOI: 10.1038/s41467-025-60092-x] [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: 04/26/2024] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Mechanical forces of the tumor microenvironment change dynamically during key events of tumorigenesis such as invasion and metastasis. These changes in compressive forces often affect the breast cancer cell phenotype. However, it is lesser known how these dynamic mechanical forces in the tumor microenvironment affect the phenotypes of tumor infiltrated leukocytes (TIL) and their subsequent anticancer activities. Here we find, in primary patient-derived explant cultures (PDEC) containing resident TILs, that low compression promotes a change in the original identity of breast cancer cells from luminal to a more mesenchymal and undifferentiated state. These altered tumor cells induce an upregulation of immunosuppressive cytokines such as interleukin-10 (IL-10) and Transforming Growth Factor Beta (TGF-β), as well as polarization of macrophages towards pro-tumor M2(Gc)-type and depletion of CD8+ effector memory T-cells. These immunosuppressive events are mediated by tumor cell derived fibroblast growth factor 2 (FGF2) and prostaglandin E2 (PGE2). We also find that FGF2 rich areas in primary tumors show enrichment in M2-like-macrophages and diminished numbers of CD8 + T and B-cells. Our results suggest that low compressive forces in the tumor microenvironment induce local immunosuppression via FGF2 secretion arising from phenotypic plasticity of tumor cells.
Collapse
Affiliation(s)
- Aino Peura
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
| | - Rita Turpin
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, Turku, Finland
| | - Ruixian Liu
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
| | - Maria Heilala
- Department of Applied Physics, Aalto University, Espoo, Finland
| | - Maria Salmela
- Finnish Genome Editing Center, HiLIFE infrastructures, University of Helsinki and Biocenter Finland, Helsinki, Finland
| | - July Aung
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
| | - Piia Mikkonen
- UPM Biomedicals, UPM-Kymmene Corporation, Helsinki, Finland
| | - Minna Mutka
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Panu E Kovanen
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Laura Niinikoski
- Breast Surgery Unit, Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tuomo Meretoja
- Breast Surgery Unit, Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Johanna Mattson
- Comprehensive Cancer Center, University of Helsinki & Helsinki University Hospital, Helsinki, Finland
| | - Päivi Heikkilä
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Palanne
- Department of Surgery, Kymenlaakso Central Hospital, KYMSOTE, Kotka, Finland
| | - Tiina Kantanen
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Mikko Kilpeläinen
- Department of Surgery, Kymenlaakso Central Hospital, KYMSOTE, Kotka, Finland
| | - Outi Ukkonen
- Department of Surgery, Kymenlaakso Central Hospital, KYMSOTE, Kotka, Finland
| | - Maija Hollmén
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, Turku, Finland
| | - Topi A Tervonen
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
- Finnish Genome Editing Center, HiLIFE infrastructures, University of Helsinki and Biocenter Finland, Helsinki, Finland
| | - Juha Klefström
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland.
- Finnish Cancer Institute, Helsinki, Finland.
- FICAN South, Helsinki University Hospital, Helsinki, Finland.
- Department of Cell & Tissue Biology, University of California, San Francisco, 513 Parnassus Avenue, UCSF Campus, San Francisco, CA, USA.
| | - Pauliina M Munne
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
9
|
Wang K, Shen X, Wu J, Bi Q, Gao Z, Sun Z, Wang W. Fibrogenesis-driven tumor progression in clear cell renal cell carcinoma: prognostic, therapeutic implications and the dual role of neuropilin-1. Cancer Cell Int 2025; 25:179. [PMID: 40380175 PMCID: PMC12082889 DOI: 10.1186/s12935-025-03801-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/24/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the predominant subtype of renal cancer, with a poor prognosis driven by therapy resistance and a propensity for recurrence. Tumor microenvironment (TME)-associated fibrosis accelerates disease progression by fostering immune evasion. Neuropilin-1 (NRP1), a key mediator in fibrotic signaling and cancer biology, has been implicated in these processes. However, the genetic correlation between fibrogenesis and ccRCC remains largely unexplored, necessitating a focused analysis of fibrogenesis-related genes (FRGs) to identify novel prognostic markers and therapeutic strategies. METHODS This study utilized an integrative bioinformatics framework to identify prognosis-associated fibrogenesis-related genes (pFRGs) and applied non-negative matrix factorization (NMF) to stratify ccRCC patients based on fibrotic signatures. A machine learning-derived prognostic model was developed to categorize patients into high-risk and low-risk groups, with tumor microenvironment (TME) features analyzed across these subgroups. The pro-tumorigenic role of NRP1 via the TGF-β/SMAD signaling pathway was validated in vitro and in vivo. RESULTS Twelve pFRGs were identified, with elevated expression correlating with reduced survival. NMF revealed two ccRCC subtypes with different fibrotic and immune profiles. The high-fibrosis subtype showed worse survival and a pro-tumorigenic TME. The risk model demonstrated robust predictive performance (AUCs: 0.738, 0.731, 0.711 for 1-, 2-, and 3-year survival). High-risk patients, marked by immune dysfunction, exhibited worse survival but greater immunotherapy sensitivity. Among the pFRGs, NRP1 was upregulated in ccRCC, and paradoxically associated with favorable prognosis in TCGA, primarily due to stromal enrichment. In vitro and in vivo experiments confirmed that NRP1 promotes ccRCC proliferation, migration, and invasion by enhancing TGF-β/SMAD-driven epithelial-mesenchymal transition (EMT). CONCLUSION Fibrosis is a critical driver of ccRCC progression, linking fibrogenesis-related genes to poor prognosis, immune suppression, and tumor aggressiveness. NRP1 was identified as a central regulator of fibrosis-induced tumor progression through the TGF-β/SMAD signaling pathway. Combining NRP1 inhibition with anti-fibrotic therapies presents a potential strategy for enhancing therapeutic outcomes in ccRCC.
Collapse
Affiliation(s)
- Kai Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Urology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xihao Shen
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qing Bi
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zihao Gao
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Wei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Porter A, Fan S, Peng Y, Lv M, Zhou Y, Alanazi A, Han L, Wang L, Lu XL. Click chemistry-based quantification of extracellular matrix turnover for drug screening and regenerative medicine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.08.652928. [PMID: 40462969 PMCID: PMC12132353 DOI: 10.1101/2025.05.08.652928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
This study presents a sensitive and cost-efficient method to quantify extracellular matrix (ECM) synthesis and degradation using copper-free click chemistry reactions to fluorescently label new ECM components. The approach enables spatial visualization and longitudinal measurement of specific ECM turnover in vitro. We validated the method across multiple platforms, including native cartilage explants and monolayer cultures of human mesenchymal stem cells and breast cancer cells. The technique also proved effective for osteoarthritis drug screening by detecting compounds that mitigate inflammation-induced ECM degradation. Compared to traditional biochemical or histological assays, this click chemistry-based technique offers higher sensitivity, lower sample requirements, and improved temporal resolution. Its versatility supports broad applications in tissue engineering, regenerative medicine, disease modeling, and high-throughput drug evaluation.
Collapse
Affiliation(s)
- Annie Porter
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Songshan Fan
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Ying Peng
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Mengxi Lv
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Yilu Zhou
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Abdulaziz Alanazi
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, 19716, USA
| |
Collapse
|
11
|
Greaney AJ, McCarthy CM, Vethil JP, Abubaker M, Reardon EC, Crowley FD, Cunnane EM, Mulvihill JJE. A comprehensive protocol for PDMS fabrication for use in cell culture. PLoS One 2025; 20:e0323283. [PMID: 40354467 PMCID: PMC12068733 DOI: 10.1371/journal.pone.0323283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 04/04/2025] [Indexed: 05/14/2025] Open
Abstract
Cells exhibit remarkable sensitivity to the mechanical properties of their surrounding matrix, particularly stiffness changes, a phenomenon known as cellular mechanotransduction. In vivo, tissues exhibit a wide range of stiffness, from kilopascals (kPa) to megapascals (MPa), which can alter with aging and disease. Traditional cell culture methods employ plastic substrates with stiffness in the gigapascal range, which does not accurately mimic the physiological conditions of most biological tissues. Therefore, employing substrates that can be engineered to span a wide range of stiffnesses, closely resembling the native tissue environment, is crucial for obtaining results that more accurately reflect cellular responses in vivo. Polydimethylsiloxane (PDMS) substrates are widely used in cell culture for their ability to simulate tissue stiffness, but their optimization presents several challenges. Fabrication requires precise control over mixing, weighing, and curing to ensure reproducible mechanical properties. Inconsistent preparation can lead to improperly cured PDMS substrates, compromising experimental outcomes. Additionally, PDMS's inherent hydrophobicity poses challenges for cell attachment, necessitating surface modifications to enhance adhesion. Moreover, the risk of contamination during the sterilization process necessitates stringent protocols to maintain cell culture integrity. These challenges are further compounded by substrate autofluorescence which can cause difficulties when imaging cells. The aim of this study is to develop a standardized method for fabricating PDMS substrates with tuneable stiffness, ranging from kPa to MPa, suitable for diverse cell types using standard laboratory equipment. This method aims to minimize the complexity and equipment required for PDMS fabrication, ensuring reproducibility and ease of use. Achieving consistent and contaminant-free PDMS substrates will facilitate a broader adoption of these substrates in mechanobiology research and improve the relevance of in vitro models to in vivo conditions. Ultimately, contributing to a more comprehensive understanding of cellular responses to mechanical cues in health and disease.
Collapse
Affiliation(s)
- Aisling J. Greaney
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| | - Clíona M. McCarthy
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| | - Jishnu Padacherri Vethil
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| | - Mannthalah Abubaker
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| | - Erin C. Reardon
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| | - Frederick D. Crowley
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| | - Eoghan M. Cunnane
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| | - John J. E. Mulvihill
- School of Engineering, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
12
|
Feng Y, Jiang Y, Yang L, Lu D, Li N, Zhang Q, Yang H, Qin H, Zhang J, Gou X, Jiang F. Targeting CAFs and extracellular matrix (ECM) in lung cancer: Potential of adjuvants and nanoparticles. Bioorg Chem 2025; 162:108586. [PMID: 40398184 DOI: 10.1016/j.bioorg.2025.108586] [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: 04/04/2025] [Revised: 05/09/2025] [Accepted: 05/11/2025] [Indexed: 05/23/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are prominent components of the lung tumor stroma and are known to foster tumor growth, invasion, and metastasis through extracellular matrix (ECM) and tumor stroma remodeling. The interactions of CAFs with cancer cells and other stromal components contribute significantly to the aggressive nature of lung cancer and pose challenges to conventional treatment approaches. Simultaneously, the ECM, which contains numerous proteins and other molecules surrounding cancer cells, serves as more than just a structural scaffold. In lung cancer, alterations in ECM composition and organization not only promote tumor cell proliferation and survival but also impact drug penetration, immune cell infiltration, and therapeutic resistance. Targeting the intricate interplay between CAFs and the dynamic ECM in lung cancer represents a crucial frontier in oncology research. This review aims to delve deeply into the pivotal roles of CAFs and the ECM in the tumorigenesis and progression of lung cancer. Then, the potential of utilizing adjuvants, phytochemicals, and nanoparticles to modulate the functions of CAFs and remodel the ECM in the lung tumor will be reviewed.
