1
|
Liu G, Shen Z, Chong H, Zhou J, Zhang T, Wang Y, Ma D, Yang Y, Chen Y, Wang H, Sack I, Guo J, Li R, Yan F. Three-Dimensional Multifrequency MR Elastography for Microvascular Invasion and Prognosis Assessment in Hepatocellular Carcinoma. J Magn Reson Imaging 2024; 60:2626-2640. [PMID: 38344910 DOI: 10.1002/jmri.29276] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Pretreatment identification of microvascular invasion (MVI) in hepatocellular carcinoma (HCC) is important when selecting treatment strategies. PURPOSE To improve models for predicting MVI and recurrence-free survival (RFS) by developing nomograms containing three-dimensional (3D) MR elastography (MRE). STUDY TYPE Prospective. POPULATION 188 patients with HCC, divided into a training cohort (n = 150) and a validation cohort (n = 38). In the training cohort, 106/150 patients completed a 2-year follow-up. FIELD STRENGTH/SEQUENCE 1.5T 3D multifrequency MRE with a single-shot spin-echo echo planar imaging sequence, and 3.0T multiparametric MRI (mp-MRI), consisting of diffusion-weighted echo planar imaging, T2-weighted fast spin echo, in-phase out-of-phase T1-weighted fast spoiled gradient-recalled dual-echo and dynamic contrast-enhanced gradient echo sequences. ASSESSMENT Multivariable analysis was used to identify the independent predictors for MVI and RFS. Nomograms were constructed for visualization. Models for predicting MVI and RFS were built using mp-MRI parameters and a combination of mp-MRI and 3D MRE predictors. STATISTICAL TESTS Student's t-test, Mann-Whitney U test, chi-squared or Fisher's exact tests, multivariable analysis, area under the receiver operating characteristic curve (AUC), DeLong test, Kaplan-Meier analysis and log rank tests. P < 0.05 was considered significant. RESULTS Tumor c and liver c were independent predictors of MVI and RFS, respectively. Adding tumor c significantly improved the diagnostic performance of mp-MRI (AUC increased from 0.70 to 0.87) for MVI detection. Of the 106 patients in the training cohort who completed the 2-year follow up, 34 experienced recurrence. RFS was shorter for patients with MVI-positive histology than MVI-negative histology (27.1 months vs. >40 months). The MVI predicted by the 3D MRE model yielded similar results (26.9 months vs. >40 months). The MVI and RFS nomograms of the histologic-MVI and model-predicted MVI-positive showed good predictive performance. DATA CONCLUSION Biomechanical properties of 3D MRE were biomarkers for MVI and RFS. MVI and RFS nomograms were established. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Guixue Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhehan Shen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huanhuan Chong
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahao Zhou
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyi Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yikun Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchen Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huafeng Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ingolf Sack
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jing Guo
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ruokun Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
O’Dowling AT, Rodriguez BJ, Gallagher TK, Thorpe SD. Machine learning and artificial intelligence: Enabling the clinical translation of atomic force microscopy-based biomarkers for cancer diagnosis. Comput Struct Biotechnol J 2024; 24:661-671. [PMID: 39525667 PMCID: PMC11543504 DOI: 10.1016/j.csbj.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The influence of biomechanics on cell function has become increasingly defined over recent years. Biomechanical changes are known to affect oncogenesis; however, these effects are not yet fully understood. Atomic force microscopy (AFM) is the gold standard method for measuring tissue mechanics on the micro- or nano-scale. Due to its complexity, however, AFM has yet to become integrated in routine clinical diagnosis. Artificial intelligence (AI) and machine learning (ML) have the potential to make AFM more accessible, principally through automation of analysis. In this review, AFM and its use for the assessment of cell and tissue mechanics in cancer is described. Research relating to the application of artificial intelligence and machine learning in the analysis of AFM topography and force spectroscopy of cancer tissue and cells are reviewed. The application of machine learning and artificial intelligence to AFM has the potential to enable the widespread use of nanoscale morphologic and biomechanical features as diagnostic and prognostic biomarkers in cancer treatment.
Collapse
Affiliation(s)
- Aidan T. O’Dowling
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Department of Hepatobiliary and Transplant Surgery, St Vincent’s University Hospital, Dublin, Ireland
| | - Brian J. Rodriguez
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- UCD School of Physics, University College Dublin, Dublin, Ireland
| | - Tom K. Gallagher
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- Department of Hepatobiliary and Transplant Surgery, St Vincent’s University Hospital, Dublin, Ireland
| | - Stephen D. Thorpe
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
3
|
Carvalho EM, Ding EA, Saha A, Garcia DC, Weldy A, Zushin PJH, Stahl A, Aghi MK, Kumar S. Viscoelastic High-Molecular-Weight Hyaluronic Acid Hydrogels Support Rapid Glioblastoma Cell Invasion with Leader-Follower Dynamics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2404885. [PMID: 39508297 DOI: 10.1002/adma.202404885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/25/2024] [Indexed: 11/15/2024]
Abstract
Hyaluronic acid (HA), the primary component of brain extracellular matrix, is increasingly used to model neuropathological processes, including glioblastoma (GBM) tumor invasion. While elastic hydrogels based on crosslinked low-molecular-weight (LMW) HA are widely exploited for this purpose and have proven valuable for discovery and screening, brain tissue is both viscoelastic and rich in high-MW (HMW) HA, and it remains unclear how these differences influence invasion. To address this question, hydrogels comprised of either HMW (1.5 MDa) or LMW (60 kDa) HA are introduced, characterized, and applied in GBM invasion studies. Unlike LMW HA hydrogels, HMW HA hydrogels relax stresses quickly, to a similar extent as brain tissue, and to a greater extent than many conventional HA-based scaffolds. GBM cells implanted within HMW HA hydrogels invade much more rapidly than in their LMW HA counterparts and exhibit distinct leader-follower dynamics. Leader cells adopt dendritic morphologies similar to invasive GBM cells observed in vivo. Transcriptomic, pharmacologic, and imaging studies suggest that leader cells exploit hyaluronidase, an enzyme strongly enriched in human GBMs, to prime a path for followers. This study offers new insight into how HA viscoelastic properties drive invasion and argues for the use of highly stress-relaxing materials to model GBM.
Collapse
Affiliation(s)
- Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Atul Saha
- Department of Neurosurgery, University of California, San Francisco, CA, 94158, USA
| | - Diana Cruz Garcia
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94158, USA
| | - Anna Weldy
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 94720, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California, San Francisco, CA, 94158, USA
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
4
|
Tavasso M, Bordoloi AD, Tanré E, Dekker SAH, Garbin V, Boukany PE. Linking Metastatic Potential and Viscoelastic Properties of Breast Cancer Spheroids via Dynamic Compression and Relaxation in Microfluidics. Adv Healthc Mater 2024:e2402715. [PMID: 39400520 DOI: 10.1002/adhm.202402715] [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: 07/23/2024] [Revised: 09/13/2024] [Indexed: 10/15/2024]
Abstract
The growth and invasion of solid tumors are associated with changes in their viscoelastic properties, influenced by both internal cellular factors and physical forces in the tumor microenvironment. Due to the lack of a comprehensive investigation of tumor tissue viscoelasticity, the relationship between such physical properties and cancer malignancy remains poorly understood. Here, the viscoelastic properties of breast cancer spheroids, 3D (in vitro) tumor models, are studied in relation to their metastatic potentials by imposing controlled, dynamic compression within a microfluidic constriction, and subsequently monitoring the relaxation of the imposed deformation. By adopting a modified Maxwell model to extract viscoelastic properties from the compression data, the benign (MCF-10A) spheroids are found to have higher bulk elastic modulus and viscosity compared to malignant spheroids (MCF-7 and MDA-MB-231). The relaxation is characterized by two timescales, captured by a double exponential fitting function, which reveals a similar fast rebound for MCF-7 and MCF-10A. Both the malignant spheroids exhibit similar long-term relaxation and display residual deformation. However, they differ significantly in morphology, particularly in intercellular movements. These differences between malignant spheroids are demonstrated to be linked to their cytoskeletal organization, by microscopic imaging of F-actin within the spheroids, together with cell-cell adhesion strength.
Collapse
Affiliation(s)
- Margherita Tavasso
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | - Ankur D Bordoloi
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | - Elsa Tanré
- École Polytechnique, Institut Polytechnique de Paris, Palaiseau, 91120, France
| | - Sanne A H Dekker
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | - Valeria Garbin
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| |
Collapse
|
5
|
Habli Z, Zantout A, Al-Haj N, Saab R, El-Sabban M, Khraiche ML. Single-Cell Fluidic Force Spectroscopy Reveals Dynamic Mechanical Fingerprints of Malignancy in Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50147-50159. [PMID: 39105773 PMCID: PMC11440459 DOI: 10.1021/acsami.4c06335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
The interplay between cancer cell physical characteristics and metastatic potential highlights the significance of cancer cell mechanobiology. Using fluidic-based single-cell force spectroscopy (SCFS), quartz crystal microbalance with dissipation (QCM-D), and a model of cells with a spectrum of metastatic potential, we track the progression of biomechanics across the metastatic states by measuring cell-substrate and cell-to-cell adhesion forces, cell spring constant, cell height, and cell viscoelasticity. Compared to highly metastatic cells, cells in the lower spectrum of metastatic ability are found to be systematically stiffer, less viscoelastic, and larger. These mechanical transformations in cells within a cluster correlate with cells' metastatic potential but are significantly absent in single cells. Additionally, the response to chemotherapy is found to be highly dependent on cell viscoelastic properties in terms of both response time and magnitude. Shifts in cell softness and elasticity might serve as mechanoadaptive mechanisms during cancer cell metastasis, contributing to our understanding of metastasis and the effectiveness of potential therapeutic interventions.
Collapse
Affiliation(s)
- Zeina Habli
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Ahmad Zantout
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Nadine Al-Haj
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Raya Saab
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Massoud L Khraiche
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| |
Collapse
|
6
|
Mowla A, Hepburn MS, Li J, Vahala D, Amos SE, Hirvonen LM, Sanderson RW, Wijesinghe P, Maher S, Choi YS, Kennedy BF. Multimodal mechano-microscopy reveals mechanical phenotypes of breast cancer spheroids in three dimensions. APL Bioeng 2024; 8:036113. [PMID: 39257700 PMCID: PMC11387014 DOI: 10.1063/5.0213077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/01/2024] [Indexed: 09/12/2024] Open
Abstract
Cancer cell invasion relies on an equilibrium between cell deformability and the biophysical constraints imposed by the extracellular matrix (ECM). However, there is little consensus on the nature of the local biomechanical alterations in cancer cell dissemination in the context of three-dimensional (3D) tumor microenvironments (TMEs). While the shortcomings of two-dimensional (2D) models in replicating in situ cell behavior are well known, 3D TME models remain underutilized because contemporary mechanical quantification tools are limited to surface measurements. Here, we overcome this major challenge by quantifying local mechanics of cancer cell spheroids in 3D TMEs. We achieve this using multimodal mechano-microscopy, integrating optical coherence microscopy-based elasticity imaging with confocal fluorescence microscopy. We observe that non-metastatic cancer spheroids show no invasion while showing increased peripheral cell elasticity in both stiff and soft environments. Metastatic cancer spheroids, however, show ECM-mediated softening in a stiff microenvironment and, in a soft environment, initiate cell invasion with peripheral softening associated with early metastatic dissemination. This exemplar of live-cell 3D mechanotyping supports that invasion increases cell deformability in a 3D context, illustrating the power of multimodal mechano-microscopy for quantitative mechanobiology in situ.