Collapse
Affiliation(s)
- Yuan Feng
- Doctoral student of Guangxi University of Chinese Medicine in grade 2022, Nanning, Guangxi 530200, China
| | - Ying Jiang
- Department of Neurology, Ruikang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi 530011, China
| | - Lin Yang
- Master student of Guangxi University of Chinese Medicine in 2022, Nanning, Guangxi 530200, China
| | - Danni Lu
- Master student of Guangxi University of Chinese Medicine in 2022, Nanning, Guangxi 530200, China
| | - Ning Li
- Master student of Guangxi University of Chinese Medicine in 2023, Nanning, Guangxi 530200, China
| | - Qun Zhang
- Master student of Guangxi University of Chinese Medicine in 2023, Nanning, Guangxi 530200, China
| | - Haiyan Yang
- Master student of Guangxi University of Chinese Medicine in 2023, Nanning, Guangxi 530200, China
| | - Huiyuan Qin
- Master student of Guangxi University of Chinese Medicine in 2024, Nanning, Guangxi 530200, China
| | - Jiaxin Zhang
- Master student of Guangxi University of Chinese Medicine in 2024, Nanning, Guangxi 530200, China
| | - Xinyun Gou
- Master student of Guangxi University of Chinese Medicine in 2024, Nanning, Guangxi 530200, China
| | - Feng Jiang
- Science and Technology Department of Ruikang Hospital affiliated to Guangxi University of Chinese Medicine, 530011, China.
| |
Collapse
|
13
|
Garemilla SSS, Gampa SC, Garimella S. Role of the tumor microenvironment in cancer therapy: unveiling new targets to overcome drug resistance. Med Oncol 2025; 42:202. [PMID: 40332723 DOI: 10.1007/s12032-025-02754-w] [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: 09/11/2024] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Cancer is a leading cause of death globally, with resistance to therapy representing a major obstacle to effective treatment. The tumor microenvironment (TME), comprising a complex network to cellular and non-cellular components including cancer-associated fibroblasts, immune cells, the extracellular matrix and region of hypoxia, is integral to cancer progression and therapeutic resistance. This review delves into the multifaceted interactions within the TME that contribute to tumor growth, survival and immune evasion. Key elements such as the role of cancer- associated fibroblasts in remodeling the extracellular matrix and promoting angiogenesis, the influence of immune cells such as tumor-associated macrophages in creating an immunosuppressive milieu and the impact of hypoxia conditions on metabolic adaptation and therapy resistance are thoroughly examined. This review evaluates current and emerging TME-targeted therapeutic strategies, including inhibitors of extracellular matrix components, modulators of immune cell activity and approached to alleviate hypoxia. Combination therapies that integrate TME-targeted agents with conventional treatments such as chemotherapy and immunotherapy are also discussed for their potential to enhance treatment efficacy and circumvent resistance mechanisms. By synthesising recent advances in TME research and therapeutic innovation, this paper aims to underscore the importance of TME in cancer therapy and highlight promising avenues for improving patient outcomes through targeted intervention.
Collapse
Affiliation(s)
| | - Siri Chandana Gampa
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India
| | - Sireesha Garimella
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India.
| |
Collapse
|
14
|
Li Y, Peng S, Xu J, Liu W, Luo Q. Integrin signaling in tumor biology: mechanisms of intercellular crosstalk and emerging targeted therapies. PeerJ 2025; 13:e19328. [PMID: 40352270 PMCID: PMC12065456 DOI: 10.7717/peerj.19328] [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: 03/25/2025] [Indexed: 05/14/2025] Open
Abstract
Integrins, a family of transmembrane cell adhesion receptors, mediate intercellular and cell-extracellular matrix crosstalk via outside-in and inside-out signaling pathways. Integrins, categorized into 24 distinct combinations of α and β subunits, exhibit tissue-specific expression and perform unique or overlapping roles in physiological and pathophysiological processes. These roles encompass embryonic angiogenesis, tissue repair, and the modulation of tumor cell angiogenesis, progression, invasion, and metastasis. Notably, integrins are significant contributors to tumor development, offering valuable insights into the potential of integrin-targeted diagnostics and therapeutics. Currently, there are various preclinical and clinical trials aiming to harness integrin antagonists that are safe, efficacious, and exhibit low toxicity. Owing to the functional redundancy across integrin types and the complexity of the mechanisms of integrin-mediated multiple key processes associated with tumor biology, challenges exist that impede advancements in integrin-targeted therapy. Nevertheless, innovative strategies focused on integrin modulation represent significant breakthroughs for improving patient care and promoting comprehensive insights into the underlying mechanisms of tumor biology. This review elucidates the impact of integrins on three distinct cell types in multiple key processes associated with tumor biology and explores the emerging integrin-targeted therapeutic approaches for the treatment of tumors, which will provide ideas for optimal therapeutic approaches in the future.
Collapse
Affiliation(s)
- Yifan Li
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shantong Peng
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiatong Xu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wenjie Liu
- The First Clinical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qi Luo
- College of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
15
|
Martinez-Marin D, Stroman GC, Fulton CJ, Pruitt K. Frizzled receptors: gatekeepers of Wnt signaling in development and disease. Front Cell Dev Biol 2025; 13:1599355. [PMID: 40376615 PMCID: PMC12078226 DOI: 10.3389/fcell.2025.1599355] [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: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Frizzled (FZD) receptors are a subset of G-protein-coupled receptors (GPCRs), the largest class of human cell surface receptors and a major target of FDA-approved drugs. Activated by Wnt ligands, FZDs regulate key cellular processes such as proliferation, differentiation, and polarity, positioning them at the intersection of developmental biology and disease, including cancer. Despite their significance, FZD signaling remains incompletely understood, particularly in distinguishing receptor-specific roles across canonical and non-canonical Wnt pathways. Challenges include defining ligand-receptor specificity, elucidating signal transduction mechanisms, and understanding the influence of post translational modifications and the cellular context. Structural dynamics, receptor trafficking, and non-canonical signaling contributions also remain areas of active investigation. Recent advances in structural biology, transcriptomics, and functional genomics are beginning to address these gaps, while emerging therapeutic approaches-such as small-molecule modulators and antibodies-highlight the potential of FZDs as drug targets. This review synthesizes current insights into FZD receptor biology, examines ongoing controversies, and outlines promising directions for future research and therapeutic development.
Collapse
Affiliation(s)
| | | | | | - Kevin Pruitt
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
16
|
Sarkar M, Hossain MT, Ewoldt RH, Laukaitis C, Johnson AW. Stiffening of a fibrous matrix after recovery of contracted inclusions. SOFT MATTER 2025; 21:3314-3330. [PMID: 40183246 DOI: 10.1039/d5sm00087d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Disordered fibrous matrices in living tissues are subjected to forces exerted by cells that contract to pull on matrix fibers. To maintain homeostasis or facilitate disease progression, contracted cells often push on matrix fibers as they recover their original sizes. Recent advances have shown that matrix geometry encodes loading history into mechanical memory independently of plasticity mechanisms such as inter-fiber cohesion or fiber yielding. Conceptualizing cells as inclusions undergoing sequential contraction and recovery, prior work documented matrix remodeling surrounding a solitary recovered inclusion. However, because the remodeling induced by the contraction of multiple inclusions differs from that caused by a single contracted inclusion, we investigate how matrix remodeling occurs when multiple contracted inclusions recover simultaneously, a scenario that more accurately reflects real tissues containing many closely spaced cells. Using mechanics-based computational models of fibrous matrices embedded with clusters of inclusions, we studied the mechanical remodeling of the matrix during the simultaneous recovery of inclusions after contraction. The results revealed permanent mechanical remodeling of the matrix within the cluster, with stiffening observed in areas of the matrix enclosed by closely spaced inclusions. This stiffening was driven by microstructural changes in matrix geometry and was corroborated in experiments, where collagen matrices permanently remodeled by the contraction and recovery of closely spaced embedded cells also exhibited stiffening. By enriching the understanding of memory formation in fibrous matrices, this study opens new possibilities for estimating cell forces on matrix substrates and refining metamaterial design strategies.
Collapse
Affiliation(s)
- Mainak Sarkar
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, USA.
| | - Mohammad Tanver Hossain
- Mechanical Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, USA
| | - Randy H Ewoldt
- Mechanical Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, USA
- Materials Research Laboratory, University of Illinois Urbana-Champaign, USA
| | - Christina Laukaitis
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, USA.
- Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, USA
- Clinical Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, USA
- Carle Health, Urbana, Illinois, USA
| | - Amy Wagoner Johnson
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, USA.
- Mechanical Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, USA
- Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, USA
- CZ Biohub Chicago, LLC, Chicago, Illinois, USA
| |
Collapse
|
17
|
Han J, Halwachs K, West T, Larsen B, Sacks MS, Rosales AM, Zoldan J. Matrix Stiffness Regulates Mechanotransduction and Vascular Network Formation of hiPSC-Derived Endothelial Progenitors Encapsulated in 3D Hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648340. [PMID: 40291699 PMCID: PMC12027365 DOI: 10.1101/2025.04.11.648340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The mechanical properties of the extracellular matrix (ECM), particularly stiffness, regulate endothelial progenitor responses during vascular development, yet their behavior in physiologically compliant matrices (<1 kPa) remains underexplored. Using norbornene-modified hyaluronic acid (NorHA) hydrogels with tunable stiffness (190-884 Pa), we investigated how hydrogel stiffness influences cell morphology, endothelial maturation, mechanotransduction, and microvascular network formation in human induced pluripotent stem cell-derived endothelial progenitors (hiPSC-EPs). Our findings reveal a stiffness-dependent tradeoff between mechanotransduction and vascular network formation. At intermediate stiffness (551 Pa), cells exhibited the greatest increase in endothelial marker CD31 expression and Yes-associated protein (YAP)/ transcriptional coactivator with PDZ-binding motif (TAZ) nuclear translocation, indicating enhanced mechanotransduction and endothelial maturation. However, this did not translate to superior plexus formation. Instead, the most compliant matrix (190 Pa) supported greater vascular connectivity, characterized by longer branches (∼0.03/volume vs. 0.015 at 551 Pa) and enhanced actin remodeling. 3D cell contraction measurements revealed a 15.6-fold higher basal displacement in compliant hydrogels, suggesting that cell-generated forces and matrix deformability collectively drive vascular morphogenesis. Unlike prior studies focusing on pathological stiffness ranges (>10 kPa), our results emphasize that vascularization is not solely driven by the most mechanotransductive environment but rather by a balance of compliance, contractility, and cell-induced remodeling. These findings underscore the need to design hydrogels that provide sufficient mechanotransduction for endothelial maturation while maintaining compliance to support dynamic vascular morphogenesis. This work provides a mechanically tuned framework for optimizing microenvironments to balance endothelial differentiation and vascular network formation in tissue engineering and regenerative medicine.
Collapse
|
18
|
Liu H, Scherpe L, Hummer LB, Snedeker JG, Zenobi-Wong M. Filamented light (FLight) biofabrication of mini-tendon models show tunable matrix confinement and nuclear morphology. Biofabrication 2025; 17:035005. [PMID: 40245877 DOI: 10.1088/1758-5090/adce35] [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: 11/19/2024] [Accepted: 04/17/2025] [Indexed: 04/19/2025]
Abstract
One hallmark of healthy tendon tissue is the high confinement of tenocytes between tightly packed, highly aligned collagen fibers. During tendinopathy, this organization becomes dysregulated, leading to cells with round-shaped morphology and collagen fibers which exhibit crimping and misalignment. The elongated nuclei in healthy tendons are linked to matrix homeostasis through distinct mechanotransduction pathways, and it is believed that the loss of nuclear confinement could upregulate genes associated with abnormal matrix remodeling. Replicating the cell and nuclear morphology of healthy and diseased states of tendon, however, remains a significant challenge for engineeredin vitrotendon models. Here we report on a high throughput biofabrication of mini-tendons that mimick the tendon core compartment based on the filamented light (FLight) approach. Each mini-tendon, with a length of 4 mm, was composed of parallel hydrogel microfilaments (2-5µm diameter) and microchannels (2-10µm diameter) that confined the cells. We generated four distinct matrices with varying stiffness (7-40 kPa) and microchannel dimensions. After 14 d of culture, 29% of tenocytes in the softest matrix with the largest microchannel diameter were aligned, exhibiting an average nuclear aspect ratio (nAR) of 2.1. In contrast, 84% of tenocytes in the stiffest matrix with the smallest microchannel diameter were highly aligned, with a mean nAR of 3.4. When tenocytes were culturedonthe FLight hydrogels (2D) as opposed to within the hydrogels three-dimensional (3D), the mean nAR was less than 1.9, indicating that nuclear morphology is significantly more confined in 3D environments. By tuning the stiffness and microarchitecture of the FLight matrix, we demonstrated that mechanical confinement can be modulated to exert control over the extent of nuclear confinement. This high-throughput, tunable platform offers a promising approach for studying the mechanobiology of healthy and diseased tendons and for eventual testing of drug compounds against tendinopathy.
Collapse
Affiliation(s)
- Hao Liu
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Lynn Scherpe
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Linnea B Hummer
- Laboratory for Orthopedic Biomechanics, Department of Health Sciences & Technology, ETH Zürich, Lengghalde 5, 8008 Zürich, Switzerland
| | - Jess Gerrit Snedeker
- Laboratory for Orthopedic Biomechanics, Department of Health Sciences & Technology, ETH Zürich, Lengghalde 5, 8008 Zürich, Switzerland
- Balgrist University Hospital, University of Zurich, Lengghalde 5, 8008 Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|
19
|
Murugan NJ, Genautis E, Voutsadakis IA. Transient Receptor Potential Channels in Prostate Cancer: Associations with ERG Fusions and Survival. Int J Mol Sci 2025; 26:3639. [PMID: 40332161 PMCID: PMC12027297 DOI: 10.3390/ijms26083639] [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: 01/28/2025] [Revised: 03/13/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Calcium movement and concentration in the cell plays significant roles in normal physiology and in diseases such as cancer. The significance of this ion in oncogenesis suggests that membrane-relevant proteins are involved in its regulation and are deregulated in various cancers. These channels and transporters could be targets for therapeutic interventions. An evaluation of the expression of transient receptor potential (TRP) channels in prostate cancer was performed using publicly available genomic and proteome data. Two TRP family members with high expression in prostate cancers, TRPML2 and TRPM4, were chosen for further analysis the uncover the associations of their level of expression with clinical and pathologic prostate cancer characteristics. Several TRP channels were expressed in prostate cancers at the protein level including TRPM4, TRPML1, TRPML2, TRPC1 and TRPP3. At the mRNA level, MCOLN2 and TRPM4 were strongly expressed in a sub-set of prostate cancers. Cases with high MCOLN2 mRNA expression were associated with frequent ERG fusions and a trend for better survival outcomes. In contrast, prostate cancer cases with high TRPM4 mRNA expression were associated with lower ERG fusion frequency than cases with low TRPM4 mRNA expression. The prognosis of prostate cancers with high TRPM4 expression was not different from the prognosis with counterparts having low TRPM4 mRNA expression. TRP channels were expressed in sub-sets of prostate cancers. The two well-expressed channels of the super family, TRPML2 and TRPM4, have divergent associations with the most prevalent prostate cancer molecular aberrations, ERG fusions. These results imply diverse regulations of the TRP channels that would have to be taken into consideration when devising therapeutic interventions targeting individual channels.
Collapse
Affiliation(s)
- Nirosha J. Murugan
- Department of Biology, Wilfrid Laurier University, Waterloo, ON N2L 6C2, Canada
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Emma Genautis
- Department of Biology, Wilfrid Laurier University, Waterloo, ON N2L 6C2, Canada
| | - Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON P6B 0A8, Canada
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
20
|
Ba J, Zheng C, Lai Y, He X, Pan Y, Zhao Y, Xie H, Wu B, Deng X, Wang N. High matrix stiffness promotes senescence of type II alveolar epithelial cells by lysosomal degradation of lamin A/C in pulmonary fibrosis. Respir Res 2025; 26:128. [PMID: 40205454 PMCID: PMC11984030 DOI: 10.1186/s12931-025-03201-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Cellular senescence is one of the key steps in the progression of pulmonary fibrosis, and the senescence of type II alveolar epithelial cells (AEC IIs) may potentially accelerate the progression of pulmonary fibrosis. However, the molecular mechanisms underlying cellular senescence in pulmonary fibrosis remain unclear. METHODS The researchers first conducted in vitro experiments to investigate whether AEC IIs cultured on high matrix stiffness would lead to cellular senescence. Next, samples from mouse pulmonary fibrosis models and clinical idiopathic pulmonary fibrosis (IPF) patients were tested to observe extracellular matrix deposition, lamin A/C levels, and cellular senescence status in lung tissue. Construct lamin A/C knockdown and overexpression systems separately in AEC IIs, and observe whether changes in lamin A/C levels lead to cellular senescence. Further explore the degradation mechanism of lamin A/C using protein degradation inhibitors. RESULTS In vitro experiments have found that high matrix stiffness promotes senescence of AEC IIs. In a mouse model of pulmonary fibrosis, AEC IIs were found to exhibit significant cellular senescence on day 21. In clinical IPF samples, it was found that senescent cells expressed low levels of lamin A/C. In the lamin A/C SiRNA knockdown system, it was further confirmed that AEC IIs with low levels of lamin A/C are more prone to cellular senescence. Under high matrix stiffness, lamin A/C in AEC IIs is degraded through the autophagy lysosome pathway. The use of chloroquine can effectively alleviate cellular senescence. CONCLUSIONS High matrix stiffness degrades lamin A/C in pulmonary fibrosis through lysosomal degradation pathways, promoting AEC II senescence. Inhibition the degradation of lamin A/C could alleviate AEC II senescence.
Collapse
Affiliation(s)
- Junhui Ba
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Changyu Zheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yimei Lai
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Xin He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yuxi Pan
- Department of Oncology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yanqiu Zhao
- Shenzhen Samii Medical Center, Shenzhen, Guangdong Province, China
| | - Huihui Xie
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Benquan Wu
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| | - Xiao Deng
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China.
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| |
Collapse
|
21
|
Wu J, He R, Xu Z, Yang H, Guan Y, Sun L, Lv W, Huang J, Wang J. Mechanical signal-mediated mitochondria-endoplasmic reticulum contacts modulate Leydig cell testosterone biosynthesis during testis development. Mol Hum Reprod 2025; 31:gaaf017. [PMID: 40419464 DOI: 10.1093/molehr/gaaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/07/2025] [Indexed: 05/28/2025] Open
Abstract
In males, 95% of testosterone is synthesized by Leydig cells, and a deficiency in this synthesis will cause metabolic disorders and multiple organ dysfunction. Testosterone deficiency is not only affected by aged or diseased Leydig cells, which have been studied extensively, but is also closely related to the development of the testis. At present, the focus on the mechanism of testis development includes epigenetic and hormone regulation. However, testicular development is constrained by the external tough tunica albuginea, suggesting that mechanical signals may also play an important role in the regulation of testis development; however, this is not yet well understood. In this in vitro study, we found that a gradual increase in extracellular substrate stiffness for testis development leads to the activation of mechanical signals to promote cytoskeleton remodeling. Eventually, the mechanical signal mediates changes in the mitochondrial-endoplasmic reticulum and affects the synthesis of testosterone in Leydig cells. Through organoid and animal experiments, we found that targeting mechanical signaling pathways that regulate testosterone biosynthesis is feasible. This provides a new angle for further exploration of testis development and new insights into how substrate stiffness affects the testis, raising new clues for clinical applications.
Collapse
Affiliation(s)
- Jiahong Wu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Ruiling He
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Zeyu Xu
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Huan Yang
- Department of Gynecology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Yupeng Guan
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Lu Sun
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Wantong Lv
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Jiayu Huang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| |
Collapse
|
22
|
Breideband L, Wächtershäuser KN, Sarkar R, Puspathasan M, Stelzer EH, Pampaloni F. Gravitational forces and matrix stiffness modulate the invasiveness of breast cancer cells in bioprinted spheroids. Mater Today Bio 2025; 31:101640. [PMID: 40124331 PMCID: PMC11930500 DOI: 10.1016/j.mtbio.2025.101640] [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: 10/13/2024] [Revised: 01/29/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
The progression of breast cancer is influenced by the stiffness of the extracellular matrix (ECM), which becomes stiffer as cancer advances due to increased collagen IV and laminin secretion by cancer-associated fibroblasts. Intriguingly, breast cancer cells cultivated in two-dimensions exhibit a less aggressive behavior when exposed to weightlessness, or microgravity conditions. This study aims to elucidate the interplay between matrix stiffness and microgravity on breast cancer progression. For this purpose, three-dimensional spheroids of breast cancer cell lines (MCF-7 and MDA-MB-231) were formed. These spheroids were subsequently bioprinted in hydrogels of varying stiffness, obtained by the mixing of gelatin methacrylate and poly(ethylene) glycol diacrylate mixed at different ratios. The constructs were printed with a custom stereolithography (SLA) bioprinter converted from a low-cost, commercially available 3D printer. These bioprinted structures, encapsulating breast cancer spheroids, were then placed in a clinostat (microgravity simulation device) for a duration of seven days. Comparative analyses were conducted between objects cultured under microgravity and standard earth gravity conditions. Protein expression was characterized through fluorescent microscopy, while gene expression of MCF-7 constructs was analyzed via RNA sequencing. Remarkably, the influence of a stiffer ECM on the protein and gene expression levels of breast cancer cells could be modulated and sometimes even reversed in microgravity conditions. The study's findings hold implications for refining therapeutic strategies for advanced breast cancer stages - an array of genes involved in reversing aggressive or even metastatic behavior might lead to the discovery of new compounds that could be used in a clinical setting.