Collapse
Affiliation(s)
| | | | | | - Danielle Vahala
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Sebastian E Amos
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Liisa M Hirvonen
- Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Perth, WA 6009, Australia
| | | | - Philip Wijesinghe
- Centre of Biophotonics, SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Samuel Maher
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | | |
Collapse
|
7
|
Stöberl S, Flommersfeld J, Kreft MM, Benoit M, Broedersz CP, Rädler JO. Nuclear deformation and dynamics of migrating cells in 3D confinement reveal adaptation of pulling and pushing forces. SCIENCE ADVANCES 2024; 10:eadm9195. [PMID: 39167661 PMCID: PMC11338266 DOI: 10.1126/sciadv.adm9195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 07/15/2024] [Indexed: 08/23/2024]
Abstract
Eukaryotic cells show an astounding ability to remodel their shape and cytoskeleton and to migrate through pores and constrictions smaller than their nuclear diameter. However, the relation of nuclear deformation and migration dynamics in confinement remains unclear. Here, we study the mechanics and dynamics of mesenchymal cancer cell nuclei transitioning through three-dimensional compliant hydrogel channels. We find a biphasic dependence of migration speed and transition frequency on channel width, peaking at widths comparable to the nuclear diameter. Using confocal imaging and hydrogel bead displacement, we determine nuclear deformations and corresponding forces during confined migration. The nucleus deforms reversibly with a reduction in volume during confinement. With decreasing channel width, the nuclear shape during transmigration changes biphasically, concomitant with the transitioning dynamics. Our proposed physical model explains the observed nuclear shapes and transitioning dynamics in terms of the cytoskeletal force generation adapting from purely pulling-based to a combined pulling- and pushing-based mechanism with increasing nuclear confinement.
Collapse
Affiliation(s)
- Stefan Stöberl
- Faculty of Physics and Center for NanoScience, Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, D-80539 Munich, Germany
| | - Johannes Flommersfeld
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of Physics, Ludwig-Maximilians-University Munich, Theresienstraße 37, D-80333 Munich, Germany
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081HV Amsterdam, Netherlands
| | - Maximilian M. Kreft
- Faculty of Physics and Center for NanoScience, Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, D-80539 Munich, Germany
| | - Martin Benoit
- Faculty of Physics and Center for NanoScience, Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, D-80539 Munich, Germany
| | - Chase P. Broedersz
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of Physics, Ludwig-Maximilians-University Munich, Theresienstraße 37, D-80333 Munich, Germany
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081HV Amsterdam, Netherlands
| | - Joachim O. Rädler
- Faculty of Physics and Center for NanoScience, Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, D-80539 Munich, Germany
| |
Collapse
|
8
|
López-Alonso J, Eroles M, Janel S, Berardi M, Pellequer JL, Dupres V, Lafont F, Rico F. PyFMLab: Open-source software for atomic force microscopy microrheology data analysis. OPEN RESEARCH EUROPE 2024; 3:187. [PMID: 39118808 PMCID: PMC11308986 DOI: 10.12688/openreseurope.16550.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Background Atomic force microscopy (AFM) is one of the main techniques used to characterize the mechanical properties of soft biological samples and biomaterials at the nanoscale. Despite efforts made by the AFM community to promote open-source data analysis tools, standardization continues to be a significant concern in a field that requires common analysis procedures. AFM-based mechanical measurements involve applying a controlled force to the sample and measure the resulting deformation in the so-called force-distance curves. These may include simple approach and retract or oscillatory cycles at various frequencies (microrheology). To extract quantitative parameters, such as the elastic modulus, from these measurements, AFM measurements are processed using data analysis software. Although open tools exist and allow obtaining the mechanical properties of the sample, most of them only include standard elastic models and do not allow the processing of microrheology data. In this work, we have developed an open-source software package (called PyFMLab, as of python force microscopy laboratory) capable of determining the viscoelastic properties of samples from both conventional force-distance curves and microrheology measurements. Methods PyFMLab has been written in Python, which provides an accessible syntax and sufficient computational efficiency. The software features were divided into separate, self-contained libraries to enhance code organization and modularity and to improve readability, maintainability, testability, and reusability. To validate PyFMLab, two AFM datasets, one composed of simple force curves and another including oscillatory measurements, were collected on HeLa cells. Results The viscoelastic parameters obtained on the two datasets analysed using PyFMLab were validated against data processing proprietary software and against validated MATLAB routines developed before obtaining equivalent results. Conclusions Its open-source nature and versatility makes PyFMLab an open-source solution that paves the way for standardized viscoelastic characterization of biological samples from both force-distance curves and microrheology measurements.
Collapse
Affiliation(s)
- Javier López-Alonso
- Universite de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CILL—Center of Infection and Immunity of Lille, Lille, F-59000, France
| | - Mar Eroles
- Aix-Marseille Univ., CNRS, INSERM, LAI, Turing Centre for Living Systems, Marseille, 13009, France
| | - Sébastien Janel
- Universite de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CILL—Center of Infection and Immunity of Lille, Lille, F-59000, France
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands
- Optics 11 B.V, Amsterdam, 1101BM, The Netherlands
| | | | - Vincent Dupres
- Universite de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CILL—Center of Infection and Immunity of Lille, Lille, F-59000, France
| | - Frank Lafont
- Universite de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CILL—Center of Infection and Immunity of Lille, Lille, F-59000, France
| | - Felix Rico
- Aix-Marseille Univ., CNRS, INSERM, LAI, Turing Centre for Living Systems, Marseille, 13009, France
| |
Collapse
|
9
|
Urbanska M, Guck J. Single-Cell Mechanics: Structural Determinants and Functional Relevance. Annu Rev Biophys 2024; 53:367-395. [PMID: 38382116 DOI: 10.1146/annurev-biophys-030822-030629] [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: 02/23/2024]
Abstract
The mechanical phenotype of a cell determines its ability to deform under force and is therefore relevant to cellular functions that require changes in cell shape, such as migration or circulation through the microvasculature. On the practical level, the mechanical phenotype can be used as a global readout of the cell's functional state, a marker for disease diagnostics, or an input for tissue modeling. We focus our review on the current knowledge of structural components that contribute to the determination of the cellular mechanical properties and highlight the physiological processes in which the mechanical phenotype of the cells is of critical relevance. The ongoing efforts to understand how to efficiently measure and control the mechanical properties of cells will define the progress in the field and drive mechanical phenotyping toward clinical applications.
Collapse
Affiliation(s)
- Marta Urbanska
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
10
|
Loginova N, Aniskin D, Timashev P, Ulasov I, Kharwar RK. GBM Immunotherapy: Macrophage Impacts. Immunol Invest 2024; 53:730-751. [PMID: 38634572 DOI: 10.1080/08820139.2024.2337022] [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: 04/19/2024]
Abstract
BACKGROUND Glioblastoma (GBM) is an extremely aggressive form of brain tumor with low survival rates. Current treatments such as chemotherapy, radiation, and surgery are problematic due to tumor growth, invasion, and tumor microenvironment. GBM cells are resistant to these standard treatments, and the heterogeneity of the tumor makes it difficult to find a universal approach. Progression of GBM and acquisition of resistance to therapy are due to the complex interplay between tumor cells and the TME. A significant portion of the TME consists of an inflammatory infiltrate, with microglia and macrophages being the predominant cells. METHODS Analysis of the literature data over a course of 5 years suggest that the tumor-associated macrophages (TAMs) are capable of releasing cytokines and growth factors that promote tumor proliferation, survival, and metastasis while inhibiting immune cell function at the same time. RESULTS Thus, immunosuppressive state, provided with this intensively studied kind of TME cells, is supposed to promote GBM development through TAMs modulation of tumor treatment-resistance and aggressiveness. Therefore, TAMs are an attractive therapeutic target in the treatment of glioblastoma. CONCLUSION This review provides a comprehensive overview of the latest research on the nature of TAMs and the development of therapeutic strategies targeting TAMs, focusing on the variety of macrophage properties, being modulated, as well as molecular targets.
Collapse
Affiliation(s)
- Nina Loginova
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Denis Aniskin
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Peter Timashev
- World-Class Research Centre "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Rajesh Kumar Kharwar
- Endocrine Research Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| |
Collapse
|
11
|
Perea Paizal J, Au SH, Bakal C. Nuclear rupture induced by capillary constriction forces promotes differential effects on metastatic and normal breast cells. Sci Rep 2024; 14:14793. [PMID: 38926422 PMCID: PMC11208511 DOI: 10.1038/s41598-024-64733-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
During metastatic dissemination, circulating tumour cells (CTCs) enter capillary beds, where they experience mechanical constriction forces. The transient and persistent effects of these forces on CTCs behaviour remain poorly understood. Here, we developed a high-throughput microfluidic platform mimicking human capillaries to investigate the impact of mechanical constriction forces on malignant and normal breast cell lines. We observed that capillary constrictions induced nuclear envelope rupture in both cancer and normal cells, leading to transient changes in nuclear and cytoplasmic area. Constriction forces transiently activated cGAS/STING and pathways involved in inflammation (NF-κB, STAT and IRF3), especially in the non-malignant cell line. Furthermore, the non-malignant cell line experienced transcriptional changes, particularly downregulation of epithelial markers, while the metastatic cell lines showed minimal alterations. These findings suggest that mechanical constriction forces within capillaries may promote differential effects in malignant and normal cell lines.
Collapse
Affiliation(s)
- Julia Perea Paizal
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
- Division of Cancer Biology, Chester Beatty Laboratories, Institute of Cancer Research, 237 Fulham Road, London, SW6 6JB, UK.
- Cancer Research UK Convergence Science Centre, Roderic Hill Building, Imperial College London, London, SW7 2BB, UK.
| | - Sam H Au
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
- Cancer Research UK Convergence Science Centre, Roderic Hill Building, Imperial College London, London, SW7 2BB, UK
| | - Chris Bakal
- Division of Cancer Biology, Chester Beatty Laboratories, Institute of Cancer Research, 237 Fulham Road, London, SW6 6JB, UK
- Cancer Research UK Convergence Science Centre, Roderic Hill Building, Imperial College London, London, SW7 2BB, UK
| |
Collapse
|
12
|
Yang Y, Li M. Side-view optical microscopy-assisted atomic force microscopy for thickness-dependent nanobiomechanics. NANOSCALE ADVANCES 2024; 6:3306-3319. [PMID: 38933861 PMCID: PMC11197429 DOI: 10.1039/d4na00153b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/06/2024] [Indexed: 06/28/2024]
Abstract
The mechanical properties of biomaterials play an important role in regulating life processes, and thus accurately delineating the mechanical properties of biomaterials is critical to understand their functionality. Particularly, atomic force microscopy (AFM) has become a powerful and standard tool for characterizing and analyzing the nanomechanical properties of biomaterials, and providing a capability to visualize the thickness of the specimen during AFM-based force spectroscopy experiments benefits the biomedical applications of AFM. Here, we present a study of side-view optical microscopy-assisted AFM based on the integration of AFM and a detachable side-view optical microscopy module, which is able to image in real time the AFM indentation process from the side-view perspective and consequently facilitates the utilization of AFM-based indentation assay to precisely detect the mechanical properties of a specimen by taking its thickness into account. The effectiveness of side-view optical microscopy-assisted AFM was confirmed on four different types of biomaterial systems, including microfabricated structures, hydrogels, living cells, and cell spheroids, and the experimental results significantly show that the mechanical properties of samples at the micro/nanoscale are closely related to their thickness, vividly illustrating side-view optical microscopy-assisted AFM as a promising approach for accurate nanomechanics of biomaterial systems. The study provides additional possibilities for measuring the thickness-dependent nanomechanical properties of biomaterials by AFM, which will enable AFM-based force spectroscopy technology to address more biological issues with enhanced precision and will benefit the field of mechanobiology.