Collapse
Affiliation(s)
- Louise Breideband
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Kaja Nicole Wächtershäuser
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Ryan Sarkar
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Melosha Puspathasan
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Ernst H.K. Stelzer
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| |
Collapse
|
23
|
Lam CD, Park S. Nanomechanical characterization of soft nanomaterial using atomic force microscopy. Mater Today Bio 2025; 31:101506. [PMID: 40018054 PMCID: PMC11867545 DOI: 10.1016/j.mtbio.2025.101506] [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: 09/27/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 03/01/2025] Open
Abstract
Atomic force microscopy (AFM) is a promising method for generating high-spatial-resolution images, providing insightful perspectives on the nanomechanical attributes of soft matter, including cells, bacteria, viruses, proteins, and nanoparticles. AFM is widely used in biological and pharmaceutical sciences because it can scrutinize mechanical properties under physiological conditions. We comprehensively reviewed experimental techniques and fundamental mathematical models to investigate the mechanical properties, including elastic moduli and binding forces, of soft materials. To determine these mechanical properties, two-dimensional arrays of force-distance (f-d) curves are obtained through AFM indentation experiments using the force volume technique. For elasticity determination, models are divided into approach f-d curve-based models, represented by the Hertz model, and retract f-d curve-based models, exemplified by the Johnson-Kendall-Roberts and Derjaguin-Müller-Toporov models. Especially, the Chen, Tu, and Cappella models, developed from the Hertz model, are used for thin samples on hard substrates. Additionally, the establishment of physical or chemical bonds during indentation experiments, observable in retract f-d curves, is crucial for the adhesive properties of samples and binding affinity between antibodies (receptors) and antigens (ligands). Chemical force microscopy, single-molecule force spectroscopy, and single-cell force spectroscopy are primary AFM methods that provide a comprehensive view of such properties through retract curve analysis. Furthermore, this paper, structured into key thematic sections, also reviews the exemplary application of AFM across multiple scientific disciplines. Notably, cancer cells are softer than healthy cells, although more sophisticated investigations are required for prognostic applications. AFM also investigates how bacteria adapt to antibiotics, addressing antimicrobial resistance, and reveals that stiffer virus capsids indicate reduced infectivity, aiding in the development of new strategies to combat viral infections. Moreover, AFM paves the way for innovative therapeutic approaches in designing effective drug delivery systems by providing insights into the physical properties of soft nanoparticles and the binding affinity of target moieties. Our review provides researchers with representative studies applying AFM to a wide range of cross-disciplinary research.
Collapse
Affiliation(s)
- Chi-Dat Lam
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Soyeun Park
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| |
Collapse
|
24
|
Mangani S, Kremmydas S, Karamanos NK. Mimicking the Complexity of Solid Tumors: How Spheroids Could Advance Cancer Preclinical Transformative Approaches. Cancers (Basel) 2025; 17:1161. [PMID: 40227664 PMCID: PMC11987746 DOI: 10.3390/cancers17071161] [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: 02/25/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Traditional 2D cell culture models present significant limitations in replicating the intricate architecture and microenvironment of in vivo solid tumors, which are essential for accurately studying cancer initiation, growth, progression, and metastasis. This underscores the need for the development of advanced preclinical models to accelerate research outcomes. Emerging 3D cell culture systems, particularly spheroid models, provide a more realistic representation of solid tumor properties by capturing the complex interactions occurring within the tumor microenvironment, including the extracellular matrix dynamics that influence cancer progression. Among solid tumors, breast cancer remains the most frequently diagnosed cancer among women globally and a leading cause of cancer-related mortality. Here we emphasize the value of breast cancer cell-derived spheroids in revealing differential molecular characteristics and understanding cancer cell properties during the early stages of invasion into adjacent tissues. Conclusively, this study underscores the urgent need to adopt 3D cell culture platforms, given their significant contributions to advanced cancer research and pharmaceutical targeting. This may well offer a transformative approach for preclinical studies and enhance our ability to test therapeutic efficiency in conditions that closely mimic the growth and progression of in vivo solid tumors.
Collapse
Affiliation(s)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
25
|
Alqosiri HM, Alqasiri HM, Alqasire SE, Nava VE, Bandyopadhyay BC, Raub CB. Breast cancer extracellular matrix invasion depends on local mechanical loading of the collagen network. J Mater Chem B 2025. [PMID: 40135428 DOI: 10.1039/d4tb01474j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Active mechanical stresses in and around tumors affect cancer cell behavior and independently regulate cancer progression. To investigate the role of mechanical stress in breast cancer cell invasion, magnetic alginate beads loaded with iron oxide nanoparticles were coated with MDA-MB-231 breast cancer cells and embedded in a three-dimensional extracellular matrix (ECM) model subjected to an external magnetic field during culture. Bead displacement, cell shape and patterns of invasion of the collagen gel, and cell proliferation were assessed over 7 days of culture. The alginate beads swelled over the first 24 h in culture, creating circumferential stress akin to that created by tumor growth, while bead magnetic properties enabled local mechanical loading (compression, tension, and relaxation) and motion within the in vitro tissue constructs upon exposure to an external magnetic field. Beads displaced 0.2-1.6 mm through the collagen gels, depending on magnet size and distance, compressing the collagen network microstructure without gel mechanical failure. Invading cells formed a spatulate pattern as they moved into the compressed ECM region, with individual cells aligned parallel to the bead surface. During the first 24 hours of compressive magnetic force loading, invading cancer cells became round, losing elongation and ability to invade out from the bead surface, while still actively dividing. In contrast, cell invasion in unloaded constructs and in loaded constructs away from the compression region invaded as single cells, transversely outward from the bead surface. Finally, cell proliferation was 1.3× higher only after external magnet removal, which caused relaxation of mechanical stress in the collagen network. These findings indicate effects on breast cancer invasion of mechanical loading of ECM, both from compressive loading and from load relaxation. Findings point to the influence of mechanical stress on cancer cell behavior and suggest that relaxing mechanical stress in and around a tumor may promote cancer progression through higher proliferation and invasion.
Collapse
Affiliation(s)
- Hanadi M Alqosiri
- Department of Biomedical Engineering, School of Engineering, Catholic University of America, Washington, DC, 20064, USA.
| | - Hadeel M Alqasiri
- Department of Biomedical Engineering, School of Engineering, Catholic University of America, Washington, DC, 20064, USA.
| | - Sara E Alqasire
- Department of Biomedical Engineering, School of Engineering, Catholic University of America, Washington, DC, 20064, USA.
| | - Victor E Nava
- Veterans Affairs Medical Center, Washington, DC, 20422, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC, 20052, USA
| | - Bidhan C Bandyopadhyay
- Department of Biomedical Engineering, School of Engineering, Catholic University of America, Washington, DC, 20064, USA.
- Veterans Affairs Medical Center, Washington, DC, 20422, USA
- George Washington University School of Medicine and Health Sciences, Washington, DC, 20052, USA
| | - Christopher B Raub
- Department of Biomedical Engineering, School of Engineering, Catholic University of America, Washington, DC, 20064, USA.
| |
Collapse
|
26
|
Liu X, Yu L, Xiao A, Sun W, Wang H, Wang X, Zhou Y, Li C, Li J, Wang Y, Wang G. Analytical methods in studying cell force sensing: principles, current technologies and perspectives. Regen Biomater 2025; 12:rbaf007. [PMID: 40337625 PMCID: PMC12057814 DOI: 10.1093/rb/rbaf007] [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: 11/04/2024] [Revised: 12/16/2024] [Accepted: 02/10/2025] [Indexed: 05/09/2025] Open
Abstract
Mechanical stimulation plays a crucial role in numerous biological activities, including tissue development, regeneration and remodeling. Understanding how cells respond to their mechanical microenvironment is vital for investigating mechanotransduction with adequate spatial and temporal resolution. Cell force sensing-also known as mechanosensation or mechanotransduction-involves force transmission through the cytoskeleton and mechanochemical signaling. Insights into cell-extracellular matrix interactions and mechanotransduction are particularly relevant for guiding biomaterial design in tissue engineering. To establish a foundation for mechanical biomedicine, this review will provide a comprehensive overview of cell mechanotransduction mechanisms, including the structural components essential for effective mechanical responses, such as cytoskeletal elements, force-sensitive ion channels, membrane receptors and key signaling pathways. It will also discuss the clutch model in force transmission, the role of mechanotransduction in both physiology and pathological contexts, and biomechanics and biomaterial design. Additionally, we outline analytical approaches for characterizing forces at cellular and subcellular levels, discussing the advantages and limitations of each method to aid researchers in selecting appropriate techniques. Finally, we summarize recent advancements in cell force sensing and identify key challenges for future research. Overall, this review should contribute to biomedical engineering by supporting the design of biomaterials that integrate precise mechanical information.
Collapse
Affiliation(s)
- Xiaojun Liu
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao 266024, China
| | - Lei Yu
- Department of Traditional Chinese Medicine, Qingdao Special Service Sanatorium of PLA Navy, Qingdao 266071, China
| | - Adam Xiao
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Wenxu Sun
- School of Sciences, Nantong University, Nantong 226019, China
| | - Han Wang
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
| | - Xiangxiu Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yanghao Zhou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Chao Li
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao 266024, China
| | - Jiangtao Li
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Yongliang Wang
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao 266024, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
- Qindao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266044, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
- JinFeng Laboratory, Chongqing 401329, China
| |
Collapse
|
27
|
Foster E, Wardhana O, Zeng Z, Lu X. Comparative Analysis of 3D Culture Methodologies in Prostate Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642589. [PMID: 40161751 PMCID: PMC11952452 DOI: 10.1101/2025.03.11.642589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Three-dimensional (3D) cell culture models are increasingly utilized in cancer research to better replicate in vivo tumor microenvironments. This study examines the effects of different 3D scaffolding materials, including Matrigel, GelTrex, and the plant-based GrowDex, on prostate cancer cell lines, with a particular emphasis on neuroendocrine prostate cancer (NEPC). Four cell lines (LNCaP, LASCPC-01, PC-3, and KUCaP13) were cultured in these scaffolds to evaluate spheroid formation, cell viability, and gene expression. The results revealed that while all scaffolds supported cell viability, spheroid formation varied significantly: Matrigel promoted the most robust spheroids, especially for LASCPC-01, whereas GrowDex exhibited limitations for certain cell lines. Gene expression analysis indicated a consistent reduction in androgen receptor (AR) expression in LNCaP cells across all scaffolds, suggesting a potential shift towards a neuroendocrine phenotype. However, the expression of neuroendocrine markers varied depending on the scaffold and culture method, with the mini-domes method in Matrigel leading to decreased expression of both castration-resistant prostate cancer (CRPC) and NEPC markers. These findings highlight the scaffold-dependent variability in 3D culture outcomes and emphasize the need for standardized methodologies to ensure consistency and relevance in prostate cancer research.