Collapse
Affiliation(s)
- Yanqi Yang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences Shenyang 110016 China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences Shenyang 110169 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences Shenyang 110016 China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences Shenyang 110169 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
13
|
Lima I, Silva A, Sousa F, Ferreira W, Freire R, de Oliveira C, de Sousa J. Measuring the viscoelastic relaxation function of cells with a time-dependent interpretation of the Hertz-Sneddon indentation model. Heliyon 2024; 10:e30623. [PMID: 38770291 PMCID: PMC11103437 DOI: 10.1016/j.heliyon.2024.e30623] [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: 02/06/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024] Open
Abstract
The Hertz-Sneddon elastic indentation model is widely adopted in the biomechanical investigation of living cells and other soft materials using atomic force microscopy despite the explicit viscoelastic nature of these materials. In this work, we demonstrate that an exact analytical viscoelastic force model for power-law materials, can be interpreted as a time-dependent Hertz-Sneddon-like model. Characterizing fibroblasts (L929) and osteoblasts (OFCOLII) demonstrates the model's accuracy. Our results show that the difference between Young's modulus E Y obtained by fitting force curves with the Hertz-Sneddon model and the effective Young's modulus derived from the viscoelastic force model is less than 3%, even when cells are probed at large forces where nonlinear deformation effects become significant. We also propose a measurement protocol that involves probing samples at different indentation speeds and forces, enabling the construction of the average viscoelastic relaxation function of samples by conveniently fitting the force curves with the Hertz-Sneddon model.
Collapse
Affiliation(s)
- I.V.M. Lima
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - A.V.S. Silva
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
- Instituto Federal do Rio Grande do Norte, Pau dos Ferros, 59900-000, Rio Grande do Norte, Brazil
| | - F.D. Sousa
- Núcleo de Biologia Experimental, Universidade de Fortaleza, Fortaleza, 60811-905, Ceará, Brazil
| | - W.P. Ferreira
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - R.S. Freire
- Central Analítica, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - C.L.N. de Oliveira
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - J.S. de Sousa
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| |
Collapse
|
14
|
Hartmann B, Fleischhauer L, Nicolau M, Jensen THL, Taran FA, Clausen-Schaumann H, Reuten R. Profiling native pulmonary basement membrane stiffness using atomic force microscopy. Nat Protoc 2024; 19:1498-1528. [PMID: 38429517 DOI: 10.1038/s41596-024-00955-7] [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: 02/20/2023] [Accepted: 11/27/2023] [Indexed: 03/03/2024]
Abstract
Mammalian cells sense and react to the mechanics of their immediate microenvironment. Therefore, the characterization of the biomechanical properties of tissues with high spatial resolution provides valuable insights into a broad variety of developmental, homeostatic and pathological processes within living organisms. The biomechanical properties of the basement membrane (BM), an extracellular matrix (ECM) substructure measuring only ∼100-400 nm across, are, among other things, pivotal to tumor progression and metastasis formation. Although the precise assignment of the Young's modulus E of such a thin ECM substructure especially in between two cell layers is still challenging, biomechanical data of the BM can provide information of eminent diagnostic potential. Here we present a detailed protocol to quantify the elastic modulus of the BM in murine and human lung tissue, which is one of the major organs prone to metastasis. This protocol describes a streamlined workflow to determine the Young's modulus E of the BM between the endothelial and epithelial cell layers shaping the alveolar wall in lung tissues using atomic force microscopy (AFM). Our step-by-step protocol provides instructions for murine and human lung tissue extraction, inflation of these tissues with cryogenic cutting medium, freezing and cryosectioning of the tissue samples, and AFM force-map recording. In addition, it guides the reader through a semi-automatic data analysis procedure to identify the pulmonary BM and extract its Young's modulus E using an in-house tailored user-friendly AFM data analysis software, the Center for Applied Tissue Engineering and Regenerative Medicine processing toolbox, which enables automatic loading of the recorded force maps, conversion of the force versus piezo-extension curves to force versus indentation curves, calculation of Young's moduli and generation of Young's modulus maps, where the pulmonary BM can be identified using a semi-automatic spatial filtering tool. The entire protocol takes 1-2 d.
Collapse
Affiliation(s)
- Bastian Hartmann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Lutz Fleischhauer
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Monica Nicolau
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas Hartvig Lindkær Jensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - Florin-Andrei Taran
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Hauke Clausen-Schaumann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany.
- Center for Nanoscience, Munich, Germany.
| | - Raphael Reuten
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany.
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
15
|
Jackson CE, Green NH, English WR, Claeyssens F. The use of microphysiological systems to model metastatic cancer. Biofabrication 2024; 16:032002. [PMID: 38579739 DOI: 10.1088/1758-5090/ad3b70] [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: 11/13/2023] [Accepted: 04/05/2024] [Indexed: 04/07/2024]
Abstract
Cancer is one of the leading causes of death in the 21st century, with metastasis of cancer attributing to 90% of cancer-related deaths. Therefore, to improve patient outcomes there is a need for better preclinical models to increase the success of translating oncological therapies into the clinic. Current traditional staticin vitromodels lack a perfusable network which is critical to overcome the diffusional mass transfer limit to provide a mechanism for the exchange of essential nutrients and waste removal, and increase their physiological relevance. Furthermore, these models typically lack cellular heterogeneity and key components of the immune system and tumour microenvironment. This review explores rapidly developing strategies utilising perfusable microphysiological systems (MPS) for investigating cancer cell metastasis. In this review we initially outline the mechanisms of cancer metastasis, highlighting key steps and identifying the current gaps in our understanding of the metastatic cascade, exploring MPS focused on investigating the individual steps of the metastatic cascade before detailing the latest MPS which can investigate multiple components of the cascade. This review then focuses on the factors which can affect the performance of an MPS designed for cancer applications with a final discussion summarising the challenges and future directions for the use of MPS for cancer models.
Collapse
Affiliation(s)
- Caitlin E Jackson
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| | - Nicola H Green
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| | - William R English
- Norwich Medical School, University of East Anglia, Norwich NR3 7TJ, United Kingdom
| | - Frederik Claeyssens
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| |
Collapse
|
16
|
Carvalho EM, Ding EA, Saha A, Weldy A, Zushin PJH, Stahl A, Aghi MK, Kumar S. Viscoelastic high-molecular-weight hyaluronic acid hydrogels support rapid glioblastoma cell invasion with leader-follower dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588167. [PMID: 38617333 PMCID: PMC11014578 DOI: 10.1101/2024.04.04.588167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Hyaluronic acid (HA), the primary component of brain extracellular matrix, is increasingly used to model neuropathological processes, including glioblastoma (GBM) tumor invasion. While elastic hydrogels based on crosslinked low-molecular-weight (LMW) HA are widely exploited for this purpose and have proven valuable for discovery and screening, brain tissue is both viscoelastic and rich in high-MW (HMW) HA, and it remains unclear how these differences influence invasion. To address this question, hydrogels comprised of either HMW (1.5 MDa) or LMW (60 kDa) HA are introduced, characterized, and applied in GBM invasion studies. Unlike LMW HA hydrogels, HMW HA hydrogels relax stresses quickly, to a similar extent as brain tissue, and to a greater extent than many conventional HA-based scaffolds. GBM cells implanted within HMW HA hydrogels invade much more rapidly than in their LMW HA counterparts and exhibit distinct leader-follower dynamics. Leader cells adopt dendritic morphologies, similar to invasive GBM cells observed in vivo. Transcriptomic, pharmacologic, and imaging studies suggest that leader cells exploit hyaluronidase, an enzyme strongly enriched in human GBMs, to prime a path for followers. This study offers new insight into how HA viscoelastic properties drive invasion and argues for the use of highly stress-relaxing materials to model GBM.
Collapse
Affiliation(s)
- Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Atul Saha
- Department of Neurosurgery, University of California, San Francisco, CA 94158, USA
| | - Anna Weldy
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley 94720, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California, San Francisco, CA 94158, USA
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
17
|
Lightsey S, Sharma B. Natural Killer Cell Mechanosensing in Solid Tumors. Bioengineering (Basel) 2024; 11:328. [PMID: 38671750 PMCID: PMC11048000 DOI: 10.3390/bioengineering11040328] [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: 03/01/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Natural killer (NK) cells, which are an exciting alternative cell source for cancer immunotherapies, must sense and respond to their physical environment to traffic to and eliminate cancer cells. Herein, we review the mechanisms by which NK cells receive mechanical signals and explore recent key findings regarding the impact of the physical characteristics of solid tumors on NK cell functions. Data suggest that different mechanical stresses present in solid tumors facilitate NK cell functions, especially infiltration and degranulation. Moreover, we review recent engineering advances that can be used to systemically study the role of mechanical forces on NK cell activity. Understanding the mechanisms by which NK cells interpret their environment presents potential targets to enhance NK cell immunotherapies for the treatment of solid tumors.
Collapse
Affiliation(s)
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 23610, USA;
| |
Collapse
|
18
|
Mittelheisser V, Gensbittel V, Bonati L, Li W, Tang L, Goetz JG. Evidence and therapeutic implications of biomechanically regulated immunosurveillance in cancer and other diseases. NATURE NANOTECHNOLOGY 2024; 19:281-297. [PMID: 38286876 DOI: 10.1038/s41565-023-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/26/2023] [Indexed: 01/31/2024]
Abstract
Disease progression is usually accompanied by changes in the biochemical composition of cells and tissues and their biophysical properties. For instance, hallmarks of cancer include the stiffening of tissues caused by extracellular matrix remodelling and the softening of individual cancer cells. In this context, accumulating evidence has shown that immune cells sense and respond to mechanical signals from the environment. However, the mechanisms regulating these mechanical aspects of immune surveillance remain partially understood. The growing appreciation for the 'mechano-immunology' field has urged researchers to investigate how immune cells sense and respond to mechanical cues in various disease settings, paving the way for the development of novel engineering strategies that aim at mechanically modulating and potentiating immune cells for enhanced immunotherapies. Recent pioneer developments in this direction have laid the foundations for leveraging 'mechanical immunoengineering' strategies to treat various diseases. This Review first outlines the mechanical changes occurring during pathological progression in several diseases, including cancer, fibrosis and infection. We next highlight the mechanosensitive nature of immune cells and how mechanical forces govern the immune responses in different diseases. Finally, we discuss how targeting the biomechanical features of the disease milieu and immune cells is a promising strategy for manipulating therapeutic outcomes.
Collapse
Affiliation(s)
- Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Valentin Gensbittel
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Lucia Bonati
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Weilin Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland.