Collapse
Affiliation(s)
- Ella Foster
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Oliver Wardhana
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Ziyu Zeng
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
- Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
28
|
Zhu Q, Zhang R, Zhao Z, Xie T, Sui X. Harnessing phytochemicals: Innovative strategies to enhance cancer immunotherapy. Drug Resist Updat 2025; 79:101206. [PMID: 39933438 DOI: 10.1016/j.drup.2025.101206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
Cancer immunotherapy has revolutionized cancer treatment, but therapeutic ineffectiveness-driven by the tumor microenvironment and immune evasion mechanisms-continues to limit its clinical efficacy. This challenge underscores the need to explore innovative approaches, such as multimodal immunotherapy. Phytochemicals, bioactive compounds derived from plants, have emerged as promising candidates for overcoming these barriers due to their immunomodulatory and antitumor properties. This review explores the synergistic potential of phytochemicals in enhancing immunotherapy by modulating immune responses, reprogramming the tumor microenvironment, and reducing immunosuppressive factors. Integrating phytochemicals with conventional immunotherapy strategies represents a novel approach to mitigating resistance and enhancing therapeutic outcomes. For instance, nab-paclitaxel has shown the potential in overcoming resistance to immune checkpoint inhibitors, while QS-21 synergistically enhances the efficacy of tumor vaccines. Furthermore, we highlight recent advancements in leveraging nanotechnology to engineer phytochemicals for improved bioavailability and targeted delivery. These innovations hold great promise for optimizing the clinical application of phytochemicals. However, further large-scale clinical studies are crucial to fully integrate these compounds into immunotherapeutic regimens effectively.
Collapse
Affiliation(s)
- Qianru Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao
| | - Ruonan Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ziming Zhao
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao; Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China.
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao; Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China.
| |
Collapse
|
29
|
de Jong MJP, van Delft F, Radstake FDW, Perik TH, Litjens G, Bisseling TM, van der Sommen F, van Geenen EJM, Hermans JJ, Siersema PD. Contrast agent dispersion visualized by CE-EUS may be a prediction tool for FOLFIRINOX chemotherapy effectiveness in patients with pancreatic adenocarcinoma. Pancreatology 2025; 25:258-265. [PMID: 39909762 DOI: 10.1016/j.pan.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/30/2024] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) still has a dismal 5-year overall survival of 13 %. Chemotherapy is increasingly used as treatment in both (neo-) adjuvant and palliative conditions. However, the overall survival benefits of chemotherapy must be weighed against significant side effects leading to a reduction in quality of life. CE-EUS and elastography could provide additional information about the vascularization and elasticity of the pancreatic tumor. The aim of this study was to investigate if contrast-enhanced endoscopic ultrasound and/or elastography could be suitable to predict the effectiveness of FOLFIRINOX. METHODS Single center, prospective proof-of-concept study in which intravenous contrast agent was administered and strain ratio was calculated in patients undergoing EUS in their regular diagnostic work-up. Directly after contrast administration, a video of 120 s was recorded and afterwards tracked and fitted by a Modified Local Density Random Walk (mLDRW) model. RESULTS We included 17 patients. Based on cross-sectional imaging based RECIST criteria, chemotherapy treatment was effective in 11 patients and not effective in 6 patients. The contrast dispersion parameter (κ1) differed significantly between both groups in favor of the responders: 2.994 (IQR 1.670-5.170) vs 1.203 (IQR 0.953-1.756), p = 0.005. The elastography strain ratio was higher in the effectively treated group (20.9 vs 13.6, p = 0.138). CONCLUSION This proof-of-concept study showed that the dispersion parameter of the first wave of contrast was 2.5 times higher in patients in whom FOLFIRINOX was effective, suggesting that this parameter could possibly be a reliable prediction tool.
Collapse
Affiliation(s)
- Mike J P de Jong
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Foke van Delft
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fer D W Radstake
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tom H Perik
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Geke Litjens
- Department of Gastroenterology and Hepatology, Isala Clinics, Zwolle, the Netherlands
| | - Tanya M Bisseling
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Erwin-Jan M van Geenen
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - John J Hermans
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter D Siersema
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
30
|
Cofre J. The first embryo, the origin of cancer and animal phylogeny. V. Cancer stem cells as the unifying biomechanical principle between embryology and oncology. MECHANOBIOLOGY IN MEDICINE 2025; 3:100110. [PMID: 40396136 PMCID: PMC12082149 DOI: 10.1016/j.mbm.2024.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 05/22/2025]
Abstract
The role of embryology in metazoan evolution is rooted deeply in the history of science. Viewing Neoplasia as an evolutionary engine provides a scientific basis for reexamining the disease cancer. Once the embryo is understood as a benign tumor with a pivotal role in the evolution of all animal forms, there will be an immediate paradigm shift in the search for cancer cure, potentially revealing insights that may be buried within the great developmental transitions of metazoans. This article discusses one of the unifying principles between embryology and oncology, namely cancer stem cells. Some considerations are also provided on the central role of physics and biomechanics in the assembly of the first embryo, which can be regarded as a differentiated benign tumor. Mechanical impregnation of the nucleus of a stem cell, culminating in a totipotent/multipotent cell, was a major event safeguarding the success of embryogenesis throughout evolution. Germ cells in the earliest ctenophore embryos underwent delayed differentiation, subsequent to the mechanical assembly of the embryo. Finally, a discussion is presented on the concept that cancer and embryogenesis (cancer and healthy stem cells) are two sides of the same coin, that is, of the same process. The only difference is that cancer stem cells reveal themselves in inappropriate contexts. Neoplasia is a free force, whereas cancer is a force contained by animal organization.
Collapse
Affiliation(s)
- Jaime Cofre
- Laboratório de Embriologia Molecular e Câncer, Federal University of Santa Catarina, Sala 313b, Florianópolis, SC, 88040-900, Brazil
| |
Collapse
|
31
|
Chavez L, Gao S, Pandey V, Yuan N, Ragab S, Li J, Hepburn MS, Smith P, Edelheit C, Corr DT, Kennedy BF, Intes X. Design and characterization of an optical phantom for mesoscopic multimodal fluorescence lifetime imaging and optical coherence elastography. BIOMEDICAL OPTICS EXPRESS 2025; 16:1006-1024. [PMID: 40109538 PMCID: PMC11919344 DOI: 10.1364/boe.549695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 03/22/2025]
Abstract
We developed a novel methodology for manufacturing multimodal, tissue-mimicking phantoms that exhibit both molecular and biomechanical contrast. This methodology leverages the immiscibility of silicone and hydrogels to create solid mesoscale phantoms with localized regions of precisely controlled fluorescence, including fluorescence lifetime properties, and adjustable stiffness, without requiring physical barriers. Mechanical, fluorescent, and optical characterization confirmed the tunability of the phantoms across a range of values relevant to biomedical applications. A macroscale 3D phantom was fabricated, and its properties were validated through fluorescence lifetime imaging (FLI) and optical coherence elastography (OCE). Validation demonstrated the successful tuning of both mechanical and fluorescence lifetime contrasts within a 3D structure, highlighting the feasibility of multimodal FLI-OCE. This new phantom manufacturing process is expected to support the development and validation of new multimodal imaging approaches to study molecular and biomechanical properties of the tumor microenvironment (TME), as well as their impact on therapeutic efficacy, and to enhance targeted therapies.
Collapse
Affiliation(s)
- Luis Chavez
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Shan Gao
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Vikas Pandey
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Nanxue Yuan
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Saif Ragab
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Matt S Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Percy Smith
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Caroline Edelheit
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - David T Corr
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, 5 Grudziądzka St., 87-100 Toruń, Poland
| | - Xavier Intes
- Center for Modeling, Simulation and Imaging for Medicine, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| |
Collapse
|
32
|
Wang D, Wu J, Xu Z, Jia J, Lai Y, He Z. Increased Matrix Stiffness Promotes Slow Muscle Fibre Regeneration After Skeletal Muscle Injury. J Cell Mol Med 2025; 29:e70423. [PMID: 39969079 PMCID: PMC11837045 DOI: 10.1111/jcmm.70423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/02/2025] [Accepted: 01/30/2025] [Indexed: 02/20/2025] Open
Abstract
The global prevalence of skeletal muscle diseases has progressively escalated in recent years. This study aimed to explore the potential role of matrix stiffness in the repair mechanisms following skeletal muscle injury. We observed an increase in muscle stiffness, a significant rise in the number of type I muscle fibres and a notable elevation in mRNA expression levels of Myh7/2 alongside a decrease in Myh1/4 on day 3 post tibialis anterior muscle injury. To replicate these in vivo changes, C2C12 cells were cultured under high matrix stiffness conditions, and compared to those on low matrix stiffness, the C2C12 cells cultured on high matrix stiffness showed increased expression levels of Myh7/2 mRNA and production levels of MYH7/2, indicating differentiation into slow-twitch muscle fibre types. Furthermore, up-regulation of DRP1 phosphorylation along with elevated F-actin fluorescence intensity and RHOA and ROCK1 production indicates that high matrix stiffness induces cytoskeletal remodelling to regulate mitochondrial fission processes. Our data also revealed up-regulation in mRNA expression level for Actb, phosphorylation level for DRP1, mitochondrial quantity and MYH7/2 production level. Importantly, these effects were effectively reversed by the application of ROCK inhibitor Y-27632, highlighting that targeting cytoskeletal dynamics can modulate myogenic differentiation pathways within C2C12 cells. These findings provide valuable insights into how matrix stiffness influences fibre type transformation during skeletal muscle injury repair while suggesting potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Physical EducationAnhui University of TechnologyMaanshanAnhuiChina
| | - Jiahong Wu
- Department of MedicineSun Yat‐Sen UniversityShenzhenGuangdongChina
| | - Zeyu Xu
- Department of MedicineSun Yat‐Sen UniversityShenzhenGuangdongChina
| | - Jinning Jia
- Department of PathologyThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Yimei Lai
- Department of PathologyThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Zhihua He
- Institute of UrologyThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| |
Collapse
|
33
|
Taufalele PV, Kirkham HK, Reinhart-King CA. Matrix Stiffness-Mediated DNA Methylation in Endothelial Cells. Cell Mol Bioeng 2025; 18:29-38. [PMID: 39949487 PMCID: PMC11813852 DOI: 10.1007/s12195-024-00836-9] [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: 01/16/2024] [Accepted: 11/26/2024] [Indexed: 02/16/2025] Open
Abstract
Purpose Altered tissue mechanics is a prominent feature of many pathological conditions including cancer. As such, much work has been dedicated to understanding how mechanical features of tissues contribute to pathogenesis. Interestingly, previous work has demonstrated that the tumor vasculature acquires pathological features in part due to enhanced tumor stiffening. To further understand how matrix mechanics may be translated into altered cell behavior and ultimately affect tumor vasculature function, we have investigated the effects of substrate stiffening on endothelial epigenetics. Specifically, we have focused on DNA methylation as recent work indicates DNA methylation in endothelial cells can contribute to aberrant behavior in a range of pathological conditions. Methods Human umbilical vein endothelial cells (HUVECs) were seeded on stiff and compliant collagen-coated polyacrylamide gels and allowed to form monolayers over 5 days. DNA methylation was assessed via 5-methylcytosine ELISA assays and immunofluorescent staining. Gene expression was assessed via qPCR on RNA isolated from HUVECs seeded on collagen-coated polyacrylamide gels of varying stiffness. Results Our work demonstrates that endothelial cells cultured on stiffer substrates exhibit lower levels of global DNA methylation relative to endothelial cells cultured on more compliant substrates. Interestingly, gene expression and DNA methylation dynamics suggest stiffness-mediated gene expression may play a role in establishing or maintaining differential DNA methylation levels in addition to enzyme activity. Additionally, we found that the process of passaging induced higher levels of global DNA methylation. Conclusions Altogether, our results underscore the importance of considering cell culture substrate mechanics to preserve the epigenetic integrity of primary cells and obtain analyses that recapitulate the primary environment. Furthermore, these results serve as an important launching point for further work studying the intersection tissue mechanics and epigenetics under pathological conditions.
Collapse
Affiliation(s)
- Paul V. Taufalele
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
| | - Hannah K. Kirkham
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
- Bioengineering Department, Rice University, Houston, TX USA
| |
Collapse
|
34
|
Keshavanarayana P, Aparicio-Yuste R, Spill F, Gomez-Benito MJ, Bastounis EE. Leveraging computational modeling to explore epithelial and endothelial cell monolayer mechanobiology. Trends Cell Biol 2025:S0962-8924(24)00282-4. [PMID: 39837738 DOI: 10.1016/j.tcb.2024.12.014] [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: 09/13/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/23/2025]
Abstract
Endothelial cells (ENCs) and epithelial cells (EPCs) form monolayers whose barrier function is critical for the maintenance of physiological processes and extremely sensitive to mechanical cues. Computational models have emerged as powerful tools to elucidate how mechanical cues impact the behavior of these monolayers in health and disease. Herein, the importance of mechanics in regulating ENC and EPC monolayer behavior is established, highlighting similarities and differences in various biological contexts. Concurrently, computational approaches and their importance in accelerating mechanobiology studies are discussed, emphasizing their limitations and suggesting future directions. The aim is to inspire further synergies between cell biologists and modelers, which are crucial for accelerating cell mechanobiology research.
Collapse
Affiliation(s)
- Pradeep Keshavanarayana
- School of Mathematics, University of Birmingham, Birmingham, UK; Centre for Computational Medicine, University College London, London, UK
| | - Raul Aparicio-Yuste
- Multiscale in Mechanical and Biological Engineering (M2BE), Engineering Research Institute of Aragon (I3A), Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain; Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Cluster of Excellence 'Controlling Microbes to Fight Infections' (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany
| | - Fabian Spill
- School of Mathematics, University of Birmingham, Birmingham, UK.
| | - Maria Jose Gomez-Benito
- Multiscale in Mechanical and Biological Engineering (M2BE), Engineering Research Institute of Aragon (I3A), Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain.
| | - Effie E Bastounis
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Cluster of Excellence 'Controlling Microbes to Fight Infections' (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany.
| |
Collapse
|
35
|
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
|
36
|
Zhang X, Al‐Danakh A, Zhu X, Feng D, Yang L, Wu H, Li Y, Wang S, Chen Q, Yang D. Insights into the mechanisms, regulation, and therapeutic implications of extracellular matrix stiffness in cancer. Bioeng Transl Med 2025; 10:e10698. [PMID: 39801760 PMCID: PMC11711218 DOI: 10.1002/btm2.10698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/19/2024] [Accepted: 06/29/2024] [Indexed: 01/03/2025] Open
Abstract
The tumor microenvironment (TME) is critical for cancer initiation, growth, metastasis, and therapeutic resistance. The extracellular matrix (ECM) is a significant tumor component that serves various functions, including mechanical support, TME regulation, and signal molecule generation. The quantity and cross-linking status of ECM components are crucial factors in tumor development, as they determine tissue stiffness and the interaction between stiff TME and cancer cells, resulting in aberrant mechanotransduction, proliferation, migration, invasion, angiogenesis, immune evasion, and treatment resistance. Therefore, broad knowledge of ECM dysregulation in the TME might aid in developing innovative cancer therapies. This review summarized the available information on major ECM components, their functions, factors that increase and decrease matrix stiffness, and related signaling pathways that interplay between cancer cells and the ECM in TME. Moreover, mechanotransduction alters during tumorogenesis, and current drug therapy based on ECM as targets, as well as future efforts in ECM and cancer, are also discussed.
Collapse
Affiliation(s)
- Ximo Zhang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Abdullah Al‐Danakh
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xinqing Zhu
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Dan Feng
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Linlin Yang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Haotian Wu
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yingying Li
- Department of Discipline ConstructionDalian Medical UniversityDalianChina
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of GlycobiologyDalian Medical UniversityDalianChina
| | - Qiwei Chen
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Zhongda Hospital, Medical School Advanced Institute Life HealthSoutheast UniversityNanjingChina
| | - Deyong Yang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of SurgeryHealinghands ClinicDalianChina
| |
Collapse
|
37
|
Chen Y, Jiang J, Yan C, Jiang J, Shi B, Xu Z, Yuan F, Zhang H, Zhang J. Prediction of tumor regression grade in far-advanced gastric cancer after preoperative immuno-chemotherapy using dual-energy CT-derived extracellular volume fraction. Eur Radiol 2025; 35:93-104. [PMID: 38981889 DOI: 10.1007/s00330-024-10737-0] [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] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVES This study examines the effectiveness of dual-energy CT (DECT) delayed-phase extracellular volume (ECV) fraction in predicting tumor regression grade (TRG) in far-advanced gastric cancer (FAGC) patients receiving preoperative immuno-chemotherapy. MATERIALS AND METHODS A retrospective analysis was performed on far-advanced gastric adenocarcinoma patients treated with preoperative immuno-chemotherapy at our institution from August 2019 to March 2023. Patients were categorized based on their TRG into pathological complete response (pCR) and non-pCR groups. ECV was determined using the delayed-phase iodine maps. In addition, tumor iodine densities and standardized iodine ratios were meticulously analyzed using the triple-phase enhanced iodine maps. Univariate analysis with five-fold cross-validation and Spearman correlation determined DECT parameters and clinical indicators association with pCR. The predictive accuracy of these parameters for pCR was evaluated using a weighted logistic regression model with five-fold cross-validation. RESULTS Of the 88 patients enrolled (mean age 60.8 ± 11.1 years, 63 males), 21 (23.9%) achieved pCR. Univariate analysis indicated ECV's significant role in differentiating between pCR and non-pCR groups (average p value = 0.021). In the logistic regression model, ECV independently predicted pCR with an average odds ratio of 0.911 (95% confidence interval, 0.798-0.994). The model, incorporating ECV, tumor area, and IDAV (the relative change rate of iodine density from venous phase to arterial phase), showed an average area under curves (AUCs) of 0.780 (0.770-0.791) and 0.766 (0.731-0.800) for the training and validation sets, respectively, in predicting pCR. CONCLUSION DECT-derived ECV fraction is a valuable predictor of TRG in FAGC patients undergoing preoperative immuno-chemotherapy. CLINICAL RELEVANCE STATEMENT This study demonstrates that DECT-derived extracellular volume fraction is a reliable predictor for pathological complete response in far-advanced gastric cancer patients receiving preoperative immuno-chemotherapy, offering a noninvasive tool for identifying potential treatment beneficiaries.
Collapse
Affiliation(s)
- Yong Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Yan
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiang Jiang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bowen Shi
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihan Xu
- Siemens Healthineers Ltd, Shanghai, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
38
|
Zhu B, Li F, Yu J, Liang Z, Ke X, Wang Y, Song Z, Li Z, Li G, Guo Y. PIEZO1 mediates matrix stiffness-induced tumor progression in kidney renal clear cell carcinoma by activating the Ca 2+/Calpain/YAP pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119871. [PMID: 39490703 DOI: 10.1016/j.bbamcr.2024.119871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE The significance of physical factors in the onset and progression of tumors has been increasingly substantiated by a multitude of studies. The extracellular matrix, a pivotal component of the tumor microenvironment, has been the subject of extensive investigation in connection with the advancement of KIRC (Kidney Renal Clear Cell Carcinoma) in recent years. PIEZO1, a mechanosensitive ion channel, has been recognized as a modulator of diverse physiological processes. Nonetheless, the precise function of PIEZO1 as a transducer of mechanical stimuli in KIRC remains poorly elucidated. METHODS A bioinformatics analysis was conducted using data from The Cancer Genome Atlas (TCGA) and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) to explore the correlation between matrix stiffness indicators, such as COL1A1 and LOX mRNA levels, and KIRC prognosis. Expression patterns of mechanosensitive ion channels, particularly PIEZO1, were examined. Collagen-coated polyacrylamide hydrogel models were utilized to simulate varying stiffness environments and study their effects on KIRC cell behavior in vitro. Functional experiments, including PIEZO1 knockdown and overexpression, were performed to investigate the molecular mechanisms underlying matrix stiffness-induced cellular changes. Interventions in the Ca2+/Calpain/YAP Pathway were conducted to evaluate their effects on cell growth, EMT, and stemness characteristics. RESULTS Our findings indicate a significant correlation between matrix stiffness and the prognosis of KIRC patients. It is observed that higher mechanical stiffness can facilitate the growth and metastasis of KIRC cells. Notably, we have also observed that the deficiency of PIEZO1 hinders the proliferation, EMT, and stemness characteristics of KIRC cells induced by a stiff matrix. Our study suggests that PIEZO1 plays a crucial role in mediating KIRC growth and metastasis through the activation of the Ca2+/Calpain/YAP Pathway. CONCLUSION This study elucidates a novel mechanism through which the activation of PIEZO1 leads to calcium influx, subsequent calpain activation, and YAP nuclear translocation, thereby contributing to the progression of KIRC driven by matrix stiffness.
Collapse
Affiliation(s)
- Biqiang Zhu
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| | - Fan Li
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| | - Jiajun Yu
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Zhulin Liang
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Xinwen Ke
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Yong Wang
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Zhengshuai Song
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Zhongyuan Li
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Guohao Li
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Yonglian Guo
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| |
Collapse
|
39
|
Zhang S, Wang H. Targeting the lung tumour stroma: harnessing nanoparticles for effective therapeutic interventions. J Drug Target 2025; 33:60-86. [PMID: 39356091 DOI: 10.1080/1061186x.2024.2410462] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Lung cancer remains an influential global health concern, necessitating the development of innovative therapeutic strategies. The tumour stroma, which is known as tumour microenvironment (TME) has a central impact on tumour expansion and treatment resistance. The stroma of lung tumours consists of numerous cells and molecules that shape an environment for tumour expansion. This environment not only protects tumoral cells against immune system attacks but also enables tumour stroma to attenuate the action of antitumor drugs. This stroma consists of stromal cells like cancer-associated fibroblasts (CAFs), suppressive immune cells, and cytotoxic immune cells. Additionally, the presence of stem cells, endothelial cells and pericytes can facilitate tumour volume expansion. Nanoparticles are hopeful tools for targeted drug delivery because of their extraordinary properties and their capacity to devastate biological obstacles. This review article provides a comprehensive overview of contemporary advancements in targeting the lung tumour stroma using nanoparticles. Various nanoparticle-based approaches, including passive and active targeting, and stimuli-responsive systems, highlighting their potential to improve drug delivery efficiency. Additionally, the role of nanotechnology in modulating the tumour stroma by targeting key components such as immune cells, extracellular matrix (ECM), hypoxia, and suppressive elements in the lung tumour stroma.