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|
19
|
Yanamandra AK, Zhang J, Montalvo G, Zhou X, Biedenweg D, Zhao R, Sharma S, Hoth M, Lautenschläger F, Otto O, Del Campo A, Qu B. PIEZO1-mediated mechanosensing governs NK-cell killing efficiency and infiltration in three-dimensional matrices. Eur J Immunol 2024; 54:e2350693. [PMID: 38279603 DOI: 10.1002/eji.202350693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/28/2024]
Abstract
Natural killer (NK) cells play a vital role in eliminating tumorigenic cells. Efficient locating and killing of target cells in complex three-dimensional (3D) environments are critical for their functions under physiological conditions. However, the role of mechanosensing in regulating NK-cell killing efficiency in physiologically relevant scenarios is poorly understood. Here, we report that the responsiveness of NK cells is regulated by tumor cell stiffness. NK-cell killing efficiency in 3D is impaired against softened tumor cells, whereas it is enhanced against stiffened tumor cells. Notably, the durations required for NK-cell killing and detachment are significantly shortened for stiffened tumor cells. Furthermore, we have identified PIEZO1 as the predominantly expressed mechanosensitive ion channel among the examined candidates in NK cells. Perturbation of PIEZO1 abolishes stiffness-dependent NK-cell responsiveness, significantly impairs the killing efficiency of NK cells in 3D, and substantially reduces NK-cell infiltration into 3D collagen matrices. Conversely, PIEZO1 activation enhances NK killing efficiency as well as infiltration. In conclusion, our findings demonstrate that PIEZO1-mediated mechanosensing is crucial for NK killing functions, highlighting the role of mechanosensing in NK-cell killing efficiency under 3D physiological conditions and the influence of environmental physical cues on NK-cell functions.
Collapse
Affiliation(s)
- Archana K Yanamandra
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Jingnan Zhang
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Galia Montalvo
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - Xiangda Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Doreen Biedenweg
- Institute of Physics, University of Greifswald, Greifswald, Germany
| | - Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Shulagna Sharma
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Franziska Lautenschläger
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - Oliver Otto
- Institute of Physics, University of Greifswald, Greifswald, Germany
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
- Chemistry Department, Saarland University, Saarbrücken, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
20
|
Cambria E, Coughlin MF, Floryan MA, Offeddu GS, Shelton SE, Kamm RD. Linking cell mechanical memory and cancer metastasis. Nat Rev Cancer 2024; 24:216-228. [PMID: 38238471 PMCID: PMC11146605 DOI: 10.1038/s41568-023-00656-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 03/01/2024]
Abstract
Metastasis causes most cancer-related deaths; however, the efficacy of anti-metastatic drugs is limited by incomplete understanding of the biological mechanisms that drive metastasis. Focusing on the mechanics of metastasis, we propose that the ability of tumour cells to survive the metastatic process is enhanced by mechanical stresses in the primary tumour microenvironment that select for well-adapted cells. In this Perspective, we suggest that biophysical adaptations favourable for metastasis are retained via mechanical memory, such that the extent of memory is influenced by both the magnitude and duration of the mechanical stress. Among the mechanical cues present in the primary tumour microenvironment, we focus on high matrix stiffness to illustrate how it alters tumour cell proliferation, survival, secretion of molecular factors, force generation, deformability, migration and invasion. We particularly centre our discussion on potential mechanisms of mechanical memory formation and retention via mechanotransduction and persistent epigenetic changes. Indeed, we propose that the biophysical adaptations that are induced by this process are retained throughout the metastatic process to improve tumour cell extravasation, survival and colonization in the distant organ. Deciphering mechanical memory mechanisms will be key to discovering a new class of anti-metastatic drugs.
Collapse
Affiliation(s)
- Elena Cambria
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Mark F Coughlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie A Floryan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni S Offeddu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
21
|
Shu J, Deng H, Zhang Y, Wu F, He J. Cancer cell response to extrinsic and intrinsic mechanical cue: opportunities for tumor apoptosis strategies. Regen Biomater 2024; 11:rbae016. [PMID: 38476678 PMCID: PMC10932484 DOI: 10.1093/rb/rbae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024] Open
Abstract
Increasing studies have revealed the importance of mechanical cues in tumor progression, invasiveness and drug resistance. During malignant transformation, changes manifest in either the mechanical properties of the tissue or the cellular ability to sense and respond to mechanical signals. The major focus of the review is the subtle correlation between mechanical cues and apoptosis in tumor cells from a mechanobiology perspective. To begin, we focus on the intracellular force, examining the mechanical properties of the cell interior, and outlining the role that the cytoskeleton and intracellular organelle-mediated intracellular forces play in tumor cell apoptosis. This article also elucidates the mechanisms by which extracellular forces guide tumor cell mechanosensing, ultimately triggering the activation of the mechanotransduction pathway and impacting tumor cell apoptosis. Finally, a comprehensive examination of the present status of the design and development of anti-cancer materials targeting mechanotransduction is presented, emphasizing the underlying design principles. Furthermore, the article underscores the need to address several unresolved inquiries to enhance our comprehension of cancer therapeutics that target mechanotransduction.
Collapse
Affiliation(s)
- Jun Shu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Huan Deng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Yu Zhang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, PR China
| | - Fang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| |
Collapse
|
22
|
Feng J, Sun Q, Chen P, Ren K, Zhang Y, Shi Y, Gao S, Song Z, Wang J, Liao F, Han D. Characterization of Cancer Cell Mechanics by Measuring Active Deformation Behavior. SMALL METHODS 2024; 8:e2300520. [PMID: 37775303 DOI: 10.1002/smtd.202300520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/17/2023] [Indexed: 10/01/2023]
Abstract
Active deformation behavior reflects cell structural dynamics adapting to varying environmental constraints during malignancy progression. In most cases, cell mechanics is characterized by modeling using static equilibrium systems, which fails to comprehend cell deformation behavior leading to inaccuracies in distinguishing cancer cells from normal cells. Here, a method is introduced to measure the active deformation behavior of cancer cells using atomic force microscopy (AFM) and the newly developed deformation behavior cytometry (DBC). During the measurement, cells are deformed and allows a long timescale relaxation (≈5 s). Two parameters are derived to represent deformation behavior: apparent Poisson's ratio for adherent cells, which is measured with AFM and refers to the ratio of the lateral strain to the longitudinal strain of the cell, and shape recovery for suspended cells, which is measured with DBC. Active deformation behavior defines cancer cell mechanics better than traditional mechanical parameters (e.g., stiffness, diffusion, and viscosity). Additionally, aquaporins are essential for promoting the deformation behavior, while the actin cytoskeleton acts as a downstream effector. Therefore, the potential application of the cancer cell active deformation behavior as a biomechanical marker or therapeutic target in cancer treatment should be evaluated.
Collapse
Affiliation(s)
- Jiantao Feng
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Quanmei Sun
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Peipei Chen
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Keli Ren
- The Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuanyuan Zhang
- Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Yahong Shi
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Songkun Gao
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100006, China
| | - Zhiwei Song
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Jigang Wang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fulong Liao
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Dong Han
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| |
Collapse
|
23
|
So WY, Wong CS, Azubuike UF, Paul CD, Sangsari PR, Gordon PB, Gong H, Maity TK, Lim P, Yang Z, Haryanto CA, Batchelor E, Jenkins LM, Morgan NY, Tanner K. YAP localization mediates mechanical adaptation of human cancer cells during extravasation in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567015. [PMID: 38076880 PMCID: PMC10705547 DOI: 10.1101/2023.11.14.567015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Biophysical profiling of primary tumors has revealed that individual tumor cells fall along a highly heterogeneous continuum of mechanical phenotypes. One idea is that a subset of tumor cells is "softer" to facilitate detachment and escape from the primary site, a step required to initiate metastasis. However, it has also been postulated that cells must be able to deform and generate sufficient force to exit into distant sites. Here, we aimed to dissect the mechanical changes that occur during extravasation and organ colonization. Using multiplexed methods of intravital microscopy and optical tweezer based active microrheology, we obtained longitudinal images and mechanical profiles of cells during organ colonization in vivo. We determined that cells were softer, more liquid like upon exit of the vasculature but stiffened and became more solid like once in the new organ microenvironment. We also determined that a YAP mediated mechanogenotype influenced the global dissemination in our in vivo and in vitro models and that reducing mechanical heterogeneity could reduce extravasation. Moreover, our high throughput analysis of mechanical phenotypes of patient samples revealed that this mechanics was in part regulated by the external hydrodynamic forces that the cancer cells experienced within capillary mimetics. Our findings indicate that disseminated cancer cells can keep mutating with a continuum landscape of mechano-phenotypes, governed by the YAP-mediated mechanosensing of hydrodynamic flow.
Collapse
Affiliation(s)
- Woong Young So
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Claudia S. Wong
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | | | - Colin D. Paul
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Paniz Rezvan Sangsari
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health
| | | | - Hyeyeon Gong
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Tapan K. Maity
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Perry Lim
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Zhilin Yang
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | | | | | - Lisa M. Jenkins
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Nicole Y. Morgan
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health
| | - Kandice Tanner
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| |
Collapse
|
24
|
Ren K, Feng J, Bi H, Sun Q, Li X, Han D. AFM-Based Poroelastic@Membrane Analysis of Cells and its Opportunities for Translational Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303610. [PMID: 37403276 DOI: 10.1002/smll.202303610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/09/2023] [Indexed: 07/06/2023]
Abstract
Cell mechanics is an emerging field of research for translational medicine. Here, the cell is modeled as poroelastic cytoplasm wrapped by tensile membrane (poroelastic@membrane model) and is characterized by the atomic force microscopy (AFM). The parameters of cytoskeleton network modulus EC , cytoplasmic apparent viscosity ηC , and cytoplasmic diffusion coefficient DC are used to describe the mechanical behavior of cytoplasm, and membrane tension γ is used to evaluate the cell membrane. Poroelastic@membrane analysis of breast cells and urothelial cells show that non-cancer cells and cancer cells have different distribution regions and distribution trends in the four-dimensional space composed of EC , ηC . From non-cancer to cancer cells, there is often a trend of γ, EC , ηC decreases and DC increases. Patients with urothelial carcinoma at different malignant stages can be distinguished at high sensitivity and specificity by analyzing the urothelial cells from tissue or urine. However, sampling directly from tumor tissues is an invasive method, may lead to undesirable consequences. Thus, AFM-based poroelastic@membrane analysis of urothelial cells from urine may provide a non-invasive and no-bio-label method to detecting urothelial carcinoma.
Collapse
Affiliation(s)
- Keli Ren
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 ZhongGuanCun BeiYiTiao, Haidian, Beijing, 100191, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou Distric, Beijing, 100190, China
| | - Jiantao Feng
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No.16, Nanxiao street, Dongzhimen, Dongcheng, Beijing, 100700, China
| | - Hai Bi
- Department of Urology, Peking University Third Hospital, 49 North Garden Rd., Haidian, Beijing, 100191, China
| | - Quanmei Sun
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 ZhongGuanCun BeiYiTiao, Haidian, Beijing, 100191, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou Distric, Beijing, 100190, China
| | - Xiang Li
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 ZhongGuanCun BeiYiTiao, Haidian, Beijing, 100191, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou Distric, Beijing, 100190, China
| | - Dong Han
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 ZhongGuanCun BeiYiTiao, Haidian, Beijing, 100191, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou Distric, Beijing, 100190, China
| |
Collapse
|
25
|
Tagay Y, Kheirabadi S, Ataie Z, Singh RK, Prince O, Nguyen A, Zhovmer AS, Ma X, Sheikhi A, Tsygankov D, Tabdanov ED. Dynein-Powered Cell Locomotion Guides Metastasis of Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302229. [PMID: 37726225 PMCID: PMC10625109 DOI: 10.1002/advs.202302229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/20/2023] [Indexed: 09/21/2023]
Abstract
The principal cause of death in cancer patients is metastasis, which remains an unresolved problem. Conventionally, metastatic dissemination is linked to actomyosin-driven cell locomotion. However, the locomotion of cancer cells often does not strictly line up with the measured actomyosin forces. Here, a complementary mechanism of metastatic locomotion powered by dynein-generated forces is identified. These forces arise within a non-stretchable microtubule network and drive persistent contact guidance of migrating cancer cells along the biomimetic collagen fibers. It is also shown that the dynein-powered locomotion becomes indispensable during invasive 3D migration within a tissue-like luminal network formed by spatially confining granular hydrogel scaffolds (GHS) made up of microscale hydrogel particles (microgels). These results indicate that the complementary motricity mediated by dynein is always necessary and, in certain instances, sufficient for disseminating metastatic breast cancer cells. These findings advance the fundamental understanding of cell locomotion mechanisms and expand the spectrum of clinical targets against metastasis.
Collapse
Affiliation(s)
- Yerbol Tagay
- Department of PharmacologyPenn State College of MedicineThe Pennsylvania State UniversityHersheyPA17033USA
| | - Sina Kheirabadi
- Department of Chemical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Zaman Ataie
- Department of Chemical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Rakesh K. Singh
- Department of Obstetrics & GynecologyGynecology OncologyUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Olivia Prince
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Ashley Nguyen
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Alexander S. Zhovmer
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Xuefei Ma
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Amir Sheikhi
- Department of Chemical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Erdem D. Tabdanov
- Department of PharmacologyPenn State College of MedicineThe Pennsylvania State UniversityHersheyPA17033USA
- Penn State Cancer InstitutePenn State College of MedicineThe Pennsylvania State UniversityHersheyPA17033USA
| |
Collapse
|
26
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
27
|
Chen MB, Javanmardi Y, Shahreza S, Serwinski B, Aref A, Djordjevic B, Moeendarbary E. Mechanobiology in oncology: basic concepts and clinical prospects. Front Cell Dev Biol 2023; 11:1239749. [PMID: 38020912 PMCID: PMC10644154 DOI: 10.3389/fcell.2023.1239749] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The interplay between genetic transformations, biochemical communications, and physical interactions is crucial in cancer progression. Metastasis, a leading cause of cancer-related deaths, involves a series of steps, including invasion, intravasation, circulation survival, and extravasation. Mechanical alterations, such as changes in stiffness and morphology, play a significant role in all stages of cancer initiation and dissemination. Accordingly, a better understanding of cancer mechanobiology can help in the development of novel therapeutic strategies. Targeting the physical properties of tumours and their microenvironment presents opportunities for intervention. Advancements in imaging techniques and lab-on-a-chip systems enable personalized investigations of tumor biomechanics and drug screening. Investigation of the interplay between genetic, biochemical, and mechanical factors, which is of crucial importance in cancer progression, offers insights for personalized medicine and innovative treatment strategies.
Collapse
Affiliation(s)
- Michelle B. Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Somayeh Shahreza
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, United Kingdom
- 199 Biotechnologies Ltd., London, United Kingdom
- Northeastern University London, London, United Kingdom
| | - Amir Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, United Kingdom
- 199 Biotechnologies Ltd., London, United Kingdom
| | - Emad Moeendarbary
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Mechanical Engineering, University College London, London, United Kingdom
| |
Collapse
|
28
|
Bao L, Kong H, Ja Y, Wang C, Qin L, Sun H, Dai S. The relationship between cancer and biomechanics. Front Oncol 2023; 13:1273154. [PMID: 37901315 PMCID: PMC10602664 DOI: 10.3389/fonc.2023.1273154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
The onset, development, diagnosis, and treatment of cancer involve intricate interactions among various factors, spanning the realms of mechanics, physics, chemistry, and biology. Within our bodies, cells are subject to a variety of forces such as gravity, magnetism, tension, compression, shear stress, and biological static force/hydrostatic pressure. These forces are perceived by mechanoreceptors as mechanical signals, which are then transmitted to cells through a process known as mechanical transduction. During tumor development, invasion and metastasis, there are significant biomechanical influences on various aspects such as tumor angiogenesis, interactions between tumor cells and the extracellular matrix (ECM), interactions between tumor cells and other cells, and interactions between tumor cells and the circulatory system and vasculature. The tumor microenvironment comprises a complex interplay of cells, ECM and vasculature, with the ECM, comprising collagen, fibronectins, integrins, laminins and matrix metalloproteinases, acting as a critical mediator of mechanical properties and a key component within the mechanical signaling pathway. The vasculature exerts appropriate shear forces on tumor cells, enabling their escape from immune surveillance, facilitating their dissemination in the bloodstream, dictating the trajectory of circulating tumor cells (CTCs) and playing a pivotal role in regulating adhesion to the vessel wall. Tumor biomechanics plays a critical role in tumor progression and metastasis, as alterations in biomechanical properties throughout the malignant transformation process trigger a cascade of changes in cellular behavior and the tumor microenvironment, ultimately culminating in the malignant biological behavior of the tumor.
Collapse
Affiliation(s)
- Liqi Bao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongru Kong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yang Ja
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengchao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongwei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengjie Dai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
29
|
Sauer F, Grosser S, Shahryari M, Hayn A, Guo J, Braun J, Briest S, Wolf B, Aktas B, Horn L, Sack I, Käs JA. Changes in Tissue Fluidity Predict Tumor Aggressiveness In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303523. [PMID: 37553780 PMCID: PMC10502644 DOI: 10.1002/advs.202303523] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Indexed: 08/10/2023]
Abstract
Cancer progression is caused by genetic changes and associated with various alterations in cell properties, which also affect a tumor's mechanical state. While an increased stiffness has been well known for long for solid tumors, it has limited prognostic power. It is hypothesized that cancer progression is accompanied by tissue fluidization, where portions of the tissue can change position across different length scales. Supported by tabletop magnetic resonance elastography (MRE) on stroma mimicking collagen gels and microscopic analysis of live cells inside patient derived tumor explants, an overview is provided of how cancer associated mechanisms, including cellular unjamming, proliferation, microenvironment composition, and remodeling can alter a tissue's fluidity and stiffness. In vivo, state-of-the-art multifrequency MRE can distinguish tumors from their surrounding host tissue by their rheological fingerprints. Most importantly, a meta-analysis on the currently available clinical studies is conducted and universal trends are identified. The results and conclusions are condensed into a gedankenexperiment about how a tumor can grow and eventually metastasize into its environment from a physics perspective to deduce corresponding mechanical properties. Based on stiffness, fluidity, spatial heterogeneity, and texture of the tumor front a roadmap for a prognosis of a tumor's aggressiveness and metastatic potential is presented.
Collapse
Affiliation(s)
- Frank Sauer
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
| | - Steffen Grosser
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
- Institute for Bioengineering of CataloniaThe Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
| | - Mehrgan Shahryari
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Alexander Hayn
- Department of HepatologyLeipzig University Hospital04103LeipzigGermany
| | - Jing Guo
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Jürgen Braun
- Institute of Medical InformaticsCharité‐Universitätsmedizin10117BerlinGermany
| | - Susanne Briest
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Benjamin Wolf
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Bahriye Aktas
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Lars‐Christian Horn
- Division of Breast, Urogenital and Perinatal PathologyLeipzig University Hospital04103LeipzigGermany
| | - Ingolf Sack
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Josef A. Käs
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
| |
Collapse
|
30
|
Cardona S, Mostafazadeh N, Luan Q, Zhou J, Peng Z, Papautsky I. Numerical Modeling of Physical Cell Trapping in Microfluidic Chips. MICROMACHINES 2023; 14:1665. [PMID: 37763828 PMCID: PMC10538085 DOI: 10.3390/mi14091665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
Microfluidic methods have proven to be effective in separation and isolation of cells for a wide range of biomedical applications. Among these methods, physical trapping is a label-free isolation approach that relies on cell size as the selective phenotype to retain target cells on-chip for follow-up analysis and imaging. In silico models have been used to optimize the design of such hydrodynamic traps and to investigate cancer cell transmigration through narrow constrictions. While most studies focus on computational fluid dynamics (CFD) analysis of flow over cells and/or pillar traps, a quantitative analysis of mechanical interaction between cells and trapping units is missing. The existing literature centers on longitudinally extended geometries (e.g., micro-vessels) to understand the biological phenomenon rather than designing an effective cell trap. In this work, we aim to make an experimentally informed prediction of the critical pressure for a cell to pass through a trapping unit as a function of cell morphology and trapping unit geometry. Our findings show that a hyperelastic material model accurately captures the stress-related softening behavior observed in cancer cells passing through micro-constrictions. These findings are used to develop a model capable of predicting and extrapolating critical pressure values. The validity of the model is assessed with experimental data. Regression analysis is used to derive a mathematical framework for critical pressure. Coupled with CFD analysis, one can use this formulation to design efficient microfluidic devices for cell trapping and potentially perform downstream analysis of trapped cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA
| |
Collapse
|
31
|
Lee YL, Mathur J, Walter C, Zmuda H, Pathak A. Matrix obstructions cause multiscale disruption in collective epithelial migration by suppressing leader cell function. Mol Biol Cell 2023; 34:ar94. [PMID: 37379202 PMCID: PMC10398892 DOI: 10.1091/mbc.e22-06-0226] [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: 01/20/2023] [Revised: 06/06/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
During disease and development, physical changes in extracellular matrix cause jamming, unjamming, and scattering in epithelial migration. However, whether disruptions in matrix topology alter collective cell migration speed and cell-cell coordination remains unclear. We microfabricated substrates with stumps of defined geometry, density, and orientation, which create obstructions for migrating epithelial cells. Here, we show that cells lose their speed and directionality when moving through densely spaced obstructions. Although leader cells are stiffer than follower cells on flat substrates, dense obstructions cause overall cell softening. Through a lattice-based model, we identify cellular protrusions, cell-cell adhesions, and leader-follower communication as key mechanisms for obstruction-sensitive collective cell migration. Our modeling predictions and experimental validations show that cells' obstruction sensitivity requires an optimal balance of cell-cell adhesions and protrusions. Both MDCK (more cohesive) and α-catenin-depleted MCF10A cells were less obstruction sensitive than wild-type MCF10A cells. Together, microscale softening, mesoscale disorder, and macroscale multicellular communication enable epithelial cell populations to sense topological obstructions encountered in challenging environments. Thus, obstruction-sensitivity could define "mechanotype" of cells that collectively migrate yet maintain intercellular communication.
Collapse
Affiliation(s)
- Ye Lim Lee
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Christopher Walter
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Hannah Zmuda
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
32
|
Zbiral B, Weber A, Vivanco MDM, Toca-Herrera JL. Characterization of Breast Cancer Aggressiveness by Cell Mechanics. Int J Mol Sci 2023; 24:12208. [PMID: 37569585 PMCID: PMC10418463 DOI: 10.3390/ijms241512208] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
In healthy tissues, cells are in mechanical homeostasis. During cancer progression, this equilibrium is disrupted. Cancer cells alter their mechanical phenotype to a softer and more fluid-like one than that of healthy cells. This is connected to cytoskeletal remodeling, changed adhesion properties, faster cell proliferation and increased cell motility. In this work, we investigated the mechanical properties of breast cancer cells representative of different breast cancer subtypes, using MCF-7, tamoxifen-resistant MCF-7, MCF10A and MDA-MB-231 cells. We derived viscoelastic properties from atomic force microscopy force spectroscopy measurements and showed that the mechanical properties of the cells are associated with cancer cell malignancy. MCF10A are the stiffest and least fluid-like cells, while tamoxifen-resistant MCF-7 cells are the softest ones. MCF-7 and MDA-MB-231 show an intermediate mechanical phenotype. Confocal fluorescence microscopy on cytoskeletal elements shows differences in actin network organization, as well as changes in focal adhesion localization. These findings provide further evidence of distinct changes in the mechanical properties of cancer cells compared to healthy cells and add to the present understanding of the complex alterations involved in tumorigenesis.