Collapse
Affiliation(s)
- Shushu Zhang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Hui Wang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
40
|
Mungai RW, Hartman RJ, Jolin GE, Piskorowski KW, Billiar KL. Towards a more objective and high-throughput spheroid invasion assay quantification method. Sci Rep 2024; 14:31007. [PMID: 39730859 DOI: 10.1038/s41598-024-82191-3] [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: 02/07/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
Multicellular spheroids embedded in 3D hydrogels are prominent in vitro models for 3D cell invasion. Yet, quantification methods for spheroid cell invasion that are high-throughput, objective and accessible are still lacking. Variations in spheroid sizes and the shapes of the cells within render it difficult to objectively assess invasion extent. The goal of this work is to develop a high-throughput quantification method of cell invasion into 3D matrices that minimizes sensitivity to initial spheroid size and cell spreading and provides precise integrative directionally-dependent metrics of invasion. By analyzing images of fluorescent cell nuclei, invasion metrics are automatically calculated at the pixel level. The initial spheroid boundary is segmented and automated calculations of the nuclear pixel distances from the initial boundary are used to compute common invasion metrics (i.e., the change in invasion area, mean distance) for the same spheroid at a later timepoint. We also introduce the area moment of inertia as an integrative metric of cell invasion that considers the invasion area as well as the pixel distances from the initial spheroid boundary. Further, we show that principal component analysis can be used to quantify the directional influence of a stimuli to invasion (e.g., due to a chemotactic gradient or contact guidance). To demonstrate the power of the analysis for cell types with different invasive potentials and the utility of this method for a variety of biological applications, the method is used to analyze the invasiveness of five different cell types. In all, implementation of this high-throughput quantification method results in consistent and objective analysis of 3D multicellular spheroid invasion. We provide the analysis code in both MATLAB and Python languages as well as a GUI for ease of use for researchers with a range of computer programming skills and for applications in a variety of biological research areas such as wound healing and cancer metastasis.
Collapse
Affiliation(s)
- Rozanne W Mungai
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | | | - Grace E Jolin
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | - Kevin W Piskorowski
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | - Kristen L Billiar
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01605, USA.
| |
Collapse
|
41
|
Dorjkhorloo G, Shiraishi T, Erkhem-Ochir B, Sohda M, Okami H, Yamaguchi A, Shioi I, Komine C, Nakazawa N, Shibasaki Y, Okada T, Osone K, Sano A, Sakai M, Ogawa H, Katayama A, Oyama T, Yokobori T, Shirabe K, Saeki H. High levels of fibrotic tumor components are associated with recurrence and intratumoral immune status in advanced colorectal cancer patients. Sci Rep 2024; 14:30735. [PMID: 39730445 DOI: 10.1038/s41598-024-80489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/19/2024] [Indexed: 12/29/2024] Open
Abstract
The importance of collagen and elastin remains incompletely understood concerning tumor immunity in cancer tissues. This study explored the clinical significance of collagen and elastin deposition on tumor immunity in advanced colorectal cancer patients. The collagen and elastin contents were assessed simultaneously using elastic van Gieson (EVG) histochemical staining. Immunohistochemical staining was performed to measure the immune cell markers CD3, CD8, CD86, and CD163 in surgically resected primary tumors from 78 pT4 colorectal cancer patients. High collagen, elastin, and EVG scores are associated with aggressive characteristics and short disease-free survival. A high EVG score was identified as an independent predictor of poor disease-free survival. Furthermore, tumors with high collagen and EVG scores exhibited significantly fewer intratumoral CD3 + and CD8 + cells. Evaluating tumor fibrosis using the classical and straightforward EVG staining method could be a reliable predictor of recurrence in high-risk colorectal cancer patients with tumor immune tolerance.
Collapse
Affiliation(s)
- Gendensuren Dorjkhorloo
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuya Shiraishi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Bilguun Erkhem-Ochir
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sohda
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Haruka Okami
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Arisa Yamaguchi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ikuma Shioi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Chika Komine
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Nobuhiro Nakazawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yuta Shibasaki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuhisa Okada
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Katsuya Osone
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Akihiko Sano
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sakai
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroomi Ogawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ayaka Katayama
- Department of Diagnostic Pathology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takehiko Yokobori
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Ken Shirabe
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroshi Saeki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
42
|
Pan H, Zhang X, Zhu S, Zhu B, Wu D, Yan J, Guan X, Huang Y, Zhao Y, Yang Y, Guo Y. Piezo1 Mediates Glycolysis-Boosted Pancreatic Ductal Adenocarcinoma Chemoresistance within a Biomimetic Three-Dimensional Matrix Stiffness. ACS Biomater Sci Eng 2024; 10:7632-7646. [PMID: 39556518 DOI: 10.1021/acsbiomaterials.4c01319] [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: 11/20/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer with a very low 5-year survival rate, which is partially attributed to chemoresistance. Although the regulation of chemoresistance through biochemical signaling is well-documented, the influence of three-dimensional (3D) matrix stiffness is poorly understood. In this study, gelatin methacrylate (GelMA) hydrogels were reconstructed with stiffnesses spanning the range from normal to cancerous PDAC tissues, which are termed as the soft group and stiff group. The PDAC cell lines (Mia-PaCa2 and CFPAC-1) encapsulated in the stiff group displayed a chemoresistance phenotype and were prominent against gemcitabine. RNA-sequencing and bioinformatics analysis indicated that glycolysis was apparently enriched in the stiff group versus the soft group, which was also validated through assays of glucose uptake, lactate production, and the expression of GLUT2, HK2, and LDHA. A rescue assay with 2-deoxy-d-glucose and N-acetylcysteine demonstrated that glycolysis is involved in chemoresistance. Furthermore, the expression of Piezo1 and the content of Ca2+ were elevated in the stiff group. The addition of Yoda1 (Piezo1 agonist) in the soft group promoted glycolysis, whereas in the stiff group, treatment with GsMTx4 (Piezo1 inhibitor) inhibited glycolysis, which showcased that Piezo1 participated in 3D matrix stiffness-induced glycolysis. Taken together, Piezo1-mediated glycolysis was involved in PDAC chemoresistance triggered by the 3D matrix stiffness. Our study sheds light on the mechanism underlying chemoresistance in PDAC from the perspective of 3D mechanical cues.
Collapse
Affiliation(s)
- Haopeng Pan
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
- Key Laboratory of Neuro-regeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuro-regeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Xue Zhang
- Key Laboratory of Neuro-regeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuro-regeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Shajun Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Biwen Zhu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Di Wu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jiashuai Yan
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaoqi Guan
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Yan Huang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Yahong Zhao
- Key Laboratory of Neuro-regeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuro-regeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yumin Yang
- Key Laboratory of Neuro-regeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuro-regeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
43
|
Dong Y, Lu M, Yin Y, Wang C, Dai N. Tumor Biomechanics-Inspired Future Medicine. Cancers (Basel) 2024; 16:4107. [PMID: 39682291 DOI: 10.3390/cancers16234107] [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: 10/30/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Malignant tumors pose a significant global health challenge, severely threatening human health. Statistics from the World Health Organization indicate that, in 2022, there were nearly 20 million new cancer cases and 9.7 million cancer-related deaths. Therefore, it is urgently necessary to study the pathogenesis of cancer and explore effective diagnostic and treatment strategies. In recent years, research has highlighted the importance of mechanical cues in tumors, which have become a new hallmark of cancer and a key factor in regulating tumor behavior. This suggests that studying the mechanical properties of tumors may open potential new avenues for understanding the pathogenesis, diagnosis, and therapeutic intervention of cancer. This review summarizes the mechanical characteristics of tumors and the development of tumor diagnostics and treatments targeting specific mechanical factors. Finally, we propose new ideas and insights for the application of mechanomedicine in cancer diagnosis and treatment in the future.
Collapse
Affiliation(s)
- Yuqing Dong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Mengnan Lu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Yuting Yin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Cong Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Ningman Dai
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
44
|
Asadi M, Zafari V, Sadeghi-Mohammadi S, Shanehbandi D, Mert U, Soleimani Z, Caner A, Zarredar H. The role of tumor microenvironment and self-organization in cancer progression: Key insights for therapeutic development. BIOIMPACTS : BI 2024; 15:30713. [PMID: 40256216 PMCID: PMC12008505 DOI: 10.34172/bi.30713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/08/2024] [Accepted: 11/20/2024] [Indexed: 04/22/2025]
Abstract
Introduction The tumor microenvironment (TME) plays a pivotal role in cancer progression, influencing tumor initiation, growth, invasion, metastasis, and response to therapies. This study explores the dynamic interactions within the TME, particularly focusing on self-organization-a process by which tumor cells and their microenvironment reciprocally shape one another, leading to cancer progression and resistance. Understanding these interactions can reveal new prognostic markers and therapeutic targets within the TME, such as extracellular matrix (ECM) components, immune cells, and cytokine signaling pathways. Methods A comprehensive search method was employed to investigate the current academic literature on TME, particularly focusing on self-organization in the context of cancer progression and resistance across the PubMed, Google Scholar, and Science Direct databases. Results Recent studies suggest that therapies that disrupt TME self-organization could improve patient outcomes by defeating drug resistance and increasing the effectiveness of conventional therapy. Additionally, this research highlights the essential of understanding the biophysical properties of the TME, like cytoskeletal alterations, in the development of more effective malignancy therapy. Conclusion This review indicated that targeting the ECM and immune cells within the TME can improve therapy effectiveness. Also, by focusing on TME self-organization, we can recognize new therapeutic plans to defeat drug resistance.