Collapse
Affiliation(s)
- Barbara Zbiral
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (A.W.)
| | - Andreas Weber
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (A.W.)
| | - Maria dM. Vivanco
- Cancer Heterogeneity Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain;
| | - José L. Toca-Herrera
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (A.W.)
| |
Collapse
|
33
|
Wohl I, Sajman J, Sherman E. Cell Surface Vibrations Distinguish Malignant from Benign Cells. Cells 2023; 12:1901. [PMID: 37508565 PMCID: PMC10378100 DOI: 10.3390/cells12141901] [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: 06/23/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
The mechanical properties of living cells, including their shape, rigidity, and internal dynamics play a crucial role in their physiology and pathology. Still, the relations between the physiological cell state and its rigidity and surface vibrations remain poorly understood. Here, we have employed AFM measurements on T cells and found a negative relation between cell surface stiffness and its vibrations. Blocking T-type Ca++-channels using Mibefradil reduced cortical actin tension in these cells and enhanced their membrane vibrations and dissipation of intracellular mechanical work to the cell surroundings. We also found increased vibrations of cell membranes in five different malignant cells lines derived from T cell leukemia, lung, prostate, bladder, and melanoma cancers, as compared to their corresponding benign cells. This was demonstrated by utilizing TIRF microscopy in single cells and dynamic laser speckles measurements in an in vitro model of multiple cells in a tissue. Our results show that cell membrane vibrations and dissipation of mechanical work are higher in malignant cells relative to benign cells. Accordingly, these properties may be used to detect and monitor cellular and tissue malignancies.
Collapse
Affiliation(s)
- Ishay Wohl
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Julia Sajman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| |
Collapse
|
34
|
Habli Z, Zantout A, El-Sabban M, Khraiche ML. Investigating malignancy-dependent mechanical properties of breast cancer cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083716 DOI: 10.1109/embc40787.2023.10340235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Cancer invasiveness significantly impacts cellular mechanical properties which regulate cell motility and, subsequently, cell metastatic potential. Understanding the adhesion forces and stiffness/rigidity of cancer cells can provide better insights into their mechanical adaptability related to their degree of invasiveness. Here, we used single-cell force spectroscopy in conjunction with quartz crystal microbalance-with dissipation measurements to compare the mechanical properties of mammary epithelial cancer cells with different metastatic potentials, namely MCF-7 (non-invasive) and MDA-MB-231 (aggressive and highly invasive). Our results showed that MCF-7 exhibits larger adhesion forces, stronger intercellular forces, and a considerably stiff/rigid phenotype, contrary to MDA-MB-231. The biomechanical properties obtained are associated with the malignant potential of these cells such that the forces of adhesion and viscoelasticity are inversely proportional to cell invasiveness. This study integrates a new quantitative tool with real-time measurements to provide better insights into the mechanics of cancer cells across metastatic stages.
Collapse
|
35
|
Liu G, Ma D, Wang H, Zhou J, Shen Z, Yang Y, Chen Y, Sack I, Guo J, Li R, Yan F. Three-dimensional multifrequency magnetic resonance elastography improves preoperative assessment of proliferative hepatocellular carcinoma. Insights Imaging 2023; 14:89. [PMID: 37198348 PMCID: PMC10192481 DOI: 10.1186/s13244-023-01427-4] [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: 12/01/2022] [Accepted: 04/14/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND To investigate the viscoelastic signatures of proliferative hepatocellular carcinoma (HCC) using three-dimensional (3D) magnetic resonance elastography (MRE). METHODS This prospective study included 121 patients with 124 HCCs as training cohort, and validation cohort included 33 HCCs. They all underwent preoperative conventional magnetic resonance imaging (MRI) and tomoelastography based on 3D multifrequency MRE. Viscoelastic parameters of the tumor and liver were quantified as shear wave speed (c, m/s) and loss angle (φ, rad), representing stiffness and fluidity, respectively. Five MRI features were evaluated. Multivariate logistic regression analyses were used to determine predictors of proliferative HCC to construct corresponding nomograms. RESULTS In training cohort, model 1 (Combining cirrhosis, hepatitis virus, rim APHE, peritumoral enhancement, and tumor margin) yielded an area under the curve (AUC), sensitivity, specificity, accuracy of 0.72, 58.73%,78.69%, 67.74%, respectively. When adding MRE properties (tumor c and tumor φ), established model 2, the AUC increased to 0.81 (95% CI 0.72-0.87), with sensitivity, specificity, accuracy of 71.43%, 81.97%, 75%, respectively. The C-index of nomogram of model 2 was 0.81, showing good performance for proliferative HCC. Therefore, integrating tumor c and tumor φ can significantly improve the performance of preoperative diagnosis of proliferative HCC (AUC increased from 0.72 to 0.81, p = 0.012). The same finding was observed in the validation cohort, with AUC increasing from 0.62 to 0.77 (p = 0.021). CONCLUSIONS Proliferative HCC exhibits low stiffness and high fluidity. Adding MRE properties (tumor c and tumor φ) can improve performance of conventional MRI for preoperative diagnosis of proliferative HCC. CRITICAL RELEVANCE STATEMENT We investigated the viscoelastic signatures of proliferative hepatocellular carcinoma (HCC) using three-dimensional (3D) magnetic resonance elastography (MRE), and find that adding MRE properties (tumor c and tumor φ) can improve performance of conventional MRI for preoperative diagnosis of proliferative HCC.
Collapse
Affiliation(s)
- Guixue Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China
| | - Di Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huafeng Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahao Zhou
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China
| | - Zhehan Shen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China
| | - Yuchen Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ingolf Sack
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jing Guo
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ruokun Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
36
|
Kurma K, Alix-Panabières C. Mechanobiology and survival strategies of circulating tumor cells: a process towards the invasive and metastatic phenotype. Front Cell Dev Biol 2023; 11:1188499. [PMID: 37215087 PMCID: PMC10196185 DOI: 10.3389/fcell.2023.1188499] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Metastatic progression is the deadliest feature of cancer. Cancer cell growth, invasion, intravasation, circulation, arrest/adhesion and extravasation require specific mechanical properties to allow cell survival and the completion of the metastatic cascade. Circulating tumor cells (CTCs) come into contact with the capillary bed during extravasation/intravasation at the beginning of the metastatic cascade. However, CTC mechanobiology and survival strategies in the bloodstream, and specifically in the microcirculation, are not well known. A fraction of CTCs can extravasate and colonize distant areas despite the biomechanical constriction forces that are exerted by the microcirculation and that strongly decrease tumor cell survival. Furthermore, accumulating evidence shows that several CTC adaptations, via molecular factors and interactions with blood components (e.g., immune cells and platelets inside capillaries), may promote metastasis formation. To better understand CTC journey in the microcirculation as part of the metastatic cascade, we reviewed how CTC mechanobiology and interaction with other cell types in the bloodstream help them to survive the harsh conditions in the circulatory system and to metastasize in distant organs.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (E LBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (E LBS), Hamburg, Germany
| |
Collapse
|
37
|
Tagay Y, Kheirabadi S, Ataie Z, Singh RK, Prince O, Nguyen A, Zhovmer AS, Ma X, Sheikhi A, Tsygankov D, Tabdanov ED. Dynein-Powered Cell Locomotion Guides Metastasis of Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535605. [PMID: 37066378 PMCID: PMC10104034 DOI: 10.1101/2023.04.04.535605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Metastasis is a principal cause of death in cancer patients, which remains an unresolved fundamental and clinical problem. Conventionally, metastatic dissemination is linked to the actomyosin-driven cell locomotion. However, locomotion of cancer cells often does not strictly line up with the measured actomyosin forces. Here, we identify a complementary mechanism of metastatic locomotion powered by the dynein-generated forces. These forces that arise within a non-stretchable microtubule network drive persistent contact guidance of migrating cancer cells along the biomimetic collagen fibers. We also show that dynein-powered locomotion becomes indispensable during invasive 3D migration within a tissue-like luminal network between spatially confining hydrogel microspheres. Our results indicate that the complementary contractile system of dynein motors and microtubules is always necessary and in certain instances completely sufficient for dissemination of metastatic breast cancer cells. These findings advance fundamental understanding of cell locomotion mechanisms and expand the spectrum of clinical targets against metastasis.
Collapse
Affiliation(s)
- Yerbol Tagay
- Department of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Sina Kheirabadi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Zaman Ataie
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Rakesh K. Singh
- Department of Obstetrics & Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Olivia Prince
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Ashley Nguyen
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Alexander S. Zhovmer
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Xuefei Ma
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Amir Sheikhi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Erdem D. Tabdanov
- Department of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
- Penn State Cancer Institute, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| |
Collapse
|
38
|
Nguyen DT, Pedro DI, Pepe A, Rosa JG, Bowman JI, Trachsel L, Golde GR, Suzuki I, Lavrador JM, Nguyen NTY, Kis MA, Smolchek RA, Diodati N, Liu R, Phillpot SR, Webber AR, Castillo P, Sayour EJ, Sumerlin BS, Sawyer WG. Bioconjugation of COL1 protein on liquid-like solid surfaces to study tumor invasion dynamics. Biointerphases 2023; 18:021001. [PMID: 36898958 PMCID: PMC10008099 DOI: 10.1116/6.0002083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 03/12/2023] Open
Abstract
Tumor invasion is likely driven by the product of intrinsic and extrinsic stresses, reduced intercellular adhesion, and reciprocal interactions between the cancer cells and the extracellular matrix (ECM). The ECM is a dynamic material system that is continuously evolving with the tumor microenvironment. Although it is widely reported that cancer cells degrade the ECM to create paths for migration using membrane-bound and soluble enzymes, other nonenzymatic mechanisms of invasion are less studied and not clearly understood. To explore tumor invasion that is independent of enzymatic degradation, we have created an open three-dimensional (3D) microchannel network using a novel bioconjugated liquid-like solid (LLS) medium to mimic both the tortuosity and the permeability of a loose capillary-like network. The LLS is made from an ensemble of soft granular microgels, which provides an accessible platform to investigate the 3D invasion of glioblastoma (GBM) tumor spheroids using in situ scanning confocal microscopy. The surface conjugation of the LLS microgels with type 1 collagen (COL1-LLS) enables cell adhesion and migration. In this model, invasive fronts of the GBM microtumor protruded into the proximal interstitial space and may have locally reorganized the surrounding COL1-LLS. Characterization of the invasive paths revealed a super-diffusive behavior of these fronts. Numerical simulations suggest that the interstitial space guided tumor invasion by restricting available paths, and this physical restriction is responsible for the super-diffusive behavior. This study also presents evidence that cancer cells utilize anchorage-dependent migration to explore their surroundings, and geometrical cues guide 3D tumor invasion along the accessible paths independent of proteolytic ability.
Collapse
Affiliation(s)
- D. T. Nguyen
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - D. I. Pedro
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - A. Pepe
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - J. G. Rosa
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - J. I. Bowman
- Department of Chemistry, College of Liberal Arts and Sciences, College of Medicine University of Florida, Gainesville, Florida 3261
| | - L. Trachsel
- Department of Chemistry, College of Liberal Arts and Sciences, College of Medicine University of Florida, Gainesville, Florida 3261
| | - G. R. Golde
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - I. Suzuki
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - J. M. Lavrador
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - N. T. Y. Nguyen
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - M. A. Kis
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - R. A. Smolchek
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - N. Diodati
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - R. Liu
- Department of Surgery, College of Medicine University of Florida, Gainesville, Florida 3261
| | - S. R. Phillpot
- Department of Materials Science and Engineering Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - A. R. Webber
- Department of Materials Science and Engineering Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - P. Castillo
- Department of Pediatrics, College of Medicine University of Florida, Gainesville, Florida 3261
| | | | - B. S. Sumerlin
- Department of Chemistry, College of Liberal Arts and Sciences, College of Medicine University of Florida, Gainesville, Florida 3261
| | - W. G. Sawyer
- Author to whom correspondence should be addressed:
| |
Collapse
|
39
|
Wong DCP, Ding JL. The mechanobiology of NK cells- 'Forcing NK to Sense' target cells. Biochim Biophys Acta Rev Cancer 2023; 1878:188860. [PMID: 36791921 DOI: 10.1016/j.bbcan.2023.188860] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 02/16/2023]
Abstract
Natural killer (NK) cells are innate immune lymphocytes that recognize and kill cancer and infected cells, which makes them unique 'off-the-shelf' candidates for a new generation of immunotherapies. Biomechanical forces in homeostasis and pathophysiology accrue additional immune regulation for NK immune responses. Indeed, cellular and tissue biomechanics impact NK receptor clustering, cytoskeleton remodeling, NK transmigration through endothelial cells, nuclear mechanics, and even NK-dendritic cell interaction, offering a plethora of unexplored yet important dynamic regulation for NK immunotherapy. Such events are made more complex by the heterogeneity of human NK cells. A significant question remains on whether and how biochemical and biomechanical cues collaborate for NK cell mechanotransduction, a process whereby mechanical force is sensed, transduced, and translated to downstream mechanical and biochemical signalling. Herein, we review recent advances in understanding how NK cells perceive and mechanotransduce biophysical cues. We focus on how the cellular cytoskeleton crosstalk regulates NK cell function while bearing in mind the heterogeneity of NK cells, the direct and indirect mechanical cues for NK anti-tumor activity, and finally, engineering advances that are of translational relevance to NK cell biology at the systems level.
Collapse
Affiliation(s)
- Darren Chen Pei Wong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore.
| | - Jeak Ling Ding
- Department of Biological Sciences, National University of Singapore, 117543, Singapore; Integrative Sciences and Engineering Programme, National University of Singapore, 119077, Singapore.
| |
Collapse
|
40
|
Edwin PERG, Kumar S, Roy S, Roy B, Bajpai SK. Anisotropic 3D confinement of MCF-7 cells induces directed cell-migration and viscoelastic anisotropy of cell-membrane. Phys Biol 2023; 20. [DOI: 10.1088/1478-3975/ac9bc1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Abstract
Tumor-associated collagen signature-3 (TACS-3) is a prognostic indicator for breast cancer survival. It is characterized by highly organized, parallel bundles of collagen fibers oriented perpendicular to the tumor boundary, serving as directional, confining channels for cancer cell invasion. Here we design a TACS-3-mimetic anisotropic, confined collagen I matrix and examine the relation between anisotropy of matrix, directed cellular migration, and anisotropy of cell membrane-the first direct contact between TACS-3 and cell-using Michigan Cancer Foundation-7 (MCF-7) cells as cancer-model. Using unidirectional freezing, we generated ∼50 μm-wide channels filled with collagen I. Optical tweezer (OT) microrheology shows that anisotropic confinement increases collagen viscoelasticity by two orders of magnitude, and the elastic modulus is significantly greater along the direction of anisotropic confinement compared to that along the orthogonal direction, thus establishing matrix anisotropy. Furthermore, MCF-7 cells embedded in anisotropic collagen I, exhibit directionality in cellular morphology and migration. Finally, using customized OT to trap polystyrene probes bound to cell-membrane (and not to ECM) of either free cells or cells under anisotropic confinement, we quantified the effect of matrix anisotropy on membrane viscoelasticity, both in-plane and out-of-plane, vis-à-vis the membrane. Both bulk and viscous modulus of cell-membrane of MCF-7 cells exhibit significant anisotropy under anisotropic confinement. Moreover, the cell membrane of MCF-7 cells under anisotropic confinement is significantly softer (both in-plane and out-of-plane moduli) despite their local environment being five times stiffer than free cells. In order to test if the coupling between anisotropy of extracellular matrix and anisotropy of cell-membrane is regulated by cell-cytoskeleton, actin cytoskeleton was depolymerized for both free and confined cells. Results show that cell membrane viscoelasticity of confined MCF-7 cells is unaffected by actin de-polymerization, in contrast to free cells. Together, these findings suggest that anisotropy of ECM induces directed migration and correlates with anisotropy of cell-membrane viscoelasticity of the MCF-7 cells in an actin-independent manner.
Collapse
|
41
|
Zills G, Datta T, Malmi-Kakkada AN. Enhanced mechanical heterogeneity of cell collectives due to temporal fluctuations in cell elasticity. Phys Rev E 2023; 107:014401. [PMID: 36797877 DOI: 10.1103/physreve.107.014401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 12/08/2022] [Indexed: 06/18/2023]
Abstract
Cells are dynamic systems characterized by temporal variations in biophysical properties such as stiffness and contractility. Recent studies show that the recruitment and release of actin filaments into and out of the cell cortex-a network of proteins underneath the cell membrane-leads to cell stiffening prior to division and softening immediately afterward. In three-dimensional (3D) cell collectives, it is unclear whether the stiffness change during division at the single-cell scale controls the spatial structure and dynamics at the multicellular scale. This is an important question to understand because cell stiffness variations impact cell spatial organization and cancer progression. Using a minimal 3D model incorporating cell birth, death, and cell-to-cell elastic and adhesive interactions, we investigate the effect of mechanical heterogeneity-variations in individual cell stiffnesses that make up the cell collective-on tumor spatial organization and cell dynamics. We discover that spatial mechanical heterogeneity characterized by a spheroid core composed of stiffer cells and softer cells in the periphery emerges within dense 3D cell collectives, which may be a general feature of multicellular tumor growth. We show that heightened spatial mechanical heterogeneity enhances single-cell dynamics and volumetric tumor growth driven by fluctuations in cell elasticity. Our results could have important implications in understanding how spatiotemporal variations in single-cell stiffness determine tumor growth and spread.
Collapse
Affiliation(s)
- Garrett Zills
- Department of Chemistry and Physics, Augusta University, 1120 15th Street, Augusta, Georgia 30912, USA
| | - Trinanjan Datta
- Department of Chemistry and Physics, Augusta University, 1120 15th Street, Augusta, Georgia 30912, USA
- Kavli Institute for Theoretical Physics, University of California, Santa Barbara, California 93106, USA
| | | |
Collapse
|
42
|
Han SJ, Kwon S, Kim KS. Contribution of mechanical homeostasis to epithelial-mesenchymal transition. Cell Oncol (Dordr) 2022; 45:1119-1136. [PMID: 36149601 DOI: 10.1007/s13402-022-00720-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Metastasis refers to the spread of cancer cells from a primary tumor to other parts of the body via the lymphatic system and bloodstream. With tremendous effort over the past decades, remarkable progress has been made in understanding the molecular and cellular basis of metastatic processes. Metastasis occurs through five steps, including infiltration and migration, intravasation, survival, extravasation, and colonization. Various molecular and cellular factors involved in the metastatic process have been identified, such as epigenetic factors of the extracellular matrix (ECM), cell-cell interactions, soluble signaling, adhesion molecules, and mechanical stimuli. However, the underlying cause of cancer metastasis has not been elucidated. CONCLUSION In this review, we have focused on changes in the mechanical properties of cancer cells and their surrounding environment to understand the causes of cancer metastasis. Cancer cells have unique mechanical properties that distinguish them from healthy cells. ECM stiffness is involved in cancer cell growth, particularly in promoting the epithelial-mesenchymal transition (EMT). During tumorigenesis, the mechanical properties of cancer cells change in the direction opposite to their environment, resulting in a mechanical stress imbalance between the intracellular and extracellular domains. Disruption of mechanical homeostasis may be one of the causes of EMT that triggers the metastasis of cancer cells.
Collapse
Affiliation(s)
- Se Jik Han
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul, Korea.,Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea
| | - Sangwoo Kwon
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
43
|
Wang Y, Guo J, Ma D, Zhou J, Yang Y, Chen Y, Wang H, Sack I, Li R, Yan F. Reduced tumor stiffness quantified by tomoelastography as a predicative marker for glypican-3-positive hepatocellular carcinoma. Front Oncol 2022; 12:962272. [PMID: 36518314 PMCID: PMC9744252 DOI: 10.3389/fonc.2022.962272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/14/2022] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Glypican-3 (GPC3) expression is investigated as a promising target for tumor-specific immunotherapy of hepatocellular carcinoma (HCC). This study aims to determine whether GPC3 alters the viscoelastic properties of HCC and whether tomoelastography, a multifrequency magnetic resonance elastography (MRE) technique, is sensitive to it. METHODS Ninety-five participants (mean age, 58 ± 1 years; 78 men and 17 women) with 100 pathologically confirmed HCC lesions were enrolled in this prospective study from July 2020 to August 2021. All patients underwent preoperative multiparametric MRI and tomoelastography. Tomoelastography provided shear wave speed (c, m/s) representing tissue stiffness and loss angle (φ, rad) relating to viscosity. Clinical, laboratory, and imaging parameters were compared between GPC3-positive and -negative groups. Univariable and multivariable logistic regression were performed to determine factors associated with GPC3-positive HCC. The diagnostic performance of combined biomarkers was established using logistic regression analysis. Area-under-the-curve (AUC) analysis was done to assess diagnostic performance in detecting GPC3-positive HCC. FINDINGS GPC3-positive HCCs (n=72) had reduced stiffness compared with GPC3-negative HCCs (n=23) while viscosity was not different (c: 2.34 ± 0.62 versus 2.72 ± 0.62 m/s, P=0.010, φ: 1.11 ± 0.21 vs 1.18 ± 0.27 rad, P=0.21). Logistic regression showed c and elevated serum alpha-fetoprotein (AFP) level above 20 ng/mL were independent factors for GPC3-positive HCC. Stiffness with a cutoff of c = 2.8 m/s in conjunction with an elevated AFP yielded a sensitivity of 80.3%, specificity of 70.8%, and AUC of 0.80. INTERPRETATION Reduced stiffness quantified by tomoelastography may be a mechanical signature of GPC3-positive HCC. Combining reduced tumor stiffness and elevated AFP level may provide potentially valuable biomarker for GPC3-targeted immunotherapy.