Collapse
Affiliation(s)
- Milad Asadi
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Venus Zafari
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Sanam Sadeghi-Mohammadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ufuk Mert
- Institute of Health Sciences, Department of Basic Oncology, Ege University, Izmir, Turkey
| | - Zahra Soleimani
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayşe Caner
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
45
|
Jamshidi MH, Karami A, Keshavarz A, Fatemi A, Ghanavati S. Magnetic Resonance Elastography for Breast Cancer Diagnosis Through the Assessment of Tissue Biomechanical Properties. Health Sci Rep 2024; 7:e70253. [PMID: 39669189 PMCID: PMC11635177 DOI: 10.1002/hsr2.70253] [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: 06/11/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 12/14/2024] Open
Abstract
Background and Aim Breast cancer and normal breast tissue exhibit different degrees of stiffness, indicating distinct biomechanical properties. Study results reveal that breast cancer tissue is several times stiffer than normal breast tissue. These variations can serve as indicative factors for imaging purposes. Depicting markers can significantly enhance the process of breast cancer diagnosis and treatment. This article provides a brief review of the biomechanical properties of breast cancer tissue, highlighting the role of the magnetic resonance elastography (MRE) technique in utilizing these properties for diagnosing breast cancer. Methods In breast MRE, low-frequency shear waves are employed to measure breast stiffness. This method not only offers a quantitative diagnosis but also generates an elastogram, determining the stiffness of each area through its colors. Results MRE represents a diagnostic technique with heightened sensitivity, based on depicting the viscoelasticity properties of breast tissue and describing tumors in terms of biomechanical properties. Combining tissue biomechanical properties, such as tissue stiffness, with contrast-enhanced breast Magnetic Resonance Imaging (MRI) leads to tumor diagnosis. The value of MRE in oncological imaging aims at the early detection of tumors and evaluating the prognosis of breast cancer. Conclusion Breast MRE can identify the reduction of interstitial pressure in tumors by detecting changes in tissue stiffness, making it an effective tool for monitoring treatment responses. This technique is safe, repeatable, and highly precise, significantly aiding in patient screening.
Collapse
Affiliation(s)
- Mohammad Hossein Jamshidi
- Department of Medical Imaging and Radiation Sciences, School of Allied Medical SciencesAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Aida Karami
- Department of Medical Imaging and Radiation Sciences, School of Allied Medical SciencesAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Amirhesam Keshavarz
- Department of Anatomical Science, Faculty of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Ali Fatemi
- Department of PhysicsJackson State UniversityJacksonMississippiUSA
- Department of Radiation OncologyGamma Knife CenterJacksonMississippiUSA
| | - Sepehr Ghanavati
- Department of Medicine, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| |
Collapse
|
46
|
Jiménez-Santos M, García-Martín S, Rubio-Fernández M, Gómez-López G, Al-Shahrour F. Spatial transcriptomics in breast cancer reveals tumour microenvironment-driven drug responses and clonal therapeutic heterogeneity. NAR Cancer 2024; 6:zcae046. [PMID: 39703753 PMCID: PMC11655296 DOI: 10.1093/narcan/zcae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
Breast cancer patients are categorized into three subtypes with distinct treatment approaches. Precision oncology has increased patient outcomes by targeting the specific molecular alterations of tumours, yet challenges remain. Treatment failure persists due to the coexistence of several malignant subpopulations with different drug sensitivities within the same tumour, a phenomenon known as intratumour heterogeneity (ITH). This heterogeneity has been extensively studied from a tumour-centric view, but recent insights underscore the role of the tumour microenvironment in treatment response. Our research utilizes spatial transcriptomics data from breast cancer patients to predict drug sensitivity. We observe diverse response patterns across tumour, interphase and microenvironment regions, unveiling a sensitivity and functional gradient from the tumour core to the periphery. Moreover, we find tumour therapeutic clusters with different drug responses associated with distinct biological functions driven by unique ligand-receptor interactions. Importantly, we identify genetically identical subclones with different responses depending on their location within the tumour ducts. This research underscores the significance of considering the distance from the tumour core and microenvironment composition when identifying suitable treatments to target ITH. Our findings provide critical insights into optimizing therapeutic strategies, highlighting the necessity of a comprehensive understanding of tumour biology for effective cancer treatment.
Collapse
Affiliation(s)
- María José Jiménez-Santos
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Santiago García-Martín
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Marcos Rubio-Fernández
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
- Lung-H120 Group, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|
47
|
Cetin M, Saatci O, Rezaeian AH, Rao CN, Beneker C, Sreenivas K, Taylor H, Pederson B, Chatzistamou I, Buckley B, Lessner S, Angel P, McInnes C, Sahin O. A highly potent bi-thiazole inhibitor of LOX rewires collagen architecture and enhances chemoresponse in triple-negative breast cancer. Cell Chem Biol 2024; 31:1926-1941.e11. [PMID: 39043186 PMCID: PMC11585458 DOI: 10.1016/j.chembiol.2024.06.012] [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: 09/14/2023] [Revised: 04/12/2024] [Accepted: 06/22/2024] [Indexed: 07/25/2024]
Abstract
Lysyl oxidase (LOX) is upregulated in highly stiff aggressive tumors, correlating with metastasis, resistance, and worse survival; however, there are currently no potent, safe, and orally bioavailable small molecule LOX inhibitors to treat these aggressive desmoplastic solid tumors in clinics. Here we discovered bi-thiazole derivatives as potent LOX inhibitors by robust screening of drug-like molecules combined with cell/recombinant protein-based assays. Structure-activity relationship analysis identified a potent lead compound (LXG6403) with ∼3.5-fold specificity for LOX compared to LOXL2 while not inhibiting LOXL1 with a competitive, time- and concentration-dependent irreversible mode of inhibition. LXG6403 shows favorable pharmacokinetic properties, globally changes ECM/collagen architecture, and reduces tumor stiffness. This leads to better drug penetration, inhibits FAK signaling, and induces ROS/DNA damage, G1 arrest, and apoptosis in chemoresistant triple-negative breast cancer (TNBC) cell lines, PDX organoids, and in vivo. Overall, our potent and tolerable bi-thiazole LOX inhibitor enhances chemoresponse in TNBC, the deadliest breast cancer subtype.
Collapse
Affiliation(s)
- Metin Cetin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ozge Saatci
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Abdol-Hossein Rezaeian
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Chintada Nageswara Rao
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Chad Beneker
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kukkamudi Sreenivas
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Harrison Taylor
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Breanna Pederson
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & Immunology, University of South Carolina, Columbia, SC 29208, USA
| | - Brian Buckley
- Small Molecule Screening Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Susan Lessner
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Peggi Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Campbell McInnes
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
48
|
Suresh M, Bhat R. Ovarian cancer cells exhibit diverse migration strategies on stiff collagenous substrata. Biophys J 2024; 123:4009-4021. [PMID: 39449201 PMCID: PMC11617636 DOI: 10.1016/j.bpj.2024.10.014] [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: 07/16/2024] [Revised: 09/17/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
In homoeostasis, the shape and sessility of untransformed epithelial cells are intricately linked together. Variations of this relationship in migrating cancer cells as they encounter different microenvironments are as yet ill understood. Here, we explore the interdependency of such traits in two morphologically distinct invasive ovarian cancer cell lines (OVCAR-3 and SK-OV-3) under mechanically variant contexts. We first established a metric toolkit that assessed traits associated with cell motion and shape, and rigorously measured their dynamical variation across trajectories of migration using a Shannon entropic distribution. Two stiffness conditions on polymerized collagen I with Young's moduli of 0.5 kPa (soft) and 20 kPa (stiff) were chosen. Both the epithelioid OVCAR-3 and mesenchymal SK-OV-3 cells on soft substrata exhibited slow and undirected migration. On stiff substrata, SK-OV-3 showed faster persistent directed motion. Surprisingly, OVCAR-3 cells on stiffer substrata moved even faster than SK-OV-3 cells but showed a distinct angular motion. The polarity of SK-OV-3 cells on stiff substrata was well correlated with their movement, whereas, for OVCAR-3, we observed an unusual "slip" behavior, wherein the axes of cell shape and movement were poorly correlated. Whereas SK-OV-3 and OVCAR-3 showed greater mean deformation on stiffer substrata, the latter was anticorrelated with variation in angular motion or the mean deviation between shape and motility axis for SK-OV-3 but poorly correlated for OVCAR-3. Moreover, on softer substrata OVCAR-3 and SK-OV-3 were relatively rigid but showed greater shape variation (with OVCAR-3 showing a higher fold change) on stiffer substrata. Our findings suggest that greater deformability on stiffer milieu allow epithelioid cells to overcome constraints on the congruence in axis of shape and motion seen for mesenchymal cells and display distinct motile behaviors across this phenotypic spectrum.
Collapse
Affiliation(s)
- Madhumitha Suresh
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | - Ramray Bhat
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India; Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
49
|
Biray Avci C, Goker Bagca B, Nikanfar M, Takanlou LS, Takanlou MS, Nourazarian A. Tumor microenvironment and cancer metastasis: molecular mechanisms and therapeutic implications. Front Pharmacol 2024; 15:1442888. [PMID: 39600368 PMCID: PMC11588459 DOI: 10.3389/fphar.2024.1442888] [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: 06/03/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and metastasis. This review summarizes the current research on how the TME promotes metastasis through molecular pathways, focusing on key components, such as cancer-associated fibroblasts, immune cells, endothelial cells, cytokines, and the extracellular matrix. Significant findings have highlighted that alterations in cellular communication within the TME enable tumor cells to evade immune surveillance, survive, and invade other tissues. This review highlights the roles of TGF-β and VEGF signaling in promoting angiogenesis and extracellular matrix remodeling, which facilitate metastasis. Additionally, we explored how metabolic reprogramming of tumor and stromal cells, influenced by nutrient availability in the TME, drives cancer progression. This study also evaluated the therapeutic strategies targeting these interactions to disrupt metastasis. By providing a multidisciplinary perspective, this study suggests that understanding the molecular basis of the TME can lead to more effective cancer therapies and identify potential avenues for future research. Future research on the TME should prioritize unraveling the molecular and cellular interactions within this complex environment, which could lead to novel therapeutic strategies and personalized cancer treatments. Moreover, advancements in technologies such as single-cell analysis, spatial transcriptomics, and epigenetic profiling offer promising avenues for identifying new therapeutic targets and improving the efficacy of immunotherapies, particularly in the context of metastasis.
Collapse
Affiliation(s)
- Cigir Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Türkiye
| | - Bakiye Goker Bagca
- Department of Medical Biology, Faculty of Medicine, Adnan Menderes University, Aydin, Türkiye
| | - Masoud Nikanfar
- Department of Neurology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
- Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
50
|
Abstract
The extracellular matrix (ECM) is the complex meshwork of proteins and glycans that forms the scaffold that surrounds and supports cells. It exerts key roles in all aspects of metazoan physiology, from conferring physical and mechanical properties on tissues and organs to modulating cellular processes such as proliferation, differentiation and migration. Understanding the mechanisms that orchestrate the assembly of the ECM scaffold is thus crucial to understand ECM functions in health and disease. This Review discusses novel insights into the compositional diversity of matrisome components and the mechanisms that lead to tissue-specific assemblies and architectures tailored to support specific functions. The Review then highlights recently discovered mechanisms, including post-translational modifications and metabolic pathways such as amino acid availability and the circadian clock, that modulate ECM secretion, assembly and remodelling in homeostasis and human diseases. Last, the Review explores the potential of 'matritherapies', that is, strategies to normalize ECM composition and architecture to achieve a therapeutic benefit.
Collapse
Affiliation(s)
- Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|