Collapse
Affiliation(s)
- Yihuan Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Guo
- Department of Radiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Di Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahao Zhou
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchen Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huafeng Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ingolf Sack
- Department of Radiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Ruokun Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Nikolić M, Scarcelli G, Tanner K. Multimodal microscale mechanical mapping of cancer cells in complex microenvironments. Biophys J 2022; 121:3586-3599. [PMID: 36059196 PMCID: PMC9617162 DOI: 10.1016/j.bpj.2022.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 08/05/2022] [Accepted: 09/02/2022] [Indexed: 02/07/2023] Open
Abstract
The mechanical phenotype of the cell is critical for survival following deformations due to confinement and fluid flow. One idea is that cancer cells are plastic and adopt different mechanical phenotypes under different geometries that aid in their survival. Thus, an attractive goal is to disrupt cancer cells' ability to adopt multiple mechanical states. To begin to address this question, we aimed to quantify the diversity of these mechanical states using in vitro biomimetics to mimic in vivo two-dimensional (2D) and 3D extracellular matrix environments. Here, we used two modalities Brillouin microscopy (∼GHz) and broadband frequency (7-15 kHz) optical tweezer microrheology to measure microscale cell mechanics. We measured the response of intracellular mechanics of cancer cells cultured in 2D and 3D environments where we modified substrate stiffness, dimensionality (2D versus 3D), and presence of fibrillar topography. We determined that there was good agreement between two modalities despite the difference in timescale of the two measurements. These findings on cell mechanical phenotype in different environments confirm a correlation between modalities that employ different mechanisms at different temporal scales (Hz-kHz versus GHz). We also determined that observed heterogeneity in cell shape is more closely linked to the cells' mechanical state. Moreover, individual cells in multicellular spheroids exhibit a lower degree of mechanical heterogeneity when compared with single cells cultured in monodisperse 3D cultures. The observed decreased heterogeneity among cells in spheroids suggested that there is mechanical cooperativity between cells that make up a single spheroid.
Collapse
Affiliation(s)
- Miloš Nikolić
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Maryland Biophysics Program, IPST, University of Maryland, College Park, Maryland
| | - Giuliano Scarcelli
- Maryland Biophysics Program, IPST, University of Maryland, College Park, Maryland; Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
45
|
Cai G, Nguyen A, Bashirzadeh Y, Lin SS, Bi D, Liu AP. Compressive stress drives adhesion-dependent unjamming transitions in breast cancer cell migration. Front Cell Dev Biol 2022; 10:933042. [PMID: 36268514 PMCID: PMC9577106 DOI: 10.3389/fcell.2022.933042] [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: 04/30/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular unjamming is the collective fluidization of cell motion and has been linked to many biological processes, including development, wound repair, and tumor growth. In tumor growth, the uncontrolled proliferation of cancer cells in a confined space generates mechanical compressive stress. However, because multiple cellular and molecular mechanisms may be operating simultaneously, the role of compressive stress in unjamming transitions during cancer progression remains unknown. Here, we investigate which mechanism dominates in a dense, mechanically stressed monolayer. We find that long-term mechanical compression triggers cell arrest in benign epithelial cells and enhances cancer cell migration in transitions correlated with cell shape, leading us to examine the contributions of cell–cell adhesion and substrate traction in unjamming transitions. We show that cadherin-mediated cell–cell adhesion regulates differential cellular responses to compressive stress and is an important driver of unjamming in stressed monolayers. Importantly, compressive stress does not induce the epithelial–mesenchymal transition in unjammed cells. Furthermore, traction force microscopy reveals the attenuation of traction stresses in compressed cells within the bulk monolayer regardless of cell type and motility. As traction within the bulk monolayer decreases with compressive pressure, cancer cells at the leading edge of the cell layer exhibit sustained traction under compression. Together, strengthened intercellular adhesion and attenuation of traction forces within the bulk cell sheet under compression lead to fluidization of the cell layer and may impact collective cell motion in tumor development and breast cancer progression.
Collapse
Affiliation(s)
- Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, United States
| | - Anh Nguyen
- Department of Physics, Northeastern University, Boston, MA, United States
| | - Yashar Bashirzadeh
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Shan-Shan Lin
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Dapeng Bi
- Department of Physics, Northeastern University, Boston, MA, United States
| | - Allen P. Liu
- Applied Physics Program, University of Michigan, Ann Arbor, MI, United States
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Biophysics, University of Michigan, Ann Arbor, MI, United States
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Allen P. Liu,
| |
Collapse
|
46
|
Chen Q, Zou J, He Y, Pan Y, Yang G, Zhao H, Huang Y, Zhao Y, Wang A, Chen W, Lu Y. A narrative review of circulating tumor cells clusters: A key morphology of cancer cells in circulation promote hematogenous metastasis. Front Oncol 2022; 12:944487. [PMID: 36059616 PMCID: PMC9434215 DOI: 10.3389/fonc.2022.944487] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Circulating tumor cells (CTCs) that survive in the blood are playing an important role in the metastasis process of tumor. In addition, they have become a tool for tumor diagnosis, prognosis and recurrence monitoring. CTCs can exist in the blood as individual cells or as clumps of aggregated cells. In recent years, more and more studies have shown that clustered CTCs have stronger metastasis ability compared to single CTCs. With the deepening of studies, scholars have found that cancer cells can combine not only with each other, but also with non-tumor cells present in the blood, such as neutrophils, platelets, etc. At the same time, it was confirmed that non-tumor cells bound to CTCs maintain the survival and proliferation of cancer cells through a variety of ways, thus promoting the occurrence and development of tumor. In this review, we collected information on tumorigenesis induced by CTC clusters to make a summary and a discussion about them. Although CTC clusters have recently been considered as a key role in the transition process, many characteristics of them remain to be deeply explored. A detailed understanding of their vulnerability can prospectively pave the way for new inhibitors for metastasis.
Collapse
Affiliation(s)
- Qiong Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jueyao Zou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yong He
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanhong Pan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pharmacy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gejun Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| |
Collapse
|
47
|
Biomechanics of cancer stem cells. Essays Biochem 2022; 66:359-369. [PMID: 35942932 DOI: 10.1042/ebc20220014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs) have been believed to be one driving force for tumor progression and drug resistance. Despite the significance of biochemical signaling in malignancy, highly malignant tumor cells or CSCs exhibit lower cellular stiffness than weakly malignant cells or non-CSCs, which are softer than their healthy counterparts, suggesting the inverse correlation between cell stiffness and malignancy. Recent years have witnessed the rapid accumulation of evidence illustrating the reciprocity between cell cytoskeleton/mechanics and CSC functions and the potential of cellular stiffness for specific targeting of CSCs. However, a systematic understanding of tumor cell mechanics and their role in CSCs and tumor progression is still lacking. The present review summarizes the recent progress in the alterations of tumor cell cytoskeleton and stiffness at different stages of tumor progression and recapitulates the relationship between cellular stiffness and CSC functions. The altered cell mechanics may mediate the mechanoadaptive responses that possibly empower CSCs to survive and thrive during metastasis. Furthermore, we highlight the possible impact of tumor cell mechanics on CSC malignancy, which may potentiate low cell stiffness as a mechanical marker for CSC targeting.
Collapse
|
48
|
Geometric trade-off between contractile force and viscous drag determines the actomyosin-based motility of a cell-sized droplet. Proc Natl Acad Sci U S A 2022; 119:e2121147119. [PMID: 35857875 PMCID: PMC9335187 DOI: 10.1073/pnas.2121147119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cell migration in confined environments is fundamental for diverse biological processes from cancer invasion to leukocyte trafficking. The cell body is propelled by the contractile force of actomyosin networks transmitted from the cell membrane to the external substrates. However, physical determinants of actomyosin-based migration capacity in confined environments are not fully understood. Here, we develop an in vitro migratory cell model, where cytoplasmic actomyosin networks are encapsulated into droplets surrounded by a lipid monolayer membrane. We find that the droplet can move when the actomyosin networks are bound to the membrane, in which the physical interaction between the contracting actomyosin networks and the membrane generates a propulsive force. The droplet moves faster when it has a larger contact area with the substrates, while narrower confinement reduces the migration speed. By combining experimental observations and active gel theory, we propose a mechanism where the balance between sliding friction force, which is a reaction force of the contractile force, and viscous drag determines the migration speed, providing a physical basis of actomyosin-based motility in confined environments.
Collapse
|
49
|
Nguyen DT, Ogando-Rivas E, Liu R, Wang T, Rubin J, Jin L, Tao H, Sawyer WW, Mendez-Gomez HR, Cascio M, Mitchell DA, Huang J, Sawyer WG, Sayour EJ, Castillo P. CAR T Cell Locomotion in Solid Tumor Microenvironment. Cells 2022; 11:1974. [PMID: 35741103 PMCID: PMC9221866 DOI: 10.3390/cells11121974] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 01/25/2023] Open
Abstract
The promising outcomes of chimeric antigen receptor (CAR) T cell therapy in hematologic malignancies potentiates its capability in the fight against many cancers. Nevertheless, this immunotherapy modality needs significant improvements for the treatment of solid tumors. Researchers have incrementally identified limitations and constantly pursued better CAR designs. However, even if CAR T cells are armed with optimal killer functions, they must overcome and survive suppressive barriers imposed by the tumor microenvironment (TME). In this review, we will discuss in detail the important role of TME in CAR T cell trafficking and how the intrinsic barriers contribute to an immunosuppressive phenotype and cancer progression. It is of critical importance that preclinical models can closely recapitulate the in vivo TME to better predict CAR T activity. Animal models have contributed immensely to our understanding of human diseases, but the intensive care for the animals and unreliable representation of human biology suggest in vivo models cannot be the sole approach to CAR T cell therapy. On the other hand, in vitro models for CAR T cytotoxic assessment offer valuable insights to mechanistic studies at the single cell level, but they often lack in vivo complexities, inter-individual heterogeneity, or physiologically relevant spatial dimension. Understanding the advantages and limitations of preclinical models and their applications would enable more reliable prediction of better clinical outcomes.
Collapse
Affiliation(s)
- Duy T. Nguyen
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elizabeth Ogando-Rivas
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Ruixuan Liu
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Theodore Wang
- College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Jacob Rubin
- Warrington College of Business, University of Florida, Gainesville, FL 32610, USA;
| | - Linchun Jin
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Haipeng Tao
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - William W. Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Hector R. Mendez-Gomez
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Matthew Cascio
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Duane A. Mitchell
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Jianping Huang
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - W. Gregory Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elias J. Sayour
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Paul Castillo
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
50
|
Tan K, Naylor MJ. Tumour Microenvironment-Immune Cell Interactions Influencing Breast Cancer Heterogeneity and Disease Progression. Front Oncol 2022; 12:876451. [PMID: 35646658 PMCID: PMC9138702 DOI: 10.3389/fonc.2022.876451] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a complex, dynamic disease that acquires heterogeneity through various mechanisms, allowing cancer cells to proliferate, survive and metastasise. Heterogeneity is introduced early, through the accumulation of germline and somatic mutations which initiate cancer formation. Following initiation, heterogeneity is driven by the complex interaction between intrinsic cellular factors and the extrinsic tumour microenvironment (TME). The TME consists of tumour cells and the subsequently recruited immune cells, endothelial cells, fibroblasts, adipocytes and non-cellular components of the extracellular matrix. Current research demonstrates that stromal-immune cell interactions mediated by various TME components release environmental cues, in mechanical and chemical forms, to communicate with surrounding and distant cells. These interactions are critical in facilitating the metastatic process at both the primary and secondary site, as well as introducing greater intratumoral heterogeneity and disease complexity by exerting selective pressures on cancer cells. This can result in the adaptation of cells and a feedback loop to the cancer genome, which can promote therapeutic resistance. Thus, targeting TME and immune-stromal cell interactions has been suggested as a potential therapeutic avenue given that aspects of this process are somewhat conserved between breast cancer subtypes. This mini review will discuss emerging ideas on how the interaction of various aspects of the TME contribute to increased heterogeneity and disease progression, and the therapeutic potential of targeting the TME.
Collapse
|